Mapping of Italian research excellence in Neurodegenerative Diseases 1. MECHANISMS OF NEURODEGENERATION A. Macromolecular interactions and Neurodegeneration 1. Public health. Therapeutic strategies for the prevention and tratment of neurodegenerative disease. Molecular basis of amyloid aggregation 2. Protein conformational transitions that trigger the aggregation processes associated to neurodegenerative diseases : a nanotechnological approach 3. Myelin proteins and demyelinating deseases 4. Conformational transitions of proteins from native to amyloid form 5. Role of altered composition of plasma membrane complex lipids in lipid-protein interactions, membrane organization and neurodegeneration 6. Alterations of proteolytic pathways in neurodegenerative diseases" 7. Optical and biomolecular methods for the functional investigation of neural circuits in vivo 8. Molecular mechanisms in neurotransmission: effects in neuron and glia phenotypes in synaptic efficiency and synapropathy 9. Presynaptic molecular machinery regulationg glutamate release an pathophysiological mechanism in amyotrophic lateral sclerosis 10. Toward exploiting the screening potential of miRNome for the identification of genes and pathways involved in neurodegenerative disorders 11. Identification of signaling pathways in neurodegenerative proteinopathies 12. Neurotensin and neurotensin antagonists in an animal model of Parkinson disease: therapeutic perspectives and role of NMDA/neurotensin interaction. B. Pier Luigi San Biagio [email protected] Bruno Samorì [email protected] Eugenia Polverini [email protected] Maria Grazia Bridelli [email protected] Sandro Sonnino [email protected] Angelo Poletti [email protected] Antonio Malgaroli [email protected] Gian Giacomo Consalez [email protected] Flavia Valtorta [email protected] Maurizio Popoli [email protected] Daniela Tardito [email protected] Maria Penuto Maria [email protected] Tanganelli Sergio [email protected] Neurotoxic stimuli 1. CSF Glycomics in AD and other neurodegenerative disease 2. Intracellular signalling during neurodegenerative events triggered upon APP overexpression Domenico Garozzo [email protected] Maurizio Taglialatela [email protected] Claudio Russo [email protected] 3. Effect of Age and neurodegenerative disorders in central neurotrasmission: focus on neurotrasmitter relase from neuron and glia Mario Marchi [email protected] 4. Developing competitive in “ vitro” and “vivo” models to study Alzheimer’s disease 5. Evaluation of mesenchymal stell cells (MSCs) effect in Alzheimer’s disease rat models competitive in “ vitro” and “vivo” studies 6. New Tratment strategies (Neurodegeneration and Neuroprotection) 7. Pathways leading to tau pathology – Role of nuclear tau in neurodegeneration in fronto-temporal dementia 8. Cholesterol and amyloid-beta: is there a relation? - Patrizia Hrelia [email protected] Fabrizio Tagliavini [email protected] Roberta Ricciarelli [email protected] Specific type 4 phosphodiestrerase inhibitors and their impact on the production of amyloid-beta Cholesterol and amyloid-beta: is there a relation? Role of non muscle myosin IIB in the processing of the amyloid precursor protein (APP) 9. Basic research - Genetic susceptibility to Alzheimer disease and genome wide association studies - Biobanking 10. Neurotoxic stimuli, synaptic dysfunction 11. Neuroprotection by chemokines 12. Phatogenesis and therapeutic targets for SBMA 13. Malattia di Alzheimer e trasduzione del segnale legata al calcio 14. Mechanisms of neurodegeneration and enhancement of adult neurogenesis to improve cognitive functions in down syndrome 15. Pathological mechanisms of neurodegeneration triggered by tau and beta-amyloid 16. Oxidative stress and neurodegeneration C. Francesca Ruberti [email protected] Mariarosaria Miloso [email protected] Dario Finazzi [email protected] Calro Sala [email protected] Cristina Limatola [email protected] Fabio Benfenati – Mara Pennuto – Chiara Scaramuzzino [email protected] Armando Genazzani [email protected] Andrea Contestabile [email protected] Laura Gasparini [email protected] Michele Mazzanti [email protected] Neuro inflammation 1. Role of vascular inflammation and leukocyte trafficking in neurodegenerative diseases 2. Regional vulmerability of the brain to neurodegeneration and immune regulatory mechanisms 3. Neurotoxic stimuli: role of astrocytes and microglia 4. Gender and hormonal/endocrine signals in neuroinflammation role in alzheimer’s disease and Gabriela Constantin [email protected] Marina Bentivoglio [email protected] Michela Matteoli [email protected] Elisabetta Vegeto [email protected] brain ageing 5. Role of pro-and anti- inflammatory cytokines in the prognosis and progression of neurodegenerative diseases 6. Study of the role of pro-inflammatory chemokines in neuroinflammatory disorders and analysis of the therapeutic potential of chemokine synthesis inhibitors 7. Role of inflammation and microglial activation in neurodegenerative diseases 8. Role of inflammation in Alzheimer’s disease: study of pro-inflammatory cytokines for the identification of new pathogenic and diagnostic targets D. Barbara Viviani [email protected] Claudio Milanese [email protected] Luisa Minghetti [email protected] Paola Bossù [email protected] Oxidative Stress and neurodegeneration 1. Dopamine and oxidative stress in the pathogenesis of Parkinson's disease 2. Mitochondria, apoptosis and neurodegeneration: characterization of mitochondrial dysfunction in neurodegenerative diseases - Characterization of the molecular mechanisms underlying aging, cell senescence and neurodegenerative diseases 3. Structural organization of the mitochondrial respiratory chain, generation of reactive oxygen species and neurodegeneration 4. Post-genomic biochemestry of neurodegenerative diseases Marco Bisaglia [email protected] Antonella Bobba [email protected] Giorgio Lenaz [email protected] Annalisa Santucci [email protected] Trophic factors involvement in the neurodegeneration process E. F. Endocrine and Metabolic Factors 1. miRNA as sensors of neuronal stress and modulators of synaptic plasticità and memory deficits 2. Developing competitive animal models to study Alzheimer’s disease Paola Fragapane [email protected] 3. Genetic susceptibility to Alzheimer’s disease (AD) in women: AD as a gender disease 4. Genetic susceptibility to Alzheimer’s disease (Epigenetic maks of susceptibility to Alzheimer disese) 5. Role of altered lipid and cholesterol metabolism in neurodegenerations Rosa Maria Corbo [email protected] Lucia Migliore [email protected] Roberto Rimondini [email protected] Laura Colombaioni [email protected] 6. Role of neuroactive steroids in the neurodegeneration process 7. Selective Alzheimer Disease Indicator -1 (Seladinò1): a liker between cholesterol, oxidative stress and Alzheimer’s disease 8. Nuclear receptors, coregulators, neuroactive steroids and epigenetics in neurodegenerative disease associated to metabolic disorders G. 4. Synaptic deficits in neurodegenerative disease Ubaldo Armato [email protected] Manuela Marcoli [email protected] Luciano Domenici [email protected] Evelina Chieregatti [email protected] Intracellular Traffiking 1. Membrane traffic and neurodegenerative diseases: role of Rab proteins and their effectors 2. Basic research. Neurodegenerative diseases I. Donatella Caruso [email protected] Cell interactions 1. The Astrocyte, not just a Bystander in Alzheimer’s disease 2. Synapse and Amyloid-beta: effects on astrocyte processes, nerve terminals and network synaptic activity 3. Alzheimer’s disease, Ageing H. Roberto Cosimo Melcangi [email protected] Roberto Maggi [email protected] Cecilia Bucci [email protected] Paolo Macchi [email protected] Animal models 1. Developing competitive animal models to study Alzheimer’s disease 2. Synaptic dysfunction in neurodegeneration: characterization of gene product in functional synaptic networks under physiological and patological conditions 3. Study of early onset Alzheimer disease in a model for Down syndrome, the Ts65Dn mouse 4. Iron and Neurodegeneration 5. Non neurotoxic activity of Beta amyloid in animal models 6. Neurochemical and morphological in vivo and in vitro models in neurodegenerative diseases. Adalberto Merighi [email protected] Monica DI Luca [email protected] Renata Bartesaghi [email protected] Sonia Levi [email protected] Stefano Govoni [email protected] Tiziana Antonelli [email protected] 2. PHARMACOLOGY Drug Design A. 1. Elementary mechanisms of neurodegenerative diseases 2. Innovative drug discovery strategies for Alzheimer’s disease 3. Development of ligands specifically targeting the Translocator Protein 18 kDa in Alzheimer disease 4. Structural studies of acetylcholinesterases and their inibitors: implicantions for the design of new antialzheimer drugs Nino Russo [email protected] Carlo Melchiorre [email protected] Federico Da Settimo [email protected] Alberto Cassetta [email protected] Drug screening B. 1. Developemt of competitive animal models for ADGLP grade animal facility and behavioural testing Laura Calzà [email protected] 2. Yeast model to seek for molecules which can reduce mitochondrial mutability Iliana Ferrero Fortunati [email protected] 3. Innovative technologies for the therapy of the neurodegenerative diseases. Luca Ferraro [email protected] Drug Delivery C. 1. Nanotechnology for health: application in the brain diseases 2. Hybrid drug containing reservoirs for long term release in brain diseases 3. New nanodevices targeting beta-amyloid and overcoming the blood-brain barrier for the therapy of Alzheimer disease MA Vandelli Giovanni Tosi [email protected] Angelo Montenero [email protected] Massimo Masserini [email protected] Novel Drugs D. 1. 2. Synthesis and screening of new AChE/BuChE inhibitors 3. Drug design and synthesis of therapeutic agent for neurodegenerative disorders, particularly for Alzheimer disease 4. Development of a novel therapeutic strategy for AD based on a natural variant of amyloid beta that Lamberto Maffei [email protected] Aldo Andreani [email protected] Olga Bruno [email protected] Fabrizio Tagliavini [email protected] hinders amyloidogenesis 5. Second generation recombinant Abeta peptide immunogens as prototype vaccines for AD treatment 6. Pediatric neurodegenerative diseases 7. Ocular NGF administration as a novel non invasive approach to protect brain-NGF target neurons that degenerative in Alzheimer’s disease 8. New symptomatic and neuroprotective terapie for parkinsons’s disease 9. Innovative neuro-reparative strategies via the implementation of endogenous neurogenesis and gliogenesis: focus on the new P2Y-like GPR17 receptor 10. Pharmacological modulation of adult neurogenesis 11. Study of the relationship between the structure/morphology of amyloid beta peptide aggregates and their toxicity in cellular, neuronal and animal models" E. Simone Ottonello [email protected] Maurizio Scarpa [email protected] Paola Tirassa [email protected] Micaela Morelli [email protected] Maria Pia Abbracchio [email protected] Maria Grazia Grilli [email protected] Fabrizio Chiti [email protected] Non-Pharmacological Therapies 1. Psychoeducational intervention for patients and care givers in ParKinson’s Disease and other neurodegenerative diseases. 2. Physical excercise and cognitive decline 3. New tratment strategies (non pharmacologic therapeutic strategies in neurodegenerative diseases) 4. Non invasive brain stimulation an integrated approach to neurorehabilitation in Alzheimer disease 5. Early cognitive and electroencephalographic markers of normal and pathological ageing 6. Transplantation of iPS-derived dopaminergic neurons for cell replacement in Parkinson's disease 7. Neural re generation in the cerebellum: development of cell replacement strategies for the management of spinocerebellar ataxias 8. Physical activity programs for elderly subjects with different grades of cognitive impairment 9. Promotion of Adapted Motor Activity among patients with Parkinson’s disease Pio Enrico Ricci Bitti [email protected] Maurizio Taglialatela [email protected] Alfonso Di Costanzo [email protected] Andrea Stracciari [email protected] Orazio Zanetti [email protected] Patrizia Bisiacchi [email protected] Vania Broccoli [email protected] Gian Giacomo Consalez [email protected] Ferdinando Rossi [email protected] Federico Schena [email protected] Enrico Granieri [email protected] [email protected] [email protected] 10. New treatment strategies in Neurodegenerative diseases with Deep brain Stimulation F. Drug Pharmacogenetics 1. Role of pharmacogenetics to identify the personalized treatment of dementia and depression. G. Mariachiara Sensi [email protected] Alberto Pilotto [email protected] Clinical studies 1. Neurocognition in Alzheimer Disease Carlo Caltagirone [email protected] 3. HUMAN/CLINICAL RESEARCH A. Human –epidemiology/risk factors 1. Pathogenic mechanisms, early diagnosis and prevention of neurodegenerative diseases following pesticide intake in animal model 2. Effects of traffic-related particulate matter on transcription of genes implicated in neurodegeneration 3. Cronobiology and neurodegeneration 4. Gene-metal interaction as determinant of neurodegenerative diseases 5. Using prevention to reduce the burden of Alzheimer’s disease 6. Construction and development of "Alzheimer-WebGIS". Geographical correlation between Alzheimer's disease and environmental parameters. Method for mapping population-based case-control studies. 7. Prospective evaluation of biomarkers of inflammation in Mild Cognitive Impairment subtypes, and their role in the conversion from cognitive impairment to dementia and Alzheimer's disease 8. Epidemiology and risk factors for Mild Cognitive Impairment, Alzheimer's disease and dementia. Population-based 7-year follow-up study. 9. [moved to B.5 Epidemiology genetics] 10. Preclinical Diagnosis 11. Interactions between the products of the Herpes Gabbianelli Rosita – Cinzia Nasuti [email protected] Micaela Caserta [email protected] Valerio Carelli [email protected] [email protected] Roberto Lucchini [email protected] Stefano Mattioli [email protected] Giuseppe Mele [email protected] Calogero Caruso [email protected] Roberto Monastero [email protected] Emilio Di Maria [email protected] Carlo Caltagirone [email protected] Manservigi Roberto simplex genome and Alzheimer's disease susceptibility genes B. Epidemiology genetics 1. Genetic susceptibility to Alzheimer’s disease and genome wide association studies 2. Genetic susceptibility to Alzheimer’s disease: biobanking C. [email protected] Luisa Benussi [email protected] Maria Del Zompo [email protected] Biomarkers and early diagnosis and imaging 1. Plasma biomarkers of oxidative stress and inflammation in early detection and follow-up of dementia. 2. Vitamins and antioxidants in aging and neurodegenerative diseases. 3. Phenotypic and genotypic markers of early Alzheimer’s disease 4. Prioritization and annotation of AD genes, their variants and their interaction networks 5. Cerebrospinal fluid cytokine and chemokine levels as potential biomarkers for early diagnosis of Alzheimer’s disease 6. Individual evaluation of cognitive decline: definition of low cost non- invasive examination battery and implementation of a predictive algorithm to identify Mild cognitive impairment 7. Impiego dei markers biologici e neuro radiologici nella diagnosi differenziale precoce 8. Biomarkers in Mild cognitive Impairment: genetics and biochemical and neuroimaging studies for early diagnosis. Patrizia Mecocci [email protected] 9. A multimodality index for early diagnosis of Alzheimer’s disease 10. Lexical semantic skills, APOE and neuroanatomical substrate of semantic memory in early diagnosis of Alzheimer's disease 11. Cognitive neuroscience of dementia: clinical and theoretical aspect 12. Mapping structural and functional brain network damage in neurodegenerative diseases 13. Studies are aimed at investigating whit PET the brain functional parameters and neurotransmission systems in neurological and psychiatric diseases 14. Global proteomics of human fibroblast for the identification of early biomarkers for dementia and Alzheimer’s disease 15. Parkinson disease and parkinsonisms Flavio Nobili [email protected] Paolo Caffarra [email protected] 16. Mild cognitive impairment Salvatore Monaco [email protected] Rita Casadio [email protected] Elio Scarpini [email protected] Paolo Maria Rossini [email protected] Orazio Zanetti [email protected] Sandro Sorbi [email protected] Stefano F. Cappa [email protected] Massimo Filippi [email protected] Daniela Perani [email protected] Maurizio Popoli [email protected] Gianfranco Spalletta [email protected] Gianfranco Spalletta 17. Electrophysiological study of striatal physiology in normal conditions and experimental parkinsonism: use of genetic and pathogenetic animal models 18. Celebrospinal fluid biomarkers in alzheimer’s disease and other neurodegenerative disease D. Lucilla Parnetti [email protected] Diagnosis: biomarkers 1. Proteomics of cognitive and movement disorders: Identification of molecular mechanisms associated with disease onset and phenotypic variability of frontotemporal lobar degeneration 2. Peripheral markers of oxidative stress, excitotoxicity and abnormal protein metabolism in neurodegenerative disorders. 3. Validation and search of CSF biomarkers for the early differential diagnosis of cognitive impairment. Development of new diagnostic approaches for prion diseases in humans. 4. Analisi Biochimica di parametri di stress ossidativo 5. Cognitive decline in multiple sclerosis 6. Search of predictive markers of AD based on amplification of disease-specific amyloid-beta oligomers 7. Mild cognitive impairment and dementias E. [email protected] Paolo Calabresi [email protected] Roberta Ghidoni [email protected] Carlo Ferrarese [email protected] Piero Parchi [email protected] Gabriele Siciliano [email protected] Laura Calzà [email protected] Fabrizio Tagliavini [email protected] Gianfranco Spalletta [email protected] Diagnosis: neuropsychology 1. Handwriting analysis as a novel diagnostic tool for Alzheimer’s disease in the clinical and forensic settings 2. Amnesic Mild Cognitive Impairment: neuropsychological features and new diagnostic criteria (Dubois et Al., 2007) 3. Predictors of conversion from MCI to dementia Maurizio Balestrino [email protected] Angiola Maria Fasanaro [email protected] Roberto Gallassi [email protected] 4. Individual evaluation of cognitive decline; validation Ildebrando Appollonio of ultrafast screening tools. [email protected] F. Biobanking Bioinformatics G. Ambient assisted living and socioeconomics 1. Home-based Empowered Living for people with dementia and Parkinson’s disease Roberto Monastero [email protected] 2. Neuro-rehabilitation and brain functional imaging: rehabilitation customization driven by best exploitation of celebral plasticity and health system sustability. 3. Role of information and communication technology (ICT) systems to improve quality of life, quality of care and safety of older patients with cognitive decline 4. A transdisciplinary orented proposal from a social sciences perspective Alessandra Pedrocchi [email protected] Alberto Pilotto [email protected] Vincenzo Maria Bruno Giorgino [email protected] 1. MECHANISMS OF NEURODEGENERATION A- Macromolecular interactions and Neurodegeneration Nome Contatti Istituto/Dipartimento Proposta di ricerca Dr. Pier Luigi San Biagio e-mail: [email protected] phone: +39 091 6809311 cell: +39 329 410 5564 Istituto di Biofisica - CNR, Via Ugo La Malfa, 153 , 90146 Palermo (Italy) Many untreatable neurodegenerative human disorders are associated with the aggregation of misfolded or natively unfolded proteins into amyloid fibrils. Our research activity concerns Alzheimer’s Parkinson’s and Albers-Schönberg diseases with the shared perspective of relating the understanding of the pathway leading to the fibrils formation to the design of appropriate drugs capable on preventing the protein aggregation. Alzheimer’s disease. Recent results suggested that the main neurotoxic species in Alzheimer’s Disease (AD) would not be the insoluble protein aggregates, made of extracellular deposits of Beta-amyloid peptide (Ab) fibrils, but rather the soluble oligomeric species, including small globular structures, 2.7 to 6.0 nm in diameter, and “protofibrils”. Conformational constrains imposed by small organic molecules could play a key role in modulating the fibrillogenesis process, so providing effective therapeutic tools to target both oligomeric and fibrillar species. In this perspective, polycyclic aromatic molecules could be of special interest as they might disrupt the molecular architectures precursors of fibrils by means of aromatic interactions, like stacking interactions with tyrosine and phenylalanine residues of Ab. In particular, natural polyphenols, composed of one or more aromatic phenolic rings, are a class of phytochemicals found in high concentrations in wine, tea, nuts, berries, fruits, cocoa, and an ample assortment of other plants. Some of them have been demonstrated to have anti-oxidant and anti-amyloidogenic effect and it has been speculated that they could prevent the development of Alzheimer's disease, not only through scavenging of reactive oxygen species, but also through directly inhibiting the deposition of fibrillar beta-amyloid in the brain. In the present project, we’ll test some natural molecules, such as ferulic acid (FA), a phenolic anti-oxidant present in fruit cell walls, hypericin (Hyp), a natural pigment extracted from Hypericum perforatum, widely used as a mild antidepressant and epigallocatechin gallate (EGCG), one of the major flavonoids of green tea. The aggregation process of Ab in the presence of these natural molecules will be studied by light scattering, steady-state and time-resolved fluorescence, FTIR and CD techniques. The analysis of the correlations between the effects of the studied compounds on the various stages of amyloid fibril formation, and their known physicochemical properties, will provide novel insights on the specific role of hydrophobic and aromatic interactions. Encouraging results have already been obtained using hypericin which has been shown to affect and to interfere with the early stages of polymerization process. The furtherance of this type of study, extended to other small molecules like FA and EGCG, hopefully will meet the goal of designing effective therapeutic tools to ideally target both oligomeric and fibrillar species, thus contributing to devise AD prevention strategies. Parkinson’s disease. Alpha-synuclein (aS) is the main constituent of Lewy bodies, intraneuronal fibrillar inclusions which are present in all patients affected by Parkinson’s disease (PD). From genetic and biochemical studies it is believed that aS is involved in the disease. AS is a natively unfolded protein with the ability to acquire secondary structure upon interaction with membranes or with itself. It is linked to PD by two evidences: the accumulation of amyloid fibrils of the protein and the autosomal dominant forms of the disease (A53T, A30P and E46K mutants). Both the biological role of this protein and the mechanisms involved in the ethiopathogenesis of PD are still unknown. The protein is located at the presynaptic terminal of neurons in all the CNS, where it exists free in the citosol or bound to synaptic vesicles. The membrane binding causes the formation of an amphipatic alpha-helix in the first part of the protein, which lies at the membrane surface without crossing the bilayer. Recent evidences show that aS is able to bind and permeabilize cell membrane. Our investigation will be devoted (i) to the characterization, at the single molecule level, of the channel like ability of aS and its architecture. The pore-forming ability of monomers and aggregates of different sizes will be investigated. Secondly, the lipid specificity will be also investigated by means of high resolution NMR and MS (both maldi-tof and esi-ms). The ability of aS to provoke or protect unsaturated acyl chains of membrane lipids from oxidation will also be investigated, by studying with MS the products of oxidation. As possible therapeutic perspectives, the organic or peptidic inhibitors of proteinprotein interaction or able to clog the already formed pores will be investigated. Furthermore, if indications of any lipid specificity will be obtained, we will develop lipid formulates suitable for sequestering aS lowering its level in solution. Albers-Schönberg disease. Mutations in the gene coding for the lysosomal Cl-/H+ exchanger CLC-7 lead to recessive osteopetrosis and Albers-Schönberg disease. Apart from the osteopetrotic phenotype the recessive disease is characterized by severe generalized neurodegeneration. Also knock-out of the plasma membrane localized CLC-2 lead to neurodegeneration (retinal degeneration and leukoencephalopathy). The mechanisms underlying these phenotypes are unclear. We are studying the functional properties of these two proteins using electrophysiological and optical methods, combined with mutagenesis, in order understand how their dysfunction may lead to neurodegeneration. In particular, we consider these proteins as potential risk factors for more generalized forms of neurodegeneration. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Public health. Therapeutic strategies for the prevention and treatment of neurodegenerative diseases. Molecular basis of amyloid aggregation Ente finanziatore Progetto strategico di ricerca finalizzata sulla malattia di Alzheimer (Art. 12/bis D.Lgs 229/99), “Beta-amyloid deposit on the cell membrane: role of metal ions and free radicals” Italian 'Ministero della Salute” Durata progetto 24 months, started 2001 Abstract del progetto Ente finanziatore Characterization of some key molecular mechanisms for the neuronal damage connected to AD. Specific objectives: (i) to identify of the key steps responsible for the amyloid depost formation on membrane model systems in vitro; (ii) to evaluate the role of the membrane lipid composition on the deposit; (iii) to understand the role of metal ions and oxidative stress on the fibril growth both in solution and on membranes; (iv) to establish an in vitro test for evaluating the ability of drugs to inhibit bA aggregation. “Animal neuropathies: molecular and functional analysis of the prionic proteins in Sicilian bovine race ” Italian 'Ministero della Salute Durata progetto 24 months, started 2003 Abstract del progetto Titolo progetto The main objective of the present proposal is to understand the mechanisms by which wild type and mutant prion proteins become misfolded in such a way as to provoke neurodegenerative diseases becoming transmissible agents. An extensive study of the aggregation process will be done by using static and dynamic light scattering and fluorescence techniques. Prin 2005 project: “The effects of metal ions on the protein aggregation processes” Ente finanziatore Italian 'Ministero Università e Ricerca' Durata progetto 24 months, started 2005 Abstract del progetto Ente finanziatore The project concerns the study of the effects of metal ions in solution on the protein aggregation processes. Main goals are (i) to understand the molecular mechanisms relevant for amyloid aggregation and to establish the relevance of metals in modulating solvent mediated protein-protein interactions. “Electrophysiological characterization of the alpha synuclein channels, a protein involved in Parkinson’s disease” Local Bank Foundation CARITRO Durata progetto 24 months, started 2008 Abstract del progetto Ente finanziatore The aim of the project is: i) to characterize the channel like activity of alpha synuclein (aS) and its role in the disease; ii) to distinguish the most active aS form; iii) to verify the ability of aS to bind to membranes and lipid requirements; iv) to investigate the role of aS in the prevention of the oxidation of unsaturated fatty acid chains Prin 2008 project: “Inhibition of beta-amyloid peptide aggregation by some natural compounds” Italian 'Ministero Università e Ricerca Durata progetto 24 months, started 2010 Abstract del progetto Ente finanziatore The aim of this project is to provide a molecular approach for preventing Abeta aggregation and toxicity through the use of some natural molecules potentially able of interact with Abeta by stabilizing or disrupt aggregates of different sizes. “Membrane transport of chloride and protons: mechanism of function of CLC family proteins involved in epilepsy and neurodegeneration” Compagnia San Paolo Durata progetto 36 months, started 2009 Abstract del progetto CLC-2 is a plasma membrane localized Cl- channel whereas CLC-7 is a lysosomal Cl-/H+ exchanger. Knock-out of CLC-2 in mice leads to retinal degeneration and leukoencephalopathy and knock-out of CLC-7 and human mutations in the CLCN7 gene Titolo progetto Titolo progetto Titolo progetto Titolo progetto lead to generalized neurodegeneration. The aim of the project is to understand better the transport function of these two proteins in order to get insight into their causative role in these forms of neurodegeneration. Nome Bruno Samorì Contatti E mail: [email protected] Istituto/Dipartimento Laboratory of Nanoscience and Nanobiotechnology, Department of Biochemistry, University of Bologna (Italy) (http://biocfarm.unibo.it/samori/) Proposta di ricerca Area di interesse identificata Basic research Abstract According to an emerging view, the aggregation-prone proteins associated to neurodegenerative, Parkinson’s, Alzheimer’s, and Creutzfeldt-Jakob diseases, can switch back and forth between functional and amyloidogenic/prionogenic conformations. Their conformational polymorphism poses tremendous challenges to standard methods in structural biology, which mask the complexity of their behaviour by recording observables as time and ensemble averages. Single-molecule methodologies can instead follow the time trajectories of individual molecules or molecular assemblies, can map their energy landscapes, and obtain distributions of molecular properties. These capabilities can provide information about the monomeric state of an amyloidogenic protein and about the very early stages of its amyloid cascade. This information is crucially needed for the design of new drugs against Parkinson’s or Alzheimer’s disease that must be capable to target upstream of the formation of the most neurotoxic oligomeric species. In fact concerns are now growing over the possibility that more harm than cure can be created whenever the deposits in the central−nervous−system, are targeted instead: in this way their dynamic equilibrium with the oligomeric forms can be shifted back to the latter. We have recently shown that Atomic Force Microscopy-based Single Molecule Force Spectroscopy (AFM-SMFS) can single out the different monomeric conformers of α-synuclein (the intrinsically unstructured protein associated to Parkinson’s disease) and of its mutants associated to genetic PD. (1,2). So far AFM-SMFS has been the only methodology that made it possible to achieve this goal. Ongoing projects: a) The integration of AFM-SMFS with Optical tweezers (OT) (in coll. with Carlos Bustamante (UC Berkeley). b) The determination of alpha synuclein aggregation step targeted by different potential drugs or chaperons, by using an integrated approach based on AFM- and OT-SMFS, AFM-imaging, Fluorescence Polarization Spectroscopy ( in collaboration with Luigi Bubacco, University of Padova) c) The conformational equilibria or prion proteins (in coll. with G. Legname SISSA Trieste, and Motomasa Tanaka, Riken, Japan) (1) Sandal, M., et al (2008). PLoS Biol 6, 999-1008. (2) Brucale, et al. (2009). Chembiochem 10, 176-83. Finanziamenti ricevuti Titolo progetto Nanomanipolazione di singole molecole delle proteine coinvolte in malattie neurodegenerative Ente finanziatore FIRB Nanobiotecnologie per dispositivi e sensori innovativi (G.U. 29.07.2005 n. 175) FIRB Metodologie e tecnologie innovative per la farmaceutica. CINECA RBNE03PX83 Durata progetto 2005-2008; 2005-2010 Abstract del progetto Sviluppo metodologie di nanomanipolazione di proteine intrinsecamente non foldate o disordinate Titolo progetto Meccanismi Neurodegenerativi nelle Malattie da Prioni (coordin. Catia Sorgato) Ente finanziatore PRIN 2009 Durata progetto 2010-2012 Nome Dr. Eugenia Polverini Contatti [email protected] Istituto/Dipartimento Department of Physics – University of Parma Proposta di ricerca Our studies are - and will be - focused on myelin proteins, to the aim of clarify their structure to function relationship and the mechanisms involved in demyelinating diseases, including multiple sclerosis. Two main proteins are under investigation: the Myelin Basic Protein (MBP) and the P2 myelin protein. MBP is a highly post-translationally modified structural component of central nervous system myelin, the multilamellar membrane wrapped around nerve axons. MBP has a high net positive charge and its principal role seems to be the adhesion of the cytosolic surfaces of the compact myelin sheath, whose structural integrity determines the speed of transmission of action potentials along the axon. However, MBP seems to be a multifunctional protein, since it also interacts with other cytoskeletal and signalling proteins, tethering some of them to the oligodendrocyte plasma membrane. The three-dimensional structure of MBP in situ is still unknown, due to its intrinsic flexibility and dependence of conformation on environment. Therefore a neighbourhood-dependent structural charcterization of fragtments is attempted. In the classic 18.5 kDa MBP sequence, there is a highly conserved fragment, constituting two regions with different putative functionality. The first region seems to associate with the membrane, and comprises the primary immunodominant epitope in multiple sclerosis; the second one is a proline-rich segment, that has been predicted to be a ligand for protein kinase SH3-domains. Due to the functional relevance of this fragment of MBP, we’re trying a structural characterization - in the absence and in the presence of protein’s post-translational modifications - by means of molecular dynamics simulations in membrane and of spectroscopic techniques (in collaboration with the University of Guelph, Ontario, Canada). P2 myelin protein is a basic peripheral nervous system protein localized to the cytoplasmic spaces in the myelin lamellae. It was recognized as the factor inducing the Experimental Allergic Neuritis, an animal model for the study of the Guillain-Barrè syndrome, a demyelinating disease of the PNS in humans. The interest for the putative autoantigenic role of the P2 protein and the need to understand its function in the myelin sheath has led to the study of its structural characteristics and behaviour, basing on the solved crystal structures. By investigating its interaction with different kinds of lipids, on the one hand we want to clarify its transport role and on the other hand we want to verify its putative involvement in the compactness of PNS myelin sheat. Different approaches are carrying out: molecular docking simulations, spectroscopical investigations and, in collaboration with the University of Oulu, Finland, crystal X-ray diffraction and small-angle X-ray scattering. Area di interesse identificata myelin proteins and demyelinating deseases Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto University local research funds (FIL) Nome Prof. ROBERTO DE RENZI - Dr. MARIA GRAZIA BRIDELLI Contatti [email protected] Istituto/Dipartimento Physics department University of Parma Parco Area delle Scienze, 7/A, Parma ITALY Proposta di ricerca The goal of our research is threefold: 1) study of the nucleation rate and the growth rate of the amyloidogenic process by controlling and modulating the solution parameters such as temperature, pH, solvent composition, and peptide concentration, not only influencing the final fibril state but also the formation kinetics. The analysis of the aggregation process will be performed in solution by employing CD and Fluorescence spectroscopies by following the change of fluorescence emission of small hydrophobic molecules, called amyloid ligands, which do not bind to specific proteins but are selective for protein aggregates with a cross β-sheet conformation. Dynamic Light Scattering measurements will allow to monitor size and shapes of the fibrils during the aggregation process. 2) study the conformational state of proteins in the native and amyloid form at very low hydration level by investigating the structural properties of the first hydration shell. It is well known that proteins are surrounded in the cell by a hydration shell formed by interacting water molecules that form patches locally distributed in dependence on the heterogeneous surface chemistry or occluded in the internal cavities. This bound water in the crowded cytoplasm may not be enough to fully cover the protein surface with a monolayer aqueous sheath, moreover about 0.4 g of water/g of protein are enough to assure protein normal functionality. Two experimental techniques can be applied to investigate this structured water to gain information on the geometry of the protein surface and secondary structure. Thermally Stimulated Depolarization Currents (TSDC) technique, applied in the past to a variety of natural and synthetic biopolymers has recently been applied to the study of lysozyme. It has allowed recognizing the main features of the hydration as a function of secondary structure, revealing a clear dominance of the βstructure in the amyloid form. Fourier Transform Infrared Spectrometry as well has proven to be highly versatile in the analysis of protein conformation by monitoring the alteration in protein secondary structure as changes in the Amide bands. 3) by means of the molecular dynamics simulation techniques, study the conformational changes that proteins undergo under the critical environmental conditions that in vitro cause the fibrils formation. This approach could be useful to investigate the pathway(s) of the transition and its molecular features. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Conformational transitions of proteins from native to amyloid form. University local research funds FIL 2008 Hydration structure analysis of Lysozyme amyloid fibrils Ministero dell’Istruzione, dell’Università e della Ricerca 2y Combined Thermally stimulated depolarization currents (TSDC) and FTIR techniques have been employed to investigate the conformation of hen egg white lysozyme in native and amyloid form, in the state of powder at very low hydration level. The spectra reveal specific features that can be attributed to water texture around the secondary structure adopted by the macromolecule. Nome Contatti Tel. E mail Dipartimento Proposta di ricerca Sandro Sonnino Via fratelli Cervi 93, 20090 Segrate (Mi) Italy, +39 0250330360, fax +39 0250330365, [email protected] Medical Chemistry, Biochemistry and Biotechnology, University of Milan Role of altered composition of plasma membrane complex lipids in lipid-protein interactions, membrane organization and neurodegeneration. Area di interesse identificata 1. MECHANISMS OF NEURODEGENERATION F. Endocrine and Metabolic Factors Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Angelo Poletti 02-5031.8215 [email protected] Dipartimento di Endocrinologia, Fisiopatologia e Biologia Applicata , Centro di Eccellenza sulle Malattie Neurodegenerative Universita' degli Studi di Milano via Balzaretti 9 - 20133 Milano Macromolecular interactions and Neurodegeneration Ente finanziatore ROLE OF ALTERED PROTEOLYSIS IN THE DEGENERATION OF MOTOR NEURONS IN in vivo AND in vitro MODELS OF FAMILIAL AMYOTROPHIC LATERAL SCLEROSIS. INVOLVEMENT OF CONSTITUTIVE AND IMMUNO PROTEASOMES AND OF THE AUTOPHAGY−LYSOSOME PATHWAY TELETHON – Italy Durata progetto three years (end 2010) Abstract del progetto Amyotrophic lateral sclerosis (ALS) is a neurological disorder primarily affecting motor neurons, i.e. the neurons responsible for the contraction of muscles that control voluntary movements, speach, swallowing, respiration. The disease, whose causes are still largely unknown, is characterized by a progressive paralysis that leads to death for respiratory failure. As in some familiar forms of ALS there are mutations in the gene that codes for the enzyme superoxide dismutase 1 (SOD1), it has been possible to create experimental models of the disease to facilitate the research on the causes that lead to motor neurons death. Both in ALS patients and in the experimental models the vulnerable neurons contain aggregates of abnormal proteins, whose accumulation can be responsible for the degeneration. It is therefore possible to hypothesize that a role in the pathology is played by dysfunctions of the proteolytic mechanisms, i.e. of the mechanisms that normally degrade intracellular protein. We propose to investigate whether motor neurons degeneration in ALS is due to a failure of protein degradation, using two experimental models (transgenic mice and a motor neuron cell line, both expressing mutant human SOD1) and analyzing two different intracellular proteolytic systems: the proteasomes and the autophagosome−lysosome Motorneuron degeneration in Spinal and Bulbar Muscular Atrophy: from the molecular mechanisms to the potential therapeutical approaches TELETHON - ITALY Three Years (End 2011) Titolo progetto Ente finanziatore Durata progetto Abstract del progetto CAG/polyglutamine (polyQ) diseases, the largest class of hereditary neurodegenerative diseases, include SpinoBulbar Muscular Atrophy (SBMA), a motorneuron disease linked to the androgen receptor (AR) mutation. SBMA is a valuable model of polyQ diseases, because ARpolyQ neurotoxicity is triggered by testosterone, the endogenous AR ligand, possibly via protein conformational changes. In SBMA animal models, the symptoms are reversed by testosterone removal suggesting that SBMA patients can be treated with drugs preventing testosterone action. The overall objectives of the study are to find the mechanism(s) by which ARpolyQ neurotoxicity can be modulated by testosterone with the aim to identify novel therapeutical approaches for SBMA and possibly indications useful for other polyQ diseases. Therefore, all the studies proposed will be carried out in absence or in presence of testosterone, and four different specific aspects, thought to be involved in SBMA, have been selected: − interactions of the two major intracellular degradative systems: the Titolo progetto Ente finanziatore Durata progetto Abstract del progetto ubiquitin−proteasome and the autophago−lysosome pathways in the removal of the neurotoxic ARpolyQ; analysis of ARpolyQ solubility and aggregation, and their modulation by selective AR modulators (SARM) and HSP−modulators. − analysis of the axonal antero−retrograde transport in presence of ARpolyQ activated by testosterone (and SARMs). This aim will include the expression analysis of some proteins related to axonal outgrowth and controlled by testosterone (neuritin or CGP15). − analysis of the possible toxicity of ARpolyQ in muscle cells, and the interaction between motorneuron and muscle cells in SBMA. − analysis of the AR promoter activity to identified factors downregulating the ARpolyQ levels in motor neuron. These studies will provide information of the mechanisms by which ARpolyQ becomes neurotoxic after testosterone interaction, that may be used to design novel therapeutical approaches for SBMA and related diseases Alterations of Axonal Functions and Neurodegeneration in Motor Neuron Diseases FONDAZIONE CARIPLO Three Years (end 2012) Motor neuron diseases (MNDs) are a group of sporadic or hereditary neurodegenerative diseases characterized by selective motor neuron death. They may affect children and adult patients with relentless progression rate leading to amyotrophic paralysis and death. Several genetic, molecular and cellular alterations have been reported in the different MNDs and interpreted as potentially relevant in the pathogenesis responsible for neuronal death. However, it is still unclear whether these alterations are cause or effects of initial insults capable to perturb cell functions. The clinical trials attempted with the aim to counteract these alterations have proven to be largely ineffective. A common aspect of most MNDs is the alteration of motor neuron axons, thus suggesting specific axonal function impairment. Axonal processes can reach a considerable length (up to 1 meter) and are fundamental for motor neuron functions and survival. Axons allow neuronal control of muscle contraction, but also collect survival/trophic factors released from the muscle, and required for motor neuron physiology. Nerve damages and axonal loss result in motor neuron death and muscle atrophy. The project will focus on three major human MNDs, spinal muscular atrophy (SMA), spino-bulbar muscular atrophy (SBMA) and amyotrophic lateral sclerosis (ALS), affecting similar motor neuron population with different rate of cell death. It will be based on advanced analytical techniques and specific expertise of five research units. The units will utilize complementary genomic, biochemical, molecular and cellular assays to study the mechanisms of axonal dysfunctions in the MNDs studied. Since gene mutations, gene expression impairment, and altered post-translational modifications of specific proteins are involved in these MNDs, the project will be dedicated to unravel such alterations at the genomic and molecular level with the final aim of identifying novel common therapeutical targets. Main objectives: i) development and/or improvement of motor neuron models of SMA, SBMA and ALS; ii) analysis at the (nuclear and mitochondrial) genomic level of alterations determined by the lack or the expression of proteins (FL-SMN/a-SMN, mutant ARpolyQ, mutant SOD1) responsible for the MNDs selected; iii) morphological, cellular, molecular and biochemical analyses of the effects of the considered proteins on axonal functions/dysfunctions in the MNDs selected.Five different research institutions (University of Milan, Center of Excellence on Neurodegenerative Diseases; IRCCS Foundation Neurological Institute C. Besta; IRCCS Italian Auxologic Institute; Mario Negri Institute for Pharmacological Research; National Institute of Molecular Genetics) with specific expertise in the basic mechanisms of neurodegeneration will be involved. The approval of this project would enable to establish a critical mass of researchers devoted to MNDs in Milan, allowing the recruitment and formation of young scientists. Nome Contatti Istituto/Dipartimento Antonio Malgaroli, Gian Giacomo Consalez 02 2643 4886 [email protected], [email protected] Università Vita-Salute San Raffaele Proposta di ricerca Optical And Biomolecular Methods For The Functional Investigation Of Neural Circuits In Vivo. Area di interesse identificata Analisi della funzione sinaptica nei tessuti nervosi normali e in modelli di neurodegenerazione Finanziamenti ricevuti Titolo progetto Optical And Biomolecular Methods For The Functional Investigation Of Neural Circuits In Vivo. Ente finanziatore Fondazione CARIPLO (pending) Durata progetto 2 anni Every human behavior depends on an activity change in one or more brain areas, often with the contribution of very small subgroups of cells and synapses. Conversely, when a brain disease whatsoever, a tumor, a vascular disorder, a grey or white matter pathology, etc., modifies some behavioral aspect, inevitably this effect arises from a change in neural activity. Hence, viewing the fine structure of the brain by the most advanced imaging techniques is certainly important but it would be more important to find some effective way to detect changes in neuronal and synaptic activity during both physiological and pathological processes. Indeed, for many different body organs, the kidney, the heart, the liver, the pancreas, etc, the early detection of signs and symptomsand the choice of an optimized therapeutic approach often depend more on the availability of a large set of chemical and molecular markers of cell function rather than on our ability to visualize fine details of the tissue anatomy. Unfortunately techniques available today to monitor brain function, such as E.E.G., fMRI, fNIRS, etc., either sample brain activity very indirectly, through changes in metabolism and blood flow, or do not have the resolution required to pinpoint changes in small networks of neuronal cells and synapses. At present, anomalous states of brain activity can be detected only when they arise from the concerted activity of large numbers of neurons and synapses, such as in epilepsy, or when cell and/or synaptic and/or axonal damage becomes very profound. For all the above reasons, the demand for a methodology that could yield a more direct readout of in vivo neuronal and synaptic activity is extremely high. In this research project we are planning to further develop a methodology that was recently designed in one of our labs to provide a very direct and sensitive readout of brain activity. This is based on a family of recombinant molecules which were designed in our labs to detect synaptic activaton via the application of exogenous markers. This methodology is capable of revealing the physiological activation of brain synaptic networks with very high sensitivity and resolution. These striking results have prompted the generation of a series of transgenic animal models and analytical tools that will be used for further in vivo validation and for the selective probing of specific subgroups of neuronal cells and pathways, both in physiological and pathological conditions. The final goal of this project is to test the applicability of this approach to the quantitative analysis of brain activity. We expect that this work will generate important information for the development of similar approaches to the human brain. Nome Contatti Tel. E mail Istituto/Dipartimento Flavia Valtorta Flavia Valtorta, Fabio Grohovaz, Daniele Zacchetti 02-2643-4826 (Valtorta) 4811 (Grohovaz), 4817 (Zacchetti) [email protected], [email protected], [email protected] San Raffaele Scientific Institute, Division of Neuroscience Via Olgettina 58 20132 Milano, ITALT Proposta di ricerca. Molecular mechanisms in neurotrasmission: effects of neuron and glia phenotypes in synaptic efficiency and synaptopathy Comprehensive goal of our research is the elucidation of the molecular mechanisms that underlie communications between cells of the brain, both in the adult and during development. This proposal involves the complementary expertise and the tight collaboration of two research units: the NEUROPSYCHOPHARMACOLOGY UNIT (Flavia Valtorta) and the CELLULAR NEUROPHYSIOLOGY UNIT (Fabio Grohovaz and Daniele Zacchetti). The main form of communication between neurons is achieved at the synaptic level through regulated secretion of neurotransmitter. This process is of paramount importance for information processing in the central nervous system and is known to undergo dysfunction in a number of pathophysiological states. Substantial advancements have been made concerning the identification of the proteins involved in the basic mechanism of neurotransmitter release. Much less is known about the molecules that participate to the assembly of the complex synaptic machinery during development. Research in Valtorta’s group will develop along two main lines: Aim 1: Diseases of synaptic origin. The main focus will be on the family of the synapsins and their link to epilepsy and autism. Synapsins are a family of brain phosphoproteins involved in neurotransmitter release through regulation of synaptic vesicle availability. Recent data from the lab show that synapsins have also an important function in the organization of synaptic contacts. Synapsin KO mice develop an epileptic phenotype, and mutations in the human synapsin genes have been recently associated with epilepsy and autism spectrum disorders. The central interest is to investigate the molecular roles of synapsin in synapse formation and function, and how these roles relate to the development of brain pathology. Another project concerns the identification of novel molecular interactors of synuclein and of their role in neurodegeneration associated with Parkinson disease. Aim 2: Membrane trafficking in neuronal development. The process that leads from undifferentiated, round neuronal precursors to fully differentiated, highly polarized neurons is extremely complex. Among the various phenomena driving this process, those associated with membrane trafficking are of fundamental importance. The group is interested in studying membrane trafficking in the very early steps of neuronal development. In this respect, they have identified a process of developmentally-regulated, high-capacity endocytosis as one of the earliest marker of neuronal polarity, and will investigate its function in neuronal commitment and axon pathfinding. This investigation will be extended to the phenotypical characterization of the early steps of neuronal commitment of neural stem/progenitor cells and iPS (induced-pluripotent stem cells). In the same cells, the late steps of neuronal development will also be studied, i.e. the formation of synapses and their functional refinement, which are essential phenomena for the proper integration of cells in neuronal networks. The work in Grohovaz’s research unit is focused on the general comprehension of the physiopathological mechanisms underlying neuroprotection or leading to neurodegeneration. The group mostly uses brain primary cultures and applys molecular biology and in vitro cellular approaches, including live cell imaging (calcium imaging and total internal reflection microscopy). Electrophysiology and animal behavioral studies are conducted in collaboration with other research groups. A main project deals with the generation of oxidative stress and the ensuing protection mechanisms in both neurons and astrocytes. In particular, the mechanisms of iron entry and its influence on the production of reactive oxygen species are investigated. On the other hand, in light of the importance of neuroinflammation, interest is also directed to the role of glia in the neurodegenerative processes. More specifically, the molecular mechanisms involved in the process of glia activation and the influence the activated phenotype has on neuronal survival are investigated. Along with the study of these general physiopathological mechanisms, the group addresses specific issues within the following disease-oriented projects: (i) Alzheimer’s disease pathogenesis: the regulation of BACE1/BACE2 expression; the implication of beta-secretase activity on amyloid-beta deposition; the cellular effects of the various amyloid-beta forms on different cell types from brain. (ii) Derangement in intracellular iron homeostasis and hereditary ferritinopathy: analysis of the mechanisms of iron entry; cytotoxic effects of iron overload; cellular effects of ferritin light chain mutations. (iii) Pathogenesis of neuronopathic (type A) Niemann-Pick disease: role of sphingosylphosphocholine on changes in astrocytic phenotype; impairment of neuronal function. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Synaptic disfunction and neuron-glia interplay Research unit in Molecular Neuroscience I.I.T. (Istituto Italiano di Tecnologia) 5 years (2008-2012); 600'000 EUR Neurotransmitter release from nerve endings is the main form of information transfer from one neuron to another or to an effector cell. Release occurs by exocytosis from storage organelles, the synaptic vesicles, and is triggered by the action potential-induced depolarization of the nerve terminal. The project pivots on the idea that synapses perform an electro-chemical computation of both local and environmental signals they receive. In this respect, a key role is played by astrocytes, the close partners of neurons that, upon stimulation, release a number of neurotransmitters and signaling molecules. The development of these processes is investigated by developing and exploiting innovative molecular imaging approaches. Nome Contatti Tel. E mail Istituto/Dipartimento Maurizio Popoli Tel: +390250318361 E-mail: [email protected] Center of Neuropharmacology, Department of Pharmacological Sciences, CEND, University of Milan Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto 1A. Presynaptic molecular machinery regulating glutamate release as pathophysiological mechanism in amyotrophic lateral sclerosis Nome Contatti Tel. E mail Istituto/Dipartimento Daniela Tardito Tel: +390250318382 E-mail: [email protected] Center of Neuropharmacology, Department of Pharmacological Sciences, CEND, University of Milan Proposta di ricerca Area di interesse identificata 1A. Toward exploiting the screening potential of miRNome for the identification of genes and pathways involved in neurodegenerative disorders Finanziamenti ricevuti Titolo progetto Ente finanziatore The regulation of neuroplasticity in the response to therapeutic drugs for mood disorders. The role of micro RNAs Cariplo Foundation Durata progetto March 2010-february 2013 Abstract del progetto Aim of this project is to investigate, by means of an integrated approach that merges preclinical and clinical studies with up-todate bioinformatic, pharmacological, genomic and genetic techniques, the role of the posttranscriptional regulators miRNAs in Mood Disorders aetiology and antidepressant response, with regard to neuroplasticity. Overall, the main objectives of the present project will be: 1. to clarify the role of miRNAs in the molecular effects of different antidepressants on the posttranscriptional modulation of genes coding for modulators of neuroplasticity with an integrate approach based on preclinical and clinical studies. 2. to discover new molecular targets for the development of innovative therapeutic strategies for the treatment of drug resistant depression 3. to identify molecular markers associated to the antidepressant response in human peripheral tissues for the optimization of drug treatment in mood disorders. The integration of data obtained in clinical and preclinical studies will permit the validation of blood leucocytes as cellular models for the study of mental disorders 4. to identify new susceptibility genes and pharmacogenetic markers for optimization of treatment in mood disorders. Nome Maria Pennuto, PhD Contatti Phone: +39 010 71781793, Fax: +39 010 720321 email: [email protected] Italian Institute of Technology, Department of Neuroscience and Brain Technology, Genoa 16163, Italy Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Mechanisms of Neurodegeneration Identification of signaling pathways in neurodegenerative propeinopathies Brain folding diseases, including Alzheimer’s disease, amyotrphic lateral sclerosis, and polyglutamine (polyQ) diseases, are neurodegenerative disorders characterized by the accumulation of toxic aggregation-prone proteins (Bertram and Tanzi, 2005; Ross and Poirier, 2004; Taylor et al., 2002). PolyQ diseases are a family of nine neurodegenerative disorders, including spinal and bulbar muscular atrophy (SBMA). These disorders are caused by expansion of glutamine tracts in nine genes, such as androgen receptor (AR). The mechanism through which the mutation causes neurodegeneration is not known. Despite the ubiquitous distribution of the disease proteins, neurons are especially vulnerable to expanded polyQ. Moreover, specific populations of neurons are affected in each polyQ disease. This suggests that the polyQ tract cannot be the sole determinant of disease pathogenesis. Among polyQ diseases, SBMA represents an example where the disease protein strikingly dictates disease features. SBMA is characterized by the loss of lower motor neurons and skeletal muscle atrophy. In SBMA, only males are fully affected. Gender-specificity is the result of the conversion of polyQ AR into a toxic species induced by binding to its natural ligand testosterone. AR is a hormone-activated transcription factor. The mechanism through which ligand binding converts polyQ AR into a neurotoxic molecule is unknown. Our long-term goal is to make a meaningful contribution to our understanding of the molecular mechanisms of neurodegeneration in brain folding diseases, so that treatment may be developed. Our objective is to characterize the impact of protein context, cell context, and native protein function on polyQ toxicity. We will test the central hypothesis that the toxicity of polyQ AR is influenced at the post-translational level by phosphorylation of specific residues in the disease protein, by tissue-specificity of disease protein expression, and by aberrant protein function, e.g. interaction with transcription co-factors and DNA. Greater insights into the molecular mechanisms underlying the pathogenesis of polyQ diseases is important to developing novel and effective therapeutic strategies for these diseases (reviewed by Pennuto and Fischbeck, 2009). Finanziamenti ricevuti Titolo progetto TARGETING ANDROGEN RECEPTOR FOR DEVELOPMENT OF SBMA THERAPEUTICS Ente finanziatore Durata progetto Abstract del progetto Muscular Dystrophy Association (USA) 01/07/2008 to 30/06/2011 The objectives of the MDA application are to identify post−translational modifications, such as phosphorylation, ubiquitylation, and SUMOylation of AR that reduce the toxicity of mutant protein, and to use this information to develop effective therapeutic interventions. We propose to accomplish these objectives by pursuing the following three specific Aims: To determine the impact of protein kinase A phosphorylation, ubiquitylation, and Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto SUMOylation on the toxicity of mutant AR. To evaluate the therapeutic efficacy of IGF−1 in vivo. To select an effective system to deliver IGF-1 to SBMA mice. POLYGLUTAMINE DISEASES: IMPACT OF PROTEIN AND CELL CONTEXT ON NEUROTOXICITY FP7−PEOPLE−2009−RG: Marie Curie International Reintegration Grant 01/04/2010 to 31/03/2014 The objectives of this proposal are to characterize the impact of three components to polyglutamine toxicity: 1) protein context; 2) aberrant protein−protein interactions; and 3) cell context. We will test the central hypothesis that the toxicity of polyglutamine AR is influenced at the post−translational level by phosphorylation of specific residues in the disease protein, by aberrant interaction with the Akt effector FOXO, and by tissue−specificity of disease protein expression. INTERPLAY BETWEEN ANDROGENIC/ANABOLIC STEROID AND IGF-1 SIGNALING IN AMYOTROPHIC LATERAL SCLEROSIS Thierry Latran Foundation (France) 01/01/2011 to 31/12/2012 The objectives of this study are to i) Determine the effect of androgens and the related anabolic steroid nandrolone in ALS pathogenesis; and ii) Characterize the molecular cross−talk between androgens and muscle−specific mIGF−1 in the maintenance of muscle phenotype. INSULIN−LIKE GROWTH FACTOR 1/AKT AND ANDROGEN SIGNALING CROSSTALK IN THE PATHOGENESIS OF SPINAL AND BULBAR MUSCULAR ATROPHY Telethon-Italy 01/01/2011 to 31/12/2013 The objective of this application is to elucidate how phosphorylation of mutant AR by Akt is regulated. The central hypothesis of this application is that phosphorylation of mutant AR by Akt is altered by polyglutamine expansion, ligand binding and age and that this modification is coordinated with other post−translational modifications, including methylation and palmitoylation. Nome Tanganelli Sergio Contatti [email protected] Istituto/Dipartimento Dept. of Clinical and Experimental Medicine Pharmacology Section, University of Ferrara, Italy Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Mechanisms of Neurodegeneration : A. Macromolecular interactions and Neurodegeneration Ente finanziatore Neurotensin and neurotensin antagonist in an animal model of Parkinson’s disease: therapeutic perspectives and role of NMDA/neurotensin interaction. Sanofi-Aventis Durata progetto 2007-2010 Abstract del progetto The tridecapeptide neurotensin (NT) significantly enhances glutamatergic signaling in different brain areas and amplifies the NMDA-receptor signaling, probably by activating the high-affinity neurotensin receptor (NTS1), as shown by the NTS1receptor antagonist SR48692-induced counteraction of this effect. These findings suggest a reinforcing action of NT on several functions exerted by glutamate in the central nervous system, in particular on the glutamatemediated excitotoxicity. NT could be implicated in the pathogenesis of chronic and acute neurodegenerative disorders, such as Parkinson’s disease. To further investigate this hypothesis, the aims of the project, which will combine biochemical, functional, morphological and behavioral experiments, will be evaluate: 1) the putative neuroprotective effects of a pretreatment with SR48692 (systemically administered) in an animal model of Parkinson’s disease [unilateral 6-hydroxydopamine (6-OHDA) lesion]; 2) the functional and physio-pathological relevance of NMDA/NT receptor interactions in the striatum of naïve and 6OHDA -lesioned rats. 1. MECHANISMS OF NEURODEGENERATION B. Neurotoxic stimuli Nome Domenico Garozzo Contatti [email protected] 0957338259 3473576202 CNR ICTP Catania Via Gaifami 18 95125 Catania Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Glycomics Finanziamenti ricevuti Titolo progetto CSF Glycomics in AD and other neurodegenerative diseases Ente finanziatore Durata progetto Abstract del progetto MIUR (Italy) 3 years Protein glycosylation is the most common among post-translational modifications (PTMs), which are key to the regulation of functional activities of proteins. Quantitative and qualitative information about PTM stages of proteins is crucial in the discovery of biomarkers of disease. Macroheterogeneity of protein glycosylation depends on the extent of glycosylation site occupancy while microheterogeneity refers to differences in the sugar chain structures. Differences of protein glycosylation may be indicative of species synthesized in the central nervous system with respect to serum liverderived glycoforms, [as already demonstrated for protein Reelin, which is implicated in Alzheimer disease and in other neurodegenerative disorders (Botella-Lopez et al., 2006)]. Recent insights into neurodegenerative disorders caused by abnormal glycosylation of protein Seipin (Seipinopathy) show that modifications of Seipin glycosylation result in improper folding leading to accumulation of the abnormal protein in the endoplasmic reticulum, formation of ubiquitinated inclusion, and activation of UPR (Ito and Suzuki, 2009). Preliminary investigations based on lectin-blotting of CSF glycoproteins revealed quantitative modifications in Alzheimer disease and altered glycosylation of CSF transferrin (Taniguchi et al. 2008). We apply mass spectrometry based glycomics of CSF as potential tool for to detect possible macro-and microheterogeneity of CSF glycoproteins associated to Alzheimer disease. The availability of CSF depository from patients with AD and related disorders and established interactions with clinical platforms will make possible to translate the results to clinical proteomics. The objective is to find out specific protein posttranslational modifications related to glycosylation which in turn might be related to the analysed cellular phenomena associated to neurodegeneration. Nome Maurizio Taglialatela, MD PhD – Professor of Pharmacology Claudio Russo, PhD – Associate Professor of Pharmacoloy Alfonso Di Costanzo MD – Associate Professor of Neurology Giovanni Scapagnini MD PhD – Associate Professor of Clinical Biochemistry Contatti Email: [email protected] Tel. +39-0874-404894/4851/4891 Istituto/Dipartimento Dept. of Health Science, University of Molise, Via De Sanctis, 8610 Campobasso - ITALY Proposta di ricerca Area di interesse identificata Intracellular signalling during neurodegenerative events triggered upon APP overexpression. The overexpression and the aberrant processing of the amyloid precursor protein (APP) is a central event in the pathogenesis of Alzheimer’s Disease (AD). Recent data suggest that the neuronal death that occurs in different neurodegenerative conditions may arise from an aberrant signal related to cell cycle re-activation in postmitotic neurons. We are currently investigating the role of APP and of Aβ peptides as signalling molecules involved in MAPK and cell cycle activation. We propose to study a specific molecular signalling involving APP and its proteolytic processing by BACE1 and Presenilins, whose failure could explain at the same time the formation of Aβ, the induction of cell cycle and neuronal dysfunction observed in the development of AD. • Venezia V. et al., Amyloid precursor protein and presenilin involvement in cell signaling. Neurodegenerative Diseases. 2007, 4 (2-3): 101-111. • Nizzari M. et al., Amyloid precursor protein and Presenilin1 interact with the adaptor GRB2 and modulate ERK 1,2 signaling. The Journal of Biological Chemistry. 2007, 282 (18): 13833-13844. Involvement of vascular factors as modulators of APP processing and signaling. The complex interaction between vascular components and tissue homeostasis is particularly relevant in the brain for the survival of vulnerable and irreplaceable postmitotic neurons. It is debated whether the presence of vascular deposits of Aβ peptides might represent a cause of oxidative stress and a predisposing condition toward the development of dementia, or rather a protective endothelial mechanism. In this context we are investigating the role of vascular components such as thrombin and apolipoproteinE as modulators of endothelial processing of APP, in Aβ formation and in Aβ-induced toxicity/protection. • Russo C. et al., Opposite roles of apolipoprotein E in normal brains and in Alzheimer’s disease. Proc. Natl. Acad. Sci, 1998, 95 15598-15602. • Russo C. et al. Presenilin-1 mutations in Alzheimer's disease. Nature. 2000; 405 (6786):531-532. • Russo C. et al., The amyloid precursor protein and its network of interacting proteins: physiological and pathological implications. Brain Res Brain Res Rev. 2005 48 (2): 257-264. Role of specific classes of ion channels in AD-related neurodegeneration. Intracellular potassium concentrations play a key role in cell survival. A decrease in cytoplasmic [K+]i, mainly caused by an increased activity of plasma membrane voltage-gated potassium channels, triggers cell death. Changes in K+channel activity play a major pathogenetic role in many neurodegenerative disorders, including Alzheimer’s disease (AD). In fact, β-amyloid fragments (Aβ) alter the properties of K+ currents in mammalian neurons. We have described a selective modulation of a neuronal K+ channel (Kv3.4) upon Aβ exposure and we have shown that the inhibition of this channel prevents cell death triggered by the neurotoxic stimulus. We are currently investigating the potential neuroprotective role of Kv3.4 blockers in several models of AD-related neurodegeneration.. • Pannaccione A., et al. Nuclear factor-kappaB activation by reactive oxygen species mediates voltage-gated K+ current enhancement by neurotoxic betaamyloid peptides in nerve growth factor-differentiated PC-12 cells and hippocampal neurones. J Neurochem. 2005 Aug;94(3):572-86. • Pannaccione A, et al. Up-regulation and increased activity of KV3.4 channels and their accessory subunit MinK-related peptide 2 induced by amyloid peptide are involved in apoptotic neuronal death. Mol Pharmacol. 2007 Sep;72(3):665-73. Oxidative stress as a pathogenetic mechanisms and therapeutic target for Alzheimer Disease. Reduction of cellular expression and activity of antioxidant proteins leading to oxidative stress are fundamental causes for brain aging and neurodegenerative diseases. Deregulation of the antioxidant protein Heme oxygenase-1 (HO-1) has been associated with the pathogenesis of Alzheimer's disease, multiple sclerosis and brain aging. Moreover, a number of experimental and epidemiological studies have recently supported the beneficial effects of some commonly used natural products such as curcumin, possibly by increasing the activity of the HO-1 protein. Curcumin has been reported to decrease oxidative damage and amyloid deposition in a transgenic mouse model of Alzheimer's disease, and to reverse Aβ-induced cognitive deficits and neuropathology in rats. We are currently investigating the potential neuroprotective role and the underlying mechanisms, of HO-1-inducing natural compounds in various in vitro and in vivo models of AD. • Scapagnini G. et al., Caffeic acid phenethyl ester and curcumin: a novel class of hemeoxygenase-1 inducers. Mol Pharmacol. 2002, 61(3):554-61. • Scapagnini G. et al., Ethyl ferulate, a lipophilic polyphenol, induces HO-1 and protects rat neurons against oxidative stress. Antioxid Redox Signal. 2004, 6(5):8118. • Scapagnini G. et al., Curcumin activates defensive genes and protects neurons against oxidative stress. Antioxid Redox Signal. 2006, 8(3-4):395-403. Physical exercise and cognitive decline. Epidemiological studies have shown that physical exercise can delay the occurrence and the progression of dementia, but also to improve physical, cognitive and psychological performances, with a positive impact on the quality of life. The mechanisms involved and the identification of potential biomarkers predictive of a positive response to physical activity are areas of intense investigation. In our group, we are currently investigating the potential preventive role of physical exercise programs on cognitive decline in subjects at risk to develop dementia. Our focus will be on subjects presenting with a subjective memory disturbance or affected by mild cognitive impairment (MCI). • Tedeschi G., et al. Brain atrophy and lesion load in a large population of patients with multiple sclerosis. Neurology. 2005 Jul 26;65(2):280-5. • Tedeschi G., et al. Correlation between fatigue and brain atrophy and lesion load in multiple sclerosis patients independent of disability. J Neurol Sci. 2007 Dec 15;263(1-2):15-9. Finanziamenti ricevuti Titolo Regulation of K+ channels by APP-interacting proteins: functional characterization and progetto implications for AD-related neurodegeneration Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Regione Campania (2000-2003) 3 years Recent experimental evidence suggest that K+ channels play a major pathogenetic role in Alzheimer disease. In the CNS, phosphorylation of voltage-gated K+ channels by tyrosine kinases (PTK) is a fundamental process regulating their functional activity; its inhibition exerts a neuroprotective action in several experimental models of neuronal damage. On the other hand, the expression of several subclasses of K+ channels can reduce cellular tyrosine kinase activity, thus suggesting the existence of a reciprocal interaction between PTKs/PTPases and K+ channels. Given that the cytosolic domain of β-APP influences, through its adaptor proteins Dab (disabled), X11 and Fe65, the tyrosine kinase activity of Abl, in the present proposal we will: A. Evaluate the potential modulation of several classes of K+ channels (Kv1.2, Kv1.3, Kv1.5, Kv2.1, ERG, KCNQ2+3) by adaptor proteins able to bind to β-APP (X11, Fe65, Dab). B. Verify the participation of tyrosine kinase activity (and in particular of Abl), in this modulation. C. Investigate the potential involvement of these channels in the cellular damage induced by the hyperexpression of β-APP adaptor proteins. D. Study the participation of K+ channels and their regulation by oxidative stress in the neuronal death triggered by various cellular models of AD-related neurodegeneration. European Community contract N° LSHM-CT-2003-503330/Apopis : Abnormal proteins in the pathogenesis of neurodegenerative disorders Study on tyrosine phosphorylation of APP and its interaction with intracellular adaptors: role in cell signaling and in the generation of amyloidogenic fragments. Ente finanziatore Durata progetto Abstract del progetto European Community 3 years (2004-2007) Within the WP 1 Generation and Turnover, our proposal is aimed to the definition of the role that posttrasductional modifications of APP, in particular its phosphorylation, may exert on its pathophysiologycal function. We will investigate both the role of phosphorylated APP/CTFs in cell signaling, and/or in the generation of amyloidogenic fragments and plaques through the interaction with intracellular adaptors, as well as the effect that site-specific tyr-phosphorylation would have on the direct amyloidogenic pathway of APP. These studies, which are focused in a completely new aspect of APP activity, would lead to the identification of early markers of the neurodegenerative process which occurs in AD, and to the definition of a potentially innovative therapeutical approach based on the findings obtained. Our proposal is aimed at: Aim 1) To study directly in human brain the tyr-phosphorylation of APP and its CTFs and their possible coupling with intracellular adaptors such as ShcA, and to correlate these data with the presence of neuropathologycal hallmarks of the disease. The comparative analysis in non-AD control, AD and/or Down’s syndrome subjects at different ages (from fetal to adult life) will allow a deeper characterization of the molecular determinants during the progression of the disease. Aim 2) To study in transgenic mice carrying APP and PS1 mutations the tyrphosphorylation of APP and CTFs, their interaction with ShcA-Grb2 adaptors and the influence that such interaction could have on plaque formation, astroglial response, neuronal death. The use of a Tg model will allow at a deeper comprehension of the possible correlation between APP phosphorylation, its cleavage, signaling activity and progressive presence of the typical hallmarks of the disease. Moreover, in this model is possible also to determine the effect that APP and PS1 mutations, which are linked with a human familial phenotype, would cause in the above mentioned parameters, and, in a second step, would allow the application of targeted therapeutical pharmacological approaches following also the indications raised from the in vitro studies (aim 3). Aim 3) To study in neuronal and/or glial cell cultures the molecular mechanisms which regulate APP phosphorylation, influence on cleavage by secretases, coupling with intracellular adaptors, and signaling activity. Using APP mutants, ShcA mutants, and APP KO cells we will define the influence that phosphorylation and interaction with ShcA would cause in signaling activity, cell proliferation and death, and in the generation of amyloidogenic fragments. Moreover, the study of the effect that proliferative and apoptotic stimuli may cause on those parameters and/or with the study of the kinases involved in the activation of APP cleavage and signaling, this would also lead to the identification of pharmacological target for a therapeutical intervention. Nome Contatti Istituto/Dipartimento MARIO MARCHI [email protected] Department of Experimental Medicine (Di.Me.S.) Section of Pharmacology and Toxicology Viale Cembrano, 4 16147 Genova ITALY +39 010 3532657 e.mail: Proposta di ricerca Effect of Age and neurodegenerative disorders on central neurotransmission: focus on neurotransmitter release from neurons and glia The effects of age and/or neurodegenerative disorders may differentially influence diverse cell types in the brain and the vulnerability of neurons may depend also on the transmitter system or the location in neural circuits. Therefore, it is of utmost importance to identify age-induced neurochemical changes in the brain in order to develop pharmacological strategies that can prevent, delay or counteract the cognitive decline. Cells may be differentially sensitive according to the function of signaling pathways that mediate cell specific processes involved in cognition (e.g. synaptic plasticity). Systemic influences of inflammation can also alter the local milieu to influence different cell populations. Our current researches are focused to assess the current status of synaptic function/dysfunction in aging and age-related neurodegenerative diseases in several regions of the brain in mammals, particularly in rodents .It is now well accepted that astrocytes integrate and process synaptic information and control synaptic transmission and plasticity being active partners in synaptic function, astrocytes are cellular elements involved in the processing, transfer and storage of information by the nervous system. Therefore our studies are aimed at investigating “ex-vivo” changes of neuronal and glial functions due to “in-vivo” aging or in the animal models of neurodegenerative disorders (i.eAlzheimer;) The pivotal concept of this approach assumes that “in-vivo” aging or the neurodegenerative disorders are able to modify the plasticity of chemical synapses which in turn could be retained, and then analyzed, in the “ex-vivo” tissue preparations. This “ex-vivo” memory has been already described to occur and studied in our laboratory to unmask neurotransmission modifications caused by “in-vivo” exposure to different stimuli.( i.e. chronic treatment with drugs, enriched environment, etc.). Our research is therefore focused to investigate whether and to what extent aging and age-related neurodegenerative diseases (i.e. Alzheimer) could modify some fundamental parameters, such as neurotransmitter release and uptake, receptor expression and functioning in plasma membranes, moreover we will investigate the effect of beta amyloid on these parameters by utilizing two “ex-vivo” functional preparations representative of neurons and astrocytes , i.e. the isolated nerve terminals (synaptosomes) and the glial subcellular particles (gliosomes). Area di interesse identificata Finanziamenti ricevuti Titolo progetto Plasticità dei recettori presinaptici nicotinici e glutammatergici che modulano la liberazione di neurotrasmettitori: studio delle variazioni delle loro risposte funzionali dopo trattamenti cronici o in seguito ad interazioni con altri sistemi recettoriali o altre condizioni sperimentali Role: Principal Investigator Ente finanziatore Durata progetto Abstract del progetto Titolo progetto MIUR-prin 24 MESI Studies on the effect of the chronic treatment with nicotine on some neurochemical presynaptic mechanisms and on the function of presynaptic receptors in the CNS. Role: Principal Investigator Ente finanziatore Durata progetto Titolo progetto COMPAGNIA DI SAN PAOLO 36 MESI Les plant aromatique, entre environment et activitè productive Role . Principal Investigator Ente finanziatore Durata progetto ALCOTRA “AROMA” 24 MESI Nome Contatti Dr. Francesca Ruberti [email protected]; INMM- CNR Istituto di Neurobiologia e Medicina Molecolare Via del Fosso di Fiorano, 64 00143 ROMA ITALY Phone: + 39 06 501 703 236 Fax: + 39 06 501 703 313 Istituto/Dipartimento Institute of Neurobiology and Molecular Medicine CNR Proposta di ricerca NEUROBIOLOGY OF ALZHEIMER’S DISEASE 1) Basic research : developing competitive in vitro and in vivo models to study Alzheimer’s Disease a) Role of microRNAs in Alzheimer’s Disease onset and progression (C. Barbato, C Cogoni, F. Ruberti) Primary neuronal cell culture and Alzheimer’s Disease (AD) mouse models, including AD11 (and transgenic mice overexpressing pathogenic variants of human APP gene, will be instrumental to investigate the function of selected miRNAs (by their overexpression and/or inhibition) on disease pathogenesis. Studies will focus on recently identified microRNAs modulating, in neuronal hippocampus cultures (Vilardo et al. 2010), APP production and on miRNAs dysregulated in human AD brain (Barbato et al 2009). Multidisciplinary approaches will be used to investigate the effect of microRNAs on molecular mechanisms underlying amyloidogenesis, neuronal degeneration, memory loss, synaptic plasticity impairment. Vilardo E, Barbato C, Ciotti MT, Cogoni C, Ruberti F. MicroRNAs regulate Alzheimer’s Amyloid Precursor Protein expression in hippocampal neurons. (2010) submitted. Barbato C, Ruberti F, Cogoni C. Searching for MIND: microRNAs in neurodegenerative diseases. J Biomed Biotechnol. 2009:871313. Barbato C, Ruberti F, Pieri M, Vilardo E, Costanzo M, Ciotti MT, Zona C, Cogoni C. MicroRNA-92 modulates K(+) Cl(-) co-transporter KCC2 expression in cerebellar granule neurons. J Neurochem. 2009 Dec 26. b) Mechanisms underlying the effect of N-terminal 26-230 tau fragment on synaptic dysfunction and neurodegeneration (N. Canu, in collaboration with V. Cestari, F. Tirone, E Mattei). The aim is is to decode the signal transduction pathway that links N-terminal 26-230 tau fragment (N-tau) expression to NMDAR activity (Canu et al 1998; Amadoro et al 2006) and their upstream and downstream effectors inducing synaptotoxicity and plasticity failure. Electrophysiological recording of NMDAR and AMPAR biochemical characterization of NMDAR complex, morphological, qualitative and quantitative analysis of synapse number, dendritic spines and behavioral analysis of conditional N-tau transgenic mice, will be performed to give an accurate and global view of the molecular events involved in N-tau modulation of neuronal plasticity. Canu N, Dus L, Barbato C, Ciotti MT, Brancolini C, Rinaldi AM, Novak M, Cattaneo A, Bradbury A, Calissano P. Tau cleavage and dephosphorylation in cerebellar granule neurons undergoing apoptosis. J Neurosci. 1998 Sep 15;18(18):7061-74. Amadoro G, Serafino AL, Barbato C, Ciotti MT, Sacco A, Calissano P, Canu N. (2004) Role of N-terminal tau domain integrity on the survival of cerebellar granule neurons. Cell Death Differ. 2004 Feb;11(2):217-30. Amadoro G, Ciotti MT, Costanzi M, Cestari V, Calissano P, Canu N. NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation.Proc Natl Acad Sci U S A. 2006 103(8):2892-7 c) Nerve Growth Factor deprivation pathways leading to Alzheimer’s Disease phenotype (C. Matrone, P. Calissano). An NGF-dependent hippocampal neuronal model in which the interruption in NGF supply quickly triggers amyloidogenesis and induces tau protein dysfunction and neuronal degeneration, mimicking essential features of in vivo AD pathology, has been recently charachterised (Matrone et al. 2008; Matrone et al 2009; Amadoro et al 2009). The main goal is to to test whether the proteins whose expression or phosphoylation is affected by NGF deprivation may represent early molecular signature and potential targets of AD. To achieve these results, studies are in progress, in collaboration with prof. Moses Chao at NYU, to characterize the phosphotyrosine-associated signalling events due to NGF deficit in primary cultures from wild type and APPk/o mice by a systematic MS/SILAC. Matrone C, Ciotti MT, Mercanti D, Marolda R, Calissano P. NGF and BDNF signaling control amyloidogenic route and Abeta production in hippocampal neurons.Proc Natl Acad Sci U S A. 2008 105:13139-44. Matrone C, Marolda R, Ciafrè S, Ciotti MT, Mercanti D, Calissano P. Tyrosine kinase nerve growth factor receptor switches from prosurvival to proapoptotic activity via Abeta-mediated phosphorylation. Proc Natl Acad Sci U S A. 2009 106: 11358-63. Amadoro G, Corsetti V, Ciotti MT, Florenzano F, Capsoni S, Amato G, Calissano P. Endogenous Abeta causes cell death via early tau hyperphosphorylation. Neurobiol Aging. 2009 Jul 21. d) Role of NH2-26-44 tau peptide in Alzheimer’s Disease (G. Amadoro P Calissano). A synthesized NH2-2644 peptide of tau, impairs the mitochondrial oxidative phosphorylation and reduces the intracellular ATP bioavailability, probably acting on adenine nucleotide translocator (ANT)- mediated ADP/ATP exchange (Atlante et al., 2008). Studies in AD human tissues are focused to evaluate if tau peptide i) is correlated with the extent of neurofibrillary degeneration and amyloid neuropathology, especially Abeta pre-fibrillar species; (ii) is linked to the synaptic changes and to the mitochondrial functional alterations. The physical interaction between ANT and NH2-derived tau fragment, in pathologically relevant conditions, will be investigated. Atlante A, Amadoro G, Bobba A, de Bari L, Corsetti V, Pappalardo G, Marra E, Calissano P, Passarella S. A peptide containing residues 26-44 of tau protein impairs mitochondrial oxidative phosphorylation acting at the level of the adenine nucleotide translocator. Biochim Biophys Acta. 2008 1777, 1289-300 e) Mitochondrial damage and genetic risk/susceptibility factors in Alzheimer’s Disease (M. Rinaldi, S. Iurescia, D. Fioretti). The aim of the research project is i) to identify and characterize early biomarkers and mechanisms of mitochondrial damage promoted by ROS/RNS; ii) to investigate the protective role of natural forms and synthetic analogues of vitamin E that can be tested in intervention studies in AD patients. Studies are in progress to evaluate post-transductional modifications (PTMs) in mitochondrial proteins, caused by ROS/RNS in in vitro model of senescence and human neuroblastoma as well as in peripheral cells from subjects with Mild Cognitive Impairment (MCI) and AD. Strong genetic risk/susceptibility factors for developing Alzheimer’s disease, such as ApoE4 that impacts on Abeta production, Abeta clearance, Abeta fibrillation and tangle formation as well as on mitochondrial functions leading to neuronal toxicity and synaptic damage, are also investigated. Iurescia S, Fioretti D, Mangialasche F, Rinaldi M.The Pathological Cross Talk Between Apolipoprotein E and Amyloid-beta Peptide in Alzheimer's Disease:Emerging Gene-Based Therapeutic Approaches. J Alzheimers Dis. 2010 In press 2) Basic research : New treatment strategies based on Neurotrophins f) Ocular NGF administration as a novel non invasive approach to protect brain-NGF target neurons that degenerate in Alzheimer’s disease (L. Aloe, P. Tirassa). Nerve growth factor and (NGF) is a soluble protein that plays a protective role on brain neurons that degenerate in age-related brain disorders, including Alzheimer’s disease (AD). We have shown that conjunctively applied NGF can protect injured brain neurons and damaged retinal ganglion neurons suggesting that topical eye NGF application might be a novel alternative for delivering NGF into the brain and for protecting brain neurons and reducing memory deficits occurring during early events of AD and glaucoma. This hypothesis is currently actively under investigation in collaboration with other research groups. Lambiase A, Pagani L, Di Fausto V, Sposato V, Coassin M, Bonini S, Aloe L. Nerve growth factor eye drop administrated on the ocular surface of rodents affects the nucleus basalis and septum: biochemical and structural evidence. Brain Res. 2007;1127:45-51. Di Fausto V, Fiore M, Tirassa P, Lambiase A, Aloe L. Eye drop NGF administration promotes the recovery of chemically injured cholinergic neurons of adult mouse forebrain. Eye drop NGF administration promotes the recovery of chemically injured cholinergic neurons of adult mouse forebrain. Eur J Neurosci. 2007;26:2473-80. Lambiase A, Aloe L, Centofanti M, Parisi V, Mantelli F, Colafrancesco V, Manni GL, Bucci MG, Bonini S, Levi-Montalcini R. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: Implications for glaucoma. Proc Natl Acad Sci U S A, 2009; 109: 1346913474,. g) Electro-acupuncture and polyphenols as novel approaches to reduce neurodegeneration ( L Manni, M Fiore). Preliminary data indicate that diabetes induced in adult rats by injection with streptozotocin causes a decrease of brain NGF and a concomitant increase of tau phosphorylation, that were all counteracted by low-frequency electro-acupuncture (EA). Using molecular and behavioural approaches the research project will be aimed at: i) Investigate the possible link between diabetes, brain NGF presence/activity and the development of AD-associated pathological features; ii) Study the effect of EA on the above mentioned diabetes-associated brain dysfunction. Alcoholism is related to neurodegeneration and diabetes. The use of polyphenols extracted by olive oil, olive leaves and olive seeds or by grapes (Fiore et al 2009) to prevent or limit mouse brain neurodegeneration due to aging or ethanol consumption will be evaluated. Manni L, Aloe L, Fiore M. Changes in cognition induced by social isolation in the mouse are restored by electro-acupuncture. Physiol Behav. 2009;98(5):537-42 Fiore M, Laviola G, Aloe L, di Fausto V, Mancinelli R, Ceccanti M. Early exposure to ethanol but not red wine at the same alcohol concentration induces behavioral and brain neurotrophin alterations in young and adult mice. Neurotoxicology. 2009;30(1):59-71. h) Human neural stem cells lentivirally transduced with human BDNF (C. Cenciarelli, P. Casalbore). A strong body of evidences show that BDNF result critically involved in Alzheimer’s and Huntington’s diseases. Recently, it has been shown that BDNF knockdown within NSC abolishes the cognitive benefits of NSC delivery. The aim is to use propagating human neural stem cells (hNSC) lentivirally transduced with human BDNF as cellular therapy for replacing degenerating neurons in disease, trauma and toxic insults. Cenciarelli C, Budoni M, Mercanti D, Fernandez E, Pallini R, Aloe L, Cimino V, Maira G, Casalbore P. In vitro analysis of mouse neural stem cells genetically modified to stably express human NGF by a novel multigenic viral expression system. Neurol Res. 2006, 28:505-12. Casalbore P, Barone I, Felsani A, D’Agnano I, Michetti F, Maira G and Cenciarelli C. Neural stem cells modified to express BDNF antagonize trimethyltin-induced neurotoxicity through PI3K/Akt and MAP kinase pathways . Accepted on J. Cell. Physiol. 2010. OTHER NEURODEGENERATIVE DISEASES Basic research, biobanking (DNA), sharing database, exchanging and pooling data i) Biobank compund by over 2000 DNAs from HD and ataxic patients and their families. Molecular basis of episodic ataxia (L. Veneziano). Research activity has been mostly devoted to CACNA1A gene, coding for the Alpha1A subunit of P/Q type Calcium channel expressed in brain, from its physical map to the refinement of the gene structure to the screening for mutations in Episodic Ataxia (EA) to the association genotype/phenotype (Veneziano et al 2009; Mantuano et al 2010). This research allowed the construction of a DNA biobank from about 100 patients affected by EAs. The project is focused on the use Comparative Genomic Hybridization (CGH) arrays, which can be used for a screening of CACNA1A gene rearrangement in a sample of 40 EA patients in which CACNA1A gene point mutations were excluded. The fulfilment of the above aim will improve the rate of molecularly diagnosed EA2 patients to be treated with known therapy or to be included in new therapeutic trials. Veneziano L, Guida S, Mantuano E, Bernard P, Tarantino P, Boccone L, Hisama FM, Carrera P, Jodice C, Frontali M. Newly characterised 5' and 3' regions of CACNA1A gene harbour mutations associated with Familial Hemiplegic Migraine and Episodic Ataxia. J Neurol Sci. 2009, 276(1-2):31-7 Mantuano E, Romano S, Veneziano L, Gellera C, Castellotti B, Caimi S, Testa D, Estienne M, Zorzi G, Bugiani M, Rajabally YA, Barcina MJ, Servidei S, Panico A, Frontali M, Mariotti C.Identification of novel and recurrent CACNA1A gene mutations in fifteen patients with episodic ataxia type 2. J Neurol Sci. 2010 Feb 1. l) Role of the Thyroid Transcription Factor 1 (TTF-1) in neurodegenerative diseases (D. Civitareale). The Thyroid Transcription Factor 1 (TTF-1) expression persists in some post-mitotic basal forebrain neurons, and mutations of TiTF-1 have been associated with the benign hereditary chorea. Our transcriptome analysis confirms that TTF-1 is involved in neuron morphogenesis, differentiation and GnRH secretion. We predict that TTF-1 could be a modifier gene involved in the pathogenesis of the neurodegenerative diseases. Therefore, we propose to study TTF-1 down-regulation via RNAi and microarray analysis in ST14A, select a mouse strain able to develop the neurodegenerative disease in absence of Titf-1, and characterize the genotype/phenotype association of TiTF-1 in a cohort of patient with neurodegenerative diseases. Provenzano C, Veneziano L, Appleton R, Frontali M, Civitareale D. Functional characterization of a novel mutation in TITF-1 in a patient with benign hereditary chorea. J Neurol Sci. 2008 264(1-2):56-62. Provenzano C, Pascucci B, Lupari E and Civitareale D. Large scale analysis of transcription factor TTF1/NKX2.1 target genes in GnRH secreting cell line GT1-7. Molecular and Cellular Endocrinology. In press. Area di interesse identificata Basic research : developing competitive in vitro and in vivo models to study Alzheimer’s Disease; new treatment strategies based on Neurotrophins. Biobanking (DNA), sharing database, exchanging and pooling data Finanziamenti ricevuti Role of microRNAs in Alzheimer’s Disease onset and progression (C. Barbato, C Cogoni, F. Ruberti,). Ongoing Research Support -3886 SD/sd 2008.2404 Programma Neuroscienze “Compagnia di San Paolo” 9/01/09-9/01/11: ‘MIND: MicroRNA In Neurodegenerative Diseases’ (to C. Barbato) -REGIONE LAZIO grant 2009-2011: ‘Studio delle basi molecolari della neurodegenerazione nella malattia di Alzheimer’- (to C.Cogoni). Completed Research Support DG.RSTL.059.012 –CNR grant-01/2008-12/2009. “Study of APP regulation by microRNAs and implication in Alzheimer’s Disease” (to F. Ruberti) Finanziamenti ricevuti Mechanisms underlying the effect of N-terminal 26-230 tau fragment on synaptic dysfunction and neurodegeration (N. Canu). Ongoing Research Support Dipartimento di Neuroscience, University of Tor Vergata Completed Research Support MIUR-PRIN 2006-2008: “Molecular and behavioural analysis of tau neurotoxic effect in cellular and animal models” -Ministero della Sanità- Ex Articolo 56 Neurodegenerativo 533F/B/1 2005-2009 “Identificazione di marcatori biologici precoci per la demenza di Alzheimer: genomica e proteomica nella neurodegenerazione” Finanziamenti ricevuti Nerve Growth Factor deprivation pathways leading to Alzheimer’s Disease phenotype (C. Matrone, P. Calissano). Ongoing Research Support MIUR-FIRB Italy / 2009-2011 “Italian Human Proteome Net” MIUR-PRIN 2009/2011 “Ruolo di Zn e Cu nella cascata amiloidogenica provocata da deprivazione di NGF” Finanziamenti ricevuti Role of NH2-26-44 tau peptide in Alzheimer Disease (G. Amadoro P. Calissano) Ongoing Research Support MIUR-FIRB 24-09-08 al 24-09-11: “Italian Human Proteome Net” MIUR 18-07-2007 to 18-07-2010: “Neurotrophic and neuroprotective drugs suitable for therapeutic applications in neuroscience: AD,HD,SLA” Finanziamenti ricevuti Mitochondrial damage and genetic risk/susceptibility factors in Alzheimer’s Disease (M. Rinaldi, S. Iurescia, D. Fioretti). Ongoing Research Support MIURPRIN 2007-2009: “Oxidative and nitrosative damage of mitochondrial proteins in models of cell senescence and in subjects with Alzheimer's disease: a pre-clinical study on natural forms and new synthetic analogues of vitamin E. Finanziamenti ricevuti, Ocular NGF administration as a novel non invasive approach to protect brainNGF target neurons that degenerate in Alzheimer’s disease (P Tirassa, L Aloe) Ongoing Research Support Progetto PRIM- 2007-2010: “Sviluppo del NGF come farmaco in collirio: Studi di farmacocinetica e di efficacia terapeutica”. (to L. Aloe) Progetto Strategico Q77, ISS- 2007-2010: “Indicazioni di indici neurobiologici di vulnerabilità a rischio di sviluppo di sindromi depressive” (to L. Aloe). Fondazione G.B. Bietti 2007-2009: “Potenziale ruolo terapeutico del NGF nel glaucoma: Studi in un modello animale e nell’uomo” (to L. Aloe) CNR funding (PORRSTL03) 2006-2008 (to P. Tirassa) Finanziamenti ricevuti, Electro-acupuncture and polyphenols as novel approaches to reduce neurodegeneration (L Manni, M Fiore) Ongoing Research Support Centro Alcologico Regione Lazio (to M. Fiore) Finanziamenti ricevuti, Human neural stem cells (hNSC) lentivirally transduced with human BDNF (C. Cenciarelli, P. Casalbore). Ongoing Research Support Funds from ATENA Onlus (2009-2010): “Transplantation of BDNF secreting-NSC in the hippocampus of adults rats treated with the neurotoxicant thrimetyltin.” Completed Research Support Grants from The Nando Peretti Foundation and ATENA Onlus (2006-2008) “Repair of lesions of Central Nervous System through cell replacement therapy by using of NSC induced towards to neuronal phenotypes.” Finanziamenti ricevuti, Biobank compund by over 2000 DNAs from HD and ataxic patients and their families and Molecular basis of episodic ataxia (L. Veneziano). Ongoing Research Support National Ataxia Foundation 2009: “Looking for gross rearrangements of CACNA1A gene in Episodic Ataxia type 2 (EA2) patients by Comparative Genomic Hybridization arrays. Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata MARIAROSARIA MILOSO [email protected] tel 0264488123 Dipartimento di Neuroscienze e Tecnologie Biomediche EVALUATION OF MESENCHYMAL STEM CELLS (MSCs) EFFECT IN ALZHEIMER’S DISEASE RAT MODELS: in vitro and in vivo STUDIES Alzheimer’s disease (AD) is the most common cause of progressive decline of cognitive function in aged humans. At present, there are no effective therapies to treat this pathology. Recently it has been hypothesized an innovative therapeutic approach based on the use of stem cells. Different sources of stem cells are being explored for potential use in repairing neurodegenerative disorders. Among them, Mesenchymal Stem Cells (MSCs) represent a good cellular source for transplantation therapy. MSCs can be easily isolated and expanded, autologously used, present a low risk of tumorigenicity and are ipoimmunogenic. Aim of our project is to study the possible effect of human and rat MSCs in an in vitro and in vivo model of Alzheimer Disease (AD). In vitro model of AD is represented by rat cortical and hippocampal neuron primary cultures exposed to 2,5 µM Aβ (1-42) fibrils. To evaluate the capacity of MSCs to improve neuronal viability and AD neuropathological changes, we will perform: a) direct and indirect cocultures of MSCs with Aβ-treated neurons; b) culture of Aβ-treated neurons in the presence of conditioned medium from MSCs. Neuronal survival will be evaluated by MTT assay, neuronal morphology and functionality will be assessed by morphological and immunofluorescence studies and electron microscopy. In vivo model of AD is represented by a transgenic rat model that develop spontaneously the AD pathology. The MSCs are prelabeled with Qdot nanocristal and than inoculated into the rat tail vein. After the MSCs injection cognitive functions will be assessed by the Morris water maze test (Morris 1984). After the behavioral test, the rats will be sacrificed and the brain will be obtained and further fixed. Sections of cortex and hippocampus will be cut in a freezing microtome and AD neuropathological changes will be evaluate by morphological and immunofluorescence studies and electron microscopy. MSC localization and differentiation will be evaluated by immunofluorescence experiments. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Nome Patrizia Hrelia, Full Professor in Toxicology, Faculty of Pharmacy Contatti Via Irnerio 48, 40126 Bologna Tel.: +39 051 2091799 e-mail: [email protected] Istituto/Dipartimento Department of Pharmacology , Alma Mater Studiorum - University of Bologna Nome Silvana Hrelia, Associate Professor in Biochemistry, Faculty of Pharmacy Contatti Via Irnerio 48, 40126 Bologna Tel.: +39 051 2091233 e-mail: [email protected] Istituto/Dipartimento Department of Biochemistry, Alma Mater Studiorum - University of Bologna Proposta di ricerca Our project is aimed at the identification of cellular and molecular targets affected by phytochemicals, to delineate pharmacological and nutritional strategies for the reduction or the prevention of neurodegeneration and neuroinflammation associated to ageing and neurodegenerative diseases. The project is innovative since it represents a joint action with the attempt: 1) to adequately understand the cellular and molecular mechanisms commanding cell death in neurodegeneration and behind neuroprotective effects of phytochemicals; 2) to identify phytochemicals having diverse pharmacological activities, such as neuroprotection and antiinflammation; 3) to confirm if these phytochemicals have a specific pharmacological effectiveness in the prevention/counteraction of one of the most important aging-related neurodegeneration and neuroinflammation hallmarks, oxidative stress, as a primary prevention mechanism; 4) to understand if these phytochemicals can act as biomodulators in neuronal cells through modulation of survival pathways, suggesting a potential therapeutic use in regenerating injured neuronal cells; 5) to establish the therapeutic window that allows an effectively impact of phytochemicals on specific targets; In longer terms, our studies will indicate whether pharmacological and nutritional strategies with phytochemicals should be effective and safe in counteracting cellular and molecular damage associated to human neurodegeneration and neuroinflammation. Area di interesse identificata New treatment strategies (Neurodegeneration and Neuroprotection) Finanziamenti ricevuti Titolo progetto Ente finanziatore Drug Research and Development (CHEM-PROFARMA-NET). Synthesis, biological and pharmacological characterization of organic molecules, bio-oligomers and natural compounds endowed with antidegenerative (neuro or cardiovascular) antiviral, and antiinfective activity. MIUR FIRB – Piattaforme/Reti (total funds euro9.768.680, euro1.274.097 assigned to P.Hrelia as node coordinator) Durata progetto 2007-2011 Abstract del progetto The vision of the project is that of a strong collaborative efforts aimed at the identification of new cellular and molecular targets affected by synthetic or natural compounds to delineate pharmacological and nutritional strategies for the prevention of human diseases such as neurodegeneration, infectious disease cancer, immune and metabolic disorders. The activities proposed will be completely performed in the Network Operative Units. This project is aimed at identifying new drugs that may be useful in preventing and/or alleviating the consequences of specific diseases, but it will focus on new therapeutic approaches for such disorders. Titolo progetto New Bioactive Molecules from Natural Sources in Neuroprotection: Opportunities In Neurodegenerative Diseases Ente finanziatore MIUR COFIN 2007 (total funds euro 63.750 assigned to G. Cantelli Forti as Scientific Coordinator, and S. Hrelia as Responsible of Research Unit) Durata progetto 2008-2010 Abstract del progetto The pathogenesis of ageing related neurodegenerative diseases, such as Alzheimer's (AD) or Parkinson's (PD), is multifactorial with the existence of a "domino" cascade of neurotoxic events, which can be initiated at any point in the cascade. Many lines of evidence suggest that oxidative stress, resulting in ROS/RNS generation, mitochondrial dysfunction and activation of death/survival pathways play a pivotal role in the age-associated neuronal loss in neurodegenerative diseases. Thus, promising future treatment of neurodegenerative diseases depends on availability of effective brain permeable, antioxidant neuroprotective drugs that would prevent the progression of neurodegeneration. An innovative approach to preventive/therapeutic intervention in oxidative stress based neurodegeneration may be the manipulation of endogenous cellular defence mechanisms by chemical inducers, such as isothiocyanates from natural sources, among which sulforaphane (SF). Molecular and cellular mechanisms behind SF neuroprotection will be initially studied in vitro, through an experimental approach which let us to evaluate neuroprotective and/or neurorescue effects by SF. Neuronal damage parameters at the cellular level will comprise the induction of neuronal death in terms of apoptosis/necrosis and the activation of cellular death/survival biochemical pathways. The scientifically important expectation of this project, by providing data on the cellular and molecular mechanisms commanding cell death in neurodegeneration and behind neuroprotection by SF, is to provide a clear rationale for delineating innovative pharmacological strategies to counteract and rescue neurodegeneration. Fondazione IRCCS Istituto Neurologico “Carlo Besta” Division of Neuropathology – Neurology 5 Director: Fabrizio Tagliavini The research group The Division comprises (i) a Clinical Unit (Dementia Center) devoted to the diagnosis and treatment of patients with degenerative dementias (over 2000 out-patients and 150 in-patients per year) and (ii) a Laboratory Unit dedicated to the analysis of genes and biomarkers associated with degenerative dementias, post-mortem characterization of the disease process and disease-specific protein, and experimental studies on disease pathogenesis and the development of therapeutic strategies. The course of action of this activity can be expressed as a “Bed to Bench to Bed” cycle, with the ultimate goal to identify disease-modifying drugs in preclinical settings and apply them to patients. In this regard, the Clinical Unit is currently coordinating a multicentre phase II clinical trial supported by AIFA, to test the effectiveness of an anti-amyloidogenic molecule identified in experimental models. This comprehensive approach has also the great advantage to collect biological samples (plasma, DNA, CSF and brain tissue) from fully characterized patients. In this regard the Laboratory Unit is part of the Network of Exellence “BrainNet” Europe devoted to biobanking, harmonization of assessment tools and standardization of diagnostic criteria of neurodegenerative diseases. The permanent staff of the Division is composed by 8 MD and two technicians. In addition, the staff comprises 13 post-doctoral fellows (7 PhD, 3 MDV, 1 MD, 1 psychologist), 2 PhD students and 1 technician who are committed to the research activities on degenerative dementias with different expertise and roles. On the overall, the team has large experience in a variety of techniques spanning from neuropathology (including immunohistochemistry, morphometry, electron microscopy and atomic force microscopy), to molecular genetics, biochemistry (purification and characterization of disease-specific proteins), cellular and molecular biology, and animal models. Research lines on Alzheimer’s disease A. Clinical research • Early diagnosis of Alzheimer’s disease (AD) We will characterize the phenotypic expression of AD and other degenerative dementias through multiplex analysis of a number of variables including neuropsychological and behavioural profile, neuroimaging, neurophysiological changes with special attention to sleep abnormalities, genetics, and plasma and CSF biomarkers. Furthermore, we will carry out longitudinal studies of presymptomatic carriers of gene mutations to detect early markers of conversion to the disease state. • Clinical trials We will carry out phase II and phase III clinical trials to assess the effectiveness of potentially disease-modifying drugs, including molecules targeted to Aβ and protein tau. • Identification of disease-specific biomarkers We are currently working on a peripheral marker of AD having high specificity and sensitivity, based on cyclic amplification of misfolded Aβ and Aβ oligomers from biological fluids, similar to the PMCA technique successfully developed by Claudio Soto for prion diseases. The same approach will be used to amplify and detect misfolded tau. • Biobanking and neuropathological and molecular characterization of patients We will collect systematically plasma, DNA and CSF from patients with different types of dementia and non-demented individuals, and perform autopsies for neuropathological characterization and biochemical and molecular studies. B. Preclinical research • Recessive A673V APP mutation: molecular mechanisms and development of a new therapeutic strategy for sporadic AD We have recently identified an APP mutation (A673V) that causes early-onset AD only in the homozygous state while the heterozygous carriers are not affected. This mutation strongly boosts the production and amyloidogenic properties of Aβ. However, the interaction of A673V-mutated and wild-type peptides inhibits amyloidogenesis and Aβ-mediated neurotoxicity (Di Fede et al., Science 2009, 323:1473-7). These findings are consistent with the observation that the A673V heterozygous carriers do not develop disease and offer grounds for a novel therapeutic strategy based on modified Aβ peptides. A research priority of our lab is to unravel the molecular mechanisms of the opposite effects of the A673V APP mutation in homo- or heterozygous state on amyloidogenesis and to develop a lead compound for AD therapy based on A673V-modified Aβ peptides or peptido-mimetic molecules. To accomplish these objectives we have generated a panel of transfected cells and transgenic C. elegans expressing human APP or Aβ with the A673V mutation, respectively, and transgenic mouse lines expressing A673V-mutated APP in the homozygous or heterozygous state. Furthermore, we have identified a prototypic lead compound corresponding to a six-mer Aβ peptide with the A673V substitution and are currently working on brain delivery systems. • Pathways leading to tau pathology A key event in AD pathogenesis is the hyperphosphorylation, misfolding and aggregation of the microtubule-associated protein tau leading to formation of neurofibrillary tangles, disruption of the neuronal cytoskeleton and neurodegeneration. Following the “Aβ cascade hypothesis” this event is triggered by aggregated Aβ species, particularly oligomeric assembly intermediates. However, the pathways linking Aβ and tau pathology are unknown. This is largely due to lack of animal models able to reproduce these two central aspects of AD. We have developed a mouse model of prion disease that shows a secondary tauopathy following deposition of PrP amyloid (Giaccone et al., Neurobiol. Aging 2008, 29:1864-73). This model will be used to investigate the molecular basis of tau pathology induced by deposition of an amyloid protein. • Role of nuclear tau in neurodegeneration in fronto-temporal dementia Tau is the major microtubule-associated protein of neurons and its primary role is to promote assembly and stabilization of microtubules required for morphogenesis and axonal transport. We have recently found that tau plays an important role also in chromosome stability, and mutations in the tau gene cause chromosome aberrations in peripheral cells in addition to formation of neurofibrillary tangles in neurons and glial cells. Our objective is to elucidate the molecular mechanisms underlying the genomic instability due to tau mutations using cellular models, and to investigate the contribution of the aneuploidy caused by mutated tau to neurodegeneration using a transgenic mouse model of tauopathy. Relevant ongoing research support • Cariplo Foundation (01/01/07-30/06/10) Title: Genoproteomics of Age Related Disorders (GuARD). PI: Roberto Sitia Role: Project leader • Italian Ministry of Health (01/01/09-30/06/11) Title: Alzheimer's disease: development and validation of a multi-factorial protocol for the diagnosis and follow-up of disease in the prodromal and incipient phase. PI: Fabrizio Tagliavini • European Union/Italian Ministry of Health (01/02/09-31/01/12) Title: Molecular mechanisms underlying synaptic dysfunction in prototypic neurodegenerative diseases related to protein misfolding (nEUROsyn). Role: PI • Italian Agency of Drug (01/09/06-31/08/10) Title: A randomized, double-blind pilot study versus placebo for the evaluation of the efficacy of doxycycline administered by oral route in patients affected by Creutzfeld-Jakob disease. Role: PI Nome Roberta Ricciarelli, Ph.D. Contatti E-mail: [email protected] Tel.: +39-010-3538838; Fax: +39-010-3538836 Istituto/Dipartimento Section of General Pathology Department of Experimental Medicine (DiMeS) University of Genoa, Italy Proposta di ricerca Dr. Roberta Ricciarelli’s ongoing research on Alzheimer’s disease: • Cholesterol and amyloid-beta: is there a relation? • Study of the nonmuscle myosin II involvement in the processing of the amyloid precursor protein (APP) • Specific type 4 phosphodiesterase inhibitors and their impact on the production of amyloid-beta Area di interesse identificata -Basic research, -Multidisciplinary projects, -New treatment strategies. Finanziamenti ricevuti Titolo progetto Molecular mechanisms of N-truncated amyloid beta peptides generation Ente finanziatore Durata progetto Abstract del progetto MIUR (Ministry of Education, University and Research) Two years (2004-2005) The amyloid precursor protein (APP) is an integral membrane protein processed by several different proteases called secretases. β-Secretase is responsible of the N-terminal cleavage of amyloid- (Aβ). The gene for βsecretase (also referred to as BACE) is located on chromosome 11, but no AD-related mutation in this gene has been identified so far. BACE mRNA is highly expressed in the brain and is also found in a variety of human tissues, consistent with the fact that Aβ is normally produced by many cell types. Recently, three new alternatively spliced transcripts of human BACE have been described, but the functional importance of the different BACE isoforms, and their implication in the pathogenesis of AD, has not yet been investigated. Our preliminary sequence analysis indicates that, as a cause of the alternative splicing, the DTG sequence, which is conserved at the active site of the enzyme, may shift to a different position within the catalytic site, affecting its substrate specificity. Since BACE is involved in the generation of the N-terminus of Aβ species, the existence of four BACE isoenzymes prompted us to investigate the possible correlation between the expression of selective BACE isoforms and the formation of neurotoxic N-truncated Aβ peptides. This hypothesis is in line with the reported absence of AD-causing mutation in BACE gene. Titolo progetto In vitro effect of PPAR-γ2 Pro12Ala polymorphism on the deposition of Alzheimer's amyloid-β peptides Ente finanziatore Durata progetto Abstract del progetto Fondazione CARIGE One year (2007) Mounting evidence suggests that peroxisome proliferator-activated receptor-γ (PPAR-γ) is involved in the modulation of pathogenic events related to Alzheimer's disease (AD). Such events would include the cerebral deposition of amyloid-β (Aβ) and the consequent local inflammatory response. PPAR-γ has been shown to act on both fronts, reducing either the secretion of Aβ or the expression of pro-inflammatory cytokines. Recently, the relatively common Pro12Ala polymorphism in exon 2 of PPAR-γ has been associated with higher risk for late onset AD. The aim of the present project is to assess the impact of PPAR-γ and its genetic variant on the secretion of Aβ in neuronal cultured cells. Titolo progetto Study of physiological function of the amyloid precursor protein Ente finanziatore Durata progetto Abstract del progetto University of Genoa One year (2008) The Alzheimer’s disease (AD) brain pathology is characterized by extracellular deposits of amyloid- (A) peptides and intraneuronal fibrillar structures. These pathological features may be functionally linked, but the mechanism by which A accumulation relates to neuronal degeneration is still poorly understood. A peptides are fragments cleaved from the amyloid precursor protein (APP), a transmembrane protein ubiquitously expressed in the nervous system. Although the proteolytic processing of APP has been implicated in AD, the physiological function of APP and the subcellular site of APP cleavages remain unknown. The overall structure of the protein and its fast anterograde transport along the axon support the idea that APP functions as a vesicular receptor for cytoskeletal motor proteins. In the current study, we will test the hypothesis that myosin II, important contributor to the cytoskeleton of neuronal cells, may influence the trafficking and/or the processing of APP. Our preliminary results demonstrated that downregulation of myosin II-B, the major myosin isoform in neurons, is able to increase A deposition, concomitantly altering the subcellular localization of APP. These new insights might be important for the understanding of the function of APP and provide a novel conceptual framework in which to analyze its pathological role. Nome Contatti Istituto/Dipartimento Patrizia Guarneri [email protected] Istituto di Biomedicina e Immunologia Molecolare – Dip- di Medicina CNR- Palermo Proposta di ricerca Rare neurodegenerative diseases: basic research Area di interesse identificata Rare diseases Finanziamenti ricevuti Titolo progetto Pathogenesis of the CLN8 form of the late-infantile neuronal ceroid lipofuscinosis Ente finanziatore 1) MIUR “PRIN” Programmi di Ricerca Scientifica di Rilevante Interesse Nazionale (DM n. 1407 del 4 dicembre 2008). “IDENTIFICATION OF BIOLOGICAL, GENETIC AND CLINICAL MODIFIERS OF THE RATE OF PROGRESSION AND SURVIVAL IN AMYOTROPHIC LATERAL SCLEROSIS ”, prot. 20082XH2Z4-001 Durata progetto Abstract del progetto 2) FIRB-MIUR, n. RBAU01E3SL, “RETINAL DEGENERATION IN MND MOUSE, AN ANIMAL MODEL OF NEURONAL CEROID LIPOFUSCINOSES OR BATTEN’S DISEASE. (dec. n. 808/Ric) 1) Two years 2) Three years The neuronal ceroid lipofuscinoses (NCLs) are a group of neurodegenerative, lysosomal diseases with onset usually in childhood, being characterized by progressive visual loss, dementia, motor decline, therapy resistant epilepsy and early death. Our team has lately focused on the pathogenic mechanisms of the CLN8 variant form of the lateinfantile NCLs (vLNCL), which is present in Finland, Turkey and Italy. The CLN8 gene encodes a transmembrane protein which is located mainly in the ER and partially in the ER-Golgi intermediate compartment of both neuronal and non-neuronal cells, and it is supposed to have a role in the ceramide synthesis. However, its exact function remains undiscovered. We have developed a protein-protein interaction study throughout the screening of a human brain cDNA library with split-ubiquitin membrane two-hybrid system in yeast, which is the only technology developed thus far that can work effectively as a in vivo screening system to find interactors of ER membrane proteins. This approach along with coimmuno-precipitation and -localization assays of transfected cells has envisaged for the first time four major protein interactors with the CLN8 protein. Through in vitro and in vivo studies, we are looking at the definition of the functional role of these interactions and particularly of the CLN8 interaction with two of the characterized proteins, which are known to have functions in synthesis and transport of lipids and are suggested to be involved in the amyothrophic lateral sclerosis. In an attempt to understand the mechanisms leading to neuronal cell death in the disease, we have been also using a spontaneous animal model of the CLN8/vNCL, the mnd mouse. We are currently investigating whether and how dysfunctions of ER/UPR system and autophagolysosomal pathway cooperate in the neurodegeneration of the retina and brain structures of the mnd model. Nome Dario Finazzi Contatti [email protected] phone and fax ++39 030 302356 Dip. Materno Infantile e Tecnologie Biomediche Università degli studi di Brescia Istituto/Dipartimento Proposta di ricerca Area di interesse identificata 1) Basic research 2) Genetic susceptibility to Alzheimer’s disease and genome wide association studies 3) Biobanking Finanziamenti ricevuti Titolo progetto 1) IRON METABOLISM IN NEURAL CELLS AND IN NEURODEGENERATIVE DISORDERS Ente finanziatore 2) ANALYSIS OF CANDIDATE GENES OF SUSCEPTIBILITY TO AD. University of Brescia Fondazione RSA Agostoni Lissone Durata progetto 3 years Abstract del progetto Our group is interested in the study of iron metabolism in the brain and its possible role in neurodegeneration. The accumulation of iron is a phenomenon often detected in the brain from patients affected by Alzheimer’s disease and by other neurodegenerative processes. Even though such accumulation is not the primary cause of neural death, it is surely involved in the progression and deterioration of the neurodegenerative process. The excess of iron can be directly linked to an increase in the production of free radicals, leading to oxidative damage to proteins and membranes, signs commonly documented in tissues with neurodegeneration. We aim to a better comprehension of such processes by developing two different and complementary lines of research: 1) Analysis of the biological mechanisms involved in the maintenance of iron homeostasis in the brain, in normal and pathological conditions; 2) Analysis of genes linked to iron metabolism as possible modifiers of the individual risk to develop AD. We intend to perform a thorough analysis of the regulation of iron metabolism in neural cells and to assess the way its malfunction may lead to iron accumulation. The experiments will be initially carried out in SH5YSY cells (either differentiated or not) and in primary hippocampal neurons with the goal to characterize the role played by the hepcidinferroportin axis in iron homeostasis of neural cells. We will determine the mechanism regulating ferroportin expression and function and describe the molecular machinery controlling hepcidin production in neuronal cells and the role of the peptide in neuronal iron trafficking. The analysis will be then extended to wild type mice and murine model of AD (APP/PS1dE9), and eventually to autopsy sections of human brain from patients and controls. In particular we will investigate the relationship between brain inflammation and the regulation of the hepcidinferroportin axis. We have preliminary evidence that BMP6 and Interleukins intervene in the transcriptional regulation of hepcidin and could be of relevance in the progressive accumulation of iron in brain. The study take advantage of a significant collaboration with Prof Paolo Arosio, one of the major expert in the study of cellular iron metabolism. The genetic part of the project will investigate the possible association between polymorphisms in genes involved in iron homeostasis (hepcidin, ferroportin, DMT1, ceruloplasmin) and the individual risk to develop AD. Promoter regions, exons and exon-intron boundary regions will be scanned for the presence of sequence variations by DHPLC and real time genotyping. The study is granted by the availability of a large set of genomic DNA samples from patients and controls and by the collaboration with research groups and clinicians with expertise in AD and genetic studies (prof. Scarpini, Dott. Galimberti, Dott. Dominici). In 2005 we set up a bank of different biological samples coming from AD patients and sex and age matched controls. The biobanking activity is continuing thanks to the collaboration with the Fondazione RSA Agostoni in Lissone and Dr Roberto Dominici. Our valid collaboration with the group of Prof Scarpini at Ospedale Maggiore in Milan allows us the access to a large and well characterized set of genomic DNA samples, as a second and different population for the confirmation of data obtained in the first set of patients and controls. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Carlo Sala Carlo Sala +390250317096 [email protected] CNR Istituto di Neuroscienze Area di interesse identificata Neurotoxic stimuli, synaptic dysfunction Finanziamenti ricevuti Titolo progetto The role of intraneuronal amyloid β(Aβ) peptide on the pathogenesis of the Alzheimer disease: functional and proteomic analysis. Ente finanziatore Fondazione CARIPLO Durata progetto 2 anni Abstract del progetto La malattia di Alzheimer rappresenta la più comune forma di demenza per la quale non esiste ancora nessun intervento terapeutico in grado di prevenire, curare o ritardare in modo sostanzaile il suo sviluppo. Recenti studi hanno dimostaro che l’accumulo intracellulare di Abeta correla con la prima comparsa di deficit cognitivi. Questo progetto si pone l’obbiettivo di identificare come Abeta intrecelluare interfereisce con la funzionalità delle sianpsi neuronali e, con tecniche proteomiche avanzate, di individuare le proteine che interagiasono con Abeta. Infine ci proponiamo di studiare i meccanismi molecolari alla base della formazione di Abeta purificando e caratterizzando la composizione proteica delle vescicole di trasporto per APP. I risultati che ci proponiamo di ottenre con questo porgetto potranno contribuire alla conoscenza dei meccanismi patogenetici dell’AD e potrebbero consentire lo sviluppo di nuovi metodi diagnostici e la eventuale messa a punto di protocolli terapeutici Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Cristina Limatola +39 06 49690243 +39 347 2924450 [email protected] Dipartimento di Fisiologia e Farmacologia, Università Sapienza Roma Tra le altre linee di ricerca di interesse del laboratorio, siamo da qualche anno coinvolti in studi di ricerca sulla sclerosi laterale amiotrofica (SLA). In particolare, in collaborazione con un gruppo di neurologi (Sabatelli, Neri) e genetisti (Zollino) dell’Università Cattolica di Roma, abbiamo descritto, in pazienti affetti da SLA, delle mutazioni a carico di diverse sub unità del recettore nicotinico neuronale, nel loop regolatorio citoplasmatico (Sabatelli et al., 2009). Queste mutazioni, in buona misura a carico di potenziali siti di fosforilazione da parte di protein chinasi, determinano delle alterazione funzionali specifiche di alcune sub unità per recettori-canale per l’acetilcolina, con una generale “gain of function” recettoriale. L’aumentata responsività alla stimolazione con acetilcolina a livello dei terminali glutamatergici potrebbe determinare una liberazione eccessiva di glutammato eccito tossico, mentre la potenziata attività dei recettori nicotinici potrebbe aumentare l’ingresso di calcio intraneuronale a livelli tossici, contribuendo all’inizio o alla progressione della malattia. Stiamo attualmente continuando questi studi molecolari, analizzando gli effetti dell’adenosina e dell’ATP, i cui livelli sono alterati nella risposta infiammatoria cronica caratteristica della SLA, sull’attività dei recettori nicotinici neuronali mutati individuati rispetto alla controparte normale. Altro punto di interesse riguarda il coinvolgimento delle cellule gliali, astrociti e microglia, nello sviluppo e nella progressione di questa patologia. Abbiamo sviluppato a tal proposito dei sistemi di co-coltura motoneuroni-cellule gliali e di fettine ottenute dal tronco cerebrale (regione del nucleo ipoglosso) per studiare il contributo della componente gliale nella alterazione dell’attività elettrica ritmica, nelle proprietà dei recettori per neurotrasmettitori, sensibilità all’eccitotossicità da glutammato, e ci proponiamo di applicare questi studi a modelli cellulari che overesprimano forme mutate di proteine associate alla SLA, come FUS, TDP-43 o SOD (in collaborazione con un gruppo di biologi molecolari della nostra università, Prof. Bozzoni). Studio funzionale di mutazioni che aumentano la suscettibilità alla Sclerosi Laterale Amiotrofica su calciatori amatoriali o professionisti. Fondazione Milan 2010-2011 La sclerosi laterale amiotrofica (SLA) è una malattia devastante che distrugge in maniera selettiva una particolare categoria di cellule nervose: i motoneuroni. Questi sono i neuroni che controllano la contrazione dei muscoli scheletrici, per cui la loro morte provoca una paralisi progressiva dei soggetti affetti da SLA e, nel giro di 2-6 anni dall’insorgenza della malattia, la loro morte (solitamente per arresto respiratorio). Solo il 5% dei casi di SLA ha chiaramente un’origine ereditaria, nel restante 95% la malattia ha un origine incerta (SLAs). Tra i giocatori di calcio, caso unico tra tutte le categorie di sportivi professionisti, il rischio di sviluppare una SLAs è ben 6 volte superiore a quello della popolazione generale. Non è ad oggi nota la ragione di questo fenomeno. Si ritiene Titolo progetto Ente finanziatore Durata progetto Abstract del progetto che i principali fattori di rischio per lo sviluppo delle SLAs, oltre all’età e al sesso maschile, siano i traumi meccanici, l’esposizione ad erbicidi, un’attività fisica molto intensa e il fumo di sigarette. Da un punto di vista cellulare, una delle cause che si ritiene essere alla base di questa malattia, è il danno prodotto dal massiccio aumento dello ione calcio all’interno del neurone. Siccome la stimolazione dei recettori della nicotina (la sostanza contenuta nelle sigarette) che sono localizzati sulla membrana dei motoneuroni consente l’ingresso del calcio all’interno del motoneurone si è pensato di studiare la frequenza delle mutazioni dei geni che codificano i recettori nicotinici neuronali. Queste mutazioni sono presenti nel 6% di pazienti affetti da SLAs e inducono tutte un’alterazione nel funzionamento dei recettori nicotinici neuronali che provoca un aumento dell’ingresso del calcio all’interno del motoneurone aumentando la probabilità che esso muoia. Data l’anormale frequenza di questa malattia nelle persone che praticano l’attività calcistica a livello agonistico (amatoriale o professionale), ci proponiamo di valutare, mediante analisi del DNA estratto con semplici prelievi di sangue, la presenza di mutazioni dei recettori nicotinici in calciatori in buone condizioni di salute e/o di coloro che si sono già ammalati di SLAs. Siamo convinti che lo studio delle modalità precise delle modificazioni del funzionamento dei recettori nicotinici neuronali dovute a queste mutazioni genetiche, possa essere il primo e fondamentale passo per sviluppare una terapia innovativa mirata a ripristinare il corretto funzionamento dei recettori con la accelerazione della loro desensibilizzazione, diminuendo significativamente il rischio dell’insorgenza della malattia, cioè la suscettibilità alla malattia. A questo scopo ci proponiamo di caratterizzare dal punto di vista funzionale le mutazioni trovate in sistemi eterologhi ricostituiti in vitro (linee cellulari trasfettate); inoltre intendiamo sviluppare un modello di topo transgenico per una delle mutazione dei recettori nicotinici associata a SLA, e utilizzarlo come modello in vivo per studiare la fisiologia del muscolo e la fisiologia neuronale. Molecular and functional approaches to investigate the physiopathological role of the chemokines and their receptors in the central nervous system. Fondazione Cenci Bolognetti 2007-2010 Chemokines and their receptors are widely expresses in CNS where they play important roles both under physiological and pathological conditions. Chemokines are implicated in the control of neuronal and glial development, both in terms of migration and maturation, modulate synaptic transmission and have trophic and protective effects on neurons and glia. The spectrum of biological activities of chemokines is broadened by their involvement in the pathogenesis and maintaining of several neuropathologies, when their expression may be altered. Aim of the proposed research project is to investigate the physiopathological activities of chemokines as modulators of neuronal functions, identifying the molecular pathways involved. In particular the effects of chemokines will be investigated in brain slices, primary neuronal cultures, cell lines and expression systems as concern synaptic transmission, short-term neuromodulatory activities and long-term plasticity, trying to correlate these events to the activation of specific molecular effectors. These investigations will be correlated with studies of chemokine-induced communication between different cell types, like glial cells and neurons. The effects of chemokines as modulators of cell migration will be investigated in glial cells and neurons freshly isolated from rodents and the signal transduction pathways involved will be investigated. The activities of chemokines as neuroprotective substances will be investigated in vitro in neurotoxicity models and in vivo in animal models of cerebral ischemia. Overall, this study aspire to help to improve the understanding of neurogenesis and neurorepair, yielding possible clues to therapeutical strategies. IIT DEPARTMENT OF NEUROSCIENCE AND BRAIN TECHNOLOGIES ONGOING PROJECTS ON NEURODEGENERATION Fabio Benfenati, MD 1. Exposing tau pathology at its earliest Team: Laura Gasparini; Axel Blau, Paolo Medini; in collaboration with M. G. Spillantini, M. Goedert Funding: Compagnia di San Paolo and IIT + pending. The study is aimed at revealing functional changes typical of initial stages of tauopathy, and elucidating the underlying molecular mechanisms. This knowledge will reveal markers suitable for early clinical detection of tau-driven degeneration and identify key pathogenetic mechanisms to be targeted by new therapies. 2. Effects of β-amyloid aggregates on neuronal viability and microglial function Team: Denise Ferrera, Claudio Canale, Fabio Benfenati, Laura Gasparini Funding: IIT + pending We have demonstrated that Ab42, but not Ab40, reproducibly aggregates into stable oligomers and protofibrils and reduces the viability of mouse hippocampal neurons. We are now investigating how distinct Aβ aggregates affect microglia viability and activate microglial neuroinflammatory response and the latter response on neuronal physiology. 3. β-amyloid aggregates and astrocytic function in Alzheimer pathophysiology Team: Tommaso Fellin, Laura Gasparini, Nadia Mazzaro Funding: IIT + pending We will use an interdisciplinary approach including two-photon microscopy, Ca2+ imaging, biochemistry and pharmacology to test the hypothesis that b-amyloid (Ab) oligomers induce reactive gliosis through the overactivation of astrocytic NMDA receptors and we will evaluate whether the NMDA open-channel blocker memantine antagonizes Ab-induced astrocytic reactive response. 4. Molecular and structural determinants for β-amyloid mitochondrial accumulation and toxicity Team: Mounia Chami, Fabio Benfenati, Dolores Del Prete Funding: IIT + pending Mitochondrial dysfunction is emerging to play a pivotal role in Alzheimer’s disease (AD). Our project is aimed at: (i) studying ER-mitochondria communication and APP and Aβ distribution in AD models; (ii) modulating ER-mitochondria communication by pharmacological (Ca2+-drugs) and genetic/molecular biology through expression of mitochondrial fission and fusion proteins and/or S1T and consequently analyse mitochondrial function, and the intracellular distribution of APP and Aβ; (iii) determining the APP/Aβ partners in the MAM fraction using a proteomic approach. 5. Calcium modulating drugs as a therapeutic tool for Alzheimer’s disease Team: Mounia Chami, Fabio Benfenati, Dolores Del Prete Funding: IIT + pending The ability of neurons to regulate Ca2+ signals is compromised during aging. Importantly, alteration of Ca2+ regulating systems has been shown to occur early in Alzheimer’s disease (AD). We will investigate the potential use of Ca2+ drugs as a mean to reduce Aβ production in different AD models in vitro, analyze the potential effect of Ca2+ drugs on beta and gamma secretase activity and apply Ca2+ drugs on animal models in vivo and consequently analyze their effect on Aβ plaque formation, synaptic activity and cognitive development and/or progression. 6. Pathogenesis and therapeutic targets for SBMA Team: Mara Pennuto, Fabio Benfenati, Chiara Scaramuzzino Funding: MDA, Marie Curie, IIT + pending Polyglutamine diseases belong to the broad family of brain folding diseases. We aim at using Spinobulbar muscular atrophy (SBMA) as a model of polyglutamine disease to identify signaling pathways that impact brain folding diseases. Recent evidence shows that the IGF-1/Akt signaling pathway impact brain folding diseases. However, the mechanisms underlying the effect of Akt signalling remain to be elucidated. By putting polyglutamine AR downstream of Akt with our previous research, we established SBMA as a good model to tease apart the molecular details of the effect of IGF-1/Akt in polyglutamine disease. 7. Interactions of Aβ and α-synuclein with the cytoskeleton Team: Evelina Chieregatti, Giuseppe Ronzitti, Anako Tsushima Funding: IIT, Cariplo Foundation + pending Both Aβ and α-synuclein are implicated in actin dynamics as well as microtubule stability through their interactions with a microtubule-binding protein, tau. Our projects concern: (i) the study of their effects on the morphology and dynamics of actin microfilaments and intermediate filaments; (ii) the possible effect of the two proteins on cytoskeletal-regulated processes, such as polarization, growth cone formation and regeneration after axotomy; (iii) the identification of proteins of interest such as enzymes or molecular motors that may be found in the APP-containing endocytic vesicles. 8. Mechanisms of neurodegeneration and enhancement of adult neurogenesis to improve cognitive functions in Down syndrome Team: Andrea Contestabile, Diego Ghezzi, Fabio Benfenati, Laura Gasparini Funding: Lejeune Foundation + IIT + pending Down syndrome (DS) patients develop neuropathological lesions typical of Alzheimer disease (AD) by the fourth decade of life with cerebral accumulation of amyloid-beta (Ab) deposits and intraneuronal inclusions made of hyperphosphorylated tau protein (neurofibrillary tangles, NFTs). Mice models of DS recapitulate many aspects of the pathology, including: APP-dependent cholinergic dysfunctions during aging and tau hyperphosphorylation. This research line aims to better characterize the neurodegeneration process driven by APP overexpression and tau hyperphosphorylation in a DS mouse model and evaluate potential therapeutic interventions to alleviate the pathology. Down syndrome is also characterized by decreased neurogenesis both during development and adulthood. Adult neurogenesis in the DG of rodents has been associated to memory formation and hippocampaldependent learning. This research line aims to elucidate the impact of adult neurogenesis on learning and memory in a mouse model of DS and assess the effect of pharmacological treatments able to enhance the neurogenesis process. Nome Armando Genazzani Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto DiSCAFF, Via Bovio 6 - Università del Piemonte Orientale, Novara 0321-375827 [email protected] DiSCAFF, Università del Piemonte Orientale Ente finanziatore Durata progetto Abstract del progetto Malattia di Alzheimer e trasduzione del segnale legata al calcio Role of calcium-dependent gene expression and protein trafficking in shaping post-synaptic activity: implications for neurodegenerative diseases Fondazione Cariplo 2 anni Alzheimer disease (AD) is a progressive neurodegenerative disease characterized by a progressive cognitive and memory decline, increasingly frequent with advancing age until it affects as many as 50% of individuals 85 years of age and older. There is no remission in the progression of the disease nor are there any truly effective pharmacological interventions available at present. In the framework of an ageing society a better understanding of pathogenic mechanisms and identification of new pharmacological targets are key strategies. For many years, AD was attributed to neuronal death induced by deposits of fibrillar Amyloid b. The production of amyloid ß (Aß) is mediated by the concerted action of two different secretases, namely ß-secretase (BACE) and gamma-secretase, showing a proteolytic action on the amyloid precursor protein (APP). Alternatively, APP can be subjected to the proteolytic cleavage by a--secretase (ADAM10), which occurs within the sequence of Aß, thus precluding the formation of the amyloidogenic fragments. Yet, a recent view suggests that Abeta production has an impact primarily on the post synaptic compartment of the excitatory glutamatergic synapse (this preceding cell death and correlating with clinical features of the disease), as demonstrated by in vitro studies, thus indicating Alzheimer disease as a synaptopathy. An additional biochemical parameter which has been linked to both synaptic plasticity events and neurodegeneration is Calcium. The link between calcium and Alzheimer has been strengthened by a number of independent groups. Calcium dyshomeostasis appears to be an integral part of the pathology, either because it can influence Abeta production or because it can be altered by amyloid beta overproduction. This project will focus primarily in dissecting the contribution of calcium dyshomeostasis to the AD synaptopathy. Thus, the main goal of the project is to investigate the link between dys-regulation of calcium signalling and early stages of AD – both at cellular and molecular level working on the hypothesis that this neurodegenerative disorder is a synaptopathy. In particular we will firstly characterize calcium dependent processes in an innovative animal model of Alzheimer disease obtained by uncoupling ADAM10 from its cargo protein SAP97 which regulates enzyme activity. Secondly, we will address the role of calcium in both trafficking and gene expression of key synaptic proteins in vitro and in in vivo models of altered amyloid cascade. The investigation of these objectives would provide novel insights in the understanding of the molecular basis of Alzheimer’s disease in early stages and would possibly contribute in identifying new targets for therapeutic intervention Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Dr. Andrea Contestabile Email: [email protected] Phone: ++39-010-71781785 FAX: ++39-010-71781230 The Italian Institute of Technology (IIT) Department of Neuroscience and Brain Technologies Via Morego 30, 16163 Genova, Italy Down syndrome (DS) or trisomy 21 is the leading cause of genetically-defined mental retardation and is characterized by decreased cognitive functions, developmental defects, impaired neurogenesis and Alzheimer (AD)-like neurodegeneration during aging. The progression of Alzheimer neuropathology in DS individuals appears to be analogous to sporadic cases of AD with cerebral accumulation of amyloid-beta (A) deposits (i.e., A plaques) and intraneuronal inclusions made of hyperphosphorylated tau protein (i.e., neurofibrillary tangles, NFTs). However, its appearance is anticipated by at least twenty-thirty years with respect to the general population. The presence of an extra-copy of the Aprecursor protein (APP) gene, located on human chromosome 21 (HSA21), is believed to be directly responsible for early deposition of A in DS brain. The gene encoding Dyrk1A is also located in the triplicated region of HSA21 and is overexpressed in DS brains. Recently it has been proposed that Dyrk1A may be involved in tau hyperphosphorylation and NFTs formation typical of DS and AD. Dyrk1A phosphorylates tau at several sites, and such sites are hyperphosphorylated in adult DS brains. In addition, phosphorylation by Dyrk1A primes further phosphorylation of tau by Glycogen Synthase Kinase-3 (GSK-3 suggesting that the extra copy of the Dyrk1A gene on HSA21 contributes to the early onset of AD neuropathology seen in DS. In fact, a role for Dyrk1A in AD neurodegeneration has been recently identified, therefore suggesting a functional link between some of the triplicated genes found in Down syndrome and Alzheimer disease. The Ts65Dn mouse is the best characterized animal model of DS and carries an extra copy of the distal segment of mouse chromosome 16 which is synthenic to human chromosome 21. The Ts65Dn mouse recapitulates many aspects of DS, including: APP-dependent cholinergic dysfunctions during aging; Dyrk1A-mediated tau hyperphosphorylation and impairment of hippocampus-dependent memory function. Ts65Dn mice also display reduced adult neurogenesis in the two known adult neurogenic niche in mammalian brain, i.e, the subventricular zone (SVZ) of the lateral ventricle and the subgranular zone (SGZ) of the dentate gyrus (DG) in the hippocampus. Adult neurogenesis has been also found to be reduced in several mouse models of AD, further supporting the notion of a functional connection between DS and AD pathological mechanisms. Adult neurogenesis in the DG of rodents has been associated to memory formation and hippocampal-dependent learning such as spatial and contextual recognition. Decreased adult neurogenesis observed in DS and AD mouse models is therefore believed to play an important role in the impaired performance of these mice in hippocampus-depended memory tasks. GSK-3 inhibition has been identified has a possible therapeutic target for AD neurophatology. Accumulating evidence also indicates that adult neurogenesis is negatively regulated by the activity of GSK3 through the inhibition of the Catenin pathway, and that adult DG neurogenesis can be stimulated by GSK3 inhibitors. In order to identify effective pharmacological treatment to improve learning and memory in mouse models of DS that could be translated into human therapy for both AD and DS we are currently evaluating the usefulness of GSK-3 inhibitors to stimulate adult DG neurogenesis and alleviate tau hyperphosphorylation in Ts65Dn mice. Finanziamenti ricevuti Titolo progetto Effects of lithium on cognitive functions, neurogenesis and Tau pathology in Ts65Dn mice. Ente finanziatore Fondation Jerome Lejeune, France Durata progetto 2 years Abstract del Compelling studies indicate that impairment of adult neurogenesis and progetto degenerative mechanisms driven by hyperphosphorylation of Tau are the main determinants in reducing cognitive and memory function in animal models of Down syndrome (DS). Indeed, adult neurogenesis is impaired in the dentate gyrus of the hippocampus in the Ts65Dn mouse, a DS model reproducing most neuropathological and cognitive deficits of the disease. We have recently shown that administration of lithium to Ts65Dn mice is able to restore neurogenesis in the subventricular zone of the lateral ventricle, another neurogenetic area of the adult brain. Lithium is an extensively used mood stabilizer and acts as an inhibitor of Glycogen Synthase Kinase-3(GSK-3). GSK-3 activity enhances tau phosphorylation and is primed by dual-specificity tyrosine phosphorylated and regulated kinase 1A (Dyrk1A), which is encoded by a gene triplicated in DS and Ts65Dn mice. Aim of the present project is to test whether lithium long-term administration ameliorates cognitive performance in Ts65Dn mice by reducing accumulation of hyperphosphorylated tau and by stimulating neurogenesis, differentiation and integration of newly-born neurons. Lithium (2.4 g/kg of Li2CO3 in the chow) will be administered to Ts65Dn for 4 or 8 weeks starting at 6 months of age. Hippocampus-dependent memory functions will be evaluated through novel object recognition, spontaneous T-maze alternation and geometric task tests performed at the end of lithium treatment. After the treatment, adult neurogenesis in the dentate gyrus of Ts65Dn mice will be assessed by BrdU labeling and evaluation of proliferation markers. The expression of neuronal differentiation markers will be analyzed to assess the maturation of newly generated neurons. The accumulation of hyperphosphorylated Tau will be quantitatively monitored by immunohistochemical and biochemical techniques. Our experimental approach will allow testing a novel therapeutic approach for DS by simultaneously targeting two known pathological features affecting cognitive functions. Nome LAURA GASPARINI Contatti [email protected] Istituto/Dipartimento Fondazione Istituto Italiano di Tecnologia, Dipartimento di Neuroscienze e Neurotecnologia, Via Morego 30, 16163 Genova Proposta di ricerca Area di interesse identificata The lab focuses on studying key neuropathological molecular mechanisms of neurodegenerative diseases, with the view of understanding the disease processes and to translate basic knowledge into potential novel therapies. Tau-driven degeneration and neurotrophic response: searching disease markers for drug screening. Accumulation of intraneuronal incusions made of tau protein is a pathological hallmark of the brain of patients with tauopathies, including frontotemporal dementia and parkinsonism linked to chromosome 17, parasupranuclear palsy, corticobasal degeneration and Alzheimer Disease (AD). Our research is aimed at providing insight on alterations of neuronal activity associated with the onset and progression of tau pathology in transgenic disease models of tauopathy in vitro and in vivo, to reveal key tau-driven pathological mechanisms, and identify potential molecular targets for drug discovery. Neuroinflammatory mechanisms in Alzheimer disease: role of β-amyloid aggregates in microglia activation. Neuroinflammation is one of the neuropathological features of Alzheimer Disease (AD) and is characterized by activation of inflammatory cells (i.e., astrocytes and microglia) in areas of the brain affected by β-amyloid (Aβ) plaques and tau pathology, with production of altered levels of inflammatory mediators. Our lab investigate the pathogenic mechanisms underlying AD-associated neuroinflammatory processes, with the main focus on understanding the role on Aβ aggregated species in microglia activation. Pathogenic mechanisms driven by laminB1 overexpression in adult onset leukodystrophy. Lamin B1 (LMNB1) is a component of the nuclear lamina. It has been recently discovered that the presence of an extra copy of the gene encoding this protein causes autosomal dominant leukodystrophy (ADLD). ADLD is an invariably fatal disease characterized by white matter loss, movement disorders and dysautonomia. The pathogenic mechanisms by which LMNB1 gene duplication leads to disease are unknown. Our research aims at clarifying such mechanisms using in vitro and in vivo experimental models. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Tau-driven degeneration and neurotrophic response: searching disease markers for drug screening Compagnia di San Paolo 3 years Background: Neurofibrillary tangles (NFTs) are neuropathological hallmarks of human tauopathies. Reduced levels of neurotrophin TrkB receptor have been found in NFT-bearing neurons. Rationale and objectives: Initial findings showed that retinal ganglion cells (RGCs) of human P301S tau transgenic (TG) mice develop NFTs and have impaired axon outgrowth upon stimulation with neurotrophins (CNTF/BDNF). We propose to investigate the mechanisms by which tauopathy affects neurotrophin signalling and neuronal activity to identify disease markers and provide a test-bed for evaluating potential neuroprotective drugs in vitro. Expected results and relevance: Reveal characteristic signatures of tau pathology, suitable to investigate potential tau-targeting therapeutics. Titolo progetto Ente finanziator Durata progetto Abstract del progetto Clinical, neuroradiological and molecular investigation of Adult-onset Autosomal Dominant LeukoDystrophy (ADLD): dissection of Lamin B1mediated pathophysiological mechanisms in cellular and mouse models. TELETHON 3 years Overall objectives: To clarify Lamin B1 (LMNB1)-mediated pathogenic mechanisms of adult-onset autosomal dominant leukodystrophy (ADLD) and provide tools to study future therapies. Background/Rationale: ADLD is a rare, progressive and fatal genetic disease generally characterized by autonomic failure at onset. We have collected two ADLD families with LMNB1 gene duplication, and a third with a variant ADLD and an unknown mutation affecting LMNB1 expression. Expected results: The project will enhance our knowledge on genetic, clinical and neuroradiological features of ADLD. It will shed light on its pathophysiology and on LMNB1-mediated pathogenic mechanisms. It will generate an ADLD mouse model and provide tools (i.e., clinical biomarkers, expression constructs, gene-reporter assays, cellular models) which will be key elements for further studies of the disease and its therapy. Nome Contatti Michele Mazzanti [email protected] +39 2 50314958 +39 2 50314959 Istituto/Dipartimento Dept. of Bimolecular Science and Biotechnology Proposta di ricerca Investigation on early markers during neurodegenerative process Area di interesse identificata D. Oxidative Stress and neurodegeneration Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Beta-amyloid effect on retina cells. Investigation on Chloride Intracellular Channel 1 (CLIC1) protein modulation to use as an early marker during neurodegenerative process induced by amyloid compounds. Merz Biotech, Germany Italian Research Ministery 1 year 2 years One of the major apprehensions of contemporary society is population aging. Paradoxically, scientific discoveries applied to medicine have significantly increased the life expectation. However, older populations create new medical problems that, once episodic, are now common in a growing number of aging individuals. Neurodegenerative diseases play a predominant role in this new scenario. A major neurodegeneration disorder is Alzheimer Disease (AD), but this is poorly understood and not only we are without a successful therapy but also a proper identification of the disease in living humans is, at the moment, problematic. The appropriate diagnosis of an AD patient can be obtained only from histological analysis of post-mortem brain tissue. AD is characterized by an accumulation in neuritic plaques of beta-amyloid (A) protein, infiltrated by reactive microglia. Although A and in particular its oligomeric forms has been shown to exert a toxic effect on neurons, the role of microglia in neuronal injury remains unclear. Microglia activation is likely to be a key element in the pathophysiology of the local inflammation that accompanies AD. Activation of microglia by A protein triggers the production of toxic free radicals and pro inflammatory cytokines which are responsible for the neuronal degeneration. Previous studies demonstrated that A protein stimulation of neonatal rat microglia specifically lead to the increase of Chloride Intracellular Channel 1 (CLIC1) expression after 48 hours of stimulation. This was not the case when microglia was activated by phorbol myristate acid (PMA), Lipopolysaccharide, colony stimulating factor or basic fibroblast growing factor, thus quoting CLIC1 as specifically up-regulated by amyloid compounds. The cellular and molecular characterization of the protein has revealed that CLIC1 is mainly cytoplasmatic and is able to shuttle between the aqueous and the membrane phase depending on the activation state of the cell. In the membrane, the protein acts as a chloride conductance, allowing ionic flow to compensate the negative charges extruded by the membrane NADPH oxidase during reactive oxygen species (ROS) production. We have recently showed that suppression of CLIC1 functional expression using (i) inhibition of the CLIC1 current, (ii) replacement of extracellular Cl with impermeant anions, (iii) transfection of microglia cells with specific siRNA, or (iv) addition of an antibody against the channel protein, significantly reduced Aβ-induced ROS production. In a triple mutant Alzheimer mouse model, CLIC1 protein is up-regulated and show a predominant membrane localization in microglia cells of cerebral cortex. Furthermore, in human brains of Alzheimer patients, CLIC1, like several other proteins, is drastically up regulated. To explore the idea that CLIC1 protein could work as a therapeutic target as well as specific early marker for amyloid based diseases, we will investigate its distribution and modulation in two other neurodegenerative processes. Prion Protein syndrome (PrPsc) and Spinocerebellar Ataxia 1 (SCA-1) share with AD the predominant -sheet structure of the presumed pathological protein. As typical amyloid compounds, A, PrP and Ataxin-1 are characterized by insoluble deposits. However, while the action mechanism of the first two takes place in the extracellular environment, Ataxin-1 accumulates in the cell nucleus. The goal of our study is to pursue CLIC1 as a molecular target for early diagnosis and possibly for a therapy of amyloid induced neurodegeneration. Since its specific reaction to amyloid peptides, CLIC1 could represent an unambiguous marker of several amyloid based neurodegenerative process. It is reasonable to think that its marked increase in the brain immune system occurs in the early stage of the pathological process following a chronic state of oxidative stress induced by the different amyloids accumulation. We will use two different strategies to assess this possibility. It is well known that during neurodegenerative states the central nervous system (CNS) is colonized by systemic macrophages recruited from the blood stream. It is likely that the up regulation of CLIC1 protein could be a general feature of all macrophages. This will be tested both in mouse models of AD, PrPsc and SCA-1 as well as in blood samples from patients. Alternatively, we will explore the possibility to detect CLIC1 increment in the most accessible part of the CNS: the retina. Activation of the CNS immune system and neurodegenerative process were described in the eyes of AD, PrPsc and SCA-1 patients. Degeneration of retinal ganglion cells (RGCs) follows the same pathways of neuronal cell death in neurodegenerative diseases. Thus, CLIC1 protein increase should be easily detected in the retina immunocompetent cells system, following a chronic condition of oxidative stress that is belive to anticipate RGC apoptosis. 1. MECHANISMS OF NEURODEGENERATION C. Neuro Inflammation Nome GABRIELA CONSTANTIN, M.D., Ph.D. Assistant Professor of Pathology Contatti Email: [email protected] Tel: 39-045 8027102; 8027120; Fax: 39-0458027127 Department of Pathology, University of Verona, Strada le Grazie 8, 37134, Italy Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Role of Inflammation mechanisms in neurodegenerative diseases In recent years growing evidence show that blood-brain barrier (BBB) breakdown and inflammatory responses, may initiate and/or contribute to a ‘‘vicious circle’’ of the disease process, resulting in progressive synaptic and neuronal dysfunction and loss in disorders such as Alzheimer’s disease (AD), Parkinson’s disease, amyotrophic lateral sclerosis, multiple sclerosis, and others (Zlokovic, Neuron 2008). In fact, it is clearly emerging that a better understanding of AD inflammatory and immunoregulatory processes may lead to the development of anti-inflammatory approaches that may help to slow-down or block the progression of this devastating disorder. Our group has established expertise in neuroinflammation/neuroimmunology field and will perform an interdisciplinary study to explore the role of vascular inflammation mechanisms and the role of leukocytes and leukocyte trafficking in the pathogenesis of AD. The following main research lines will be developed: 1. Characterization of vascular inflammation and leukocyte-endothelial interactions in animal models of AD. Our team has previously shown that leukocyte adhesion to brain endothelium leads to BBB damage and contribute to neuronal loss (Fabene et al., Nat. Med. 2008). We will first study BBB leakage and vascular inflammation by identifying adhesion mechanisms able to mediate leukocyte-endothelial interactions in brain vessels. Then we will study the selectins, mucins, integrins and chemokines involved in rolling and firm arrest of leukocyte subpopulations such as neutrophils, monocytes, Th1 and Th17 cells, which were able to interact with endothelium in brain microcirculation of AD mice in preliminary studies performed by our team. To this aim, we will use our established expertise in: in vivo staining, in vivo Imaging System IVIS® 200 (Caliper Life Sciences) and intravital microscopy in cortical microcirculation (Constantin et al., Immunity, 2000; Battistini et al., Blood 2003; Pluchino et al., Nature 2005; Fabene et al., Nat. Med. 2008; Bolomini-Vittori et al., Nat. Immunol., 2009; Deban et al., Nat. Immunol., in press). Finally, we will study the effect of adhesion mechanism blockade whether by inhibitory antibodies or genetic deficiency on the induction and evolution of AD and will identify new potential therapeutic targets for AD. 2. Study of neuro-imuno interactions by using two-photon laser scaning microscopy (TPLSM) in AD cortex. Leukocyte motility behavior inside the cortex of transgenic animals with AD will be studied in relation to the vicinity of blood vessels, amyloid deposition and presence of neurofibrillary tangles. Taking into account our preliminary confocal microscopy studies showing that monocyte, Th1 and Th17 cells migrate into the cortex of AD mice, we will study the cell-cell contacts between immune cells and neural cells using transgenic mice expressing fluorescent neurons or glial cell populations. This research line will be developed based on our TPLSM expertise (Rossi et al., submitted) and our in vivo confocal microscopy facility, which consists of a customized upright Leica TCS SP5 AOBS system formed by: microscope DM6000 equipped with a complete set of PlanApo objectives, ultrafast scanning head SP5 AOBS coupled to Ar- HeNe sources and a mode-locked Ti:Sapphire Chameleon Ultra II laser (Coherent Inc). Overall these studies represent a novel and original contribution to the understanding of AD pathogenesis and have the potential to identify new therapeutic targets in AD. Finanziamenti ricevuti Titolo progetto Ente finanziatore Leukocyte recruitment: molecular mechanisms and pathological aspects Fondazione Cariverona Durata progetto February 1, 2009 - January 30, 2011 Abstract del progetto The research activity includes 3 tasks: Task 1. Mechanisms involved in the control of granulocyte and monocyte recruitment by Src tyrosin kinases; Task 2. Role of Rap1, cdc42 and PIP5KC in the control of the modalities of integrin activation by chemiotactic factors in T and B lymphocytes; Task 3. Role of Src-family tyrosin kinases in the migration of leukocytes in inflamed brain vessels and in the induction of neurodegenerative diseases Adipose-derived stem cells (ADSC) therapy in experimental autoimmune encephalomyelitis National Multiple Sclerosis Society, New York, USA July 1, 2009- June 30, 2011 This project has two objectives. The first objective is focused on the study of the clinical and neuropathological effect of ADSC on experimental autoimmune encephalomyelitis (EAE), which represents the animal model of multiple sclerosis, a neurodegenerative disease. The second objective is to study the immunomodulatory mechanisms controlling the effect of ADSC on EAE. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Name Contacts Istituto/Dipartimento Marina Bentivoglio, MD, Professor Office: +39-045-8027158; fax: +39-045-8027163; e-mail: [email protected] Department of Neurological, Neuropsychological, Morphological and Motor Sciences – Medical Faculty – University of Verona Strada Le Grazie 8, 37134 Verona, Italy Research proposal Regional vulnerability of the aging brain to neurodegeneration and immune regulatory mechanisms Background and aims: A wealth of evidence in the last years has indicated that normal aging is hallmarked by low-grade chronic inflammatory activity, with increased production of proinflammatory cytokines both peripherally and in the brain, and decreases of anti-inflammatory mediators. Data from our laboratory (e.g. Deng et al 2006; Sadki et al., Neurobiol. Aging, 28, 296-305, 2007. Xu et al., Brain Behav. Immun. 1, 138-152, 2010) have shown in mice an increase of the immune responsiveness of the aging brain (at the molecular and cellular levels, the latter including glial cells and T-cell recruitment). This is in contrast with the known peripheral decline of innate immunity during senescence, and could be involved not only in cognitive decline but also in the vulnerability of the aging brain to neurodegeneration. On the other hand, and importantly, not only aging is the primary risk factor for neurodegenerative diseases, including Alzheimer’s disease (AD), but also these diseases are hallmarked by regional vulnerability in the brain, which is still elusive. It is, therefore, of primary interest to assess whether immune regional changes in the aging brain are implicated in its susceptibility to neurodegeneration. To this purpose, the current proposal aims at investigating: i) regional immune-regulatory changes in the aging brain, and ii) the significance of such changes in terms of vulnerability of different brain regions to neurodegenerative insults during senescence. Plan of work: Step 1: Analyses of the following parameters in mice of young age versus an advanced age, and in transgenic mice which provide AD models: gene profiling (by means of microarrays and quantitative real-time RT-PCR) of immune-regulatory genes (including adhesion molecules, complement, cytokine and chemokine receptors, MHC antigens, Toll receptors, etc.), and immunophenotyping of cells (microglia, astrocytes, perivascular macrophages, T-cell subtypes, dendritic cells) in different dissected brain regions (neocortex, hippocampus, hypothalamus, midbrain, etc); magnetic resonance imaging with ultra-small superparamagnetic particles of iron oxide (USPIO) to visualize regional cellular infiltration. Step 2: A: Application of challenges with different potentially beneficial or detrimental approaches (proinflammatory stimuli, like lipopolysaccharide chronic administration; anti-inflammatory therapy; enriched environment, which is known to exert a beneficial effect; chronic sleep restriction -frequent during normal aging and known to affect cognitive performance), and test the outcome of these challenges on immune-regulatory gene expression, neurodegeneration and behavior/cognitive performance. B: Investigate the eventual occurrence of neurodegeneration during aging in mice with targeted deletion of immune molecules (e.g. STAT-I signaling pathway knockout, interferon-gamma or its receptor knock-out, etc.) versus wild-type littermates. Identified area of interest Current funding Project title Prediction of cognitive properties of new drug candidates for neurodegenerative diseases in early clinical development (Acronym: PHARMA-COG). IMI/115009 - IP (Integrated project) Funding agency Project duration European Commission 01/24/2010- 01/23/2014 (note: murine models of AD are available in the framework of this project) Project abstract Recently the EU Council of Ministers for Health underlined the importance of generating novel therapeutic agents both for symptomatic and disease modifying treatment of Alzheimer’s disease (AD). However, despite the increase in translational medicine activities attrition rates still remain high and progress in bringing these biomarkers and models to a state of readiness as effective decision making tools is slow as each academic and pharmaceutical company work in isolation. Bringing together European experts in technologies fully translatable from animal to human, experts in translational medicine, drug discovery and mathematical modelling, PHARMA-COG proposes to accelerate this validation using a ‘MATRIX’ approach i.e. conducting parallel experiments in animals and human using a comprehensive and standardised battery of behavioural, neurophysiological, morphological/functional imaging, and biochemical endpoints to: o o o o develop models with greater predictive capacity for the clinics develop and validate translatable pharmacodynamic markers to support dose selection develop challenge models to support early hint of efficacy studies identify and validate of markers of disease progression and patient stratification. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Michela Matteoli 02 50317097 [email protected] Farmacologia Chemioterapia e Tossicologia medica Toxic effects of beta-amyloid mediated by microglial cells Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Neurotoxic stimuli Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Elisabetta VEGETO Via Balzaretti, 9 02 50318263 [email protected] Dept. Pharmacological Sciences, University of Milan Gender and hormonal/endocrine signals in neuroinflammation: role in Alzheimer’s disease and brain aging Publications from the laboratory demonstrated a key role for estrogens in regulating microglia cells reactivity in an experimental model of Alzheimer’s disease and in other neuroinflammatory conditions. Accordingly, we showed that hormonal manipulations, achieved through experimental models of menopause and estrogen therapy, impinge on neuroinflammatory signs in the aging female brain. Considering that menopause is associated with a higher risk to develop AD in women and that estrogens are consistently associated with neuroprotective effects, our research is focused on the identification of the cellular and molecular targets of estrogens that modulate the susceptibility to neuroinflammation. Since estrogens action is mediated through the interaction with specific intracellular receptors, our studies are aimed at identifying selective estrogenic drugs and appropriate biomarkers of hormone signaling in neuroinflammation. Estrogen activity in microglia as a potential pharmacological target for AD therapy (GP0127Y01) Telethon; 126,000 Eu 3 yrs (2001-2003) This study was aimed at evaluating estrogens activity on the activation of microglia cells that is evoked by beta-amyloid deposition in the brain of APP23 mice, an animal model of AD. Results show that withdrawal of circulating estrogens through ovariectomy leads to an increased number of amyloid deposits that show reactive microgliosis, whereas estrogens replacement reduce this event to control levels. These results support the hypothesis that sicrulating estrogens are beneficial signals that acting also on the neuroinflammatory component of AD delay pathological signs of this disease. Estrogens and Women Ageing (Contract no. 518245) Specific Targeted Research or Innovation Project European Commission; 510,000 Eu 3 yrs (2005-2007) This project investigated on the efficacy of estrogenic drugs in reducing the neuroinflammatory response in the brain of aging female mice, with particular emphasis on the timing of drug administration in relation with menopause induction. Results show that menopause potentiates the neuroinflammatory response in brain and that the anti-inflammatory activity of several estrogenic compounds is reduced in parallel with increasing time of hormone deprivation. These results implicate that hormone replacement therapy might benefit from the anti-inflammatory activity of estrogenic compounds in brain only when therapy initiates Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto closer in time with the onset of menopause. MADRI (Menopause: decreased response to increasing inflammation) 5 R01 AG027713-02 NIH; 200,000 Eu 2007-2011 This project investigates more directly on the effect of menopause and aging on the inflammatory response of the brain. Preliminary results show that menopause modifies the susceptibility of brain inflammatory cells that is already altered by the aging process. DIMI (Diagnostic Molecular Imaging ) A Network of Excellence for Identification of new imaging markers for diagnostic purpose European Commission; 376,000 Eu 2006-2010 This project is aimed at challenging imaging techniques for improving studies on brain reactivity, including inflammation. Several reporter and transgenic animal models were generated under this program; the main results from this project show that it is now feasible to trace estrogen transcriptional activity in brain Role of estrogens in brain vulnerability to neuroinflammatory signals associated with aging and neurodegenerative diseases Ministero della Salute; 20,000 Eu 2009 Aim of the study was to analyse the reactivity of macrophage cells from male and female mice in response to pathological signals that arise in the aging brain. Nome Dr. Barbara Viviani Prof. Marina Marinovich Dr. Mariaserena Boraso Prof. Corrado L. Galli Contatti Tel: +39 (0)2 50318356 Istituto/Dipartimento Proposta di ricerca Mail: [email protected] [email protected] [email protected] [email protected] Department of Pharmacological Sciences, University of Milan Role of pro- and anti-inflammatory cytokines in the prognosis and progression of neurodegenerative diseases Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore MOLECULAR MECHANISMS OF INFLAMMATION AND REPAIR IN STROKE: NEURORADIOLOGY AND BIOCHEMICAL ENDPOINTS TO DESIGN A NOVEL THERAPEUTIC AND PROGNOSTIC APPROACH Regione Lombardia Durata progetto 24 mesi Abstract del progetto Stroke remains a leading cause of adult death and disability, which still demand an appropriate therapeutical approach. Some degree of spontaneous behavioral recovery is usually seen in the weeks after stroke onset. In general, the best outcomes are associated with the greatest return toward the normal state of brain functional organization. Reorganization of surviving central nervous system elements supports behavioral recovery. In this framework, the development of therapeutic approaches whose goal is to promote repair and restoration of function after stroke could be strategic. Two of the most prominent molecular events occurring in brain ischemia are inflammation and overactivation of the NMDA subtype of glutamate receptors (excitotoxicity), which lead to the progression of neuronal damage. Although this, an endogenous neuroprotective response, at least partially mediated via induction of neuroprotective factors, is also elicited by ischemic injury. Actually, the great potential of the brain for reorganization, plasticity and repair after injury is a critical issue to be taken into consideration, that has previously been understimated. Knowing the pathophysiological mechanisms that follow ischemia and their time course is essential in order to be able to identify and use biomarkers in monitoring the patients with acute stroke. Although still in the research phase, some biomarkers have been shown to correlate with the clinical evolution and could be important in foreseeing the clinical response to the treatment of the acute phase and to the different rehabilitation strategies in the post-acute and chronic phases. In addition, the inflammatory and neuroprotective response during the evolution of stroke can be monitored in the patient through suitable techniques such as neuroimaging. Up to now research on molecular targets in cerebral ischemia for developing novel therapeutics has mainly focused on single pathways, being inflammation or excitotoxicity or oxidative stress. The step forward, to go beyond the current state of the art, of the present project is to try to consider a group of different concurrent factors elicited during stroke. The project aims at investigating the cross-talk between inflammation, excitotoxicity, and endogenous neuroprotective/neuroregenerative pathways. In collaboration with the Neurological Institute C. Mondino, levels of selected biomarkers representative of inflammation and neuroprotection will be monitored in the serum of patients with stroke at different time points and then will be correlated with neuroimaging data and the clinical outcome. This will provide a clinical basis to finalize the basic research focussed on the evaluation of the molecular mechanisms recruited in the cross-talk between inflammation, excitotoxicity, and endogenous neuroprotective/neuroregenerative pathways. To this purpose we will evaluate in glial and neuronal population (i) the impact of inflammation on expression of neuroprotective factors relevant to brain ischemia (i) the effect of neuroprotective factors on inflammation, (ii) the effect of cytokines and neuroprotective factors on excitotoxicity driven by the NMDAR The project is designed to identfy new prognostic markers and new targets for therapy, aimed at boosting or superinducing the endogenous neuroprotective response directly or interfering with the mechanisms that blunt this response in conditions of injury. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto ANGELINI - ACRAF SpA Claudio Milanese 06-91045290 [email protected] Angelini Research Center- S.Palomba (Rome), Italy Study of the role of pro-inflammatory chemokines in neuroinflammatory disorders and analysis of the therapeutic potential of chemokine synthesis inhibitors Abstract Chemokines and their receptors play a crucial role in the trafficking of leukocytes and other immune system cells and are of particular interest in the context of the unique immune responses elicited in the central nervous system (CNS). Within this particular district, most data focus on the role of CCL2 and the related factors CCL7 and CCL8, since they have been implicated in a wide range of neuropathologies, including trauma, ischemic injury and multiple sclerosis (Semple BD et al., 2010, J Cerebral Flow & Metabolism 30:459). Furthermore, members of this chemokine family and their receptors recently emerged as key modulators in nociceptive influx transmission in neuropathic and inflammatory chronic pain models (Gosselin RD et al., 2008, Curr Med Chem 15:2866), providing evidence of the involvement of chemokines as important mediators of the initiation and maintenance of pain hypersensitivity. Angelini has a large experience in the study of chemokine synthesis inhibitors. A small original Angelini molecule able to inhibit the synthesis of CCL2, bindarit, has recently successfully concluded a phase II clinical study in diabetic nephropathy and it is currently undergoing another phase II study in restenosis. The aim of the present project is the identification and development of new molecules able to inhibit the synthesis of chemokines and endowed with physicochemical and ADME characteristics useful to target CNS disorders. The evaluation of the pharmacological activity of such compounds in appropriate models will allow to assess their potential as therapeutic agents useful for the treatment of CNS disorders and chronic pain. ISTITUTO SUPERIORE DI SANITA’ (Referente, Luisa Minghetti) The Istituto Superiore di Sanità is the leading technical and scientific body of the Italian National Health Service. Its activities include research, clinical trials, control and training in public health. The Institute conducts scientific research in a wide variety of fields and also serves as a major source of information relating to public health and biomedicine in Italy through online connections to national and international scientific databases and data banks. The institute has been actively involved in research in ageing and related neurodegenerative diseases over the last decade. It is partner in the European Research Area in Ageing network (ERA-AGE and ERA-AGE2) funded under the Sixth and the Seventh Framework Programme. The research on Alzheimer’s disease in ISS takes advantage of multidisciplinary expertise and resources including proteomics, molecular and cellular biology, neuroimmunology, behavioral analysis, in vitro and in vivo models, genetic, clinical and epidemiological studies, advanced statistical analysis, data and bio-banks. Main topics of research are: in vitro toxicity of amyloid proteins; excitotoxicity and free radical-related alterations of signaling pathways and post-translational modifications of post-synaptic machinery; analysis of protein aggregate characteristics responsible for the induction of amyloid formation; characterization of relevant rodent models by pre-clinical imaging with MRI scanning; characterization of behavioral and cognitive alterations in relation to neurochemical/neuroanatomical markers in AD murine models; effects of diet, pharmacological agents, or neurotoxicants on disease susceptibility and progression in AD murine models; early environmental factors in modulating onset and progression of neurodegenerative diseases; self antigens and autoantibodies in the pathogenesis of AD and as biomarkers of the disease; peripheral oxidative stress and antioxidant defense related biomarkers as prognostic and diagnostic tools; identification of mutations and polymorphisms affecting clinical phenotype and progression; descriptive, analytical, clinical epidemiological studies; longitudinal data bases for cohorts of elderly persons with cognitive deficits (IPREA and ILSA). Research projects currently active include: • Role of Genetic Determinants on the Progression of Clinical Phenotype from MCI to Alzheimer’s Disease. PI: A.Confaloni (National coordinator F.Tagliavini), Italian Ministry of Health .Eur 60,000 • Determinanti genetici e fattori modulatori nelle malattie neurodegenerative: modelli clinici ed animali P.I. A. Confaloni (National coordinator A. Bruni), Italian Ministry of Health, Eur 70,000 • Risk Assessment of iatrogenic transmission of neuro-AMYloidoses (RAAMY). PI: M.Pocchiari (in collaboration with E.Comoy, CEA, France), Alliance BioSecure, Fondation de recherche reconnue d’utilité publique. • Early diagnosis of Alzheimer’s diseases through the combination and correlation of molecular, functional and clinical biomarkers PI: M.Pocchiari, Italian Ministry of Health. Eur. 364,000 • Innovative approaches to the study of induction/repair of DNA oxidative damage in models of neurodegenerative diseases: molecular basis and identification of possible therapeutic targets. PI: P.Popoli, Italian Ministry of Health. Eur 364,000 • Effects Of Dog-Assisted Therapies On Physical And Psychological Well Being In The Institutionalized Elderly. PI: F.Cirulli, ISS and Nando Peretti Foundation, Eur 100,000 • Therapeutic effects of human-animal interactions in a rural environment on neuropsychiatric diseases and brain aging. PI: F.Cirulli, INEA Eur 62,000 • Nuovi farmaci ad azione neurotrofica e neuroprotettiva per applicazioni terapeutiche nelle neuroscienze: malattia di Azheimer, malattia di Huntington e Sclerosi Laterale Amiotrofica. PI: D. Merlo, (National coordinator A. Cattaneo). MIUR – FIRB “Idee Progettuali RBIP063ANC” 2007, Eur 32,000 • Ruolo neuroprotettivo della Timosina beta4 in modelli sperimentali in vitro di eccitotossicità, ischemia e Alzheimer Disease”. PI: D. Merlo, Tecnogen Spa, Eur 125,000 • Role of recurrent herpetic infections in neuronal damage. PI D. Merlo (National coordinator A.T.Palamara), Ministero della Salute, Eur 70,000 • An integrated approach to identify functional, biochemical and genetic markers for diagnostic and prognostic purposes in the elderly, in the centenarians and in people with dementia, Alzheimer’s disease, mild cognitive impairment. PI: N.Vanacore, Italian Ministry of Health, Eur 291,000 • Alzheimer's Disease (AD) and antipsychotics: a long term, multicentre, double blind, randomised clinical trial. PI: R.Raschetti, Italian Agency of Drug, Eur 1,950,000. • EC 7th Framework Programme “ERA AGE 2 Project – European Reasearch Area in Ageing Extension” . E. Scafato. EC 7th Framework Programme “ FUTURAGE Project – A Roadmap for Ageing Research” . E. Scafato • EC 2nd Programme of Community Action in the Field of Health “VINTAGE Project – Good Health into Older Age”. E. Scafato. • EC 5th Framework Programme “DESCRIPA Project - Development of screening guidelines and criteria for predementia Alzheimer’s disease”. E. Scafato. Nome Paola Bossù Contatti Via Ardeatina 306, I-00179, Rome Tel. 06-51501520 E mail [email protected] Istituto/Dipartimento IRCCS Fondazione Santa Lucia/Clinical and Behavioral Neurology Proposta di ricerca Role of inflammation in Alzheimer’s Disease: study of pro-inflammatory cytokines for the identification of new pathogenic and diagnostic targets Area di interesse Inflammation and neurodegeneration identificata Finanziamenti ricevuti Titolo progetto Genetic Risk factors and peripheral biological markers of conversion from Mild Cognitive Impairment to Alzheimer’s Disease Ente finanziatore Italian Ministry of Health Durata progetto 01/2009 - 01/2011 Abstract del progetto Several studies have demonstrated that inflammation plays an important role in mediating glial alterations and neurodegeneration that occur in AD patients. This phenomenon includes the accumulation of reactive microglia and astrocytes in damaged regions of AD brain and increased pro-inflammatory cytokine expression that may further contribute to the development and progression of pathological state. Accordingly, in several studies pro-inflammatory cytokines such as Interleukin (IL)-1, IL-6, Tumor Necrosis Factor (TNF)-α and IL-18 have been found to be peripherally increased in AD patients and substantially impaired in the late stage of the disease. However, the temporal definition of their implication in respect to disease progression, as well the extent to which inflammatory factors participate to the neuronal damage are issues of fundamental importance, which are to date still unclear. Recent studies performed by this participant unit have identified a possible role for the proinflammatory cytokine IL-18 in AD, by means of genetic data suggesting that IL-18 gene promoter polymorphisms can predict risk and outcome of AD, and by demonstrating that IL-18 is overexpressed by blood cells of AD patients and correlates with their cognitive decline. In the present study, by using a longitudinal clinical approach, the previous results regarding IL-18 will be expanded also to other inflammatory mediators with potential role in the disease progression and pathogenesis. In fact, this project is aimed to investigate the impact of pro-inflammatory and anti-inflammatory parameters during disease progression in patients at the higher risk of developing AD, i.e. MCI patients. These objectives are relevant for the identification of new diagnostic markers of the AD preclinical form and for the comprehension of inflammatory mechanisms, likely participating in the earliest pathogenic processes of AD. 1. MECHANISMS OF NEURODEGENERATION D. Oxidative Stress and neurodegeneration Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Elisa Greggio, Marco Bisaglia, Luigi Bubacco Via Ugo Bassi 58/b, 35129 Padova, ITALY Tel. +39 049 8276244 Dipartimento di Biologia, Università di Padova Indagine dei meccanismi molecolari alla base della malattia di Parkinson Mitochondrial dysfunction and axonal transport in genetic models of Parkinson’s disease MIUR 4 years Mitochondrial dysfunction and oxidative stress have been implicated in the pathogenesis of Parkinson’s disease (PD). Defective axonal transport, including abnormal accumulation of misfolded proteins and organelles and damage of transport of cargoes through axons, is also observed in many neurodegenerative diseases. Few lines of evidences suggest that both alpha-synuclein and LRRK2, proteins involved in dominantly transmitted forms of familial PD, may play a role in mitochondrial function, microtubulo stability/dynamics and axonal transport. The aim of this project is to use cellular and, in part, animal models of !-synuclein and LRRK2 to explore the physiological and pathological role of this two genes in mitochondrial functionality and in the transport of mitochondria and synaptic vesicle along the axons. Biochemical characterization of full length human recombinant LRRK1 and LRRK2 Michael J Fox Foundation 1+1 years Mutations in one gene, LRRK2, are a common cause of Parkinson’s disease. However, the protein product of this gene is large and complex and, to date, no one has been able to make highly pure, functional LRRK2 that can be easily used for biochemical experiments. The primary objective of this project is to make purified protein by combining expertise in multiple technologies from each of our collaborating laboratories and to share our methods with other researchers. If successful, our secondary objective is then to perform an exhaustive search for other proteins that may interact with LRRK2. Molecular mechanisms that involve the oxidation product of dopamine in the pathogenesis of Parkinson disease. Prin- MIUR 1+1 years Several hypotheses have been proposed in the literature to describe the mechanisms that lead to neurodegeneration in PD. This project will focus on the ossidative stress associated to the redox chemistry of dopamine. The rational in this choice is that dopamine oxidation may reasonably account for the specificity observed for dopaminergic neuron degeneration in PD. Although the oxidation of DA and DOPA lead to the formation of toxic ROS, the main cytotoxic species which could potentially explain the specific damage to dopaminergic neurons remain the highly reactive quinone species (DAQ) that have many potential protein targets for chemical modifications. The identification of several proteins, whose functions are affected by the mutations associated to familiar PD, triggered the hypothesis that late onset idiopathic forms of PD may derive from similar functional effects induced by chemical modifications produced by DAQ. This project will be focus on the definition of the damages induced by DAQ on two cytoplasmatic (DJ-1 and α-Synuclein) and two mitochondrial (PINKI and Superoxide Dismutase 2) possible protein targets. Nome Contatti Antonella Bobba Tel: 080 5442412 Fax: 0805443317 E-mail: [email protected] Istituto/Dipartimento Istituto di Biomembrane e Bioenergetica (IBBE), CNR Via Amendola 165/A 70126 Bari, Italy Proposta di ricerca Area di interesse identificata MITOCHONDRIA, APOPTOSIS AND NEURODEGENERATION: MITOCHONDRIAL DYSFUNCTION IN NEURODEGENERATIVE DISEASES CHARACTERIZATION OF There is compelling evidence for the direct involvement of mitochondria in neurodegenerative disorders. In most cases, increased oxidative stress appears to be the crucial initiating event that affects respiratory chain function and starts a vicious cycle finally leading to neuronal cell death via apoptosis or necrosis. In addition a functional interaction exists in the cells between cytoplasm and mitochondria and the cross-talk ability of these cellular components is involved in cell homeostasis and death. In the light of this, the goal of this topic is to ascertain whether and how, during neurodegeneration, the energy metabolism in the cytoplasm changes as a result of mitochondrial alterations and vice-versa and the mechanism by which certain compounds can prevent cell death. CHARACTERIZATION OF THE MOLECULAR MECHANISMS UNDERLYING AGING, CELL SENESCENCE AND NEURODEGENERATIVE DISEASES Aberrant modulation of cell signaling pathways involved in cell death regulation underlies several human pathological conditions, such as neurodegeneration. Although the mechanistic aspects of cell death and growth have been extensively investigated, there is still lack of knowledge about the early events that lead to deregulation of cell homeostasis and about the mechanisms of cell sensitization to growth and death signals in response to changes in cell environment. Thus, it is worthwhile to characterize the progression of the biochemical events and in particular of those occurring in the early phase of neurodegeneration with the aim to identify the critical steps that once blocked could allow for the restore, even if partial, of the impaired cellular function/activity. The characterization of the main signalling pathways that are activated and/or impaired during neurodegeneration and the close definition of the cause-effect relationship between them are powerful tools which are potentially usable to design new diagnostic, prognostic and therapeutic approaches. To this aim it is of great relevance an approach in which functional genomics and whole-genome expression profiling are combined in order to efficiently identify key players in biological response pathways. This is possible if different cellular systems are complementarily used in the experimental approach. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Neurotrofine e meccanismi relativi a malattie neurodegenerative. UO3 - Stress ossidativo e bioenergetica mitocondriale nella patogenesi delle malattie neurodegenerative MIUR (Progetto CNR/MIUR - Fondo FISR) 2 years (11/06/2003 – 1/10/2005) Studies on the involvement of mitochondria and role of oxidative stress in the ethiopathogenesis of neurodegenerative diseases by using primary neuronal cell cultures as in vitro model. The main results can be summarized as follow: i) early impairment of the energetic metabolism; ii) released cytochrome c exerts a crucial role by acting both as a ROS scavenger and an electron donor other than a caspase activator; iii) ATP level increases in the early phase of the death process and iv) ROS and thereafter activated caspases induce the progressive impairment of the Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto mitochondrial ANT. Malattie neurodegenerative come conseguenza di un alterato processamento di proteine neuronali, modelli animali e di colture cellulari in vitro MIUR (Progetto FIRB cod. RBNE01ZK8F) 3 years (08/01/2003 – 10/01/2007) Both animal and in vitro cultured cellular models have been used to study the molecular mechanisms which are at the basis of some neurodegenerative diseases and to identify new, potential therapeutical tools. Studies have been mainly focused on a thorough analysis of the mitochondrial function impairment with collapse of oxidative phosphorylation and increased production of reactive oxygen species. Bioenergetic and apoptotic systems of mitochondria. Genomics, proteomics, cellular homeostasis and physiopathology. WP2- Mitochondria role in animal, plant and microorganism cell apoptosis MIUR (Progetto MIUR N. 157) 3 years (2006-2009) The aims of the research project are concerning with: a)The role of mitochondria in the progression of neuronal apoptosis; b)The role of mitochondria in the apoptosis of a plant model system; c)The role of mitochondria in yeast apoptosis; d)The role of mitochondria in the induction/resistance to apoptosis in neoplastic cells Molecular bases in ageing-related degenerative syndromes MIUR - Progetto MERIT - Protocollo: RBNE08HWLZ (UR n 11: Protocollo: RBNE08HWLZ_012) 3 years (approved but not yet funded) The aim of this project is to discover specific molecular pathways and/or targets for the developing of new therapeutic strategies. Research activities are grouped into four workpackages: 1) Aggregation states and cytotoxic activity: molecular and in vivo/in vitro biological studies. 2) Oxidative stress and ageing: biochemical processes and regulation of gene expression. 3) Dismetabolic factors and neurodegeneration: molecular, biophysical and biochemical studies. 4)Exogenous and endogenous molecules against ageing and neurodegeneration: application for the diagnosis and therapy. Biotechnology applied to the study and utilization of Apulian extra-virgin olive oil and its minor components in aging and in diseases associated to oxidative stress Regione Puglia (Progetto Reti di Laboratorio Pubblici di Ricerca) 3 years (approved but not yet funded) Constitution of a Laboratories Network aimed to identify the nature and efficacy of minor components of Apulian extra virgin olive oil in the prevention of oxidative stress-associated pathologies Nome Lenaz Giorgio and Giancarlo Solaini Contatti 051-2091229; 333-2383218; fax: 051-2091217 [email protected] 051-2091215 ; 339 1948613 ; fax 051-2091224 [email protected] Istituto/Dipartimento Dipartimento di Biochimica Proposta di ricerca The involvement of mitochondria in neurodegenerative diseases, including Alzheimer disease (AD) is widely documented, although no clear causal relationships are available for a great number of scattered observations. Both a decrease of mitochondrial respiration, in particular of cytochrome oxidase and an enhanced production of Reactive Oxygen Species (ROS) has been reported in autoptic samples of AD patients and in neuronal cells of animal models of AD. Most researchers in the field believe these findings being the consequence of toxic amyloid peptides (A-beta) interacting with mitochondrial structures, and several studies have recently shown that A-beta and tau proteins, characteristic of AD, interact with mitochondrial respiratory complexes, opening novel hints to understand the pathogenesis of the disease. A significant investigation on the above issue has been carried out by some of us, but it’s noteworthy that our research group has a world wide reputation on the study of mitochondrial structure and function and has already been involved in research on aging and neurodegenerative diseases including AD. In particular we have shown the functional consequences of the recently discovered super-association of respiratory complexes and demonstrated that these super-complexes are easily dissociated by a number of changes in proteinprotein and lipid-protein interactions. We intend to propose and test the following working hypothesis. Under conditions likely present in brain regions of AD patients, the interaction of A-beta and tau with mitochondria at the level of respiratory chain is able to disrupt super-complex associations with consequent destabilization of the individual complexes, in particular Complex I (NADH Coenzyme Q reductase). This would necessarily determine an enhancement of production of ROS, thus explaining the observed oxidative stress in AD mitochondria. Loss of efficient electron transfer and altered assembly of Complex I would determine decreased oxidative phosphorylation and at the same time the oxidative stress could represent a sufficient stimulus for triggering cell death by necrosis or apoptosis. Our laboratory has the expertise and technology for all investigations required to test this hypothesis, and we are currently setting conditions considered likely to occur in brain regions of AD patients. The study will be performed on animal model systems and in human cells cultures. On the other hand, a better understanding of AD pathogenesis might establish the basis for attempts of therapeutic interventions, including metabolic therapy as recently shown in our laboratory for diseases featuring alterations of ATP synthesis. Area di interesse identificata Finanziamenti ricevuti Basic research/new treatment strategies Titolo progetto Ruolo dell’organizzazione sopramolecolare della fosforilazione ossidativa mitocondriale nella produzione di specie reattive dell’ossigeno e in alterazioni patologiche in sistemi modello e in cellule umane. Ente finanziatore MIUR (PRIN 2008) Durata progetto 2 years Abstract del progetto In this project we will test the working hypothesis that an initial enhanced ROS generation due to different possible reasons and originating in different districts of the cell besides mitochondria would induce disorganization of the supercomplex assemblies of the oxidative phosphorylation system, eventually leading to instability of Complex I quaternary structure; both the lack of efficient electron channelling and the decrease of Complex I would cause a fall of NAD-linked respiration and ATP synthesis, leading to pathological changes. This hypothesis will then be tested in two pathologies in which a major pathogenic factor is oxidative stress, i.e. the NARP syndrome (neuropathy with ataxia and retinitis pigmentosa) due to mitochondrial DNA mutations in the ATP6 gene, and diabetic peripheral neuropathy, a common and severe complication of diabetes mellitus. (Details follow) Nome Contatti Annalisa Santucci Tel. +390577234958 Fax +390577234903 Email [email protected] Department of Molecular Biology, University of Siena, ITALY Istituto/Dipartimento Proposta di ricerca Amyloid β peptide (Aβ) is directly involved in the pathogenesis of Alzheimer Disease (AD) or tightly correlated with other primary pathogenic factors. The predominant forms of Aβ in the human brain are Aβ(1-40) and Aβ(1-42), but Aβ(25-35) fragment, physiologically present in elderly people [1], is the more toxic region and has been recently found to play a relevant role in AD, due to its peculiar aggregation properties. In fact, actually, the ability to affect cognitive processes is typical not only of the full-length peptide, but also of several Aβ fragments, in particular just the undecapeptide Aβ(25-35). This peptide is regarded as the functional domain of Aβ, responsible for its neurotoxic properties and represents the actual biologically active region of Aβ. In vivo, Aβ(25-35) is present in neurons of subiculum and entorhinal cortex of AD brains, and it was also observed in Inclusion-Body Myositis (IBM) muscle. Self-assembly of Aβ(25-35) is a key player in AD, since it could nucleate the assembly of Aβ monomers and a bent in this region could be the rate-limiting step in Aβ fibril formation. In virtue of this, there is increased interest in the role of Aβ(25-35) fragment in free radical-associated neurotoxicity in AD brain. Age-dependent racemisation of Ser residue at the 26th position in Aβ(1-40) is critical in AD pathogenesis. In vivo conversion of non-toxic [DSer26] Aβ(1-40) to toxic [D-Ser26]Aβ(25-35) has been finally demonstrated and the presence of truncated and toxic [DSer26] Aβ(25-35) in AD brains, but not in age-matched control brains, adds a major value to in vivo neurodegeneration mechanism. Given the suggested centrality of Aβ(25-35) to the pathogenesis of AD, this peptide should deserve major attention also as a therapeutic target. In fact, diagnosis and monitoring of sporadic AD have long depended on clinical examination of individuals with end-stage disease. However, upcoming anti-AD therapies are optimally initiated when individuals show very mild signs of neurodegeneration. Our research group provided for the first time an exhaustive overview about Aβ(25-35) [2] and during the last few years it has deepened the aggregation properties of this peptide according to solution and environmental conditions [3]. Actually, we are dedicating attention to this peculiar peptide in the light of its great oxidative stress generation capacity and extreme toxicity in neuronal cells and synaptosomes. The evidence points toward Aβ(25-35) being primarily responsible for neurodegeneration observed in AD. The question remains on how this can induce such degenerative effects. Strong of our experience in the field of the study of oxidative stress [4-9], we propose ourselves to estimate the mechanisms been involved in the ROS generation Aβ(25-35)-mediated in different cell models. We can use SELDI technology to study [DSer26] Aβ(25-35) as a core biomarker for the mild cognitive impairment stage of AD. From a longer perspective, establishment of the most effective combinations of different biomarkers and other diagnostic modalities may be foreseen. Moreover, due to our well established experience in Proteomics [10-15], we will apply proteomics techniques to study cellspecific early markers of brain aging-related degeneration in human astrocytes and neurons that may contribute to neurodegenerative damage. The protein expression patterns of the AD neuron and astrocyte cultures will be compared with those obtained from healthy people. Differentially expressed spots will be identified by matrix-assisted laser desorption/ionization-time of flight peptide map fingerprinting and database search. A number of the protein alterations have been previously reported in the brain tissue proteome of animal models, aged brain or AD brain, but are still quite unexplored in neuron and astrocyte proteome and our project could help to clarify if both cell types are involved in the brain degenerative changes as well as to study neuronal changes having a greater influence in the pathology progress. These findings will be discussed in reference to the effect of specific protein oxidation and changes of expression on potential mechanisms of abnormal alterations in metabolism and neurochemicals, as well as to the learning and memory deficits in AD. References [1] Kubo, T.; et al. A possible mechanism of neurodegeneration in AD - Involvement of racemized βamyloid. Soc. Neurosci., 1999, Abstr. 25, p. 838. [2] Millucci L, et al. Conformations and biological activities of Amyloid Beta Peptide 25-35. Curr Protein Pept Sci. 2009 Sep 15. [3] Millucci, L., et al. Rapid aggregation and assembly in aqueous solution of Aβ(25-35) peptide. J. Biosci. 2009, 34(2), 293-303. [4] Braconi D, et al. Proteomics and redox-proteomics of the effects of herbicides on a wild-type wine Saccharomyces cerevisiae strain. J Proteome Res. 2009;8(1):256-67. [5] Braconi D, Possenti S, Laschi M, Geminiani M, Lusini P, Bernardini G, Santucci A. Oxidative damage mediated by herbicides on yeast cells. J Agric Food Chem. 2008 May 28;56(10):3836-45. [6] Braconi D, et al. Comparative analysis of the effects of locally used herbicides and their active ingredients on a wild-type wine Saccharomyces cerevisiae strain. J Agric Food Chem. 2006 19;54(8):316372 [7] Desideri G, et al. Effects of bezafibrate and simvastatin on endothelial activation and lipid peroxidation in hypercholesterolemia: evidence of different vascular protection by different lipid-lowering treatments. J Clin Endocrinol Metab. 2003;88(11):5341-7. [8] Desideri G, et al. Angiotensin II inhibits endothelial cell motility through an AT1-dependent oxidantsensitive decrement of nitric oxide availability. Arterioscler Thromb Vasc Biol. 2003;23(7):1218-23. [9] Trabalzini L, et al. Proteomic response to physiological fermentation stresses in a wild-type wine strain of Saccharomyces cerevisiae. Biochem J. 2003;370(Pt 1):35-46. [10] Bernardini G, et al. Postgenomics of Neisseria meningitidis for vaccines development. Expert Rev Proteomics. 2007;4:667-77. [11] Bernardini G, et al. The analysis of Neisseria meningitidis proteomes: Reference maps and their applications. Proteomics. 2007;7(16):2933-46. [12] Spreafico A, et al. A proteomic study on human osteoblastic cells proliferation and differentiation. Proteomics. 2006;6(12):3520-32. [13] Mini R, et al. Comparative proteomics and immunoproteomics of Helicobacter pylori related to different gastric pathologies. J Chromatogr B Analyt Technol Biomed Life Sci. 2006;833(1):63-79. [14] Bernardini G, et al. Proteome analysis of Neisseria meningitidis serogroup A. Proteomics. 2004;4(10):2893-926. [15] Magnani A, et al. Two-step elution of human serum proteins from different glass-modified bioactive surfaces: a comparative proteomic analysis of adsorption patterns. Electrophoresis. 2004;25(14):2413-24. Area di interesse identificata Post-Genomic Biochemistry of Neurodegenerative Diseases Finanziamenti ricevuti Titolo progetto Characterization of peptide oligomers in Alzheimer Disease Ente finanziatore Siena Biotech s.p.a. Durata progetto 1/07/2006 - 30-04-2009 Abstract del progetto Aβ(25-35) is the shortest peptide sequence that retains biological activity of full-length Aβ(1-42) and exhibits large β-sheet aggregated structures. Because of these features, it has often been chosen as a model for fulllength Aβ in structural and functional studies but kinetic and structural studies of this peptide are indeed strongly hampered by its tendency to quickly assemble into insoluble aggregates. The aim of the project was to study the uncommon rapidity with which Aβ(25-35) self-assembles at different pH. The obtained results support the hypothesis that the early Aβ(25-35) aggregated forms, that develop at pH 3, may not enhance the growth of dangerous fibrils generated at pH 7.4. We conclude that the mechanism by which Aβ forms toxic fibrils is initiated in vivo and probably does not involve low pH compartments, confirming the hypothesis of Kubo et al. (2002) on the origin and toxicity of Aβ(25-35). 1. MECHANISMS OF NEURODEGENERATION E. Trophic factors involvement in the neurodegeneration process 1. MECHANISMS OF NEURODEGENERATION F. Endocrine and Metabolic Factors Country: Italy Contact person: Paola Fragapane, Cecilia Mannironi (CNR) Date: 28/01/2010 I) Strategic Issues Stress is a term to describe experiences that are challenging both emotionally and physiologically. A feature of the stress response is the activation of the autonomic nervous system and hypotalamus-pituitary-adrenal (HPA) axis with the production of glucocorticoids hormones. This response is needed to survive dangerous situation but excessive adrenocortical and autonomic function is deleterious for health and survival. The central nervous system in response to such activation elaborates the appropriate behavioral and physiological responses. As regards to the neuroanatomical substrate the hypothalamus and the brain stem are essential in the response to stressors, however, other brain regions are also well known targets of stress hormones. Among such regions, research has focused its attention on the prefrontal cortex, the hippocampus and the amygdala. Interestingly these brain structures have been related also to functions such as memory, and decision making but also to anxiety and other psychiatric diseases. Acute (single stress) and chronic (repeated) stress induce prominent changes in neuronal activity and synaptic functions. These changes are thought to be due to neuronal remodelling such as dendrites shortening and debranching (Mc Ewen, 2007) in the hippocampus and in the medial prefrontal cortex. These stress-induced changes are mediated by modification of gene expression at transcriptional levels. However, recent studies are pointing out the contribution of posttranscriptional regulation to stress associated responses (De Rijk et al., 2003). An important post-transcriptional mechanism of gene regulation involves microRNA (miR). A range of evidence suggests that these small RNAs form complex networks involved in the regulation of differentiation and development, via regulation of chromatin modification, transcription, translation and RNA stability. They are small RNA molecules (about 22 nucleotide long) that inhibit mRNA translation or induce mRNA degradation by sequence specific pairing with mRNA 3' untranslated regions (UTRs). In the vertebrate nervous system miR have been shown to play a role during development and neurogenesis (Kosik, 2009; Lagos-Quintana et al., 2001). MiR are also abundantly expressed in the adult brain and appear to regulate the maintenance of mature trait and synaptic plasticity (Mehler and Mattick, 2006). MiR134 was reported to be a regulator of dendritic spine development in brain (Schratt et al., 2006). Perturbation in miR are associated with a number of neuronal diseases as X-linked mental retardation, Parkinson’s disease and schizophrenia. II) Priority areas The molecular mechanism underlying brain response to stress are still incompletely understood. The aim of this project is to identify region-specific stress-induced changes in miR expression. The expression profiles of miRs in brain regions known to be involved in stress response such as the hippocampus, the amygdala and the prefrontal cortex will be analyzed after the exposure to aversive stimuli. The analysis will be performed in rodents exposed to single or repeated restraint stress and gene expression in each brain regions will be compared with that of matched control mice. LNA (locked nucleic acid) microarrays will generate a limited set of candidate miRNAs, which will be validated by Northern blots and qPCR. In order to have a better neuroanatomical definition of miR expression selected miR will be analyzed by in situ hybridization. mRNA targets it will be predicted through the use of freely available algorithms and will be validated by reporter construct expression in cell system. Moreover bioinformatic analysis of miR upstream promoter sequences will allow us to identify transcription factors involved in their modulation in response to stress stimuli. III) Impact The hippocampus, the amygdala and the prefrontal cortex are interconnected and influence each others via direct and indirect neuronal activity. It is well established their involvement in the stress response and in the plastic changes occurring in response to acute and repeated stress exposure. Our analysis of miR expression pattern will allow us to understand possible region specific expression pattern and/or common molecular networks. Translation of the information on stress effects from animal models to the human organism is an important challenge. Indeed maladaptive response to stress is considered causal to many psychiatric diseases such as major depression, anxiety as well as schizophrenia. The understanding of the contribution of non coding RNAs might allow on the one hand a better understanding of the neural mechanisms at the basis of stress response on the other hand might help identify possible targets for the development of new future therapies. Nome Roberto Rimondini-Giorgini Contatti [email protected] Istituto/Dipartimento Department of Pharmacology Via Irnerio 48 40126 Bologna Italy Proposta di ricerca Area di interesse identificata Developing competitive animal models / basic research Finanziamenti ricevuti Titolo progetto MECHANISMS BEHIND THE ASSOCIATION OF APOE4 GENOTYPE AND LIFE STYLE RELATED FACTORS IN ALZHEIMER`S DISEASE Ente finanziatore Fondazione Cassa di Risparmio di Bologna Durata progetto 2 anni Abstract del progetto Alzheimer`s disease (AD) is a devastating neurodegenerative disease and the most common cause of dementia. The current estimate number of people with dementia in the world stands at 24.3 million, with an estimated 4.6 million new cases every year in the world. There are no definitive diagnostic tests, biological markers of AD or pharmaceutical treatments. At central nervous system (CNS) level, the pathological signs include deposits of extracellular amyloid-β peptide (Aβ) in plaques. Loss of neurons and synapses is also widespread. Whilst some cases have a genetic component (familiar AD) the majority of cases (90-95%) have unknown cause and are named sporadic AD. Familiar AD seems to be correlated to mutations in key genes such as in the amyloid-precursor-protein (APP) gene and in the presenilin genes (1 and 2). On the contrary, sporadic AD seems to be consequence of a complex interaction between both genetic and environmental risk factors. The major genetic risk factor is an allelic variant of apolipoprotein E (apoE) called apoE4. Nowadays, no specific environmental risk factor has been definitively identified as being associated with AD. Anyway, AD is associated with a history of depressive illness, traumatic head injury, cardiovascular disease, smoking and stroke. In addition, several evidences highlight the correlation between high alcohol consumption, saturated fats and cholesterol intake. The apolipopoteins are cholesterol transporters of high importance for neuronal plasticity, CNS glucose utilization and mitochondrial functions. The apoE isoforms (2, 3 and 4) differ in their abilities to accomplish these critical tasks. Approximately 20% of the population carries one or two apoE4 alleles. ApoE4’s involvement in neuropathology is well documented factor for AD in many populations. In some, 40–80% of patients with AD possess at least one apoE4 allele. While ApoE3 and apoE2 are effective in maintaining brain homeostasis, apoE4 seems to have an opposite rule. Impaired cognition in non-dement individuals carrying the apoE4 allele worsens with age, suggesting a global detrimental effect on the CNS. Although apoE4 is strongly linked to AD pathology, its mode of action is unknown. Insights into the role of apoE in neuropathology have come from studies of transgenic mice expressing human apoE3 or apoE4 in neurons or astrocytes. Features of AD pathology in these animals include reduced numbers of presynaptic terminals in mice expressing apoE4 with or without expression of human amyloid precursor protein (APP), increased plaque deposition in apoE4 mice expressing APP, increased phosphorylation of tau, impaired learning and memory, and altered long-term potentiation. No studies are available on the correlation between life style and apoE4 genotype in AD incidence. Aim of this project is to study the impact of different life conditions in relation to the apoE genotype on neurodegeneration. Wild and transgenic mice expressing human apoE3 or apoE4 are exposed to different diet conditions such as a normal diet, high saturated fats and cholesterol intake. After 6 months of diet mice will be tested to evaluate cognition and memory impairments. In the same subjects, gene expression (cDNA microarrays) will be studies in discrete brain areas. RT-PCR assay will be used to confirm the results. Moreover, selected genes will be further analyzed with classical biochemical procedures. In addition, other AD proteins such as Aβ, APP and Tau, and several markers for apoptosis, necrosis and neurodegeneration, will be investigated with biochemical methods in order to determine their quantity, their activity and their distribution in the brain. The combination of high throughput methods with the classical biochemical assays will allow a deep understanding of the molecular mechanisms behind the studied risk factors involvement in neuropatology in relation with apoE genotype. The obtained results will provide important information to define life guidelines for people carrying the apoE4 allele, as prevention for AD and neurodegenerative disorders. This could be of relevance in public health strategies. Furthermore, results could help to identify new therapeutic targets and early diagnostic markers. Moreover, this new experimental approach will develop a new animal model for AD that has a more natural onset of the pathology. Rosa Maria Corbo Nome Contatti Istituto/Dipartimento Proposta di ricerca [email protected] Dip. Genetica e Biologia Molecolare, Università La Sapienza, Roma Sporadic Alzheimer’s disease (AD) is a complex neurodegenerative disease caused by the interaction of genetic and nongenetic factors. The e * 4 allele of the apolipoprotein E gene (APOE) is the only well-established genetic susceptibility factor for sporadic AD, though numerous risk alleles in other genes are also thought to be involved in AD development. Gender is a relevant AD risk determinant and there is evidence for a higher prevalence of AD in women. However, whether this is due to the longer life expectancy of women or to biological gender- specific risk factors for the disease is unclear. It has been hypothesized that a declining estrogen level following menopause and subsequent reduction in its neuroprotective action may contribute to the development of the disease in women. In line with this hypothesis, the possible role of estrogen nuclear receptor genes (ESR1 and ESR2) as AD risk factors has been largely investigated. In addition there is evidence that past fertility is associated with a higher AD risk and a reduced AAO of the disease, probably because of the lifelong decrease in estrogen levels that parity seems to produce in women. The aim of the present investigation is to extend previous investigations (1-3) and in particular 1) to collect data on the reproductive life of AD women to identify those parameters which could influence the risk of AD and/or AD onset age; 2) To study the genetic variability of genes involved in estrogen metabolism or women fertility. - Corbo RM, Gambina G, Ruggeri M, Scacchi R. Association of estrogen receptor α (ESR1) PvuII and XbaI polymorphisms with sporadic Alzheimer’s disease and their effect on apolipoprotein E concentrations. Dement Geriatr Cogn Disord, 22:67-72, 2006- Combined effect of Apolipoprotein E (APOE) genotype and past fertility on age at onset of Alzheimer’s disease in women. Dement Geriatr Cogn Disord, 24: 82-85, 2007 - Genetic variation of CYP19 (aromatase) gene influences age at onset of Alzheimer’s disease in women. Dement Geriatr Cogn Disord. 2009, 27, 513-518. Area di interesse identificata Genetic susceptibility to Alzheimer’s disease Alzheimer’s disease in women: AD as a gender disease A. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Basic research Investigation on common and rare variation of genes involved in sporadic Alzheimer’s Disease susceptibility Università di Roma La Sapienza 5 years At present the genetic component of the sporadic late-onset form of Alzheimer disease (LOAD) has been only partially elucidated, since only the apolipoprotein E (APOE) gene, specifically the e*4 allele, has been reportedly associated with the disease. Since the APOE polymorphism alone does not entirely explain the genetic predisposition to sporadic AD, investigation into whether any variation in further candidate genes could account for the remaining part may offer important clues. However, investigations on AD candidate genes have usually examined only common polymorphisms in accordance with the hypothesis “common disease /common variants” . The additional hypothesis suggesting the presence of a relevant genetic heterogeneity due to rare alleles (“common disease/rare variants” ) has been scarcely investigated . The aim of the present study was 1) to examine the common variation of numerous candidate genes (ACT, PSEN1, LDL-R, APOC1, alfa2M, PON1, LPL, AChE, BChE, Chat, CYP19); 2) to carry out a mutation screening in the exons of APOE, PSEN1, APP genes by means of DHPLC (denaturing high performance liquid chromatography). (product: 12 publication, see pubmed) Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Prof. LUCIA MIGLIORE University of Pisa Department of Human and Environmental Sciences Via S. Giuseppe, 22 56026 PISA Italy E-mail: [email protected] Fax: +39 050 2211034 Tel: +39 050 2211029-28 - Genetic susceptibility to Alzheimer’s disease (Epigenetic marks of susceptibility to Alzheimer disease) Abstract AD is characterized by high Hcy and low folate blood levels. Many of the genes participating in folate metabolism are polymorphic, meaning that this pathway can be altered by low dietary folate intake, by the polymorphic variants and/or by combinations of both. In vitro studies showed that in conditions of altered folate metabolism the status of methylation of the promoter of the PSEN1 gene underwent a variation, causing a deregulation of presenilin 1, BACE and APP proteins. The study is designed to evaluate the contribution of folate metabolism (including polymorphisms of metabolic genes and plasma levels of folate, homocysteine (Hcy) and VitB12) in both early (MCI) and late stages of dementia, and its possible contribution to the methylation status of the promoter of genes that participate in Aβ processing. The study will be performed in blood samples with the goal of identifying easily detectable epigenetic biomarkers of disease susceptibility. Finanziamenti ricevuti Titolo progetto A PILOT STUDY ON THE EFFECTS OF FPP ON THE COGNITIVE FUNCTION AND ON OXIDATIVE STRESS PERIPHERAL BIOMARKERS IN MILDCOGNITIVE IMPAIRMENT PATIENTS (MCI) Ente finanziatore Durata progetto Abstract del progetto OSATO Research Institute, Gifu, Japan Project ongoing 30+30 amnesic MCI patients divided into two main groups. One treated with fermented papaya preparation (FPP), the other without treatment (placebo: PLC). The study will be performed within 12 months for a total treatment time of six months. After six and twelve months a report will analyze the results. At the beginning of the pilot study all the subjects recruited will be screened for the Total Antioxidant Status (TAS) that will include the determination of the following values in blood: selenium, folic acid, vitamin B12, C, E together with the evaluation of the GSH/GSSG ratio. The groups will be analyzed for assessing primary and oxidative DNA damage levels (Comet assay) and chromosome damage also due to oxidative damage (Cyt-B micronucleus assay) in peripheral blood leukocytes. Moreover 8OH-dG levels in urines samples will be evaluated. We plan also to evaluate the following biochemical parameters of oxidative stress: advanced oxidation protein products (AOPP); glutathione; hydroxynonenal; lipoperoxide (basal and under stress conditions); lactate (basal and under stress conditions); mitochondrial metabolite (by Mass Spectroscopy). The decrease (if any) of oxidative stress biomarkers due to the FPP subministration will be evaluated by comparing after12 months the two MCI groups (FPP treated and placebo controls) data by means of statistical analyses. At the same time the cognitive impairment evolution will be monitored by means of the neuropsychological evaluations also within the treatment period (0, 6 and 12 months). Moreover for all the subjects involved, DNA extraction will be performed and the samples will be stored for further genotype analysis (e.g.: ApoE, ..). Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto LAURA COLOMBAIONI [email protected]> Istituto di Neuroscienze CNR, Pisa Role of altered lipid and cholesterol metabolism in neurodegenerations Ente finanziatore Associazione Italiana Niemann-Pick Durata progetto 3 years Abstract del progetto Deregulated lipid metabolism is of particular importance in pathogenesis of CNS disorders and injuries. For this reason the role of cholesterol and lipid metabolism as susceptibility factors in neurodegenerations needs to be further investigated. Cholesterol is an important regulator of lipid organization in neuronal membranes and is the precursor for neurosteroid biosynthesis. Low levels of neurosteroids are related to poor outcome in many brain pathologies. The deregulation of the principal cholesterol carrier protein in the brain, the Apolipoprotein E, is a significant risk factor for Alzheimer's disease. Similarly, Parkinson's disease is, in some degree, caused by an enhancement of lipid peroxidation. Also Niemann-Pick A-B diseases are caused by sphingomyelin accumulation, while Niemann-Pick disease C is due to mutations in either the NPC1 or NPC2 genes, resulting in defective cholesterol and sphingolipid transport. Sphingolipid metabolites are emerging as members of a novel class of lipid mediators, acting both as intracellular second messengers and as ligands of cell surface receptors, capable of controlling neuronal proliferation, differentiation and apoptosis. In my laboratory, the effects, on neuronal cells, of an altered lipid metabolism are examined. The goal is to understand how the altered lipid metabolism produces neuronal injury, what subcellular compartments are involved and how this occurs. An interdisciplinary approach is used employing cell biology, molecular biology and live cell imaging methods. By using confocal microscopy, some key events associated to altered cholesterol trafficking or to sphingolipid unbalance are characterized, such as mitochondrial deregulation, release of pro-apoptotic effectors and alterations of calcium homeostasis in specific subcellular compartments. Cellular mechanisms of neuronal dysfunction due to altered lipid metabolism. In particular, the effects on neuronal calcium homeostasis of some sphingolipid metabolites such as Glucosyl-ceramide Sphingosine and Sphingosine 1-phosphate are analyzed. Our results indicate that the calcium balance in mitochondria and in Endoplasmic Reticulum is regulated by cholesterol and sphingolipid metabolites and that this is critically involved in control of neuronal physiology. Understanding the impact of lipid metabolism changes in neuronal cell functions will promote novel concepts and will reveal new therapeutic targets for a variety of nervous system disorders. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Roberto Cosimo Melcangi 02-50318238 [email protected] DEFIB-Università degli Studi di Milano F. Endocrine and metabolic factors Sex-specific therapeutic strategies based on neuroactive steroids for Alzheimer's disease. partial support by Bayer-Pharma 2 years The importance of sex in the incidence of Alzheimer's disease (AD) has been studied. Indeed, epidemiological studies support a higher prevalence and incidence of AD in women. Moreover, as observed in several experimental models and in AD patients, parameters such as oxidative damage, plaque load, amyloid precursor protein, β-amyloid production, seizure activity, locomotion, hippocampal neurodegeneration, astrocyte proliferation, level of brain-derived neurotrophic factor, cognition are differently affected in a sex-dimorphic way. Although sex differences in the brain may be in part the consequence of the genetic complement of the cell, differentiation of the neural tissue is also influenced by the developmental actions of sex hormones. Both genetic sex and developmental actions of sex hormones contribute to the generation of sexually dimorphic neuronal networks in the brain and the spinal cord, and both factors may influence the outcome of pathological insults to the nervous system. Thus, some sex differences in brain pathology may be the consequence of the functional and morphological differences in neural structure between the sexes. However, it is important to remember that CNS is not only a target for steroid hormones coming from peripheral glands but also of those directly synthesize in loco (i.e., neurosteroids). Moreover, steroid hormones are also actively CNS converted into metabolites that are more active and in some cases utilize a different mechanism of action. Both steroid hormones and neurosteroids may be included in the neuroactive steroid family. Therefore, the assessment of the levels of neuroactive steroids directly in male and female brain areas of experimental models of AD might be extremely important. Using our previous experience in other experimental models of neurodegeneration, we intend to evaluate the levels of neuroactive steroids by liquid chromatography tandem mass spectrometry and to confirm possible changes analyzing by real time PCR the gene expression of molecules and enzymes involved in their synthesis. These observations might be very useful to identify possible pharmacological target useful to design sex-specific therapeutic approaches based on neuroactive steroids. Nome Contatti Tel. E mail Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Roberto Maggi 02 50318233 – 3293718540 [email protected] Endocrinology, Physiopatology, Applied Biology Selective Alzheimer Diserase Indicator-1 (Seladin-1): a linker between cholesterol, oxidative stress and Alzheimer’s disease. PRIN – MIUR 2006069900_002 2 anni Alzheimer's disease: correlation with the Gonadotropin Hormone Releasing Hormone (GnRH). Amyloid beta (Abeta) plaques represent one of the primary pathological features that appear in the brains of Alzheimer's patients. Recent findings suggest that the complete proteolytic machinery required for Abeta generation is located within lipid rafts. Membrane cholesterol content is crucial for lipid raft organization and it seems to regulate the membrane localization and the activity of secretases involved in the metabolism of APP. Epidemiological and biochemical studies indicate that all changes in hypothalamic-pituitary gonadal (HPG) hormones (sex steroids, LH, Gonadotropin releasing Hormone or GnRH) levels following menopause/andropause are involved in the cognitive and neuropathological changes observed in AD. Sex steroids have been shown to be potent neuroprotective agents and a decreased incidence and delay in the onset of AD has been described among women on hormone replacement therapy (HRT). Direct roles of GnRH in AD have received scant attention, even if phase III clinical trials are currently underway to establish the role of a GnRH agonist in lowering the incidence of dementia and in improving cognitive functions of AD patients. Recently, it has been discovered that estrogens are able to up-regulate the new gene seladin-1 (Selective Alzheimer disease indicator 1). Seladin1 is a gene down-regulated in the brain areas involved in AD and its overexpression confer protection against Abeta mediated toxicity. Seladin-1 was found to be identical to DHCR24, the enzyme that catalyzes the last step of cholesterol biosynthesis. Therefore seladin-1 could represent a key regulator of membrane cholesterol and it could be the molecular mediator of the neuroprotective effect of estrogens. This project is devoted to study the role of two hormones of the HPG axis, estrogen and GnRH, in AD pathogenesis. Moreover, we will investigate the molecular mechanisms that can link these hormonal actions with the cholesterol content of plasma membrane and with the cholesterol-rich microdomains organization and functionality, and finally the possible role for seladin-1 as a molecular mediator of this biological mechanisms. For this research proposal we will utilize different appropriate humane (FNC-B4 and SHSY5Y) and murine (GT1-7, GN11, and primary hippocampal neurons) cellular models. The experimental plan aims to clarify three points: 1.The involvement of membrane cholesterol and lipid membrane organization in the processing of the APP and its modulation by estrogens. We will understand if estrogen stimulation is able to alter the organization of membrane lipids and proteins and the proteolytic processing of the APP, and if these modifications correlate to changes of the cholesterol content and of seladin-1 expression. 2.The role of GnRH in the modulation of the cytotoxic action of Abeta and the APP processing and in neuronal excitability. We will clarify how GnRH affect APP processing, Abeta deposition and Abeta toxicity and how GnRH modulate excitability in hippocampal neurons. 3.The hormonal regulation of seladin-1, its ability to modulate the HPG hormone action and its involvement in Abeta induced cytotoxicity. We will evaluate the role of seladin-1, as a regulator of membrane cholesterol, in the amyloidogenic pathway, in the amyloid-induced neurotoxicity and in lipid rafts organization and functionality as "signaling platforms". Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto PRIN – MIUR 2006069900_002 PUR – MIUR 2008 2 anni Alzheimer's disease: correlation with the Gonadotropin Hormone Releasing Hormone (GnRH). Amyloid beta (Abeta) plaques represent one of the primary pathological features that appear in the brains of Alzheimer's patients. Recent findings suggest that the complete proteolytic machinery required for Abeta generation is located within lipid rafts. Membrane cholesterol content is crucial for lipid raft organization and it seems to regulate the membrane localization and the activity of secretases involved in the metabolism of APP. Epidemiological and biochemical studies indicate that all changes in hypothalamic-pituitary gonadal (HPG) hormones (sex steroids, LH, Gonadotropin releasing Hormone or GnRH) levels following menopause/andropause are involved in the cognitive and neuropathological changes observed in AD. Sex steroids have been shown to be potent neuroprotective agents and a decreased incidence and delay in the onset of AD has been described among women on hormone replacement therapy (HRT). Direct roles of GnRH in AD have received scant attention, even if phase III clinical trials are currently underway to establish the role of a GnRH agonist in lowering the incidence of dementia and in improving cognitive functions of AD patients. Recently, it has been discovered that estrogens are able to up-regulate the new gene seladin-1 (Selective Alzheimer disease indicator 1). Seladin1 is a gene down-regulated in the brain areas involved in AD and its overexpression confer protection against Abeta mediated toxicity. Seladin-1 was found to be identical to DHCR24, the enzyme that catalyzes the last step of cholesterol biosynthesis. Therefore seladin-1 could represent a key regulator of membrane cholesterol and it could be the molecular mediator of the neuroprotective effect of estrogens. This project is devoted to study the role of two hormones of the HPG axis, estrogen and GnRH, in AD pathogenesis. Moreover, we will investigate the molecular mechanisms that can link these hormonal actions with the cholesterol content of plasma membrane and with the cholesterol-rich microdomains organization and functionality, and finally the possible role for seladin-1 as a molecular mediator of this biological mechanisms. For this research proposal we will utilize different appropriate humane (FNC-B4 and SHSY5Y) and murine (GT1-7, GN11, and primary hippocampal neurons) cellular models. The experimental plan aims to clarify three points: 1.The involvement of membrane cholesterol and lipid membrane organization in the processing of the APP and its modulation by estrogens. We will understand if estrogen stimulation is able to alter the organization of membrane lipids and proteins and the proteolytic processing of the APP, and if these modifications correlate to changes of the cholesterol content and of seladin-1 expression. 2.The role of GnRH in the modulation of the cytotoxic action of Abeta and the APP processing and in neuronal excitability. We will clarify how GnRH affect APP processing, Abeta deposition and Abeta toxicity and how GnRH modulate excitability in hippocampal neurons. 3.The hormonal regulation of seladin-1, its ability to modulate the HPG hormone action and its involvement in Abeta induced cytotoxicity. We will evaluate the role of seladin-1, as a regulator of membrane cholesterol, in the amyloidogenic pathway, in the amyloid-induced neurotoxicity and in lipid rafts organization and functionality as "signaling platforms". Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Laboratory of Biochemistry & Molecular Biology of Lipids - Mass Spectrometry "Giovanni Galli" Donatella Caruso, Maurizio Crestani, Emma De Fabiani, Nico Mitro 0250318344 – 0250318393 [email protected], [email protected], [email protected], [email protected]. Department of Pharmacological Sciences Nuclear receptors, coregulators, neuroactive steroids and epigenetics in neurodegenerative diseases associated to metabolic disorders. 1. MECHANISMS OF NEURODEGENERATION G. Cell interactions Nome Contatti Istituto/Dipartimento Prof. Ubaldo ARMATO, MD [email protected] Histology & Embryology Unit, Dept. of Life and Reproduction Sciences, University of Verona Medical School, Verona, Venetia, I-37134, Italy (in collaboration with Balu Chakravarthy e James F. Whitfield, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ont., K1A 0N6, Canada) Proposta di ricerca The Astrocyte, not just a Bystander in Alzheimer’s Disease Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto - Multidisciplinary projects Basic research -------The Astrocyte, not just a Bystander in Alzheimer’s Disease There are two kinds of Alzheimer’s disease (AD). There is the rare early onset AD (EOAD) which includes less than 5% of the cases and is due to mutation-induced hyperproduction of the Aβ42 (amyloid β-(1-42) fragment of the membrane-associated amyloid precursor protein (APP). Then there is the slowly developing, late-onset AD (LOADAD that includes 95-99% of the cases. The mutant driver genes of EOAD have been relatively easy to identify. But the key LOAD drivers have been far harder to find in the host of changes associated with the progressive breakdown of neuronal-glial circuits starting in the entorhinal cortex and spreading through the hippocampus and into the cerebral hot-spots of the cerebral DMN (default mode neuronal network ) ending in catastrophic cognitive failure with the loss of memory formation by the hippocampus being an outstanding feature. So far the attention of workers in the AD field focused on neurons. But things are changing. Now we know that certain (radial) astrocytes are the stem cells for adult neurogenesis in the hippocampal dentate gyrus and therefore are key components of the memory-forming machinery. We are learning that massive astrocyte syncytial networks connect to, and collaborate with, neurons in sustaining and driving brain functions. And they are co-conspirators with neurons in the cognitive catastrophe of AD. Therefore we have developed a method of culturing normally functioning (minimally altered) astrocytes directly following their removal from adult human cerebral cortices. We are using these cells, which we call NAHAs (normal human astrocytes), to explore the possible roles of astrocytes in AD. There is an ever-escalating list of things that might cause of help cause LOAD. At first it was thought that the striking fibrillar amyloid plaques in AD brains were the neuronal circuit breaking killers. But plaque-rich brains of cognitively normal or even above-normal 90- years old nuns and the failure of plaque formation in transgenic mice to correlate with neuronal and cognitive loss argue that this is probably not true. It is now suspected that it is a build-up of invisible pre-plaque Aβ42 oligomers that start destroying synapses and disconnecting cognitive circuits. We have been and will keep working on three things that seem to be part of the LOAD process: the induction of VEGF production by astrocytes; the expression and function of p75NTR, an Aβ42 and neurotropin receptor. VEGF Production We have so far found that both Aβ42 and a combination of three glial-induced cytokines (IL-1β, IFN-γ and TNF-α) that appear in the inflammatory environment of an AD brain can induce NAHAs to produce and secrete mainlythe VEGF-A165 isoform. Interestingly none of the three cytokines can significantly stimulate VEGF production, but together they are more potent stimulators than Aβ35-42 (an active fragment of Aβ42). We must now find out how Aβ42 (i.e., Aβ35-42) by itself and how the three cytokines together produce the HIF-1α•HIF-1β heterodimer that stimulates VEGF production in the NAHAs. Is VEGF just a minor player in AD? What might VEGF do in an AD brain? The release of angiogenic VEGF from the Aβ42/cytokine ensemble-stimulated astrocytes of the neurobarrier-coupling units with their end-feet attached to blood vessels damaged by deposited Aβ42 would be expected to reverse, or at least minimize, the Aβ42-induced vascular damage widely believed to be responsible for hypoxia, glucose shortages, breached blood-brain barrier and failing Aβ42 drainage. But then it may not. Hypoxic endothelial cells in the hypoperfused regions of an AD brain downregulate their MEOX-2 homeobox gene and its GAX protein product. This causes the endothelial cells to upregulate the AFX1 forkhead transcription factor that downregulates their anti-apoptogenesis Bcl-XL gene by stimulating the BCL-6 transcriptional repressor. The downregulation of MEOX-2 and its GAX protein product also lowers the level of the Aβ42-clearing LRP1. Therefore while the increased VEGF production in an AD brain would indeed stimulate angiogenesis, the new blood vessels would be dysfunctional with endothelial cells unable to clear Aβ1-42 and hyper-prone to apoptogenesis. But VEGF may have another impact on memory formation. It is a major factor in the maintenance of the vascular-based the radial glial (astrocyte-like) neuronal stem cell niche in the dentate gyral subgranular zone, a center of adult neurogenesis. VEGF stimulates the production of the niche’s endothelial cells that produce the neurostimulatory BDNF factor. Thus, angiogenesis is linked to neurogenesis in the niche to continuously supply the new granular cells needed to start the recording the memory of novel polymodal inputs from the entorhinal cortex. Therefore the increased expression of VEGF in a normal brain or injection of VEGF into such a brain might promote the survival and proliferation of neural progenitor cells as indicated by the reported response of hippocampal neuronal progenitor cells to infusion of the protein into the lateral ventricles of adult rats. But this may be unlikely in an AD brain even with its elevated VEGF expression, because it has been reported that the core AD pathology prevents any VEGF-stimulated immature granule cells from differentiating into mature granule cells. p75NTR, the villainous Darth Vader of the AD brain? p75NTR combines with the TrkA neurotrophin receptor to drive brain development when activated by BDNF and NGF. But it fades away after development only to re-emerge in the aging, Aβ42 –accumulating AD brain along with the downregulation of TrkA. Now without restraint by TrkA it becomes dangerous because it has a cytoplasmic apoptogenic DD (‘death domain’) which is turned on when the receptor is activated by Aβ42 binding to its neurotrophin binding site. When its DD is not opposed by associated by TrkA and it meets Aβ42 on a neuronal surface it kills the cell via its DD. We have found that accumulating Aβ42 actually stimulates the expression of p75NTR by neurons and NAHAs. Obviously this could accelerate AD development. Aβ42 and neurotropin receptor. But then there are also indications of a Aβ42/ p75NTR vicious cycle. Activated p75NTR receptors have been shown to stimulate caveolar sphingomyelinase to release ceramide from a pool of sphingomyelin in the cell membrane. The released ceramide post-transcript-ionally stabilizes BACE1 which, by β-cleaving membraneassociated AβPP, accelerates the drive to full-blown AD by increasing Aβ42 production. This Aβ42would then switch back and bind to p75NTR receptors and increase the calpain-driven cleavage of the receptor’s C-terminal membrane-associated domains which ultimately induce the receptors’ cytoplasmic death domains and trigger a cytocidal apoptogenic caspase 8/caspase 3 cascade. We are now trying to confirm the contrapuntal rise and fall of p75NTR and TrkA in a key region of the human AD brain such as the hippocampus. And along with this we are trying to find out with whether NAHAs can start an Aβ42/ p75NTR vicious cycle. If they can, it will be a strong indication that astrocytes have a major role in the cognitive collapse of the AD brain. Nome Contatti Manuela MARCOLI Dipartimento di Medicina Sperimentale (DIMES), Sezione di Farmacologia e Tossicologia – Università degli Studi di Genova Viale Cembrano, 4 - I-16148 Genova, Italy Phone: +39 010 3532656; Fax: +39 010 3993360 E-mail: [email protected] Istituto/Dipartimento Dipartimento di Medicina Sperimentale (DIMES), Sezione di Farmacologia e Tossicologia – Università degli Studi di Genova The research strategy we propose addresses the need for better knowledge of early molecular events leading to neuronal dysfunction in Alzheimer’s disease (AD). We propose to focus on the effects of soluble amyloid-beta peptides (Aβ) on mechanisms sustaining neuron-astrocyte cross-talk at glutamatergic synapses and on network synaptic activity in Multi Electrode Array (MEA)-coupled neuron/astrocyte cultures, in both wild type and triple transgenic AD mouse (3xTg-AD mouse). Combined use of isolated models (nerve terminals and astrocyte processes) and an integrated model (MEA-coupled neuron/astrocyte cultures) will provide complementary data to understand Aβ-related dysruption of glutamatergic transmission in AD. Assessment of changes of synaptic transmission at the level of astrocyte processes and nerve terminals (exposed to Aβ) together with analysis of the effects of (endogenously produced and exogenously added) Aβ on neuron/astrocyte network activity may shed new light on the molecular and cellular mechanisms underlying early dysfunction of glutamatergic transmission in AD, helping to discover new therapeutic targets to improve nerve cell signal transduction and ultimately preventing neuronal dysfunction. Background and rationale: Astrocytes regulate neuron function through nonoverlapping domain organization: one astrocyte processes contact thousands of synapses, promoting neuronal synchrony and regulating synaptic transmission; the neuro/gliotransmitter glutamate mediate bidirectional neuronastrocyte communication. Although altered glutamate transmission and astrocyte global perturbation are reported in AD, scarce attention is focused on the potential role of astrocyte processes in synaptic dysfunction in AD. Soluble Aβ appears involved in synaptic dysfunction; focus on the impact of Aβ on mechanisms sustaining neuron-astrocyte cross-talk at glutamatergic synapses might unravel key steps in synaptic impairment and in the cascade of events causing specific sets of neurons becoming dysfunctional. Experimental models and Methods: Aβ effects on [Ca2+]i and transmitter release will be directly investigated on astrocyte processes (gliosomes) and nerve terminals (synaptosomes) from adult wild type and 3xTg-AD mouse brains. Gliosomes and synaptosomes stem from astrocytes and neurons obtained from adult mouse brain; in situ maturation of neurons and astrocytes would assure that the impact of intracellularly accumulating Aβ would also be accounted for when studying glutamate release and Ca2+ signaling in nerve terminals and astrocyte processes prepared from 3xTg-AD mouse brain. Integrated effects of Aβ on network activity and synaptic connection maturation will be analyzed by non-conventional electrophysiology on cultured neurons/astrocytes from wild type and 3xTg-AD mouse brain on MEAs, allowing long-term, non invasive, multielectrode detection of neuronal network activity and synchronization; the bioelectronic system has proven very sensitive in detecting early neuron damage and loss of synaptic transmission. Human resources: key persons, experience and know-how that can be accessed without additional funds: Manuela Marcoli, Guido Maura (University of Genova, Department of Experimental Medicine DIMES; Center of Excellence for Biomedical Research CEBR; National Institute of Neuroscience INN): transmitter release from CNS preparations (nerve terminals, astrocyte processes) and cell cultures Sergio Martinoia (University of Genoa, Department of Biophysical and Electronic Engineering DIBE; Italian Institute of Technology IIT): Micro electrode arrays technology; MEA-based systems and in-vitro neural interface and neuro-robotics Mario Nobile (National Research Council, Institute of Biophysics, Genova): [Ca2+]I imaging on single nerve terminals and astrocyte processes Area di interesse identificata First challenge: scientific; understanding the mechanisms of the disease Priorites: - Basic research - Multidisciplinary projects - New treatment strategies Finanziamenti ricevuti - ongoing research support Titolo progetto Neuronal differentiation: functional evaluation of neurotransmission Ente finanziatore MIUR – PRIN mod B Anno 2008 - prot. 20088JEHW3_005 (to Guido Maura) Durata progetto 2 years Abstract del progetto To understand the molecular mechanisms associated to the differentiation of neuron precursor cells, we plan to use Neuroblastoma N18TG2 clones, transfected with Choline Acetyl Transferase (ChAT) and overexpressing early growth response factor-1 (EGR-1), that mimic neuron precursor cells. The research program plans to investigate on the possibility to induce a fully developed neuron phenotype in N18TG2 clones transfected with ChAT and/or overexpressing EGR-1, by inhibiting the expression of the Repressor element-1 Silencing Transcription (REST), coding for a transcriptional repressor of neurogenesis. Our analyses will focus on evaluation of the cell ability to communicate through neurotransmitters and regulated exocytosis upon REST knock down; if this occurs the gene expression profile will be determined. The project will reveal gene networks relevant for complete maturation of the neuroblastoma cell line, and hopefully will be of help in understanding the neuron maturation process. Finanziamenti ricevuti - ongoing research support Titolo progetto Mechanisms of glial and neuronal glutamate release as specific therapeutic targets in brain ischemia Ente finanziatore Fondazione Cassa di Risparmio di Genova, 2008 (to Mario Nobile, role of MM: participant) Durata progetto 2 years Abstract del progetto The aim of the project is to get new light on molecular and cellular mechanisms activated during/after ischemia to find out pharmacological targets for neuroprotection. Brain ischemia results from multiple related mechanisms involving both neurons and glial cells. The neuron-glia crosstalk, subserved by mechanisms including glutamate is of fundamental importance to the synapse physiology and to the synaptic transmission dysregulation in neurodegenerative diseases; increased extracellular glutamate and intracellular calcium overload lead to cell death. Multiple mechanisms are responsible for glutamate release: vesicular release, glutamate transporter reversal, and possibly activation of ATP P2X7receptors, of volume-regulated anion channels or of connexin/pannexin. hemichannels. These ways for glutamate release can function both on neurons and astrocytes, although the role for glial cells in ischemic neuron damage is still debated. As the pharmacological intervention based on glutamate ionotropic receptor antagonism was unsatisfactory, knowledge of the glutamate modes of exit during ischemia might help to find new specific therapeutic targets. The research strategy will be based on the investigation at synapse level, at the level of single cell and of the cell network (slices), on the participation of P2X7 receptors, VRAC channels and pannexin in ischemic damage in vitro. As we previously found that adenosine controls astrocyte P2X7 receptors, we will investigate on the relationship between adenosine and P2X7 receptors during ischemic insult. Finanziamenti ricevuti - submitted proposal Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Synapse and Amyloid-beta: effects on astrocyte processes and nerve terminal Alzheimer’s Association 2010 Investigator-Initiated Research Grant Proposal Identifiers LOIID: 173512: approved Full proposal: under evaluation (role of MM: principal investigator) 3 years Understanding of neuron-astrocyte interaction at synapses is crucial to successful therapeutic approach to neurodegenerative diseases. Astrocytes regulate neuronal function through nonoverlapping domain organization: the processes of one astrocyte contact thousands of synapses, promoting neuronal synchrony and regulating neuron excitability and synaptic transmission. The neuro/glio transmitters glutamate and ATP mediate bidirectional neuron-astrocyte communication at synapses. Although altered glutamate or ATP transmission and astrocyte global perturbation are reported in AD models, scarce attention has been focused on the potential role of astrocyte processes in synaptic dysfunction in AD. Soluble amyloid-beta peptides are involved in synaptic dysfunction, an early event in the pathogenesis of AD before synapse loss. Our proposal focus on the effects of soluble amyloid-beta on the mechanisms subserving bidirectional communication between astrocyte processes and nerve terminals. Focus on the impact of amyloid-beta peptides on astrocyte process might unravel key steps in synaptic impairment and in the cascade of events causing specific sets of neurons becoming dysfunctional. How? Astrocyte processes (gliosomes) and nerve terminals (synaptosomes) from wild type and transgenic AD mouse brains will be used, allowing direct investigation on the amyloid-beta effects on intracellular calcium and transmitter release. It is worth noting that gliosomes and synaptosomes stem from astrocytes and neurons obtained from adult mouse brain; in situ maturation of gliosome-producing astrocytes and of synaptosomeproducing neurons provides experimental models suitable to investigate on amyloid-beta effects in adult life. Integrated effects of amyloid-beta on network activity and synaptic connection maturation will be analyzed on co-cultured neurons/astrocytes from wild type and transgenic AD mouse brain on multielectrode arrays, by non-conventional electrophysiology (allowing long-term, non invasive, multielectrode detection of spontaneous and evoked firing activity and neuronal network synchronization). This bioelectronic system has proven very sensitive in detecting early neuronal damage and loss of synaptic transmission. Why? Assessment of changes of synaptic transmission at the level of astrocyte processes and nerve terminals (exposed to amyloid-beta) together with analysis of the amyloid-beta effects on neuron/astrocyte network activity may shed new light on the molecular and cellular mechanisms underlying early neurodegenerative processes and cognitive dysfunction in AD, helping to discover new therapeutic targets to improve nerve cell signal transduction and ultimately preventing neuronal dysfunction and synapse loss. Finanziamenti ricevuti - completed research support Titolo progetto Feasibility of a prototypical system development for in vitro testing for biomedical, toxicological and ambiental purpose Ente finanziatore Scientific and Technological Park of Liguria Program Objective 2 20002006 (to Sergio Martinoia, role of MM: responsible of Unit) Durata progetto 2 years Abstract del progetto The goal of this project was to set up neuron cultures on multielectrode arrays allowing neuropharmacological/neurotoxicological analysis of the integrated effects of neuroactive substances on network activity Combining the study of the network firing activity with the study of the release of the excitatory transmitter glutamate and of the activation of the intracellular pathways following ionotropic receptor (NMDA, AMPA/kainate) activation and involved in the excitotoxic neuron cell damage or death would allow an integrated mechanistic-based approach to neurotoxicity evaluatio in neuron network. Non conventional electrophysiological and neurochemical approach allow neuropharmacological /neurotoxicological analysis of the integrated effects of neuroactive substances on the network activity and dissection of the mechanisms underlying complex network responses to the substances. Finanziamenti ricevuti - completed research support Titolo progetto Pharmacological control of extracellular excitatory amino acid accumulation and cellular calcium overload in neonatal ischemia/reperfusion Ente finanziatore Mariani Foundation (Milan) R-07-66 (to Mario Nobile, role of MM: participant) Durata progetto 2 years Abstract del progetto The study evaluates acute and long-term (60 days) effects of perinatal hypoxia, in order to find new pharmacological targets to be used in intervention devoted to prevention/protection against neonatal ischemic neuron damage. Perinatal hypoxia-ischemia is a major cause of neonatal morbidity and mortality. A number of evidences indicate that ischemic neuron damage can depend on increase of extracellular K+ accumulation of excitatory aminoacids and of other transmitter (such as adenosine/ATP), and intracellular Ca2+ overload, triggering excitotoxicity and apoptotic/necrotic nerodegeneration. Such events might play different roles depending on the developmental stage of central nervous system. Scarce reports are available on specific models, allowing dissection of diverse mechanisms possibly involved and temporal sequences of neonatal ischemia. Relative contributions of neurons and glia and of their bi-directional communication in the control of glutamate synaptic level and in excitotoxicity triggering and execution, are not clearly defined. Knowledge of the modes for efflux of excitatory aminoacid and their regulation, in early and late phases of ischemia/reperfusion, could open the possibility to reduce the extracellular excitatory aminoacid levels in different neonatal ischemia phases. As intracellular Ca2+ accumulation seems prerequisite for neuron damage cascade, dampening of Ca2+ influx through ionotropic glutamate or purinergic receptors (e.g. P2X7) could significantly reduce neuron damage. We therefore propose investigation on glutamate efflux and cellular Ca2+ levels in physiological conditions and during ischemia in vitro models of neonatal brain (preparations from neonatal and 60 days-old rats, models for premature infant and adulthood brain, respectively). Parallel studies will be conduced with the aim to unravel whether glutamate release from astroglial cells can be modulated by endogenous signaling molecules through the regulation of swelling-activated Clchannels. New pharmacological targets to be used in intervention devoted to prevention/protection against neonatal ischemic neuron damage, will be searched for through the following intermediate steps: better knowledge of the functional role played by ionotropic glutamate and purine receptors on neuron and glial cells; - better knowledge of the modes responsible for extracellular glutamate accumulation and of their relative importance in the temporal sequence of events during ischemia/reperfusion; -better knowledge of the mechanisms regulating activity of swelling-activated Cl- channels involved in the pathologic release of excitatory amino acids; - identification of receptors whose activation (or blockade) might affect vesicular excitatory aminoacid efflux or glutamatergic pathways or neuron-glia activation in different phases of ischemia/reperfusion. Finanziamenti ricevuti - completed research support Titolo progetto Neuron networks coupled in vitro to microtransducers and artificial body: a new potent device to study neuronal interfaces and cerebral plasticity in vitro Ente finanziatore Durata progetto Abstract del progetto Italian Ministry of University and Scientific Research (FIRB 2002) RBAU012KF8_005 (to Guido Maura, role of MM: participant) 3 years The goal of the project was to investigate on spontaneous and electrically-evoked activity of neuronal networks on microtransducer arrays for a better knowledge of the cell mechanisms underlying burst generation and propagation. To this end, non conventional electrophysiology on MEA will be applied to detect the effects of chemical interference on neuron-specific features linked to neuron functioning (electrophysiological events at level of single cells and at the network level) and will be combined with neurochemical analysis for neuron-specific features linked to neuron functioning (neurotransmitter processing and release; presence and functioning of neurotransmitter receptors and linkage to transduction mechanisms) combined with the electrophysiological analysis. Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Domenici Luciano [email protected]; [email protected] Neuroscience Institute, CNR, Via G. Moruzzi 1, 56100 Pisa; Dep. STB, University of L’Aquila, L’Aquila. Alzheimer’s disease, Ageing Active projects: 1) RAGE, MAP kinases and Abeta induced synaptic dysfunction. 2) From brain ischemia to beta amyloid toxicity in Alzheimer’s disease; neurobiological mechanisms, new biomarkers and therapeutical approach. Submitted projects: - Vascular triggering of beta amyloid toxicity in AD. - BDNF and beta amyloid toxicity, building a bridge from neurobiological mechanisms to treatment of brain disorders 1) American Health Assistance Foundation (AHAF). 2) Regione Toscana, Regional Health Research Program 2009 (first step approval passed, final approval and funding pending). 1) Two years. 2) Two years. 1. Abstract project AHAF. Beta amyloid protein (Aβ) is the principal component of senile plaques, a neurophatological prominent feature of Alzheimer's disease (AD). Increasing evidence suggests that loss of neuronal function and cognitive impairment precede the accumulation of Aβ plaques in AD. We hypothesize that oligomeric Aβ is involved in neuronal dysfunction during an early phase of AD. We will investigate the mechanisms underlying oligomeric Aβ induced synaptic dysfunction in entorhinal cortex (EC) whose impairment contributes to cognitive decline in AD. We will address three specific aims: 1. To test the hypothesis that Aβ affects cortical synaptic plasticity. The effects of different concentrations of oligomeric Aβ on synaptic transmission and efficacy will be investigated in EC slices using extracellular and single cell recordings. 2. To verify the identity of receptors and intracellular pathways transducing Aβ signal. Receptor for Advanced Glycation Endproducts (RAGE) is a cell surface binding site for Aβ. We will investigate the effects of oligomeric Aβ on synaptic transmission and efficacy in EC slices: i) in the presence of anti-RAGE antibodies, ii) in the transgenic (Tg) mice with targeted neuronal expression of the wild type form of RAGE and in RAGEdeficient mice; iii) in the Tg mice with a dominant negative form of RAGE targeted to neurons (DN-RAGE). Finally, we will investigate the involvement of different kinases associated with RAGE activation, such as p38 MAPK, p42-p44 MAPK and JNK. 3. To test the role of neuronal RAGE in vivo using animal models of Aβmediated synaptic dysfunction. We will employ Tg mice overexpressing mutant human amyloid precursor protein (mAPP); Aβ-rich environment in Tg mAPP mice induces synaptic dysfunction. We expect that synaptic dysfunction observed in Tg mAPP mice does not occur in Tg mAPP mice crossed with Tg DN-RAGE mice. Finally, we will determine whether increased expression of neuronal RAGE in Tg mAPP mice exaggerates synaptic dysfunction. 2. Abstract Project Regione Toscana. Alzheimer’s disease (AD) is a progressive neurodegenerative disorder of the elderly characterized by senile plaques composed of beta amyloid (Aβ), neurofibrillary tangles, synaptic loss and impairment of cognitive functions. AD is the most common cause of dementia followed by vascular dementia (VaD). The incidence of neurodegenerative disorders in AD is increased by vascular pathologies such as stroke, clogging arteries and hypertension inducing brain hypoperfusion and hypoxia. However, a causal relationship between AD and brain ischemia at the molecular level has not been established. The present project will contribute to elucidate whether Aβ/RAGE signalling may be viewed as part of a neural system that controls synaptic responses in vascular pathology and AD. We are interested in investigating this issue as many patients with AD also have hypertension and vascular infarctions. Designing new biomarkers of neurodegeneration and interfering with Aβ/RAGE signalling is expected to have high impact to ameliorate cellular dysfunction in AD and vascular pathology. Patentability of a new diagnostic method of AD based on biomarkers associated with RAGE activation and deposition of Aβ represents the goal of our plan. Nome Evelina Chieregatti Contatti [email protected] Istituto/Dipartimento Dept. of Neuroscience and Brain Technologies, IIT, Genoa, Italy Proposta di ricerca Synaptic deficits in neurodegenerative diseases The main lines of our research concern the investigation of the early mechanisms underlying synaptic dysfunctions in Parkinson’s and Alzheimer diseases. These neurodegenerative diseases are characterized by the deposition of proteinacious aggregates in the neurons and in the brain tissue. However it has been shown that most neurons that undergo cell death do not contain aggregates and that, whether they contain aggregates or not, are similarly affected by morphological dendritic abnormalities or biochemical changes. It may thus be conceivable that, rather than being caused by the accumulation of aggregates with neuronal cell bodies, which could be an epiphenomenon, neurodegenerative diseases may be characterized by dying back mechanisms that begin at the synapse and lead to axonal degeneration. Both β amyloid (Aβ) and α-synuclein (Syn) are pathogenetic proteins that upon mutations or increase in their levels may induce alterations in the synaptic function and neuronal cell death. A key element in the regulation of neurotransmitter release and in the fine tuning of synaptic efficacy is the neuronal cytoskeleton. Both proteins are implicated in actin dynamics as well as microtubule stability through their interactions with a microtubule-binding protein, tau. We recently demonstrated a disrupting effect of the mutated form of Syn on actin dynamics in neurons. We are studying their effects on the morphology and dynamics of actin microfilaments and intermediate filaments by the use of purified proteins and neurons derived from knock-out and transgenic mice. The long term goal of this project is to investigate the effect of the two proteins on cytoskeletal-regulated processes, such as polarization, regeneration after axotomy and neuronal plasticity. Another project concerns the characterization of the molecular interactors of Syn that may have a role in neurodegeneration associated with αsynucleopathies. The intracellular localization of the cleavage of APP by BACE1 has long been a topic of debate. Early endosome is the first site of APP endocytic sorting, where APP has been shown to co-localize with BACE1. Altered endosomal structures have been found in AD brains. Our aim is to identify proteins of interest such as enzymes or molecular motors that may be found in the APP-containing endocytic vesicles. It is expected that the results will shed light on the role of Syn and Aβ in synaptic function, and on the molecular interactors mediating their effect, providing cues useful to reach appropriate therapeutic endpoints based on the modulation of the signalling pathways involved in the pathogenic effect of Syn and Aβ. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Macromolecular interactions and Neurodegeneration The role of intraneuronal amyloid β(Aβ) peptide on the pathogenesis of the Alzheimer disease: functional and proteomic analysis. Fondazione CARIPLO – role: Partner 2 years (end 2009) Alzheimer disease (AD), one of the neurodegenerative diseases of the nervous system characterized by a chronic process of cell death, is the most frequent form of dementia, but there is still no therapeutic intervention capable of preventing, delaying or treating it. Intraneuronal Aβ accumulation precedes the deposition of amyloid plaques and the appearance of neurofibrillary tangles, and correlate with the first appearance of cognitive deficits. The role of intraneuronal Aβ in the AD pathogenesis is still unclear, but recent data suggest a possible involvement in cytoskeletal functions and neurotransmitter release. The goal of this project is to understand how Aβ may interfere with synaptic function and, by the use of advanced proteomic techniques, to identify the proteins that interact with Aβ. The results we expect to obtain will shed light on the pathogenetic mechanisms in AD and will allow the development of advanced diagnostic and therapeutic protocols. Titolo progetto Pathogenetic mechanisms of familial Parkinson’s disease: wt and A30P α-synucleins affect the structure of microfilaments and intermediate filaments. Pathways and effects on cytoskeletal dynamics. Ente finanziatore Durata progetto Abstract del progetto Telethon-Italy – role: PI 3 years (end 2013) Overall objectives. To understand a physiological role of alpha-synuclein (Syn) and its role in the pathology of Parkinson's disease (PD) by comparative studies of wt and A30P Syn mutant interaction with microfilaments and intermediate filaments. To understand Syn role in cellular processes that require active remodeling of these two cytoskeletal elements. Specific aims. Analysis of the regulation of actin-Syn interaction in living cells by fluorescence energy transfer (FRET). Analysis of the composition of actin/A30P Syn aggregates formed during cytoskeleton remodeling. Analysis of Syn effects on intermediate filaments turnover and on neuronal regeneration after axotomy. Investigation of the pathway(s) that mediates the effect of extracellular Syn on actin cytoskeleton morphology. Background. Mutations in the Syn gene are responsible for familial PD. Syn knock-out mice suggest a role of Syn in the regulation of the presynaptic vesicular pools in neurons. Syn has been involved in neuronal regeneration occurring during differentiation and repair. Interestingly, actin microfilaments and the intermediate filaments are actively involved in every one of these processes. Description of the project. Based on our data demonstrating interaction between Syn and actin we propose to study the regulation and the subcellular localization of their interaction by FRET. In neurons in culture from knock-out and from conditional transgenic mice we plan to investigate the role of Syn in regeneration after axonal injury. We propose to investigate the transduction pathway that is activated by extracellular Syn. Anticipated output. We expect to identify the specific step at which Syn function at the pre-synapse and the possible loss of function of the A30P Syn mutant. These results will be of great relevance in the understanding of PD pathogenesis. 1. MECHANISMS OF NEURODEGENERATION H- Intracellular Traffiking Nome Contatti Istituto/Dipartimento Cecilia Bucci Tel. +39-0832-298900; Fax +39-0832-298626 e-mail: [email protected] Department of Biological and Envirnmental Sciences and Technologies (DiSTeBA), University of Salento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Membrane traffic and neurodegenerative diseases: role of Rab proteins and their effectors The laboratory of Applied Biology directed by Cecilia Bucci is focused on the role of endocytic Rab proteins. Rab proteins are small GTPases that control membrane traffic in the endocytic and exocytic pathways. Indeed, they are responsible, through the recruitment of several specific effectors, for several aspects of membrane transport as vesicle formation, vesicle movement onto cytoskeletal tracks, vesicle docking and tethering to the target compartment, and, finally, vesicle fusion with the target compartment. There are about 70 rab genes in the human genome. Alterations of membrane traffic, and, in particolar, of Rab proteins or of their effector proteins, are responsible for many human genetic and acquired diseases including cancer. In particular, Rab dysfunction has been linked to several neurodegenerative diseases. For instance mutations in Rab7 protein cause the Charcot-Marie-Tooth type 2B disease, a inherited neuropathy of the peripheral nervous system. Late endocytic dysfunction has been demonstrated in Alzheimer’s disease and presenilin (a protein mutated in Alzheimer) interacts with the endocytic Rab11 and with the Rab regulatory protein GDI (GDP-dissociation inhibitor). In addition, presenilin influences localization and function of at least other two Rab proteins, Rab6 and Rab8. Rab6 expression is increased in Alzheimer’s disease brain. Mutations in alpha-synuclein are a cause of familial Parkinson disease; mutated alphasynuclein interacts abnormally with Rab3a, Rab5 and Rab8. These are only some examples that demonstrate the close connection between membrane traffic (and in particular Rab proteins function) and neurodegenerative diseases. We propose therefore to study different aspects of membrane traffic at the molecular level, with a particular focus on Rab proteins, in order to understand the molecular mechanism underlying degenerative diseases as Alzheimer, Parkinson, Charcot-Marie-Tooth, etc. In particular, we could investigate the role of the proteins found mutated in these diseases (i.e. presenilin, alpha-synuclein, Rab7, etc.) in membrane traffic, and the role of their interaction with regulators of membrane traffic. Molecular basis of Charcot-Marie-Tooth type 2B disease Telethon Italy 3 years (2009-2012) The inherited neuropathies of the peripheral nervous system show clinical and genetic heterogeneity. CharcotMarieTooth type 2B (CMT2B) is a ulceromutilating neuropathy clinically characterized by prominent sensory loss, marked distal muscle weakness and wasting, high frequency of foot ulcers, infections, and amputations of the toes because of recurrent infections. Four missense mutations in the small GTPase late endosomal protein Rab7 have been identified in autosomal dominant ulceromutilating neuropathy families. These missense mutations cause the CMT2B phenotype and target highly conserved amino acid residues. It is not known how these mutations affect specifically peripheral neurons leading to an axonal pathology in CMT2B. This issue is particularly challenging considering that Rab7 is a ubiquitous protein. This proposal plans to continue investigating the molecular mechanism responsible for the development of the disease. We have previously biochemically characterized the Rab7 mutated proteins causative of CMT2B in HeLa cells, determining that they are mechanistically similar. Indeed, we have demonstrated that activated forms of Rab7 are responsible for the disease.Within this project we plan to: i) do functional studies using the mutated proteins on PC12, SH−SY5Y, Neuro2a cells and neurons ii) screen DRG and motor neuron cDNA libraries with different methods to search for new Rab7 interacting proteins, looking in particular at effectors expressed specifically in the peripheral nervous system. iii) do functional assays to establish the role of these newly identified Rab7 interacting proteins and to establish if and how Rab7 mutations affect their function. Understanding how dysfunction of Rab7 causes CMT2B will be instrumental to gain insights in the pathogenesis of this group of disorders, and will open the way to future identification of therapeutic approaches. Nome Contatti Istituto/Dipartimento Prof. Paolo Macchi, PhD Phone: +39 0461-883819/883095 Fax: +39 0461-883937 Email: [email protected] Molecular and Cellular Neurobiology Lab CIBIO – Centre for Integrative Biology, University of Trento. Via delle Regole 101 3060 Mattarello, Trento – Italy WEB: http://www.unitn.it/cibio Research proposal “Comparative analysis of the transport and translational RNPs complexes in neurons: an integrate transcript- and proteomic approach.” RNA localization is emerging as an important process to restrict certain proteins to specific subcellular domains and thus spatially control the expression of genes within cells. One key feature of messenger RNA (mRNA) localization is that it precedes translation. As a consequence, mRNA has to be kept translationally silent during its transport towards the proper target compartment. Localized mRNAs are packaged into ribonucleoprotein particles (RNPs), composed of RNAs and proteins. The localized RNAs contain specific cis-acting sequence elements, which are recognized by proteins, called trans-acting factors. The importance of RNA subcellular localization in the formation and function of the nervous system is also gaining increasing acceptance. In highly polarized cells, such as in neurons, a functional compartmentalization is crucial. RNA localization is not only involved in establishing the structural polarity of neurons, but is also assumed to play an important role in synaptic plasticity1. It has been estimated that approximately 200 different mRNAs encoding for proteins important for synaptic plasticity as well as various housekeeping genes, are found in dendrites of hippocampal neurons2. Interestingly, the loss-offunction of one component of the RNA transport machinery and/or the misexpression of specific mRNAs causes abnormalities during neuronal development. For example, defects in axonal and dendritic outgrowth and/or in dendritic spine development have been described. These phenotypes have recently been correlated with different neurological disorders, such as FXS (fragile X mental retardation syndrome), SCA (spinocerebellar ataxia) and SMA (spinal muscular atrophy)3. However, the composition of the transport as well as the translation control machinery is still poorly characterized. Our research focuses on dissecting the molecular composition of the neuronal transport RNPs under physiological as well as pathological conditions (e.g. neurodegeneration, brain tumours and epilepsy). In detail, we are trying to answer the following biological questions: 1 2 Is the local protein synthesis and/or mRNA trafficking altered in neurons undergoing degeneration? What is the molecular composition of the neuronal RNPs (RNA and protein interactome) in neurons affected by degeneration compared to normal cells? The goal of purifying transport RNPs to identify novel components has been previously attempted, but has not yielded the precise mechanistic details of the process that are sought. Our laboratory has successfully demonstrated the feasibility of maintaining endogenous transport complexes containing Staufen, Barentsz and Pumilio proteins, RNA (RIP-chip analysis), and motor proteins in cell-free extracts of brains and cultured neurons4,5. Using our collection of affinity-purified antibodies to immunoprecipitate localization RNPs from brain extract fractions, the composition of RNPs isolated from wild type mice will be assessed and compared with RNPs isolated from animal model for neurological diseases. We will then test the individual functions of both known (or suspected) and novel identified components of neuronal RNPs in a variety of in vivo and in vitro assays to characterize their roles in localization and associated mechanisms. Finally, since the activation of translation of dendritically localized mRNAs upon specific inputs, such as synaptic activity, allows for the modulation of existing synapses, notably the morphological and functional changing of dendritic spines, 3 Are there possible drugs able to affect mRNA targeting (and/or turnover) and local protein synthesis that can be used to block neurodegeneration? We are currently setting up a high throughput drug screening assay on neuronal cells at CIBIO (University of Trento). RIP-Chip experiments will be functional for the development of pharmacological assays where the level of expression of mRNAs identified will be evaluated through the use of a reporter protein (luciferase). Through a bioluminescence assay we will be able to quantify the effect of each chemical's inhibitory activity or promoter of the complex proteins/ mRNA-luciferase. We propose that a multidisciplinary study to underline the mechanisms that lead to correct regulation of both RNA and RNA-binding proteins will be crucial for a better understanding of the pathogenesis of neuronal diseases. Interdisciplinary internal collaborations, with other institutions in Italy and abroad have been already established to take full advantage of different experimental systems and areas of expertise. 1. Kuhl D. & Skehel P. (1998) Curr. Opin. Neurobiol. 8, 600-606. 2. Job C. & Eberwine J. (2001) Nat. Rev. Neuroscie. 2, 889-898. 3. Dahm R. & Macchi P. (2007). Biol. Cell 99, 649-61. 4. Mallardo M. et al., (2003). PNAS 100, 2100-2105 5. Vessey J. et al., (2010). PNAS 107, 3222-3227 Area di interesse identificata Basic research. Neurodegenerative diseases. Finanziamenti ricevuti Titolo progetto Electrophysiological characterization of alpha-synuclein forming channels, a protein involved in the neurodegeneration of Parkinson's disease Ente finanziatore CARITRO Foundation Durata progetto 2 years Abstract del progetto The aggregation of aberrant proteins is involved in the pathogenesis of neurodegenerative diseases associated with aging but the mechanisms leading to neuronal death are unknown. The α-synuclein (αS) is the main component of Lewy bodies, namely fibrillar intraneuronal inclusions present in all patients with Parkinson's disease (PD), and is strongly indicated as the cause of the disease. The αS is not structured in a "native" form while in vitro form amyloid fibrils that are rich in beta sheet. Nell'αS mutations found in patients with PD promote the formation of annular and tubular structures of protofibrille. The protofibrille of αS, but not the monomer or mature fibrils, bind to structures such as lipid vesicles, planar and cellular membranes. Here, they assume a new structural conformation similar to the pores formed by the bacterial toxins that cross the membrane. These "amyloid pores" may be the cause of neuronal death, through an uncontrolled and nonspecific permeabilization of the cell membrane. We intend to investigate in detail the electrophysiological characteristics of the pores of αS formed in model systems of increasing complexity: in lipid planar membranes, synaptic vesicles from liposomes and/or mitochondria and eventually in cultured neurons. In particular an innovative analysis technique suitable for noise characterization of pores will be developed. We intend to investigate, with techniques of analysis of noise, EEG from healthy patients with PD and to highlight similarities with anomalies recorded in single cells. We believe that this multidisciplinary approach will allow 'early diagnosis and more precise onset and evolution of PD as well as of other neurodegenerative diseases. 1. MECHANISMS OF NEURODEGENERATION I- Animal models Nome Contatti Istituto/Dipartimento Adalberto MERIGHI Phone:++390116709118 Fax: ++390112369118 e-mail: [email protected] Istituto Nazionale di Neuroscienze e Dipartimento di Morfofisiologia Veterinaria, università degli Studi di Torino Via Leonardo da Vinci 44 I-10095 GRUGLIASCO (TO) Italy Proposta di ricerca Effects of over-expression of the anti-apoptotic proteins BCL-2 and survivin on the endoplasmic reticulum stress induced by β amyloid: study of neuroprotection on animal models Area di interesse identificata Basic research Developing competitive animal models to study Alzheimer’s disease The etiology and mechanisms responsible for neurodegeneration in Alzheimer’s disease (AD) are multifaceted, but recent studies indicate that activation of an intrinsic gene-regulated program of cell death (PCD = programmed cell death) is a common end-point of the various intracellular pathways leading to neuronal loss. Multiple intracellular pathways activated during PCD, but, among them, apoptosis plays a major role. The post-natal cerebellar cortex is an ideal model for the study of PCD, because of the massive apoptosis of the granule cells, a population of cerebellar excitatory interneurons formed by billions of cells (see for example Lossi L. and Merighi A. Prog Neurobiol. 2003. 69:287-312). β-amyloid (Aβ) is directly involved in neurodegeneration observed in AD, where neurodegenerative changes are due, at least in part, to activation of an apoptotic programme (see for example Wisniewski T. Neurobiol Dis. 1997. 4:313-28). In primary cortical neuron cultures, the toxic effects of synthetic peptides corresponding to specific sequences of Aβ have been shown to be consequent to release of calcium across ryanodine receptor (RyR) channels and/or inositol-3-phosphate channels (IP3) on the membrane of the endoplasmic reticulum (Ferreiro E. Neurobiol Dis. 2006. 23:669-78). We have recently demonstrated that the block of RyR channels in organotypic cultures obtained from mouse cerebellum interferes with the increase in density of cerebellar granule cells expressing the BCL-2 protein (one of the most widely characterized antiapoptotic factors in Eukaryotic cells) fused with a fluorescent reporter marker (EYFP = enhanced yellow fluorescent protein) after KCl depolarization (Lossi et al. Dev Neurobiol. 2009. 69:855-870). These observations suggest that BCL-2 could modulate the apoptotic program triggered by Aβ as a consequence of endoplasmic reticulum stress. Survivin is one member of a family of proteins commonly referred to as apoptosis inhibiting proteins. It is expressed during mitosis, and in tumors, but also during normal development of neurons in vivo, as demonstrated in conditional transgenic mice using the Cre-loxP technology (Jiang Y. et al. J Neurosci. 2005. 25:6962-70). In these animals the conditional deletion of the survivin gene at P10.5 leads to massive apoptosis in several areas of the central nervous system. As a consequence, newborn mutants display a marked reduction of brain size, with severe multifocal apoptosis affecting the cerebral and cerebellar cortices, the brainstem , the spinal cord and the retina. Caspase 3 and 9 (the main effector molecules of apoptosis)activities are increased, whereas expression of the anti-apoptotic protein BAX is unaltered. This project consists of two phases carried out in sequence. The first phase will consist of: - Pharmacological manipulation of organotypic cultures by addition to culture medium of Aβ peptide fragments; - Qualitative and quantitative evaluation of the effects the fragments after labeling of live/dead cells by fluorescent probes (Syto10/Dead Red, Invitrogen, USA or iodidium propide); - Immunocytochemical analysis of apoptosis with special reference to activation of caspases/PARP1 and cell cycle pathways (retinoblastoma protein, chk1, cdc2) - Analysis of relationship between proliferation and apoptosis following visualization of proliferating cells with immunofluorescence (H3 histone, BrdU) and apoptotic cells with the TUNEL technique. In parallel cleavage of apoptosis related proteins will be analyzed by Western blotting. The second phase of the project will consist of: - Evaluation of the effects of Aβ after biolistic transfection of cultures with the following vectors: ApoAlert Caspase 3 sensor (Clontech, USA) to monitor the activation of caspase 3 in alive cells; pEYFP-N1 (Clontech, USA) with insertion of the cDNA for human BCL-2; pHcRed-C1 (Clontech, USA) with insertion of the cDNa for survivin (Obtained from Dr. Rachel Altura, Ohio, USA); - Analysis of apoptosis and cell cycle kinetics in transfected cells transfection (as for control cultures above); - Analysis of cell calcium dynamics by real time calcium imaging. Specific experimental procedures Gene gun technology: We will employ a biolistic transfection procedure (gene gun) to artificially introduce one or more genes of interest in tissue slices. The procedure involves shooting of slices with gold microcarriers coated with cDNAs of different constructs encoding for molecule(s) under study and for a reporter protein (GFP/EYFP/DsRed). Gold microcarriers will be accelerated into tissue by a helium blast (180 psi) so that they will penetrate cell membranes and reach the cytoplasm of target cells. Transfection will be performed after 2-5 days in vitro, by a commercial apparatus (Helios Gene Gun System, BioRad, USA). Confocal microscopy: Control and transfected cultures will be incubated inside a custom made microscope stage incubator (Tokai Hit, Japan) and monitored at variable intervals of time under a Leica SP5 laser confocal microscope. We have already verified that by this set up it will be possible to monitor the cultures for 4 days or longer under the microscope, and it will be thus possible to study short to medium term effects on apoptosis. In certain experiments a cell permeant calcium indicator will be used (Oregon Green Bapta 1AM, Invitrogen, USA)to monitor the intracellular calcium concentration in individual neurons. Visualization of proliferating cells: a technique based upon incorporation of bromodeoxyuridine (BrdU) during S phase will be used by adding BrdU directly to culture medium. M and G2 phase cells will be labeled by immunocytochemistry using a primary antibody directed against phosphorylated H3 histone. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Effects of over-expression of the anti-apoptotic proteins BCL-2 and survivin on the endoplasmic reticulum stress induced by prion protein and βamyloid: study of neuroprotection on animal models MIUR PRIN 2007 (to Laura Lossi) 24 mesi Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Several neurodegenerative diseases are characterized by an initial endoplasmic reticulum stress followed by the activation of an apoptotic program leading to cell death and neuronal loss. The two main goals of this project are: - analysis of apoptosis induced by prion protein (PrP) e da β-amyloid (Aβ) in an ex vivo model consisting of murine organotypic cerebellar cultures obtained from P7-P20 mice. This developmental stage has been proved to be particularly suitable to our purposes since it is characterized by a massive apoptotic neuronal death. - analysis of potentially neuroprotective effects of two anti-apoptotic proteins (BCL-2 and survivin) onto neuronal death induced by PrP and Aβ. The project has duration of 24 months, and is organized into two phases, each corresponding to one of the two main objectives outlined above. We will employ a multidisciplinary approach consisting of in vitro culturing techniques, biolistic transfection (gene-gun), confocal microscopy (medium to long term analysis of transfected cell, reat time imaging of calcium influx), in situ immunocytochemistry and molecular biology (immunofluorescence, labeling of proliferating/apoptotic cells), biochemistry (Western blotting). All these procedure will eventually aim to obtain a direct correlation between morphological and functional data. We expect to obtain a better characterization of the basic biology of PrP and Aβ in relation to modulation of programmed cell death. Development of an in vitro system to study the neuroprotective effects of ghrelin and its derivatives on central neurons MIUR PRIN 2008 24 mesi Background and rationale: Ghrelin is a peptide hormone produced by the endocrine cells of the gastric mucosa. Its most relevant biological action is the regulation of feeding behavior in the presence of a negative energetic balance. After discovery at periphery, the hypothalamus has been identified as the main source of ghrelin in the central nervous system (CNS). Recent studies show that the hormone has indeed a wide range of biological effects in central neurons (see Ferrini et al. 2009, Current Neuropharmacology. 7:37). Among these, we have recently characterized the role of ghrelin in spinal cord pain neurotransmission (Vergnano et al. 2008. Endocrinology. 149:2306). However, data have been obtained in support to the notion that ghrelin also plays a role in neuronal proliferation ,differentiation, protection against apoptosis and maintenance of a differentiated status (see Chung et al. 2007. Endocrinology. 148:148). To understand whether or not the effects of ghrelin in cultured cells are indeed of relevance in vivo it is necessary to respond to the following question: is the hormone produced in the stomach capable to cross the blood brain barrier? The importance of this question appears immediately obvious if one considers that the physiological levels of ghrelin expression in CNS are relatively low. General layout and timetable of project: This is a two year project. During the first year we will first test the antiapoptotic effects of ghrelin and its derivatives on organotypic cerebellar slices. Then we will develop an in vitro model based on our previous experience (postnatal cerebellar slices cultured in vitro under static conditions - see Lossi et al. 2009. Prog Neurobiol. 88:221) where the integrity of the blood brain barriers is maintained. This model will be derived according to a recently developed protocol (Bendfeldt et al. 2007. J Neurosci. 27:3260) where it was demonstrated that the cultivation of cortical slices in the presence of FGF2 (basic fibroblast growth factor 2) is ideal to the preservation of the blood brain barrier. During the second year of the project we will assess the effects of ghrelin on cell death in relation to integrity of the blood brain barrier. Aims and methods: The project focuses on the study of the antiapoptotic effects of ghrelin in a widely established model of developmental cell death, the post-natal cerebellar cortex. The effects of the hormone will be compared in the presence/absence of the blood brain barrier to understand whether or not peripheral ghrelin is capable to influence the survival of central neurons. To do so we will use a multidisciplinary approach consisting of in vitro cultures, cell transfection (gene-gun), confocal microscopy (mid-to-long term analysis of cell calcium fluxes), immunocytochemistry, molecular biology in situ (immunofluorescence, labeling of proliferating/dead cells), and biochemistry (Western blotting). The concurrent employment of all these procedures will allow obtaining a direct correlation between morphological and functional data. Project phases: 1. Development of FGF2 stimulated cerebellar organotypic cultures and assessment of integrity of the blood brain barrier. 2. Pharmacological manipulation of cultures with ghrelin, agonists and antagonists. 3. Analysis of cell death by means of fluorescent probes (Syto 10/Dead Red, Invitrogen, USA). 4. Immunocytochemical analysis of the molecules involved in the apoptotic cascade (retinoblastoma protein, cell cycle-related kinases chk1, cdc2), 5. Analysis of the correlation between apoptosis and cell proliferation (after labeling with fluorescent probes and/or specific markers). 6. Western blotting of relevant proteins to study their cleavage in the course of cell death. Nome Contatti Monica DiLuca Universita’ degli Studi di Milano Via Balzaretti, 9 02 50318374 [email protected] Istituto/Dipartimento Proposta di ricerca Area di interesse identificata CEND and Dept of Pharmacological Sciences Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Synaptic dysfunction in neurodegeneration: characterization of gene products in functional synaptic networks under physiologicaò and pathological conditions. This includes characterization of mechanisms involved in subsynaptic targeting of proteins and protein complexes. These studies will allow for identificationa nd validation of targets as molecules responsible for synaptopathies using pharmacological approaches, siRNA, mimetic peptides and small synthetic molecules. In addition, these approaches will lead to model stages of neurodegenaration in vivo by obtaining innovative animal models for Alzheimer diseases and other neurodegenerative disorders. Role of calcium dependent gene expression and protein trafficking in shaping post synaptic activity: implication for neurodegenerative diseases. Cariplo foundation April 2009 – March 2011 the main goal of the project is to investigate the link between dyshomeostasis of calcium signalling and early pathogenic events of AD – both at cellular and molecular level - working on the hypothesis that this neurodegenerative disorder is a synaptopathy. As an addendum to this, we do not wish to focus on the role of calcium in neuronal loss but on the role of initial small changes of calcium homeostasis in synaptic deficits, as in vivo this correlates better with early neurological disturbances and cognitive impairment. Molecular mechanism and behavioural aspects in experimental models of Parkinson’s disease. Italian Minister of Health November 2008 – November 2010 Despite abundant evidences support the role of NMDA receptor complex in Parkinson’s Disease, only scattered if any information are available at single subunit level, although this is a much-needed information in order to identify possible new molecular pathways that can represent a pharmacological target for the disease as well as strategies to tackle dyskinesia. Thus, specific aim of the project will be to characterize in striatal neurons the molecular events responsible for the correct synaptic localization of NR2A and NR2B subunits of NMDA receptor and to study the functional consequences of such an event in experimental parkinsonism and in L-DOPA induced dyskinesia. “REPLACES” - Restorative Plasticity At Corticostriatal Excitatory Synapses EU VII Framework Programme November 2008- October 2012 The main ideas underlying this proposal is that the long-term efficacy of Titolo progetto Ente finanziatore Durata progetto Abstract del progetto new treatments for Parkinson’s disease (PD) will be conditioned by their ability to restore, both structurally and functionally, the “synaptic wiring” of striatal neurons and physiological synaptic plasticity. Accordingly, the proposal will address the potential restorative effects of either novel pharmacological treatments or neuronal transplants on the corticostriatal microcircuitry. Since chronic treatment with the DA precursor L-DOPA induces in the majority of PD patients a maladapTative plasticity causing involuntary movements (dyskinesia), innovative strategies should prevent the development of this disabling condition. The project will characterize corticostriatal synaptic plasticity from molecular aspects to clinical neurophysiology involving also behavioural as well as morphological analysis of the basal ganglia system. “cPADs” - Cell permeable peptides as drug delivery system: a way towards innovative therapeutic strategies for neurodegeneration. EU VII Framework Programme April 2008- March 2012 The project aims to foster a dynamic pathway between academic research organizations and private commercial enterprises, based on a long-term co-operation programme in the field of innovative therapeutic strategies for neurodegenerative disorders. Through inter-sectoral industry-academia collaboration, the proposal aims to define appropriate technologies to target key protein-protein interactions and key intracellular pathways mediating neurodegeneration and cell death, addressing both the creation of innovative models and the design of innovative drugs by means of exploiting the use of Cell-Permeable Peptides. Nome Prof. Renata Bartesaghi Contatti Tel +39 051 2091727, +39 051 2091747 Fax +39 051 2091737 e-mail: [email protected] Istituto/Dipartimento Dipartimento di Fisiologia Umana e Generale, Università di Bologna Proposta di ricerca Study of early onset Alzheimer disease in a model for Down syndrome, the Ts65Dn mouse Area di interesse identificata: DEVELOPING COMPETITIVE ANIMAL MODELS TO STUDY ALZHEIMER’S DISEASE Area di interesse identificata: BASIC RESEARCH Individuals with Down syndrome (DS) develop neuropathological features similar to Alzheimer disease (AD) early in life, including dementia, accumulation of beta-amyloid, and irregular phosphorylation of tau proteins. Ts65Dn mice have an extra copy of the distal aspect of mouse chromosome 16, a segment which is homologous to human chromosome 21 and which contains much of the genetic material responsible for the DS phenotype. Ts65Dn mice show developmental delay during the postnatal period as well as abnormal behaviors in both young and adult animals that may be analogous to mental retardation. Though the Ts65Dn brain is normal on gross examination, there is age-related degeneration of septohippocampal cholinergic neurons and astrocytic hypertrophy, markers of the AD pathology present in elderly DS individuals. Moreover, Ts65Dn mice undergo cognitive deterioration beginning at 6-8 months of age. All this evidence suggests that Ts65Dn mice may provide a unique model which is suitable for following the progression of AD. While AD develops only in some individuals of the general population, it is the invariable hallmark of DS, implying that trisomic genes play a paramount role in development of the disease. Indeed, recent evidence shows that the presence of an extra copy (due to trisomy 21) of the dual-specificity tyrosine-phosphorylated and regulated kinase 1A (Dyrk1A) gene results in overexpression of Dyrk1A and elevated kinase activity in the DS brain. Dyrk1A phosphorylates tau at several sites, and these sites are hyperphosphorylated in adult DS brains. Phosphorylation of tau by Dyrk1A primes its further phosphorylation by glycogen synthase kinase-3beta (GSK-3beta). These findings strongly suggest a novel mechanism by which the overexpression of Dyrk1A in DS brains causes neurofibrillary degeneration via hyperphosphorylating tau. The putative involvement of Dyrk1A in the etiology of AD implies that de-regulation of Dyrk1A may be an age-related phenomenon that ultimately leads to the pathology. In the Ts65Dn mouse brain, an extra copy of the Dyrk1A gene causes increased expression and activity of Dyrk1A and results in increased tau phosphorylation. Consequently, Ts65Dn mice may represent a good model for the elucidation of the role of Dyrk1A (and other trisomic genes) in the development of AD. Area di interesse identificata: NEW TREATMENT STRATEGIES AD is histopathologically characterized by intraneuronal neurofibrillary degeneration of the abnormally hyperphosphorylated tau and extracellular β-amyloidosis. The neurofibrillary degeneration is apparently required for the clinical expression of AD, and in related tauopathies it leads to dementia in the absence of amyloid plaques. While normal tau promotes assembly and stabilizes microtubules, the abnormally hyperphosphorylated tau disrupts microtubules. Tau is phosphorylated by a number of protein kinases. Glycogen synthase kinase-3 (GSK-3) is one of the key kinases required for AD-type abnormal hyperphosphorylation of tau, which is believed to be a critical event in neurofibrillary tangle formation. Thus, GSK-3 inhibition represents a very attractive drug target in AD and other neurodegenerative disorders. The inhibition of abnormal hyperphosphorylation of tau is one of the most promising therapeutic targets for the development of disease modifying drugs. By exploiting the Ts65Dn mouse it will be possible to establish whether treatments interfering with tau phosphorylation ameliorate the AD phenotype. More importantly, it will be possible to establish whether pharmacotherapy during the early life stages that precede the overt manifestations of AD is able to prevent/delay the onset of the pathology Finanziamenti ricevuti Titolo progetto PRIN 2008 - Effect of prenatal and early postnatal treatment with fluoxetine on neurogenesis and dendritic maturation in the Ts65Dn mouse model for Down's syndrome Ente finanziatore MIUR Durata progetto 2 anni Abstract del progetto Cognitive decline in Down mice; pharmacological treatment to prevent cognitive decline by stimulating neurogenesis Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Sonia Levi +39 0226434755-53 [email protected] San Raffaele Scientific Insitute/Division of Neuroscience Neurodegeneration Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Functional and epidemiological study of neuroferritinopathies: implications for the role of iron in neurodegenerative disorders. Telethon Three years Iron participates in the oxidative damage common to various neurodegenerative disorders, such as Alzheimer and Parkinson diseases and Hallevorden−Spatz syndrome, and iron accumulations have been frequently observed in the areas of the brain that degenerate. Genetic abnormalities of some proteins strictly involved in the regulation of iron metabolism are associated to neurodegeneration. Among them, neuroferritinopathies are rare autosomal dominant disorders associated with mutations of the FTL gene that lead to alterations of the C−terminus of ferritin L−chain. They are characterized by abnormal iron deposition in the basal ganglia of the brain and by the appearance of ferritin aggregates. L−ferritin is dedicated to iron storage, and preliminary results indicate that one of the mutations alters iron availability in cellular models. The project is aimed at studying the structural and functional properties of the ferritin mutants, by developing various cellular models, including primary neurons and glial cells. By this approach, we expect to provide indications on the molecular mechanisms that are responsible for both intracellular iron accumulation and protein aggregate formation. In parallel, we plan to produce knock−in mice models expressing the mutated ferritins, to study the progress of neurodegeneration as well as the formation of ferritin aggregates in basal ganglia. Possible therapeutic approaches, including the use of RNA interference, will be evaluated on both cellular and animal models. Since DNA variations in FTL gene are not uncommon, we plan to analyse its mutations, as well as those of other iron−related genes, in patients with neurological disorders. This study is aimed to elucidate how iron acts as a cofactor in neurodegenerative processes. Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Tiziana Antonelli [email protected] Dept. of Clinical and Experimental Medicine Pharmacology Section, University of Ferrara, Italy Mechanisms of Neurodegeneration : I. Animal models. Neurochemical and morphological in vivo and in vitro models in neurodegenerative diseases BioPharmaNet: Laboratory of research and innovation for the Life Sciences sector industries (Emilia Romagna, Italy) 2007-2010 In vitro models of neurodegenerative diseases have been developed and optimized. Cortical cell cultures, mainly containing GABAergic and glutamatergic neurons, and hippocampal cell cultures, especially containing glutamatergic neurons, can be used to study the cell degeneration following neurotoxic insults. The neuronal damage may be 3 3 evaluated by analysing neurochemical ([ H]GABA and [ H]glutamate uptake and release, biochemical (MTT assay, LDH levels) and morphological (Hoechst 33258 nuclear staining, MAP-2 immunoreactivity) parameters. These in vitro experiments, performed in primary neuronal cultures, cerebral tissue slices and/or synaptosomes, are complementary to the in vivo microdialysis experiments and by combining them it is possible to investigate on the efficacy as well as the neurochemical and molecular mechanisms of new drugs against neurodegenerative pathologies. In vivo microdialysis technique in unilaterally 6-hydroxydopamine (6-OHDA) lesioned rats allows to understand the possible relevance of new pharmacological approaches in the treatment of this pathology. The simultaneous evaluation of the release of several neurotransmitters such as dopamine, acetylcholine, glutamate and GABA in the striatum, globus pallidus and/or other close related brain nuclei of the indirect and direct pathways of the basal ganglia, gives the opportunity to evaluate the efficacy of new therapeutic and genetic strategies in the control of the “motor” pathways both in physiological and pathological conditions. 2. PHARMACOLOGY A- Drug Design Nome Contatti Istituto/Dipartimento Nino Russo [email protected] Dipartimento di Chimica, Università della Calabria, via P. Bucci, cubo14c, 87036 Rende (CS), Italy Proposta di ricerca Alzheimer_s disease (AD) is characterized by the deposit of extracellular amyloid plaques, which consist predominantly of the aggregated peptide amyloid-b (Ab). The aggregation mechanism is not very well understood, but it is proposed that aggregated forms of Ab are neurotoxic because of the production of reactive oxygen species (ROS). Copper and other metal ions (e.g. Zn) have been found to accumulate in amyloid plaques. Moreover, Cu ions have been proposed to be linked to the aggregation of the Ab peptide and/or to the increased generation of ROS. In this context, Cu II coordination to Ab becomes a key feature, because it will directly affect the structure of the peptide and, hence, the aggregation behaviour. The catalytic ROS production of the Cu II ions coordinated by Ab will also be governed by the binding mode. Therefore, this system has been extensively studied. However, the unambiguous identification of the Cu II ligands has remained difficult and no real consensus has emerged in the literature yet. It has been shown that all of the ligands of the high affinity Cu II site are contained in the Ab16 N-terminal domain (DAEFRHDSGYEVHHQK); hence, this soluble fragment has been widely used as a model for the CuII binding site in the native Ab40/42 peptides. Although a lot of experimental studies performed with a wide range of methods, the analysis of the results difficult and impaired the unambiguous identification of the Cu II ligands. Modern theoretical and computational methods can give a significant contribution to the knowledge of the elementary mechanisms at atomistic level on a series of open questions such as: i. unambiguous identification of the metal coordination site; ii. the role of the metal coordination in the conformational change of the metallated protein; iii. The effect of the environment on the complex formation; iv. Design of new and specific drugs for the metal chelation and sequestration. For the development of this research program, sophisticated computational protocols as well as theoretical methods will be implemented and used. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Basic research Development of computational and theoretical methods based on the Density Functional theory and their application on systems with biological significance. Università della Calabria Due anni New algorithms and computational protocols based on the density functional theory should be proposed and tested. The applications should be essentially devoted to the study of complex biological systems Nome Carlo Melchiorre Contatti e-mail: [email protected] ph: 051-2099706 Istituto/Dipartimento Dipartimento di Scienze Farmaceutiche Università di Bologna Via Belmeloro, 6, 40126 Bologna Proposta di ricerca Innovative drug discovery strategies for Alzheimer’s disease Area di interesse identificata New treatment strategies Alzheimer’s disease (AD) is a major neurodegenerative disease affecting a substantial proportion of elderly population and imposing a significant burden on public health. For AD there is still no cure, and only few symptomatic treatment options are available to the 30 million patients worldwide. Driven by the clear unmet medical need and a better understanding of the AD biology and pathophysiology, several lead candidates have progressed to preclinical testing in the last decade. However, to date none has been successful in late-stages clinical trials. Currently, AD is best characterized as a multidysfunctional condition, where interrelated molecular events result in amyloid formation, tau abnormalities, amyloid accumulations, and loss of acetylcholine neurotransmission. Currently available drugs and most of the drugs under development target only one of these mechanisms. This may explain why they exhibit limited effectiveness. Therefore, therapeutic tools able to confront this complexity are urgently needed. Another issue to take into account in AD drug discovery is that AD patients are susceptible to a wide range of concomitant medical conditions (co-morbidity), including hypertension, vascular diseases, diabetes, which can often be associated. There is therefore a need for therapeutic tools to be tailored to their specific conditions. In this respect, we have recently proposed that multitarget ligands, i.e. single chemical entities able to modulate simultaneously multiple targets, might represent the best pharmacological option for both tackling the multifactorial nature of AD and simplifying AD patient therapeutic regimen. Furthermore, from a chemical biology perspective, we envisaged that rationally designed multitarget chemical probes have promise for studying the complex mechanisms underlying neurodegeneration. Finanziamenti ricevuti Titolo progetto Principal Investigator: “Neuroreceptors machinery: design and synthesis of molecules useful for their characterization and modulation and for the treatment of related dysfunctions” (20073EWPF9_003;) Ente finanziatore Ministero dell’Istruzione, dell’Università e della Ricerca (€ 490,000) Durata progetto 22/09/2008-21/09/2010 Abstract del progetto The present project regards the design, synthesis and preliminary pharmacological evaluation of ligands and modulators of neurotransmitter receptor systems. The knowledge of structure and functions of neurotransmitter receptor systems is critical to design and develop drugs useful to treat the numerous pathologies that depend on central and peripheral nervous transmission dysfunctions. As a matter of fact, it is well known that severe pathologies such as pain, anxiety, mental conditions, and neurodegeneration are due to receptor dysfunctions. Some of these pathologies have a tremendous social impact as they involve large population segments, in particular aged people. Titolo progetto Principal Investigator: “Development of Innovative Methodologies for the Identification and Synthesis of New Bioactive Molecules: application to the Alzheimer's disease” (FIRB 2003 RBNE03FH5Y;) € Ente finanziatore Ministero dell’Istruzione, dell’Università e della Ricerca (€ 2,871,000) Durata progetto 12/09/2005-11/09/2008 Abstract del progetto The modern Medicinal Chemistry is nowadays requiring an actual multidisciplinary approach and the development of new technologies either to accelerate the whole discovery process, or to provide better and safer candidate drug. In this context, the goals of the present project are both to develop innovative technological frameworks for molecular recognition and combinatorial chemistry, and to validate the applicability of such techniques through the production of new therapeutics in the field of the Alzheimer's disease (AD) Nome Contatti Istituto/Dipartimento Prof. Federico Da Settimo, Prof. Claudia Martini [email protected], [email protected] Department of Pharmaceutical Science, Department of Psychiatry, Neurobiology, Pharmacology, and Biotechnology - University of Pisa Proposta di ricerca Development of labelled ligands specifically targeting the Translocator Protein 18 kDa in Alzheimer Disease. The Translocator Protein (TSPO) is a mitochondrial transmembrane protein involved in a variety of biological processes, such as steroidogenesis, apoptosis induction and inflammation, and it is differently expressed under physio/pathological conditions, in brain cells (astrocytes and microglia). A dramatic increase in TSPO levels occurs in glial cells in response to brain injury and inflammation, so that TSPO can be considered as a biomarker of ongoing brain insults. The activation of microglial cells and the consequent up-regulation of the TSPO sites have been demonstrated in both normal ageing and in brains of patients affected by Alzheimer disease (AD), stroke and multiple sclerosis. Increased TSPO expression has been observed In this view, the use of TSPO ligands could efficiently reveal microglia activation, so representing a useful tool in brain imaging, both in vivo and in post-mortem analysis. Our group has gained experience in the synthesis of heterocyclic compounds acting at the mitochondrial translocator protein (TSPO). As well documented by several international publications, over the course of several years the research group has also gained experience in the evaluation of the biological activity of new synthesised compounds, through high yield methodologies (Primofiore, G., et al. J. Med. Chem. 2004, 47, 1852; Chelli, B., et al Chembiochem. 2005, 6, 1082; Taliani, S., et al. J. Med. Chem. 2007, 50, 404; Da Settimo, F., et al. J. Med. Chem. 2008, 51, 5798; Chelli, B., et al. J. Cell. Biochem. 2008, 105, 712.) The gained experience have generated significant efforts of the group to apply TSPO ligands as marker of “active” brain pathology, in line with multiple research groups throughout the world. In the present project we aim to design, synthesise and evaluate novel specific TSPO probes featuring the indolglyoxylamide scaffold, suitable as useful tools in brain imaging. A number of radiolabelled ligands targeting TSPO will be developed as powerful tools to image and measure the expression levels of the protein in humans, opening up the opportunity for the discovery of specific PET ligands for TSPO. PET studies of TSPO could offer quantitative follow-up of neuroinflammation, an advantage for biomarker-type measurement during drug development and early clinical development. Preliminarily, studies are in progress with monkeys. Moreover, fluorescent ligands, that represent a safer, faster, and less expensive alternative to radioligands, will be developed for usage in biomedical research. In particular we will investigate a series of fluorescent probes characterized by the presence of a chemoreactive group able to bind the receptor protein irreversibly and covalently, in order to overcome the fact that tissue experimental procedures using a labelled ligand often cause the alteration of the chemical equilibrium and the subsequent loss of the bound fluorescent ligand. The presence of these two devices (a chemoreactive group and a fluorescent chromophore) on a single molecule may offer a multiplicity of advantages, both in protein purification/characterization, and in protein cell visualization/density determination in both cell cultures and tissue slices derived from post-mortem brains. Pursuing of these goals will be realized in strict collaborations with the research groups of Prof. Silvia Selleri (University of Florence). Area di interesse identificata Finanziamenti ricevuti Drug development for AD Diagnosis, Monitoring and Brain imaging Titolo progetto New fluorescent tools targeting the translocator protein (TSPO) as diagnostics for cancer and neurodegenerative diseases. Ente finanziatore Durata progetto University of Pisa 24 months Abstract del progetto The proposed research is addressed to develop innovative fluorescent probes targeting the 18 kDa Translocator Protein (TSPO) (previously known as the peripheral-type benzodiazepine receptor) as promising molecular imaging (MI) tools to be widely applied in studying TSPO location, turn-over and interaction as well as in diagnosing TSPO related pathologies. TSPO is a protein mainly expressed in mitochondria, which is involved in cell proliferation, apoptosis, immunomodulation and steroidogenesis. TSPO basal density is altered in pathologies in which a perturbation of cell survival/death processes has been evidenced such as cancer and neurodegenerative disorders and alterations in TSPO subcellular distribution have been correlated with the malignancy grade of cancer cells. Therefore, TSPO has been suggested as a promising drug target as well as a valid diagnostic marker of state and progression of such pathologies and has become an interesting target in MI. Currently, MI has emerged as an important multidisciplinary area, which involves radiology, biology, biochemistry, physics, engineering, medicine and synthetic chemistry, essential in providing potent imaging agents. Based on our previous experiences, the aim of this research program is to design, synthesize and biologically characterize/validate new reversible and irreversible fluorescent 2-phenylindol-3-ylglyoxylamide ligands with high affinity and selectivity for TSPO. The development of these innovative fluorescent TSPO ligands will offer a wide range of advantages for their applications in molecular basic studies as well as in diagnosis. Nome Alberto Cassetta Contatti [email protected] - Tel: ++39 040 226881 Istituto/Dipartimento CNR – Istituto di Cristallografia UOS Trieste (Dip. Progettazione Molecolare) Proposta di ricerca Structural studies of acetylcholinesterases and their inhibitors: implications for the design of new anti-Alzheimer drugs Acetylcholine esterase (AChE) plays a crucial role in the regulation of signal transmission, by terminating the action of acetylcholine at the cholinergic synapse and muscular junctions. AChE inhibitors are used in the symptomatic treatment of pathologies such as Alzheimer's disease. The 3D crystal structures of a number of AChE complexes with reversible and irreversible inhibitors have been determined. The structural information accompanied by kinetic studies will allow a detailed understanding of the molecular determinants underlying the mechanism of action of this enzyme. Furthermore these results are expected to offer a rational framework for the design and synthesis of compounds with optimal pharmacokinetic and pharmacodynamic properties. Structural studies of neurotrophins (NGF), proneurotrophins(proNGF) and their interactions with their receptors TrkA, p75, sortilin) and a panel of neutralizing antibodies(MNAC13, αD11) Neurotrophins such as nerve growth factor (NGF) are involved in the development and maintenance of neurons both in the central and peripheral nervous system, and have been implicated as potential therapeutic target molecules in the Alzheimer's disease. NGF exerts its biological function by binding to TrkA receptor promoting dimerization, catalytic auto-phosphorylation of intracellular tyrosine residues and triggering of the signal transduction cascade. Until recently, no clear biological role had been attributed to the proneurothrophin (proNGF) peptide, except helping in folding and secretion of mature NGF. Moreover, proNGF was found to be the predominant form of NGF in brain and to be increased in Alzheimer's disease and to induce p75NTR dependent apoptosis in cultured neurons. The specific receptor for proNGF is sortilin. We are investigating the structural and functional bases of the biological differences between NGF and proNGF. The monoclonal antibodies MNAC13 and αD11 are potent antagonists that prevent the NGF-TrkA interactions in vivo systems. In order to gain deeper insight into the molecular basis of the observed high affinity of NGF for TrkA, we are investigating the 3D structures of both neutralizing antibodies and of the respective complexes. Detailed structural information on their antigenic recognition is expected to aid in the development of analogs as antagonists or agonists of neurotrophins that may have greater affinity or specificity for further experimental and therapeutic applications. Structural studies of Hydroxysteroid Dehydrogenases and their interactions with phytoestrogens offlavonoid nature. 17β-Hydroxysteroid dehydrogenases (17β-HSDs) are enzymes which play a crucial role in steroid hormones action by acting on the final steps of estrogen or androgen biosynthesis. Given their ability to interconvert a hormonally inactive (or less active) steroid to its active form, these enzymes operate a pre-receptorial regulation of steroid action. Several distinct 17β-HSDs isozymes can be found in human, principally distinct by their tissue type and cellular localization, homology sequence and metabolic pathway. In humans different 17β-HSDs have been linked to several pathological forms: prostate and breast cancer (17β-HSD type 1) and Alzheimer’s disease (17β-HSD type 10) among others. We have recently undertaken a structural study on a fungal 17β-HSD which share a remarkable structural similarity with both type 1 and 10 of human 17β-HSD. Our studies were aimed to investigate the use of the fungal enzyme as possible model for the understanding at the molecular level the interaction mechanism of 17β-HSDs with specific and less-specific inhibitors. In particular we are investigating the inhibitory action of phytoestrogens, principally of flavonoid nature, towards 17β-HSD type 1, which have been demonstrated to be inactivated at the μmol level by certain flavonoids and coumestans. Moreover, given the lack of biochemical and structural data about the effects of flavonoids on type 10 17β-HSD, we intend to further investigate this subject by a biochemical/biophysical approach combined with crystallographic methods by studying the inhibition properties of the above compounds toward the human 17β-HSD homologue. Because both mutations of the gene encoding this enzyme and its overexpression give raise to a distinct type of mental retardation (17--β-Hydroxysteroid Dehydrogenase X Deficiency; OMIM #300438) and it has been shown to be a molecular link between mitochondrial dysfunction and Alzheimer’s disease by direct binding of the β-amyloid peptide, our approach will provide a valuable understanding of the role played by this enzyme in the occurrence of these neuropathies. LUSTBADER J, CIRILLI M., LIN C., XU H.W., CASPERSEN C., WANG N., TAKUMA K., CHEN X., CHANEY M. TRINCHESE F., LIU S., KUPPUSAMY P., ZEWIER Z., ARANCIO O., STERN D., YAN S.D., WU H.: “ABAD Directly Links Aβ to Mitochondrial Toxicity in Alzheimer’s Disease”. Science (2004) 304, 448-52 Paoletti F, Covaceuszach S, Konarev PV, Gonfloni S, Malerba F, Schwarz, E,Svergun DI, Cattaneo A, Lamba D. “Intrinsic structural disorder of mouse proNGF”. Proteins. 2009 Jun;75(4):990-1009. Covaceuszach S, Cassetta A, Konarev PV, Gonfloni S, Rudolph R, Svergun DI, Lamba D, Cattaneo A. “Dissecting NGF interactions with TrkA and p75 receptors by structural and functional studies of an anti-NGF neutralizing antibody”. J Mol Biol. 2008 Sep 12;381(4):881-96. Bartolucci C, Haller LA, Jordis U, Fels G, Lamba D. “Probing Torpedo californica acetylcholinesterase catalytic gorge with two novel bis-functional galanthamine derivatives”. J Med Chem. 2010 Jan 28;53(2):745-51. Cattaneo, S. Covaceuszach, D. Lamba: “Method for the humanization of antibodies and humanized antibodies thereby obtained”. Patent WO/2005/061540 Area di interesse identificata Structural Neurobiology Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Not Applicable 2. PHARMACOLOGY B-Drug screening Nome Laura Calzà Contatti [email protected] Tel. +39 051 2097947 Cell. +39 335 310979 Istituto/Dipartimento DIMORFIPA Proposta di ricerca Area di interesse identificata Development of competitive animal models for AD GLP grade animal facility and behavioural testing for cognitive performance in AD animal models using video tracking and computer analysis; screening of new molecules after acute administration Basic research Brain metabolism of thyroid hormone: a risk factor for AD? We are interested in exploring possible connections between thyroid hormone content, receptor and activating enzyme expression, APP metabolism and Ab toxicity in the brain in normal and AD mice. Recent advances in knowledge on thyroid hormone brain metabolism and molecular biology of nuclear and membrane thyroid hormone receptors and transporters offer new possible targets to explore this topic. New treatment strategies Nome Contatti Iliana Ferrero Fortunati [email protected] Phone: +39-0521905600 Fax: +39-0521905604 Mobile : +39-3480030063 Istituto/Dipartimento Dipartimento di Genetica, Biologia dei microrganismi, Antropologia, Evoluzione – Università degli Studi di Parma Parco Area delle Scienze 11/A - Campus 43100 Parma Proposta di ricerca “Yeast model to seek for molecules which can reduce mitochondrial mutability” Mitochondrial dysfunction is a prominent feature of Alzheimer’s Disease (AD) and there is growing body of evidence that mitochondrial dysfunction plays a crucial role in the pathogenesis or influences the risk of AD. The Yeast Genetics Group of University of Parma, coordinated by of Prof. Iliana Ferrero, has a long lasting experience in the use of Saccharomyces cerevisiae as a model system to study mitochondrial mutability to address the question of whether there is a molecular relationship between the pathology and the specific mutation and to study the pathogenic mechanisms of human disease associated to mitochondrial DNA mutations. The yeast group has a forty years experience on yeast genetics and a ten years experience in the use of yeast model of mitochondrial diseases (in collaboration with Dr. Massimo Zeviani, Unit of Molecular Neurogenetics, IRCCS Foundation Neurological Institute FC. Besta, and with Prof. Giacomo Comi, Department of Neurological Sciences and Centre of Excellence on Neurodegenerative Diseases, University of Milan). We have created yeast models of several mitochondrial pathologies due to mutations in a relevant number of genes involved in mitochondrial DNA stability. Moreover, we have already used the S. cerevisiae model system to study the effect of molecules on mitochondrial DNA mutability and we have identified molecules able to reduce mitochondrial mutability. Specific aims The main scope of our proposal is the use of S. cerevisiae models already existing in our collection and, when necessary, create new yeast models to test a pharmacological approach to therapy. To this aim we propose to subject yeast model to chemicals (chemical library) screening to prospect new therapeutic drugs able to reduce either extended or point mutability or both. Publications of Iliana Ferrero on yeast model of mitochondrial disease Dallabona, C., Marsano, R.M., Arzuffi, P., Ghezzi, D., Mancini, P., Zeviani, M., Ferrero, I., Donnini, C. (2010). Sym1, the yeast ortholog of the MPV17 human disease protein, is a stress-induced bioenergetic and morphogenetic mitochondrial modulator. Hum Mol Genet 2010 Jan 15. [Epub ahead of print] Ghezzi D, Goffrini P, Uziel, G, Horvath R, Klopstock T, Lochmaller H, D'Adamo P, Gasparini P, Strom TM, Prokisch H, Invernizzi F, Ferrero I, Zeviani M (2009). SDHAF1, encoding a LYR complex-II specific assembly factor, is mutated in SDH-defective infantile leukoencephalopathy. Nature Genetics, 41, 654-656. Di Fonzo, A., Ronchi, D., Lodi, T., Fassone, E., Tigano, M., Lamperti, C., Corti, S., Bordoni, A., Fortunato, F., Nizzardo, M., Napoli, L., Donadoni, C., Salani, S., Saladino, F., Moggio, M., Bresolin, N., Ferrero, I., Comi, G.P. (2009). The mitochondrial disulfide relay system protein GFER is mutated in autosomal-recessive myopathy with cataract and combined respiratory-chain deficiency. Am J Hum Genet 84 594-604. Ghezzi, D., Goffrini, P., Uziel, G., Horvath, R., Klopstock, T., Lochmaller, H., D'Adamo, P., Gasparini, P., Strom, T.M., Prokisch, H., Invernizzi, F., Ferrero, I., Zeviani, M. (2009). SDHAF1, encoding a LYR complex-II specific assembly factor, is mutated in SDH-defective infantile leukoencephalopathy. Nature Genetics, 41, 654-656 Goffrini, P., Ercolino, T., Panizza, E., Giachè, V., Cavone, L., Chiarugi, A., Dima, V., Ferrero, I., Mannelli, M. (2009). Functional study in a yeast model of a novel succinate-dehydrogenase subunit B gene germline missense mutation (C191Y) diagnosed in a patient affected by a glomus tumor. Hum Mol Genet 18, 1860-1868. Spinazzola, A., Invernizzi, F., Carrara, F., Lamantea, E., Donati, A., Dirocco, M., Giordano, I., MeznaricPetrusa, M., Baruffini, E., Ferrero, I., Zeviani, M. (2009). Clinical and molecular features of mitochondrial DNA depletion syndromes. J Inherit Metab Dis 32 143-158. Galassi, G., Lamantea, E., Invernizzi, F., Tavani, F., Pisano, I., Ferrero, I., Palmieri, L., Zeviani, M. (2008). Additive effects of POLG1 and ANT1 mutations in a complex encephalomyopathy. Neuromuscul Disord 18 465-70. Massa V, Fernandez-Vizarra E, Alshahwan S, Bakhsh E, Goffrini P, Ferrero I, Mereghetti P, D'Adamo P, Gasparini P, Zeviani M. (2008). Severe Infantile Encephalomyopathy Caused by a Mutation in COX6B1, a Nucleus-Encoded Subunit of Cytochrome C Oxidase. Am J Hum Genet. 82 1281-1289. Massa, V., Fernandez-Vizarra, E., Alshahwan, S., Bakhsh, E., Goffrini, P., Ferrero, I., Mereghetti, P., D'Adamo, P., Gasparini, P., Zeviani, M. (2008). Severe infantile encephalomyopathy caused by a mutation in COX6B1, a nucleus-encoded subunit of cytochrome c oxidase. Am J Hum Genet 82 12811289. Barberio, C., Bianchi, L., Pinzauti, F., Lodi, T., Ferrero, I., Polsinelli, M., Casalone, E. (2007). Induction and characterization of morphologic mutants in a natural Saccharomyces cerevisiae strain. Can J Microbiol 53 223-230. Baruffini, E., Ferrero, I., Foury, F. (2007). Mitochondrial DNA defects in Saccharomyces cerevisiae caused by functional interactions between DNA polymerase gamma mutations associated with disease in human. Biochim Biophys Acta 1772 1225-35. Fernandez-Vizarra E, Bugiani M, Goffrini P, Carrara F, Farina L, Procopio E, Donati A, Uziel G, Ferrero I, Zeviani M. (2007). Impaired complex III assembly associated with BCS1L gene mutations in isolated mitochondrial encephalopathy. Hum Mol Genet. 16 1241-1252. Valente, L., Tiranti, V., Marsano, R.M., Malfatti, E., Fernandez-Vizarra, E., Donnini, C., Mereghetti, P., De Gioia, L., Burlina, A., Castellan, C., Comi, G.P., Savasta, S., Ferrero, I., Zeviani, M. (2007). Infantile encephalopathy and defective mitochondrial DNA translation in patients with mutations of mitochondrial elongation factors EFG1 and EFTu. Am J Hum Genet 80 44-58. Baruffini, E., Lodi, T., Dallabona, C., Puglisi, A., Zeviani, M., Ferrero, I. (2006). Genetic and chemical rescue of the Saccharomyces cerevisiae phenotype induced by mitochondrial DNA polymerase mutations associated with progressive external ophthalmoplegia in humans. Hum Mol Genet 15 2846-55. Spinazzola, A., Viscomi, C., Fernandez-Vizarra, E., Carrara, F., D'Adamo, P., Calvo, S., Marsano, R.M., Donnini, C., Weiher, H., Strisciuglio, P., Parini, R., Sarzi, E., Chan, A., DiMauro, S., Rötig, A., Gasparini, P., Ferrero, I., Mootha, V.K., Tiranti, V., Zeviani, M. (2006). MPV17 encodes an inner mitochondrial membrane protein and is mutated in infantile hepatic mitochondrial DNA depletion. Nat Genet 38 570-5. Fontanesi, F., Viola, A.M., Ferrero, I. (2006). Heterologous complementation of the Klaac null mutation of Kluyveromyces lactis by the Saccharomyces cerevisiae AAC3 gene encoding the ADP/ATP carrier. FEMS Yeast Res 6 414-20. Lodi, T., Bove, C., Fontanesi, F., Viola, A.M., Ferrero, I. (2006). Mutation D104G in ANT1 gene: Complementation study in Saccharomyces cerevisiae as a model system. Biochem Biophys Res Commun 341 810-5 Palmieri L, Alberio S, Pisano I, Lodi T, Meznaric-Petrusa M, Zidar J, Santoro A, Scarcia P, Fontanesi F, Lamantea E, Ferrero I, Zeviani M (2005). Complete loss-of-function of the heart/muscle-specific adenine nucleotide translocator is associated with mitochondrial myopathy and cardiomyopathy. Hum Mol Genet 14 3079-88. Fontanesi F, Palmieri L, Scarcia P, Lodi T, Donnini C, Limongelli A, Tiranti V, Zeviani M, Ferrero I, Viola AM (2004). Mutations in AAC2, equivalent to human adPEO-associated ANT1 mutations, lead to defective oxidative phosphorylation in Saccharomyces cerevisiae and affect mitochondrial DNA stability. Hum Mol Genet 13 923-34. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Yeast model for human health: Neurodegenerative diseases; Alzheimer’s disease “Identification and characterization of nuclear genes responsible for human mitochondrial disorders” Project coordinated by Massimo Zeviani PhD, Unit of Molecular Neurogenetics, IRCCS Foundation Neurological Institute FC. Besta. Telethon Italy – Tree years project – 2008-2010 Three years Albeit still insufficiently acknowledged, mitochondrial disease encompasses a substantial fraction of human inherited disorders. The inclusive term "mitochondrial medicine" has recently been coined to account for the broad functional implications of mitochondria in normal and disease conditions. Mitochondrial disorders are very heterogeneous, can affect any organ, at any age, and responsible genes are still missing in most cases. These features make the diagnosis of mitochondrial disease difficult, and in many instances cause these disorders to be overlooked. Effective therapy is missing as well. Nevertheless, great progress has been made in this field in the past decade, thanks to the discovery of new genes relevant to disease, and the investigation of the physiopathology of these disorders through a multidisciplinary approach. In recent years we have been finding several new disease genes, which are responsible for well−defined mitochondrial syndromes. However, the function of these genes is still completely or largely unknown. We shall also create disease models user the friendly systems yeast recombinant strains. The study of yeast model will help us understand the physiopathology of the disorders caused by the defective genes, gain insight on their function and related metabolic pathways, and devise sensible and specific therapeutic strategies. Nome Luca Ferraro Contatti [email protected] Istituto/Dipartimento Dept. of Clinical and Experimental Medicine Pharmacology Section, University of Ferrara, Italy Proposta di ricerca Area di interesse identificata Pharmacology: B. Drug screening . Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Innovative technologies for the therapy of the neurodegenerative diseases NeuroBioTech: Innovative technologies for the therapy of the neurodegenerative diseases (Emilia Romagna, Italy) 2009-2012 The aim of NeuroBioTech is the development of a precompetitive research activity to find new therapeutic strategies for the treatment of Alzheimer disease (AD) and other correlated neurodegenerative pathologies. The experimental activity of our research unit is aimed to develop and optimize in vivo models (microdialysis, behavioural tests) and ex vivo models (cerebral tissue), which will be applied in preclinical testing activity for the screening of molecules with antiAlzheimer activity and for the identification of new therapeutic targets in AD. The eventual alterations of endogenous neurotransmitter levels (glutamate, GABA, noradrenaline, dopamine) will be evaluated in the above reported in vivo and ex vivo experimental models. In particular, the eventual modifications of neurochemical signal will be analysed in mice with mutations of APP and/or PS and/or tau. Furthermore, it will be studied if the CHF5074 (new modulator of gamma secretases) administration will be able to correct these neurochemical alterations. Then, the effect of CHF5074 on the APP metabolism and on APP molecular interactions will be studied. 2. PHARMACOLOGY C- Drug Delivery Country: Italy Contact person: Prof. MA Vandelli, Dr. G Tosi ([email protected]) Università di Modena e Reggio Date: 14-01-2009 I) Strategic Issues (Justification of the importance of the issue) NANOTECHNOLOGY FOR HEALTH: THE APPLICATION IN THE BRAIN DISEASES The application of nanotechnology to health raises high expectations for a more efficient and affordable healthcare and has the potential of delivering promising solutions to many illnesses. Even if several areas of medical care could benefit from the advantages that nanotechnology can offer, a selective CNS drug delivery and targeting could improve the therapy of brain diseases which have a tremendous negative impact not only on the patient himself but also on the whole society and linked social and insurance systems. The project could be also the answer to the fragmentation in nanomedicine research in Europe establishing a clear strategic vision in the area. In order for CNS drugs to penetrate to the brain tissue, they must pass through the blood brain barrier (BBB). Many of the compounds that otherwise would be effective in treating CNS diseases are excluded from reaching a sufficient concentration in the brain tissue and producing the desired therapeutic effect. It has been estimated that only 2% of the possible CNS therapeutic compounds can cross the BBB. Thus, drug delivery to the brain is a challenge, because this tissue benefits from a very efficient protective barrier. Even if several disorders and diseases that affect the brain can lead to some loss of BBB integrity, this barrier represents an insurmountable obstacle for many drugs able to treat pathologies in which the BBB integrity is preserved. The same mechanisms that protect the brain from foreign substances also restrict the entry of many potentially therapeutic agents. The blood brain barrier is the major barrier to the passage of active molecules from the blood compartment to the brain. It is located at the level of the brain capillaries, where there is a convergence of different cell types: endothelial cells, pericytes, astrocytes and microglias. The brain microvessel endothelial cells that form the BBB, display important morphological characteristics such as the presence of tight junctions between the cells, the absence of fenestrations and a diminished pinocytic activity, that together help to restrict the passage of compounds from the blood into the extracellular environment of the brain. A promising strategy for the delivery of therapeutics to CNS could be to associate drugs without any modification (prodrugs, for example) to colloidal carriers. These vehicles could deliver numerous drug molecules at specific sites by coupling ligands to the surface of the colloids which can be administered intravenously for chronic treatment. Moreover, owing to their poor stability in biological fluids, rapid enzymatic degradation, unfavourable pharmacokinetic properties and lack of diffusion towards the CNS, biotech drugs (peptides, proteins, genes and antisense drugs) may be advantageously formulated in brain targeted protective nanocontainers. Colloidal drug carriers include micelles, emulsions, liposomes and nanoparticles (nanospheres and nanocapsules). So far, only liposomes and nanoparticles have been largely exploited for brain drug delivery. Recently, the first liposomal preparation of a drug reached the market: it is a formulation of the antitumor drug Doxorubicin loaded into PEGyilated liposomes for the i.v. treatment of brain tumors (commercial formulation Caelyx). In contrast to liposomes, and despite the abudance of experimental works and achievements in the field of polymeric nanoparticle (Np) technology that show the ability of properly functionalized nanoparticles to cross the BBB, no nanoparticle-based drug formulations have been marketed so far. Drugs encapsulated into poly(butylcyanoacrylate) (PBCA) Np covered by the surface active agent polysorbate 80 (Doxorubicin. Loperamide, Dalargin, the anticonvulsivant MRZ 2/576), on the surface of HSA Np (the model drug loperamide), unable to cross BBB when administered alone, were able to exert an appropriate pharmacological effect. Despite interesting results, PBCA Np have limitations that may preclude or at least limit their potential clinical applications; its use for humans is at the moment not approved by FDA. Thus there is a need for the development of nanoparticles based for example on the polymer poly(D,L-lactide-co-glycolide (PLGA), which is approved by FDA for human use, and very promising results were found in the literature. So far, only a low amount of injected nanoparticle dose is able to reach the brain (in the order of 1%); thus active targeting of the BBB represents a promising non invasive strategy for improving drug delivery to brain. In fact, the use of ligands specific for receptors present in high percentage on the BBB could represent the way to realize a selective delivery of nanoparticles, and thus of the embedded drugs, to CNS. With this aim, positive results were obtained when nanocarriers have been conjugated to ligands (OX26 mAb; transferrin, apolipoproteins or surface-active agents (polysorbate-80) able to bind some of the apolipoproteins present in blood that confer the ability to cross the BBB, insulin, folic acid) for active targeting, but other ligands have to be discovered and tested for their ability to carry into CNS drug-loaded nanoparticles. The application of nanotechnology to health raises high expectations for a more efficient and affordable healthcare and has the potential of delivering promising solutions to many illnesses. Even if several areas of medical care could benefit from the advantages that nanotechnology can offer, a selective CNS drug delivery and targeting could improve the therapy of brain diseases which have a tremendous negative impact not only on the patient himself but also on the whole society and linked social and insurance systems. The project could be also the answer to the fragmentation in nanomedicine research in Europe establishing a clear strategic vision in the area. II) Priority areas Health; Neurodegenerative Diseases Innovative strategies for the treatment of brain pathologies Nanotechnology applied to Health, Nanomedicine III) Impact The pharmaceutical treatment of central nervous system (CNS) disorders is the second largest area of therapy, following cardiovascular disease. U.S. sales for CNS drugs exceeded $ 53 billion in 2002 to 2003. CNS disorders are five of the top 10 causes of disability. Stroke is the third leading cause of death and costs the economy $ 40 billion annually. Fifteen million people suffer from Alzheimer’s disease, which is the second most expensive disease to the economy at $ 100 billion annually. Many brain diseases do not have satisfactory treatments. Clearly, CNS disorders are an important current and future priority for the pharmaceutical industry. Moreover, the development of new drug molecules is expensive and time-consuming. The average cost and time for the development of a new chemical entity are much higher (approximately $ 500 million and 10-12 years, respectively) than those required to develop a novel drug delivery system. Hence, there is a need for evolving an existing drug molecule from a conventional form to a novel delivery system that can significantly improve its performance in terms of patient compliance, safety and efficacy by targeting to the specific site. Partner involved: • Italy (ACADEMY) (PROPONENT) (Prof. Vandelli, Department of Pharmaceutical Sciences, Dean of Faculty of Pharmacy, University of Modena and Reggio Emilia, T.e.Far.T.I.) o Nanotechnology, nanoparticles, liposome, surface modification, derivatization of biodegradable polymers, in vivo experience on animals o In vivo pharmacology expertise on CNS drug efficacy o Preparation and loading of engineered Np with selected drugs or active substance • Switzerland (ACADEMY) o Nanotechnology, Coupling methodology with preformed Np , in vitro tests on BBB culture • England (ACADEMY) o Nanomedicines based on novel nanomaterials of synthetic and biological nature • France A (SME) o In vivo models of CNS disease (Parkinson’s Disease, Brain Tumor Glioblastoma, Stroke models) o In vivo toxicity o PK, Biodistribution studies • France B (ACADEMY) o in vitro BBB (astrocyte/endothelial cell co-cultures), models of pathological BBB (inflamed BBB) o HTS platform for the development of peptide-vectors for Np, drug and imaging agent targeting to the CNS across the BBB via receptor mediated trancytosis (RMT). • France C (ACADEMY) o Gene therapy Delivery of siRNA targeted against the prion protein (in vitro and in vivo) Delivery of growth factors (bdnf etc..). in vivo and in vitro experimental models. • France D (SME) o Imaging techniques o Optical Imaging - Scintigraphy - Autoradiography • Portugal (SME) o Synthesis of peptides and antibodies o Gene therapy • Czech Republic (ACADEMY) o Toxicity of Nanoparticles o Interaction with drug metabolising enzymes • Denmark (ACADEMY) o In vivo experiments on olivocerebellar degeneration and Purkinje cell death and excitotoxic model of epileptic seizures o Novel peptides for the engineering of the Np (15 amino acids (aa) derived from the (61 aa) endogenous protein metallothionein (MT). • Israel (SME) o Imaging o Amyotrophic Lateral Sclerosis Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Angelo Montenero [email protected] 0521905553 Dipartimento di Chimica G.I.A.F. – Università degli Studi di Parma Our proposal in this study is the use of sol–gel technique to synthesize mesoporous and nanostructured reservoirs which could be implanted giving a sustained release of the drug molecules. The devices will be constituted by a matrix that contains the drug dispersed or suspended in a fine state. The mechanism, as well as the kinetics and rate of drug release will depend upon the physicochemical properties of the drug, the materials, and the device construction. They will be basically made of oxides derived from Titanium, Silicon (or other metals) alkoxides. The inner structure of the inorganic matrix will be tailored (playing with the conditions of reaction and the relative amounts of the alkoxides, for example) to the specific peculiarity of the substance and to the length of the administration period. The use of alkyl substituted alkoxides together with non-substituted alkoxides could be useful to modify the internal network structure, which depends on the respective rates of hydrolysis and condensation, and therefore on steric hindrance, inductive effects (of alkyl substituents) and functionality (number of alkoxy groups) on the central atom as well as on catalysis and solvent. In cooperation with some European and Mexican research centers we are working with sol-gel devices used to release anticonvulsant drugs, such as phenytoin and valproic acid; similar reservoirs could be employed with drugs for Alzheimer’s treatment. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Basic Research in Advanced Innovative Nanomaterials: Applications to the Solution of Brain Disorders (Identifier: FP7-NMP-2007-LARGE-1) Università Autonoma Metropolitana di Città del Messico - CEE Two years The aim of the group is to study the treatment of three serious neurological disorders: Epilepsy, Parkinson’s and Alzheimer’s, using an interbrain nanodevice to deliver the drug in front of damaged membrane neurons, in order to obtain a potential effect and avoid the decomposition of the molecule. Nanostructured materials biocompatible with brain tissue will be developed. The solids will include ordered silica (SiO2) and ordered titania (TiO2), nanostructured manganese oxides (MnOx) cryptomelane type. Also the sol-gel nanomaterials: SiO2, TiO2, ZrO2, HfO2, hybrid nanostructured materials and monoliths will be prepared using a variety of alkoxides and by thoughtfully varying synthesis conditions. All the inert matrices will be designed using soft chemistry and will require functionalization in order to be biocompatible with the surrounding neural membranes. The drugs to be encapsulated within the devices include for Epilepsy, valproic acid, sodium phenytoine, leviracetam and topiramate; for Parkinson, L-Dopa, and dopamine. For Alzheimer the aim is to quantify the change in concentration of Filaments tangles Helicoidally (PHFs) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), in presence of selective serotonine adsorbent nanomaterial. Nome Contatti Istituto/Dipartimento MASSERINI MASSIMO 0264488203 [email protected] Dip. Medicina Sperimentale Via Cadore 48 20052 Monza Università Milano Bicocca Proposta di ricerca New nanodevices overcoming the blood-brain barrier, for the therapy of Alzheimer Disease Nanodevices offer an attractive mean of performing therapy and diagnosis with their high potential for multi-task functionalization. Theoretically, multi-task functionalization may confer on nanodevices a wide array of properties, such as stealth characteristics once they enter the bloodstream (ability to avoid the reticulo-endothelial system, RES), ability to cross the blood-brain barrier (BBB) and, of course, ability to reach a target. In addition, since it is possible to confer on them features of biocompatibility, lack of toxicity and immunogenicity, biodegradability, simple preparation and high physical stability, the nanotechnology -based systems are intelligent tools for diagnostics, prognostics, and controlled and sustained delivery of therapeutic agents to specific targets . However, the BBB represents a formidable challenge to the delivery of therapeutic agents to the central nervous system, and in particular for the treatment of Alzheimer disease, due to its complex, high selectivity, to the passage of desired molecules into the brain parenchyma. This fact represents an handicap also for testing new and future drugs. These issues has not yet been solved with the existing nanotechnologies, and need strong investments in the future years. For this, completely new approaches are necessary. The proposal is to realize new nano-devices, for overcoming the BBB, for the therapy of the Alzheimer disease. For this purpose, new approaches should be sought,, such as the possibility of nanodevices to carry molecular factories able to perform small tasks; example are the ability to enzymatically modify the architecture of the neuronal membrane modifying the activity of resident molecules involved in the mechanisms of the disease. Area di interesse identificata Finanziamenti ricevuti 2.930.387,00€ Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Nanomedicine Nanoparticles for therapy and diagnosis of Alazheimer Disease EC 5 y (2008-2013) A hallmark of the disease in the AD brain is extracellular aggregates (plaques) of the cytotoxic peptide β-amyloid (Aβ). This project intends to use nanoparticles (NPs) for therapy and diagnosis, singly or combined (theranostics), focusing on brain Aβ as the target. Brain and blood Aβ are in equilibrium across the blood-brain barrier (BBB), so the project also considers blood Aβ as a target. Different NPs will be multiple-functionalized with: i) molecules interacting with Aβ, ii) molecules stimulating BBB crossing, ii) PET or MRI contrast agents. Artificial and cellular models will be used ; the efficacy of NPs will eventually be evaluated in rodent models of AD. Different routes of administration (i.v., oral, nasal) or different protocols (two-step, NP cocktails, aerosols) will be employed to boost NP brain delivery. The prediction is that NPs will range, detect, disaggregate, and remove brain Aβ. Besides this action, NPs will interact with blood Aβ, drawing out the excess of brain peptide by a “sink” effect. 2. PHARMACOLOGY D-Novel Drugs Nome Contatti Lamberto Maffei [email protected] Istituto/Dipartimento Istituto di Neuroscienze del CNR, Pisa Proposta di ricerca In human subjects, building on the results obtained in subjects with Mild Cognitive Impairment (MCI) or in the very initial stages of Alzheimer’s Disease (AD) with the ongoing project “Train the Brain”, we plan to: extend the intervention to a second group of subjects with MCI assess the efficacy of the training employed in “Train the Brain” in subjects in more advanced stages of AD determine the extent of the training beneficial effects by means of extensive follow up of the subjects included in “Train the Brain” determine the most effective components of the training In murine AD models, we plan to: characterize the cellular and molecular mechanisms underlying the action of Intranasal administration (IA) of Neurotrophins and of Environmental Enrichment (EE) in preventing the onset or in ameliorating the progression of the disease. develop new non invasive strategies aimed at preventing or rescuing cognitive deficits and at preventing the onset or reducing the progression of the neurodegeneration We shall also evaluate whether exposure to an “impoverished” environment accelerates the cognitive decline in animal models of AD. We have already exploited IA of NGF (De Rosa et al., 2005) to rescue visual memory deficits in a mouse model of NGF deprivation which exhibits all hallmarks of human AD and we have recently shown in the same model that EE prevents the onset of memory deficits and ameliorates AD hallmarks (Berardi et al., 2007). We have also recently shown that EE and fluoxetine treatment enhances adult visual cortical plasticity to the point of promoting recovery from defective visual development (Sale et al., 2007; Maya Vetencourt et al., 2008; Sale et al., 2009); cortical plasticity enhancement seems highly effective in rescuing cognitive deficits in models of neurodegeneration even in advanced stages of neuronal loss (Fischer et al, 2007). Area di interesse identificata - New treatment strategies - Translational research Finanziamenti ricevuti Titolo progetto Train the brain: clinical and experimental study of the efficacy of cognitive training and physical exercise in dementia Ente finanziatore Fondazione Cassa di Risparmio di Pisa Durata progetto May 2009-May 2012 Abstract del progetto Age-dependent cognitive decline is destined to become a highly impacting problem at the clinical, economical and assistance level. Age, indeed, is the major risk factor for dementia. In Italy, there are around 700.000 patients diagnosed with dementia and around 100.000 new cases every year. Among the pathologies which may lead to dementia, Alzheimer's Disease (AD) and vascular dementia (VaD) are by far the most frequent. At present, there are no effective therapeutic strategies for AD or VaD, which are still pathologies orphan of treatment. It is therefore more and more apparent the need of testing, validating and implementing new strategies aimed at preventing and/or slowing down cognitive decline, starting from the early stages of the disease. We propose to assess the efficacy of a combination of physical exercise and cognitive training in slowing down or arresting the progression of symptoms in subjects at risk of or at the very initial stages of AD and VaD. The scientific rationale for this proposal is described below. a): several studies in humans have demonstrated that the exposure to a cognitively and socially stimulating environment and physical exercise have beneficial effects on brain functioning, especially in the elderly, and reduce the risk of developing dementia (e.g. Laurin et. al, 2001; Fratiglioni et al., 2004; Podewils et al., 2005; Marx, 2005; Kramer and Erickson, 2007). The estimated reduction in the risk of developing dementia varies from study to study but generally ranges between 20 and 50%. b) In parallel, a large number of studies performed in animal models has demonstrated that physical exercise and expossure to a cognitive and socially stimulating environment (a combination known as “enriched environment”, EE) ameliorates cognitive performance, slow down the decline associated with aging, are neuroprotective, and enhance neural plasticity (Cotman and Berchtoldt, 2002; Nithianatharajah and Hannan, 2006). EE ameliorates cognitive deficits in animal models of AD (Adlard et al., 2005; Jankowsky et al., 2005) and is also capable to revert cognitive deficits when they are already well evident. These results show the EE potential as non pharmacological therapeutic strategy not only to prevent the onset of cognitive deficits but also to rescue them. Studies on the efficacy of a combined physical and cognitive intervention in subjects with dementia are very few and present methodological limits. AIMS Primary: To assess the efficacy of protocols of physical exercise and cognitive stimulation on the progression of the disease in subjects at risk of, or with AD and VaD in the initial stages of the disease, identified through an advanced battery of diagnostic tests. Secondary: To develop a nonpharmacological preventive/therapeutic strategy easily applicable to humans and exploitable by the National and Regional Health Service and to study the underlying cellular and molecular mechanism of action in animal models. Nome Prof. Aldo Andreani Prof. Aldo Roda Contatti Istituto/Dipartimento [email protected] 2099734 Tel. +39 051 2099714 Fax: +39 051 [email protected] 343398 Tel. +39 051 343398 Fax: +39 051 Dipartimento di Scienze Farmaceutiche - Università di Bologna Via Belmeloro 6 40126 Bologna Proposta di ricerca Synthesis and screening of new AChE/BuChE inhibitors The cholinergic transmission in the central nervous system is important for the regulation of memory and learning process. Numerous studies have highlighted that the cholinergic neurotransmission system is profoundly compromised in the brain affected by Alzheimer’s disease (AD), with losses of cholinergic neurons and synapses occurring in forebrain, cortex, and hippocampus. The efficacy of cholinergic therapies in AD supports the cholinergic hypothesis and validates this neurotransmitter system as a therapeutic target. Donepezil inaugurated a new class of acetylcholinesterase (AChE) inhibitors with longer and more selective action with manageable adverse effects. It binds both the active-site gorge and the peripheral site through benzylpiperidine and indanone moieties. In an attempt to obtain analogues of donepezil that could maintain its main spatial and physicochemical characteristics while being more compact and less flexible, we applied a strategy to synthesize and rapidly evaluate a series of molecules to be eventually developed as AChE inhibitors for use in AD. We designed a small library of benzylpiperidinone derivatives, where the indanone group was replaced by a bioisosteric indole or pyrrole ring bearing different substituents. The biochemical evaluation of the newly synthesized series was performed in collaboration with the research group coordinated by Prof. Aldo Roda by using a chemiluminescent (CL) method suitable for high-throughput screening (HTS) of AChE inhibitors [1]. The new derivatives showed a rather low degree of potency against the enzyme compared to donepezil, but the molecular skeleton allows for structural modifications which could be done to explore the potential of the series toward new cholinergic agents useful in the treatment of AD [2]. To give a further insight into cholinesterase inhibitors, we also prepared new imidazo[2,1-b]thiazole derivatives [3,4], which were also subjected to CL screening for the evaluation of AChE and butyrylcholinesterase (BuChE) inhibitory activity. These studies allowed the identification of some analogues displaying a peculiar AChE-selective inhibitory activity in the micromolar range [4]. Both AChE and BuChE hydrolyze ACh, albeit with slightly different kinetics, and coexist ubiquitously in humans. BuChE is widely distributed in the body of vertebrates and is implicated in neurogenesis and regulation of cell proliferation and differentiation. AChE and BuChE inhibitors could reverse the cognitive deficits associated with damaging of cholinergic neurons. These enzymes are encoded by different genes and clearly differ in substrate specificity and sensitivity to inhibitors, likely due mainly to structural differences in the active site gorge. In brain affected by severe AD, the reduction of AChE is accompanied by an up to 2-fold increase in BuChE levels. Moreover both enzymes are involved in aggregation of beta-amyloid and in formation/maturation of senile plaques, therefore the use of reversible cholinesterase inhibitors is considered an attractive therapeutic approach. Unlike AChE, the physiologic function of BuChE in normal and diseased humans is not completely clear yet, although a role in neurodegenerative disorders has been suggested. Ongoing elucidation of the properties and behavior of BuChE in normal and AD brains supports a role for BuChE in AD pathology as well as normal cognition. In AD clinical studies the cognitive improvement was correlated with rivastigmine (Exelon) treatment. This drug is a well known AChE/BuChE inhibitor and these studies support a role for central BuChE in addition to AChE inhibition in modulating cholinergic function. Observation made in these studies indicates that a good balance of inhibition of both AChE and BuChE may be beneficial in treating the cognitive decline in AD. The research we propose concerns the design and synthesis of small libraries of molecules, whose AChE/BuChE inhibitory activity will be assayed using the HTS CL method for AChE/BuChE inhibitors described above. The availability of a simple, rapid, and sensitive HTS CL method will facilitate the study of the biological activity of the designed compounds and the identification of the most promising molecules. In addition, the effect of AChE inhibitors could be evaluated “ex vivo” in experiment animals by ultrasensitive CL microscope imaging. In fact, the CL system can be also employed for the localization and quantification of AChE in brain tissue sections [5]. Overall, these studies could furnish new molecular tools useful in elucidating structural and functional differences between AChE and BuChE and may contribute to the development of new potential drugs for the treatment of AD. [1] Guardigli M., Pasini P., Mirasoli A., Leoni A., Andreani A., Roda A.: Chemiluminescent highthroughput microassay for evaluation of acetylcholinesterase inhibitors. Anal. Chim. Acta, 535, 139144, 2005. [2] Andreani A., Cavalli A., Granaiola M., Guardigli M., Leoni A., Locatelli A., Morigi R., Rambaldi M., Recanatini M., Roda A.: Synthesis and screening for antiacetylcholinesterase activity of (1-benzil-4oxopiperidin-3-ylidene)methylindoles and –pyrroles related to donezepil. J. Med. Chem. 44, 40114014, 2001. [3] Andreani A., Granaiola M., Guardigli M., Leoni A., Locatelli A., Morigi R., Rambaldi M., Roda A.: Synthesis and chemiluminescent high throughput screening for inhibition of acetylcholinesterase activity by imidazo[2,1-b]thiazole derivatives. Eur. J. Med. Chem., 40, 1331-1334, 2005. [4] Andreani A., Burnelli S., Granaiola M., Guardigli M., Leoni A., Locatelli A., Morigi R., Rambaldi M., Rizzoli M., Varoli L., Roda A.: Chemiluminescent high-throughput microassay applied to imidazo[2,1b]thiazole derivatives as potential acetylcholinesterase and butyrylcholinesterase inhibitors. Eur. J. Med. Chem., 43, 657–661, 2008. [5] Pasini P., Musiani M., Russo C., Valenti P., Aicardi G., Crabtree J.E., Baraldini M., Roda A.: Chemiluminescence imaging in bioanalysis, J. Pharm. Biomed. Anal., 18, 555-564, 1998. Area di interesse identificata Multidisciplinary projects Nome Contatti Istituto/Dipartimento Olga Bruno [email protected] Tel: +39 010 353 8367 Dipartimento di Scienze Farmaceutiche – Università degli Studi di Genova Viale Benedetto XV,3 16132 Genova Proposta di ricerca In the past few years, it has been suggested that dysfunctions of the cAMP/PKA/CREB pathway could be a major cause of the cognitive deficits characterizing Alzheimer Disease (AD). Indeed, in vitro experiments have shown that Aβ42 is able to reduce the function of this pathway in hippocampal neurons, thus leading to impairment of long term potentiation (LTP) and both effects can be reversed by rolipram, a well known PDE4 inhibitor. More interestingly, rolipram has been shown to restore LTP and to ameliorate cognitive deficits in a double transgenic (APP/PS1) murine model of AD. Recently, sub-chronic rolipram treatment was reported to improve long-term memory performances also in normal rodents. Despite PDE4 inhibitors have been indicated as promising therapeutical agents for AD, rolipram cannot be clinically used due to its adverse effects, particularly sedation and nausea. Among the different PDE4s, PDE4D is highly expressed in hippocampal regions and some isoforms seem to be augmented in the hippocampus of AD patients. Recently we have synthesized a series of innovative selective PDE4D2, PDE4D3 inhibitors,1 which increase cAMP levels in the hippocampus and improve cognitive functions in preliminary in vivo pre-clinical studies. Surprisingly, in vitro experiments showed that our compounds, as well as rolipram, augment Aβ levels in neuronal cultured cells, a result in agreement with a recent study showing that dimebolin, a drug known to ameliorate cognitive deficits in aged rodents and in people suffering from mild AD, causes an acute increase of Aβ brain levels in experimental animals. Our project is aimed at studying new PDE4D selective inhibitors as novel therapeutic agents in AD. These compounds could be also useful pharmacological tools to increase the knowledge on the role of different PDE4 isoforms in CNS under physiologic as well as pathologic conditions, and to verify the relationship between PDE4D inhibition, A levels and memory improvement. Our research unit skilful in medicinal chemistry will work to modify the chemical structure of previously synthesized compounds in order to increase its selectivity, potency and in vivo activity. Our first goal is to obtain a beneficial effect on memory performances without emesis. The molecular modifications will be performed basing on a preliminary computational study aimed to show what structural features could discriminate between different PDE4D splicing variants. Thus docking studies and 3D-QSAR analysis will be used to filter the large number of new molecular entities deriving from various chemical modifications on our previously synthesized compounds. 1) Bruno O, Romussi A, Spallarossa A, Brullo C, Schenone S, Bondavalli F, Vanthuyne N, Roussel C. New selective phosphodiesterase 4D inhibitors differently acting on long, short, and supershort isoforms. J Med Chem. 2009 52(21), 6546-57. Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Drug Design and synthesis of therapeutic agents for neurodegenerative disorders, particularly for Alzheimes Disease. Heterocycle compounds as neuroprotective agents (MIUR, Italian Minister of University Research) 2005032713_005 Ronsisvalle Giuseppe (Principal Investigator) Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Bruno Olga Role: Co-investigator 1/01/06-31/12/07 This study was aimed at synthesizing new molecules acting by different pathways in neurodegenerative pathologies. Heterocycle compounds as neuroprotective agents (MIUR, Italian Minister of University Research) 2007E8CRF3_003 Ronsisvalle Giuseppe (Principal Investigator) Bruno Olga Role: Genoa Unit PI 22/09/08-22/09/10 This grant is a renewal of previously financial support for a large study aimed to synthesize new molecules acting by different pathways in neurodegenerative pathologies. Nome Contatti Istituto/Dipartimento Fabrizio Tagliavini Tel +39 02 2394 2260; Fax +39 02 2394 2101 Email: [email protected] Sito web: http://www.istituto-besta.it UO Neuropatologia – Neurologia 5 Fondazione IRCCS Istituto Neurologico “Carlo Besta” Via Celoria ,11 - 20133 Milano- ITALY Proposta di ricerca The research group: The Division comprises (i) a Clinical Unit (Dementia Center) devoted to the diagnosis and treatment of patients with degenerative dementias (over 2000 out-patients and 150 inpatients per year) and (ii) a Laboratory Unit (certified ISO 9001:2000 - Registration Number 26080, dedicated to the analysis of genes and biomarkers associated with degenerative dementias, postmortem characterization of the disease process and disease-specific protein, and experimental studies on disease pathogenesis and the development of therapeutic strategies. The course of action of this activity can be expressed as a “Bed to Bench to Bed” cycle, with the ultimate goal to identify disease-modifying drugs in preclinical settings and apply them to patients. In this regard, the Clinical Unit is currently coordinating a multicentre phase II clinical trial supported by AIFA, to test the effectiveness of an anti-amyloidogenic molecule identified in experimental models. This comprehensive approach has also the great advantage to collect biological samples (plasma, DNA, CSF and brain tissue) from fully characterized patients. In this regard the Laboratory Unit is part of the Network of Excellence “BrainNet” Europe devoted to biobanking, harmonization of assessment tools and standardization of diagnostic criteria of neurodegenerative diseases. The permanent staff of the Division is composed by 8 MD and two technicians. In addition, the staff comprises 13 post-doctoral fellows (7 PhD, 3 MDV, 1 MD, 1 psychologist), 2 PhD students and 1 technician who are committed to the research activities on degenerative dementias with different expertise and roles. On the overall, the team has large experience in a variety of techniques spanning from neuropathology (including immunohistochemistry, morphometry, electron microscopy and atomic force microscopy), to molecular genetics, biochemistry (purification and characterization of disease-specific proteins), cellular and molecular biology, and animal models. The Division of Neuropathology is provided with fully equipped laboratories for histology, immunohistochemistry, electron microscopy, atomic force microscopy, biochemistry, and molecular and cellular biology, and a 60 sqm BL3 facility. Moreover an animal facility composed by a 140 sqm environmental controlled area, provided with a HVAC system and heap-filtered ventilated racks, with separate rooms for maintaining and breeding mouse lines, and carrying out surgical procedures, collection of samples and post-mortem examination is available. B. Preclinical research • Recessive A673V APP mutation: molecular mechanisms and development of a new therapeutic strategy for sporadic AD We have recently identified an APP mutation (A673V) that causes early-onset AD only in the homozygous state while the heterozygous carriers are not affected. This mutation strongly boosts the production and amyloidogenic properties of Aβ. However, the interaction of A673V-mutated and wild-type peptides inhibits amyloidogenesis and Aβ-mediated neurotoxicity (Di Fede et al., Science 2009, 323:1473-7). These findings are consistent with the observation that the A673V heterozygous carriers do not develop disease and offer grounds for a novel therapeutic strategy based on modified Aβ peptides. A research priority of our lab is to unravel the molecular mechanisms of the opposite effects of the A673V APP mutation in homo- or heterozygous state on amyloidogenesis and to develop a lead compound for AD therapy based on A673V-modified Aβ peptides or peptido-mimetic molecules. To accomplish these objectives we have generated a panel of transfected cells and transgenic C. elegans expressing human APP or Aβ with the A673V mutation, respectively, and transgenic mouse lines expressing A673V-mutated APP in the homozygous or heterozygous state. Furthermore, we have identified a prototypic lead compound corresponding to a six-mer Aβ peptide with the A673V substitution and are currently working on brain delivery systems. Area di interesse identificata CLINICA/PRECLINICA - Genetic susceptibility to Alzheimer’s disease (AD) Early diagnosis Biomarkers Developing competitive animal models to study AD Studying early onset forms of AD to follow-up disease progression Cinical trials with or without involvement of pharmaceutical companies Basic research Biobanking (blood sample, CSF, brain repository…) Standardization of diagnostic criteria New treatment strategies - Translational research Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Simone OTTONELLO +39 0521 905646 (office) +39 0521 905148 (lab) [email protected] Department of Biochemistry and Molecular Biology Viale G.P. Usberti 23/A University of Parma 43100 Parma (Italy) -“Biomarkers” / “Early diagnosis” Implementation of high-resolution techniques for Aoligomer detection and quantification in brain samples from Tg-mouse models of Alzheimer disease and in body fluids, including human CSF. -“Basic research” Yeast mutant strains (selected from a genome-wide mutant collection) conditionally expressing Alzheimer-related pathogenic polypeptides as tools to unravel new fundamental aspects of AD pathogenesis (e.g., mitochondrial function impairment) and potential treatments thereof. -“New treatment strategies” Second generation, recombinant A peptide immunogens as prototype vaccines for AD treatment/prevention (active immunization approach) and as primary antigens for the production of conformation-sensitive therapeutic monoclonal antibodies (passive immunization approach). Finanziamenti ricevuti Titolo progetto Ente finanziatore “Novel anti-A immunization approaches for the treatment of Alzheimer disease” Pharmaceutical Company Durata progetto 2 years Titolo progetto Ente finanziatore “Further studies on the Trx-A antigen and its potential as an anti-AD vaccine” Pharmaceutical Company Durata progetto year Titolo progetto Ente finanziatore “Target selectivity of NSAID -secretase modulators evaluated by oligomacroarray analysis” Pharmaceutical Company Durata progetto year Titolo progetto "Large-scale production of recombinant TrxA for a preclinical study in Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Tg-AD mice” Pharmaceutical Company months "Immunoelectrophoretic analysis of A oligomers in brain samples from Tg-AD mice treated with a NSAID -secretase modulator” Pharmaceutical Company 6 months (ongoing) Nome Contatti Istituto/Dipartimento Proposta di ricerca MAURIZIO SCARPA Area di interesse identificata Finanziamenti ricevuti Titolo progetto PEDIATRIC NEURODEGENERATIVE DISEASES [email protected] DEPARTMENT OF PEDIATRICS UNIVERSITY OF PADOVA ITALY Ente finanziatore BRAINS4BRAIN – Treating Pediatric Neurodegenerative Diseases: From Laboratory Bench to Bedside EUROPEAN SCIENCE FOUNDATION Durata progetto Exploratory Workshop March 3-5 2010 Frankfurt Abstract del progetto Neurodegenerative diseases are most prevalent in the elderly, but can in rare cases also affect individuals early in life. In children, neurodegeneration leads to severe mental retardation and premature death with devastating consequences on their immediate environment and relatively high costs for society. Within the EU pediatric neurodegenerative disorders are considered “Rare”. Because of the low-prevalence of these disorders, there is often a striking lack of information, research, treatment and expert availability. There is also often a significant delay before a definitive diagnosis is achieved. BRAINS4BRAIN calls for a collaborative research effort to focus the available multidisciplinary European brainpower with relevance to tackling pediatric neurodegenerative diseases. BRAINS4BRAIN (www.brains4brain.eu) is a European task force aimed at developing and implementing new therapies for pediatric neurodegenerative diseases for which, at the moment, no effective therapy is available. The task force wishes to create a coordinated European effort towards i) the comprehension of pathophysiology processes of these pediatric neurological disorders, ii) the implementation of knowledge on the BBB, iii) the development of new molecular and/or biochemical strategies to overcome the BBB for brain-targeted drug delivery, and iv) the development and implementation of diagnostics and clinical therapies to timely detect and treat these devastating CNS disorders. Importantly, it is foreseen that such knowledge and insights will also be valuable for understanding and treating other important neurological diseases such as Alzheimer’s, Parkinson’s Disease, and epilepsy. LSDs are metabolic disorders, caused by the lack of certain (lysosomal) enzymes or lysosome components, thus preventing the complete degradation of macromolecules and the recycling of their components. The accumulation of intermediate degradation products affects the appropriate functioning of lysosomes and other cellular organelles. Accumulation starts immediately after birth and progressively worsens, often affecting several organs, including the Central Nervous System (CNS). CNS pathology causes mental retardation and progressive neurodegeneration that ultimately ends in early death of these young patients. LSDs are the only group of pediatric neurodegenerative diseases for which therapy that can reverse the natural history of the disease in peripheral organs is available (Enzyme Replacement Therapy, ERT). Unfortunately, ERT is currently unable to effectively reach the CNS to stop the lethal progression of the neurodegeneration. Nonetheless, ERT in combination with i) the advanced knowledge of the (genetic- and biochemical) causes for the development of neurodegeneration in LSDs and ii) the availability within Europe of well established in vitro and animal models, now provides the unique opportunity to decipher the cascade of events leading to loss of brain plasticity and mental retardation, as well as its possible reversal. For instance, suppressing the primary cause of neurodegeneration (e.g. by ERT) in a young brain –that at this stage of development retains considerable plasticity– maximizes the potential for neurological repair. Important to note is that common secondary events in both pediatric and adult neurodegenerative diseases lead to neurodegeneration. Studies on LSDs therefore have the ability to provide unique insights into the pathophysiology and restorative capacities of neurodegenerative diseases in general. Therapies based on ERT can modify the natural history of some LSDs in the peripheral organs (liver, spleen, joint mobility etc.). However, the BBB prevents the therapeutic enzymes used from reaching the CNS and modifying the course of neurodegeneration 1. This limitation can be circumvented by non-pharmaceutical therapy in which sources of enzyme (e.g. a viral vector) are created by surgical injection directly into the CNS. It might eventually also be achieved to some extend by pharmaceutical therapies based on BBB traversing small molecules aimed at reducing the accumulation of molecules requiring degradation (e.g. Substrate Reduction Therapy (SRT):inhibiting early enzymes in the biosynthetic pathway, or Chemical Chaperones: assisting the affected enzyme to attain its correct, active conformation2).3,4 However, ERT is likely to be the most efficient strategy with the least side effects. As neurodegeneration in LSDs affects the entire brain, the technological challenge is to safely deliver ERT efficiently to all affected areas. This might be achieved only by developing strategies enabling the therapeutic enzyme to cross the BBB. Tools to enable the entry of curative molecules across the BBB into the CNS will represent a major breakthrough for the long-term therapy of many (if not all) CNS diseases. Several cutting edge initiatives using different approaches to specifically deliver molecules across the BBB (e.g. exploiting certain transporters or manipulating BBB permeability) exist within Europe. Nome Contatti Paola Tirassa [email protected]; INMM- CNR Istituto di Neurobiologia e Medicina Molecolare Via del Fosso di Fiorano, 64 00143 ROMA ITALY Phone: + 39 06 501 703 236 Fax: + 39 06 501 703 313 Istituto/Dipartimento Proposta di ricerca Institute of Neurobiology and Molecular Medicine CNR 2) Basic research : New treatment strategies based on Neurotrophins f) Ocular NGF administration as a novel non invasive approach to protect brain-NGF target neurons that degenerate in Alzheimer’s disease (L. Aloe, P. Tirassa). Nerve growth factor and (NGF) is a soluble protein that plays a protective role on brain neurons that degenerate in age-related brain disorders, including Alzheimer’s disease (AD). We have shown that conjunctively applied NGF can protect injured brain neurons and damaged retinal ganglion neurons suggesting that topical eye NGF application might be a novel alternative for delivering NGF into the brain and for protecting brain neurons and reducing memory deficits occurring during early events of AD and glaucoma. This hypothesis is currently actively under investigation in collaboration with other research groups. Lambiase A, Pagani L, Di Fausto V, Sposato V, Coassin M, Bonini S, Aloe L. Nerve growth factor eye drop administrated on the ocular surface of rodents affects the nucleus basalis and septum: biochemical and structural evidence. Brain Res. 2007;1127:45-51. Di Fausto V, Fiore M, Tirassa P, Lambiase A, Aloe L. Eye drop NGF administration promotes the recovery of chemically injured cholinergic neurons of adult mouse forebrain. Eye drop NGF administration promotes the recovery of chemically injured cholinergic neurons of adult mouse forebrain. Eur J Neurosci. 2007;26:2473-80. Lambiase A, Aloe L, Centofanti M, Parisi V, Mantelli F, Colafrancesco V, Manni GL, Bucci MG, Bonini S, Levi-Montalcini R. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: Implications for glaucoma. Proc Natl Acad Sci U S A, 2009; 109: 1346913474,. g) Electro-acupuncture and polyphenols as novel approaches to reduce neurodegeneration ( L Manni, M Fiore). Preliminary data indicate that diabetes induced in adult rats by injection with streptozotocin causes a decrease of brain NGF and a concomitant increase of tau phosphorylation, that were all counteracted by low-frequency electro-acupuncture (EA). Using molecular and behavioural approaches the research project will be aimed at: i) Investigate the possible link between diabetes, brain NGF presence/activity and the development of AD-associated pathological features; ii) Study the effect of EA on the above mentioned diabetes-associated brain dysfunction. Alcoholism is related to neurodegeneration and diabetes. The use of polyphenols extracted by olive oil, olive leaves and olive seeds or by grapes (Fiore et al 2009) to prevent or limit mouse brain neurodegeneration due to aging or ethanol consumption will be evaluated. Manni L, Aloe L, Fiore M. Changes in cognition induced by social isolation in the mouse are restored by electro-acupuncture. Physiol Behav. 2009;98(5):537-42 Fiore M, Laviola G, Aloe L, di Fausto V, Mancinelli R, Ceccanti M. Early exposure to ethanol but not red wine at the same alcohol concentration induces behavioral and brain neurotrophin alterations in young and adult mice. Neurotoxicology. 2009;30(1):59-71. h) Human neural stem cells lentivirally transduced with human BDNF (C. Cenciarelli, P. Casalbore). A strong body of evidences show that BDNF result critically involved in Alzheimer’s and Huntington’s diseases. Recently, it has been shown that BDNF knockdown within NSC abolishes the cognitive benefits of NSC delivery. The aim is to use propagating human neural stem cells (hNSC) lentivirally transduced with human BDNF as cellular therapy for replacing degenerating neurons in disease, trauma and toxic insults. Cenciarelli C, Budoni M, Mercanti D, Fernandez E, Pallini R, Aloe L, Cimino V, Maira G, Casalbore P. In vitro analysis of mouse neural stem cells genetically modified to stably express human NGF by a novel multigenic viral expression system. Neurol Res. 2006, 28:505-12. Casalbore P, Barone I, Felsani A, D’Agnano I, Michetti F, Maira G and Cenciarelli C. Neural stem cells modified to express BDNF antagonize trimethyltin-induced neurotoxicity through PI3K/Akt and MAP kinase pathways . Accepted on J. Cell. Physiol. 2010. Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Micaela Morelli Department of Toxicology, University of Cagliari New symptomatic and neuroprotective therapies for Parkinson’s disease 1) Development and dissemination of innovative methodologies for planning, synthesis and determination of biological activity of new ligands for membrane receptors (FIRB:RBNE03YA3L_004) 2) Evaluation of behavioural and biochemical changes in 'Engrailed' mutant mice (PRIN 2006) 3) : Evaluation of neuroprotective effects of ST1535 in animal model of Parkinson ‘s disease (SIGMA-TAU industry) 1-2) MURST 3 ) SIGMA-TAU Industry 1) 3 years 2) 2 years 3) 1 year 1) To determine the in vivo and ex vivo biological activity of adenosine A2A antagonists compounds of new synthesis. Three compounds symptomatically active on models of Parkinson’s disease have been selected on the basis of the results obtained. Moreover their potential neuroprotective effects has been recently investigated in models of Parkinson’s disease. 2) The engrailed strain of mice has shown behavioural deficits and biochemical changes superimposible of Parkinson’s disease models. This strain of mice is now used as a model of this disease. 3) Evaluation of neuroprotective and anti-inflammatory effects of new adenosine A2A receptor antagonist ST1535 in animal model of Parkinson ‘s disease. ST1535 has shown positive symptomatic improvement in motor functions and also neuroprotective and anti-inflammatory effects in the MPTP mouse model of Parkinson’s disease. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Maria P. Abbracchio Dept Pharmacol Sci, Univ Milan, Italy +39-02-50318310/355 [email protected] Area di interesse identificata Innovative neuro-reparative strategies via the implementation of endogenous neurogenesis and gliogenesis: focus on the new P2Y-like GPR17 receptor Finanziamenti ricevuti Titolo progetto Ente finanziatore PURINOCEPTORS AND NEUROPROTECTION: FOCUS ON THE NEW PURINERGIC RECEPTOR GPR17 COFIN-MIUR 2008; Durata progetto 2 anni Titolo progetto Ente finanziatore IMPLEMENTATION AND GLIOGENESIS VIA THE PURINERGIC SYSTEM: A NEW STRATEGY TO REPAIR ACUTE NEURODEGENERATIVE DISEASE Ministero della Salute 2007 Durata progetto 2 anni Abstract delle ricerche in Deciphering the molecular mechanisms controlling the behaviour of corso endogenous neural stem cells is of paramount importance to understand the processes at the basis of brain recovery and to foster the brain’s ability to repair itself in acute and chronic neurodegenerative diseases. We have recently identified a new P2Y-like G-protein-coupled purinoceptor GPR17, activated by both uracil nucleotides and cysteinylleukotrienes (cysLTs) (Ciana P et al., The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J 25:4615-27, 2006). We have established a national consortium (the GPR17 research team) that specifically studies the role of this receptor in cerebral diseases. Initial data in rodent models of focal brain ischemia have shown that GPR17 participates to both the propagation of brain damage and to the subsequent post-injury remodelling and repair (Lecca D et al., The recently identified P2Y-like receptor GPR17 is a sensor of brain damage and a new target for brain repair. PLoS ONE 3:e3579, 1-15, 2008). Specifically, GPR17 was found to activate the quiescent oligodendrocyte precursor cells that are still present in the adult brain, inducing them to resume myelination, thus re-establing neuron-toneuron communication. GPR17 was also found to be expressed by the ependymal cells lining spinal cord’s central canal (Ceruti S et al., The P2Ylike receptor GPR17 as a sensor of damage and a new potential target in spinal cord injury. Brain 132:2206-18, 2009) that are believed to represent the true adult stem cells in this part of the central nervous system. Upon spinal cord mechanical injury, these cells started proliferating and re-expressing markers of multipotent stem-like cells (ibidem). Interestingly, in the adult brain, GPR17 specifically decorates a precursor cell (NG2-positive neural precursors) that normally differentiates to oligodendrocytes, but that, under appropriate conditions, can be addressed to generate functional new neurons and new astrocytes. These data make GPR17 an interesting new target not only for stroke but also for chronic human neurodegenerative diseases characterized by demyelination or neuronal pathology, such as multiple sclerosis (MS) and Alzheimer’s disease (AD). Preliminary data in animal models indeed support this hypothesis: activated GPR17-expressing adult precursor cells were found to accumulate, respectively, around demyelinated wounds in an Experimental Autoimmune Encephalomyelitis (EAE) MS animal model, and around brain plaques in a transgenic murine model of AD (APPPS1 mice). Our current efforts aim at: 1. We want to develop an in silico tridimensional pharmacophore model of GPR17 to screen large virtual chemical libraries and to design new specific ligands. The newly synthesized agonists/antagonists (including new “dual” ligands acting at both the nucleotide and the cysLT binding sites) will be tested in our established in vitro assays and then addressed to their in vivo validation 2. “Old” and new GPR17 ligands will be used to unveil the exact role of the receptor during oligodendrocyte differentiation and myelination, in both in vitro and in vivo models, and to identify the factors regulating its transcription in precursor cells. 3. To set the basis for a possible wider exploitment of GPR17 in adult neurogenesis/gliogenesis, by using both isolated precursor cells and neurospheres from control and diseased brains, we shall also develop pharmacological protocols to address GPR17-positive precursor cells to neurons or astrocytes. 4. The role of GPR17 in the EAE MS model will be studied in detail. Initial information on the changes of GPR17 expression in these models will be obtained via an in silico analysis of microarray data; then, the regional localization, cell types, and kinetics of GPR17 expression during disease evolution will be assessed. Finally, we shall determine the effects of a “preventive” or “therapeutic” in vivo administration of GPR17 ligands on animals’ weight, neurological score and disease course. Globally, results will contribute to finding new GPR17-based regenerative medicine approaches for human neurodegenerative diseases, allowing for substantial improvement of “endogenous neural stem cell”-based in vivo therapy. Nome Contatti Tel. E mail Istituto/Dipartimento Mariagrazia Grilli/Pier Luigi Canonico Mariagrazia Grilli 0321375828 3280587577 [email protected] DiSCAFF, University of Piemonte Orientale, Novara Italy Proposta di ricerca Area di interesse identificata Novel drugs (D). “Pharmacological modulation of adult neurogenesis” Finanziamenti ricevuti Titolo progetto Ente finanziatore Pharmacological modulation of endogenous neural stem cells for amelioration of cognitive impairment in neuropsychiatric disorders Cariplo, Regione Piemonte, PRIN, Fondazione Comunità del Novarese Durata progetto 3 anni Abstract del progetto As of today, the pharmacological treatment of cognitive deficits associated with several neuropsychiatric disorders is still very limited. The essential requirement for an ideal anti-dementia drug would be the ability to re-organize the neuronal network in the injured brain so as to prevent/slow down the neurodegenerative process and possibly to partially recover brain function. Structural and functional plasticity, often referred to as “neuroplasticity”, a fundamental property of the brain, comprises chemical, electrical, molecular and cellular responses by which connections within a brain region and/or between brain regions are reorganized. It is maximal during brain development but it persists to a relevant extent in adulthood, conferring to neuronal networks a higher degree of circuit adaptation to the novel experiences we are exposed to for our entire life. Neuroplasticity in adulthood is in fact the neurobiological substrate of learning and memory. A better understanding of molecular events underlying neuroplasticity may lead to innovative therapeutic approaches in a variety of neuropsychiatric disorders characterized by learning and memory deficits. Different forms of neuroplasticity are known to exist in the adult brain, including neurogenesis. A large body of experimental work has recently established that newborn neurons continue to be added to discrete regions of the adult nervous system including the hippocampus, an area implicated in learning and memory which form the basis for complex behaviour patterns in vertebrates. Several studies support the hypothesis that experience- or activity-dependent integration of new neurons into hippocampal pre-existing neural networks represents a functional mechanism of brain plasticity subserving specific types of learning and memory functions. Several human disorders affecting memory and cognition, including neurodegenerative and psychiatric disorders, are characterized by abnormalities in synaptic structure/number as well as in neurogenesis, including Alzheimer’s disease (AD). The present project aims to investigate whether the pharmacological manipulation of neuroplasticity in pathological conditions may be effective in modulating different memory stages and systems. This would be a starting point for developing and testing novel pharmacotherapies for cognitive disorders. In particular, the contribution of NFkappaB proteins to this process and in cognitive disorders is under investigation. Indeed a vast body of information points at the role of the NF-kappaB family in synaptic plasticity, learning and memory and at neuropychiatric disorders arising when it is deranged (Grilli & Meneghini, in press, 2010). Recently, we showed that NF-kappaB p50KO mice displayed a very selective defect in hippocampal dependent short-term memory which correlated with the disruption of hippocampal adult neurogenesis (Denis-Donini et al., 2008). More specifically, in the absence of p50 protein, defective neurogenesis resulting in memory defects appeared to possibly occur at the transition between distinct maturation steps in newly generated neurons. Intriguingly, this is the stage characterized by high synaptic plasticity when in the adult newborn neurons with extended axons and dendrites have to be selected so as to become integrated into the preexisting functional network. Morover, we were also able to show that several drugs promoting in vitro and in vivo neurogenesis actually involve the NF-kappa signalling pathway. Altogether, these data identify NF-κB signaling components as critical mediators of adult neurogenesis and suggest that that they may represent previously undescribed therapeutical targets for neuropsychiatric disorders, including AD. The overall goal of this research proposal is evaluate whether a disregulated NF-kappaB signalling may affect mechanisms regulating neuronal maturation and differentiation of adult generated neurons and, as a consequence, contribute to cognitive impairment associated with relevant CNS disorders. To this aim, we will use a relevant animal model of AD such as the TgCRND8 mouse line as well as the p50KO mice, both characterized by cognitive impairment. Experimental strategies will be applied to allow to prove the concept that this signaling pathway can be pharmacologically modulated to promote adult neurogenesis and, in turn, ameliorate cognitive impairment. Our project has the potential to increase our understanding of the molecular participants in the modulation of adult neurogenesis in physiology and pathology and it also holds the potential for identifying novel strategies aimed at ameliorating cognitive impairment associated with CNS disorders of high medical need but with no or unsatisfactory therapy. Nome Contatti Tel. E mail Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Fabrizio Chiti Dip. di Scienze Biochimiche, Univ. di Firenze, V.le Morgagni 50, Firenze. Tel: +39-055-4598319 e-mail: [email protected] Dip. di Scienze Biochimiche, Università di Firenze, Viale Morgagni 50, Firenze We intend to focus on the relationship structure-function of oligomers of the β amiloid peptide and its analogues. We will use a number of methodologies to gain structural information on the oligomers at the molecular level, including site-directed mutagenesis, site-directed labelling with molecular probes to be followed in fluorescence, NMR and EPR. We will also use cultured cells and animal models, evaluating the effect of structural changes of the oligomers on their toxicity, of inhibitors of toxicity such as chaperones and small molecules, of changes of the membrane lipid content, of the presence of glycosaminoglycans and other components of the extracellular matrix, and of protective agents such as extrogens, IGF-1 and others that mediate neuroprotection via the activation of the seladin-1 gene. We will use common cellular cultures but also cultures of primary neurons of rat and human ippocampi to evaluate, for example, interactions between oligomers and synapses or oxidative stress of the membrane. We will also use animal models such as transgenic mice. The following research groups will participate: Prof. Fabrizio Chiti Dipartimento di Scienze Biochimiche Prof. Massimo Stefani Dipartimento di Scienze Biochimiche D.ssa Cristina Cecchi Dipartimento di Scienze Biochimiche Prof. Fiorella Casamenti Dip. di Farmacologia Precl. e Clinica Prof. Alessandro Peri Dipartimento di Fisiopatologia Clinica The role of cholesterol in the pathogenesis of Alzheimer's disease Fondazione Cassa di Risparmio di Pistoia e Pescia and Centro Internazionale delle Malattie neurodegenerative (CIMN) 12 months (January 2010 - December 2010) We will investigate the relationship between membrane cholesterol content and cytotoxicity induced by Aβ oligomers on human neuroblastoma cells and primary fibroblasts from familial AD patients bearing genetic mutations. Cytotoxicity of amyloid aggregates in neuronal cells and in Alzheimer fibroblasts: effect of putative anticytotoxic molecules. MIUR and Università di Firenze (cofinanziamento PRIN 2008) 24 months (March 2010 – March 2012) We will study the ability of antioxidant compounds, such as glutathione thioesters (S-acyl-GSH) derivatives, to prevent the degenerative effects triggered by Aβ accumulation on primary cortical neurons, human neuroblastoma cells and primary skin fibroblasts from familial AD patients. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Effect of the extracellular matrix on the formation, stability and cytotoxicity of amyloid fibrils and their oligomeric precursors MIUR and Università di Firenze (cofinanziamento PRIN 2008) 24 months (March 2010 – March 2012) We will study the effect of components of the extracellular matrix where amyloid accumulate (chaperones, glycosaminoglycans, collagen fibres, etc.) on the formation, stability and toxicity of amyloid fibrils and their precursors Titolo progetto Study of the structure-toxicity relationship of protein oligomers Ente finanziatore Durata progetto Abstract del progetto EMBO Young Investigator Programme From 2005 to end of funding (still available to date) We will investigate the relationship between the structure of protein oligomers formed by a model proteins at the molecular level and their biological effect on cultured cells, primary neurons and animal models Titolo progetto Structural and function aspects of lipid bilayers in molecular medicine: involvement of lipid rafts in human pathology Cassa di Risparmio Firenze 36 months (2008-2010, but estende to one more year) The project will investigate the role of natural or synthetic llipid surfaces with different lipid composition and physicochemical features as modulators of peptide/protein misfolding and aggregation as well as of amyloid toxicity. Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Integrative approach to the interplay between Aß, oxidative stress, the proteasome complex and the autophagic pathway. Effect of neuroprotective molecules MIUR and Università di Firenze (cofinanziamento PRIN 2008) 24 months (March 2010 – March 2012) To characterize in vivo in the TgCRND8 mouse model of ß-amyloid deposition the molecular bases correlating oxidative damage and autophagy and ubiquitin-proteasome dysfunctions to the gradual deposition of Aß and to evaluate the effects of neuroprotective drugs. Design of small molecule therapeutics for treatment of Alzheimer's disease based on the discovery of innovative drug targets. European commission (6FP, ADIT) (project LSHB-CT-2005-511977) 5 years (June 2005-November 2010) To analyze in vivo the expression and phosphorylation state of new protein targets in selected brain regions of mouse models of Aβ overproduction using standard immunohistochemical methodologies. To analyze phenotype of target over-expression in transgenic mice, generate inside the project, with respect to neurodegenerative and cognitive parameters Oleuropein aglycon and hydroxythyrosol as anti-aggregating molecules potentially exploitable in Alzheimer’s disease prevention and therapy Ente finanziatore Durata progetto Abstract del progetto Regione Toscana 4 months (November 2010 – October 2012) The research will study both in vitro and in vivo the anti-aggregating effect on Abeta42 of oleuropein aglycone and its derivative hydroxytyrosol, to assess the possible use of these molecules for prevention and treatment of Alzheimer’s disease. 2. PHARMACOLOGY E- Non-Pharmacological Therapies Country: Italy Contact person: Pio E. Ricci Bitti, M.D. (Dept. of Psychology, University of Bologna) [email protected] Date: January 29, 2010 I) Strategic Issues ( Justification of the importance of the issue ) 2.2.1 Brain and brain-related diseases An increasing number of Parkinson disease (PD) patients are undergoing deep brain stimulation DBS). It is now important to evaluate not only the benefits concerning the main symptoms related to movement, but also the psychological (cognitive, affective and behavioural) impact of DBS. II) Priority areas In relation to our experience with PD patients, the most important area to be evaluated by a psychological point of view is the possible influence of DBS on cognitive processes (such as memory, spatial orientation, time perception and perspective…). III) Impact of the research area The results of this kind of studies could be used in order to propose specific psychoeducational interventions to improve the cognitive efficacy of PD patients that underwent DBS Nome Contatti Istituto/Dipartimento Maurizio Taglialatela, MD PhD – Professor of Pharmacology Claudio Russo, PhD – Associate Professor of Pharmacoloy Alfonso Di Costanzo MD – Associate Professor of Neurology Giovanni Scapagnini MD PhD – Associate Professor of Clinical Biochemistry Email: [email protected] Tel. +39-0874-404894/4851/4891 Dept. of Health Science, University of Molise Via De Sanctis, 8610 Campobasso - ITALY Proposta di ricerca Area di interesse identificata Physical exercise and cognitive decline. Epidemiological studies have shown that physical exercise can delay the occurrence and the progression of dementia, but also to improve physical, cognitive and psychological performances, with a positive impact on the quality of life. The mechanisms involved and the identification of potential biomarkers predictive of a positive response to physical activity are areas of intense investigation. In our group, we are currently investigating the potential preventive role of physical exercise programs on cognitive decline in subjects at risk to develop dementia. Our focus will be on subjects presenting with a subjective memory disturbance or affected by mild cognitive impairment (MCI). • Tedeschi G., et al. Brain atrophy and lesion load in a large population of patients with multiple sclerosis. Neurology. 2005 Jul 26;65(2):280-5. • Tedeschi G., et al. Correlation between fatigue and brain atrophy and lesion load in multiple sclerosis patients independent of disability. J Neurol Sci. 2007 Dec 15;263(1-2):15-9. Finanziamenti ricevuti Titolo progetto Regulation of K+ channels by APP-interacting proteins: functional characterization and implications for AD-related neurodegeneration Ente finanziatore Regione Campania (2000-2003) Durata progetto 3 years Abstract del progetto Recent experimental evidence suggest that K+ channels play a major pathogenetic role in Alzheimer disease. In the CNS, phosphorylation of voltage-gated K+ channels by tyrosine kinases (PTK) is a fundamental process regulating their functional activity; its inhibition exerts a neuroprotective action in several experimental models of neuronal damage. On the other hand, the expression of several subclasses of K+ channels can reduce cellular tyrosine kinase activity, thus suggesting the existence of a reciprocal interaction between PTKs/PTPases and K+ channels. Given that the cytosolic domain of β-APP influences, through its adaptor proteins Dab (disabled), X11 and Fe65, the tyrosine kinase activity of Abl, in the present proposal we will: Titolo E. Evaluate the potential modulation of several classes of K+ channels (Kv1.2, Kv1.3, Kv1.5, Kv2.1, ERG, KCNQ2+3) by adaptor proteins able to bind to β-APP (X11, Fe65, Dab). F. Verify the participation of tyrosine kinase activity (and in particular of Abl), in this modulation. G. Investigate the potential involvement of these channels in the cellular damage induced by the hyperexpression of β-APP adaptor proteins. H. Study the participation of K+ channels and their regulation by oxidative stress in the neuronal death triggered by various cellular models of AD-related neurodegeneration. European Community contract N° LSHM-CT-2003-503330/Apopis : Abnormal proteins in the progetto pathogenesis of neurodegenerative disorders Study on tyrosine phosphorylation of APP and its interaction with intracellular adaptors: role in cell signaling and in the generation of amyloidogenic fragments. Ente finanziatore European Community Durata progetto 3 years (2004-2007) Abstract del progetto Within the WP 1 Generation and Turnover, our proposal is aimed to the definition of the role that posttrasductional modifications of APP, in particular its phosphorylation, may exert on its pathophysiologycal function. We will investigate both the role of phosphorylated APP/CTFs in cell signaling, and/or in the generation of amyloidogenic fragments and plaques through the interaction with intracellular adaptors, as well as the effect that site-specific tyr-phosphorylation would have on the direct amyloidogenic pathway of APP. These studies, which are focused in a completely new aspect of APP activity, would lead to the identification of early markers of the neurodegenerative process which occurs in AD, and to the definition of a potentially innovative therapeutical approach based on the findings obtained. Our proposal is aimed at: Aim 1) To study directly in human brain the tyr-phosphorylation of APP and its CTFs and their possible coupling with intracellular adaptors such as ShcA, and to correlate these data with the presence of neuropathologycal hallmarks of the disease. The comparative analysis in non-AD control, AD and/or Down’s syndrome subjects at different ages (from fetal to adult life) will allow a deeper characterization of the molecular determinants during the progression of the disease. Aim 2) To study in transgenic mice carrying APP and PS1 mutations the tyr-phosphorylation of APP and CTFs, their interaction with ShcA-Grb2 adaptors and the influence that such interaction could have on plaque formation, astroglial response, neuronal death. The use of a Tg model will allow at a deeper comprehension of the possible correlation between APP phosphorylation, its cleavage, signaling activity and progressive presence of the typical hallmarks of the disease. Moreover, in this model is possible also to determine the effect that APP and PS1 mutations, which are linked with a human familial phenotype, would cause in the above mentioned parameters, and, in a second step, would allow the application of targeted therapeutical pharmacological approaches following also the indications raised from the in vitro studies (aim 3). Aim 3) To study in neuronal and/or glial cell cultures the molecular mechanisms which regulate APP phosphorylation, influence on cleavage by secretases, coupling with intracellular adaptors, and signaling activity. Using APP mutants, ShcA mutants, and APP KO cells we will define the influence that phosphorylation and interaction with ShcA would cause in signaling activity, cell proliferation and death, and in the generation of amyloidogenic fragments. Moreover, the study of the effect that proliferative and apoptotic stimuli may cause on those parameters and/or with the study of the kinases involved in the activation of APP cleavage and signaling, this would also lead to the identification of pharmacological target for a therapeutical intervention. Nome Dr. Andrea Stracciari, dr.ssa Maria Guarino, prof. Fabio Cirignotta Contatti Istituto/Dipartimento Unità Operativa di Neurologia Azienda Ospedaliero-Universitaria S. Orsola-Malpighi, Bologna Dipartimento Scienze Neurologiche - Università di Bologna Proposta di ricerca Area di interesse identificata New treatment strategies (non pharmacologic therapeutic strategies in neurodegenerative diseases) Finanziamenti ricevuti Titolo progetto Cognitive training in Parkinson’s disease Ente finanziatore Fondazione del Monte di Bologna e Ravenna Durata progetto 24 mesi Abstract del progetto Background. Patients with Parkinson’s disease (PD) frequently present cognitive disturbances, which impair their autonomy and quality of life and are correlated with a significantly increased death rate. Objectives. The primary objective is to assess improvements in quality of life in PD patients undergoing cognitive training. Secondary end-points will be changes in cognitive capacity, autonomy, mood, clinical judgement and burden to caregivers. Methods. This is a prospective randomized, 8-week, single blind, 2arm, multicenter, parallel-group study to evaluate the effect of task specific cognitive training on quality of life in patients with cognitive impairment, but without dementia (MMSE> 24), due to idiopathic PD. Fifty patients per arm will be randomized to detect a 10 point difference in the PDQ-39 overall score. Expected results. An improvement in quality of life obtained by enhancing cognitive functions, namely executive functions, is expected a the main result, being considered a clinically relevant outcome. Nome Contatti Istituto/Dipartimento Proposta di ricerca Orazio Zanetti [email protected] IRCCS Centro San Giovanni di Dio Fatebenefratelli- Brescia Non invasive brain stimulation an integrated approach to neurorehabilitation in Alzheimer Disease Alzheimer’s Disease (AD) is a progressive disorder that impacts memory, language and several other cognitive functions. Given the limited effectiveness of pharmacological treatments, non-pharmacological interventions in AD have gained attention in recent years, and there are currently many different approaches under study, ranging from multi-strategy approaches to cognitive training (Cotelli et al 2006). Despite the potential therapeutic impact of the non-pharmacological approaches, the neural mechanisms underlying the beneficial effects of behavioral interventions remain largely unknown. Functional neuroimaging studies have shown that rehabilitation in patients with developmental and acquired cerebral damage may lead to functional cortical reorganization, a process mediated by activity-dependent plasticity mechanisms (Warburton et al 1999; Buckner et al 2004). These “plastic” mechanisms may also play a role in the aging brain and in Alzheimer Disease (Cotelli et al 2006). An increase in the activation of areas involved in memory, together with the recruitment of new areas during memory intensive tasks, has been confirmed using neuroimaging techniques in AD patients who underwent behavioral interventions (Cotelli et al 2008; Manenti et al 2008; Cotelli et al 2010 in press). Recent studies have reported enhanced performance on specific cognitive tasks in patients with several types of neurological disease, after receiving non invasive brain stimulation (BS), i.e., repetitive Transcranial Magnetic Stimulation (rTMS) or transcranial Current Stimulation (tCS) to specific cortical areas (Miniussi et al., 2008). Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto New treatment strategies Nome Contatti Istituto/Dipartimento PATRIZIA BISIACCHI 049 8276587 DIPARTIMENTO DI PSICOLOGIA GENERALE Proposta di ricerca Neurodegeneration occurring during normal ageing affects several cognitive processes, particularly memory, attention, executive functions. Within these functions there are specific aspects that are particularly susceptible to impairment due to unhealthy ageing, such as working memory, control mechanisms and sustained attention. These are every day cognitive functions that are a result of a sequence of complex neurological processes. The bio-signals (i.e. EEG, NIRS etc) recorded from a set of specific cognitive tasks have important roles in the assessment of these processes. Discovering features in signals of speech and visual processing, which are modalities that are must less open to subjective interpretation, would remove the subjective nature of some existing assessments. These modalities provide signals that can be collected remotely without significant amounts of resources and enable large research corpora of data, of great clinical significance, to be gathered effortlessly Specific focus of our researches is on the assessment of cognitive decline in older persons through the use of multimodal signal processing (i.e. behavioural, EEG, ERPs, fNIRS, Eye movements etc.) . Current clinical measures of cognitive status and general wellness of the elderly have several limitations. Most must be administered by trained professionals, the results of these measures can be affected by many external factors and the ratings can be affected by subjective scoring. Discharging elderly patients from hospital can often rely on a clinician’s subjective opinion about the patient’s ability to function in their own home. The possibility to utilize a remote multimodal assessment will bypass such difficulties. Area di interesse identificata - Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Early diagnosis Imaging techniques and protocols Standardization of diagnostic criteria and diagnostic instruments, harmonization and assessment tools New treatment strategies Ambient Assisted Living, Home automation, smart homes and domotics Cognitive Ergonomics, ICT for elderly/Gerontechnology ELDERGAMES (Development of High Therapeutic Value IST-based Games for Monitoring and Improving the Quality of Life of Elderly People) PRINCIPAL INVESTIGATOR: PROF L. GAMBERINI EU FP6 € 100.373,00 01/09/06 - 28/02/09 Motivating and amusing environments could help elderly people to preserve their cognitive functions and to improve their social life. Based on these premises, ELDERGAMES project Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore developed a mixed reality table-top solution on which users have to exploit and train their cognitive abilities through mind-challenging games in both collaborative and competitive settings SENIORCHANNEL (ICT based solutions for the advancement of social interaction of elderly people) PRINCIPAL INVESTIGATOR: PROF L. GAMBERINI EU- AAL/ MIUR € 303.000,00 2010-2012 Successful aging requires the involvement of elderly people in activities such as interacting and sharing their knowledge, opinions and aspirations with the wider community. SENIORCHANNEL project aims at supporting these activities through the development of a low-cost interactive IPTV and TV studio, through which elderly people will be able to create, interact and enjoy contents exploiting their creativity and knowledge. EARLY COGNITIVE AND ELECTROENCEPHALOGRAPHIC MARKERS OF NORMAL AND PATHOLOGICAL AGEING PRINCIPAL INVESTIGATOR: PROF P. BISIACCHI CARIPARO (Cassa di Risparmio di Padova e Rovigo) € 45.000,00 January 2009 – December 2011 The main goal of the project is to investigate cognitive and electrophysiological alterations in healthy old individuals and in MCI patients, in order to early identify markers of pathological ageing. LIFESPAN CHANGES IN ELECTROPHYSIOLOGICAL PATTERNS ASSOCIATED WITH TEMPORAL DISCRIMINATION PRINCIPAL INVESTIGATOR: PROF P. BISIACCHI Bial Foundation € 45.000,00 January 2009 – December 2011 The present project is aimed at clarifying the specific contribution of frontal and parietal brain regions in time processing, and their development and decline with age by means of new EEG and ERPs methodologies of signal analyses. Results might further have clinical application in diagnosis and rehabilitation of Parkinson’s disease and dementia. AN INTEGRATED APPROACH TO NEUROCOGNITIVE ASSESSMENT OF OLD PEOPLE PRINCIPAL INVESTIGATOR: PROF P. BISIACCHI FSE (Fondo Sociale Europeo) Regione Veneto € 55.372,00 Durata progetto March 2009 – February 2010 Abstract del progetto The present project, thanks to the integration of psychological, medical and bioengineering competencies, was aimed at developing systems of analysis that optimize the use of ERP/EEG methodology in the screening of executive functions in old people. Titolo progetto IMPLEMENTATION OF NEW EEG SIGNAL ANALYSES FOR THE DETECTION OF SUBCLINICAL COGNITIVE IMPAIRMENT PRINCIPAL INVESTIGATOR: PROF P. BISIACCHI FSE REGIONE VENETO € 49.418,18 01/04/10 - 31/03/11 Ente finanziatore Durata progetto Abstract del progetto Titolo progetto Ente finanziatore Durata progetto Abstract del progetto The goal of the project is to create a new integrate approach to detect subclinical cognitive alterations in many pathologies. Cross Ages (Inter-generational learning. From diagnostic to impact evaluation) PRINCIPAL INVESTIGATOR: PROF R. DE BENI EU € 126.649,00 01/12/07 - 30/11/09 Promoting successful active ageing and social inclusion of older people in the local communities through the elaboration of an integrated lifelong learning model based on the intergenerational exchange. Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Vania Broccoli 02 26434616 [email protected] Neuroscience Division iPS technology offers also an unprecedented opportunity to recapitulate both normal and pathologic human tissue formation in vitro, thereby enabling disease investigation and drug development. PD might greatly benefit from an approach with iPS, since it is completely missing a valuable in vitro model of this pathology. In fact, up to now, primary skin fibroblasts and lymphocytes have been the only human cells available for investigating the pathogenetic mechanisms of PD. Although, they provided some insights on the disease, their completely different nature and metabolism prevents gaining information on the neuronal-specific degeneration processes. To overcome this limitation, disease affected neuronal cells would provide a completely new experimental system where to investigate the molecular roots of PD. This is even more important in PD, since also the animal models used for many years for this pathology have shown some important limitations failing to recapitulate the normal evolution of the pathological process observed in the patients. In fact, most of the studies have relied on mouse models of PD, which might be divided in genetic or symptomatic. Mutant animals for the PD-genes have permitted the investigation of specific processes associated to neuronal death such as oxidative stress and mitochondrial dysfunctions, but usually lack key features of the human disease as for instance selective degeneration of dopaminergic neurons. On the other hand, neurotoxins (6-hydroxydopamine, Rotenone, MPTP, Paraquate) have been widely used to efficiently induce dopaminergic neurodegeneration in animals, but again, some of the specific pathological signs of the human disease are not recapitulated as, for instance, the formation of Lewy bodies, the dynamics of neuronal death and a comparable development of the neurobehavioral deficits. For these reasons, the establishment of an iPS model system for PD might result particular significant. Indeed, there are some important features, which might enable this technology to be particular informative in PD research. First of all, some different procedures have been proved rather efficient in generating dopaminergic neurons starting from human ES and iPS cells. In particular, a treatment with FGF8 and Sonic Hedgehog (Shh) acts a strong enhancer of the in vitro dopaminergic commitment. These factors could be associated with different in vitro conditions such as coculture systems with stromal cells or SMAD inhibitors that strongly induce neuronal specification (Barberi et al., 2003; Chambers et al., 2009). Moreover, the identification of genes whose mutations cause dopaminergic neuronal loss in a mainly cell-autonomous fashion allows to generate lines of iPS cells with a known genetic alteration and compare them to those derived from PD sporadic cases. Furthermore, we plan to use iPS-derived dopaminergic neurons as a source of transplantable cells in 6-hydroxidopamine-treated rats and mice. Neurons will be grafted in either the striatum or the substantia nigra and their survival, integration and functional activity will be analyzed. Behavioral studies will assess whether transplanted cells will be able to rescue the neurological impairment in these animals. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Modeling Parkinson’s disease by iPS technology: generation of human affected dopaminergic neurons and gene disease correction by site-specific integration ERANET-NEUON- EU 2010-2013 Induced pluripotent stem (iPS) cells derived from somatic cells of patients represent an innovative tool for in vitro modeling of diseases and may provide a source for replacement therapies. In Parkinson disease (PD), the identification of the pathophysiological mechanisms behind this common neurodegenerative pathology has been particularly hampered by the lack of genuine in vitro models and animal models faithful to the human pathology. Exploiting new generations of viral vectors expressing the reprogramming (Oct4, Sox2 and Klf4) genes in fibroblasts from skin biopsies of the patients, we aim to establish faithful pluripotent iPS cell ines from sporadic and various monogenic forms of PD carrying mutations in either SNCA, PINK1, Parkin, LRRK2, DJ-1 or UCH-L1. In order to create an unprecedented in vitro culture system for PD affected human dopaminergic (DA) neurons, iPS cells will be differentiated in vitro through the Shh/Fgf8 stepwise induction protocol. Such patient-specific neurons will provide a system where we will investigate the mechanisms of genetic PD such as neuronal morphology, Lewy body formation, mitochondrial dysfunction, cell viability after oxidative and other stressors, interactions between different disease proteins, as well as electrophysiological deficits. For those experiments requiring a pure population of DA neurons, Pitx3-GFP iPS cell lines will be generated by means of BACtransgenesis enabling specific labelling of nigral DA neurons. Thus, pure populations of GFP+ dopaminergic neurons will be utilized for microarray based transcriptome profiling comparing PD versus control neuronal populations. These results promise to add precious information on both cellular and molecular pathological mechanisms leading ultimately to understand PD progression. Further, the establishment of human PD affected neurons will offer a remarkable cell platform for future systematic drug screenings. Finally, we will exploit an innovative strategy for site-specific integration of the therapeutic gene based on zinc finger nucleases mediated site-specific integration. This procedure will complement Parkin mutated iPS cells by targeting the therapeutic gene in a safe and transcriptionally competent genomic harbour. Overall, this project has the ambitious task to pioneer new approaches for iPS cell generation and combining them to develop safe gene correction strategies. These new procedures represent a big advantage for a broad functional characterization of iPS-derived dopaminergic neurons affected by PD. The successful achievements of our goals will disclose new opportunities for disease modelling, drug-screenings and cell replacements strategies in PD. Nome Contatti Tel. E mail Istituto/Dipartimento Neural regeneration in the cerebellum: development of cell replacement strategies for the management of spinocerebellar ataxias. Gian Giacomo Consalez, Ferdinando Rossi 02 2643 4838 [email protected], [email protected] Istituto Scientifico San Raffaele, Università degli Studi di Torino Proposta di ricerca Optical And Biomolecular Methods For The Functional Investigation Of Neural Circuits In Vivo. Area di interesse rigenerazione in modelli di malattia neurodegenerativa identificata Finanziamenti ricevuti Neural regeneration in the cerebellum: development of cell replacement strategies for the management of spinocerebellar ataxias. Ataxia UK Ente finanziatore 2 anni Durata progetto The project will use the postnatal and adult mouse cerebellum as a model system to set up cell replacement protocols to treat degenerative disorders of the central nervous system and, namely, spinocerebellar ataxias. Titolo progetto In this project, we plan to: 1. determine the ability of PC progenitors isolated from the embryonic cerebellum to proliferate in the host tissue and integrate into the wildtype and mutant cerebellar cortex 2. analyze the spatio-temporal distribution of cues guiding PCs in their migration into the developing and adult cerebellar cortex 3. manipulate the system to restore developmentally regulated cues guiding PC progenitor homing and connectivity The cerebellum is a highly plastic system whose development is completed after birth. Numerous observations show that exogenous PCs grafted to the cerebellum may integrate productively and establish proper synaptic connections with their afferents and post-synaptic targets. Several Mendelian cerebellar disorders, metabolic or neurodegenerative, feature a selective loss of PCs, fully warranting an attempt to develop cell replacement strategies. We plan to analyze the ontogenetic mechanisms that direct the integration of PC progenitors in the cerebellar cortex of wild type and spinocerebellar ataxia type 2 mice, and determine whether they are still available (or they can be re-activated) in the adult to promote repair. Our strategy for cerebellar regeneration will be driven by this analysis, in an attempt to exploit existing cues and to enhance those that are lost in time, eventually favoring the integration of exogenous precursors in the mature cerebellum. Protocols suited to promote the anatomical and functional integration of early PC progenitors into the wt or mutant mouse cerebellum. Nome Federico Schena Contatti [email protected] Istituto/Dipartimento Dipartimento di Neuroscienze-Sezione Scienze Motorie Università di Verona & Centro di ricerca in Bioingegneria e Scienze Motorie, Rovereto Proposta di ricerca Area di interesse identificata Physical activity programs for elderly subjects with different grades of cognitive impairment. B. Finanziamenti ricevuti Titolo progetto Walking program for Alzheimer patients. Ente finanziatore Comune di Rovereto – Assessorato alle attività sociali Fondazione Mazzali - Mantova 3 years Durata progetto Abstract del progetto Today, one of the main purposes of institutional long-term care is to offer well-organized therapy that also enhances the patients’ functional abilities. New integrated clinical approaches concerning physical exercise and Alzheimer’s disease (AD) have received attention in the scientific literature. Apart from individual rehabilitation, in the special care unit, patients rarely do physical activity alone; this phenomenon has been seen to cause a decline in the patients’ functional progress. Motor inactivity and asthenia become evident in the later stages of AD, including the ability to step, walk, and sit up independently. There is evidence, however, that specific physical activities can slow down this inexorable decline. For example, Fiatarone et al. demonstrated that in very old subjects high intensity resistance training and nutritional supplementation increased gait velocity, stair climbing power, spontaneous activity level, cross-sectional thigh area and maximal leg strength. Moreover, reports in the literature and recent guidelines suggest that the oldest-old can enhance cardiovascular function, flexibility, balance and strength through systematic exercise training. Most researchers in this field agree that also patients with Alzheimer’s disease can benefit from a targeted exercise program, because exercise has additional benefits for elderly dementia patients. [8-10] This notion has been proven in studies on AD patients; which have reported numerous positive effects of physical exercise and walking programs: improvement in walking endurance, better urinary continence, enhanced communication, reduced depression and an increase in activities of daily life (ADLs). However in this institutional scenario, caregiver intervention and interaction with the medical staff and patients are rather difficult. In a nursing home, however, these important positive relationships were easier to manage. The aim of this program is to determinate whether an institution-based walking program carried out together with caregivers would reduce the functional, cognitive and physical decline of patients in the later stages of Alzheimer’s disease. The walking program will be compared with routine care in a randomized controlled clinical trial. We hypothesized that patients in the walking program would show improvements in physical capacity, cognitive performance and biomedical outcomes compared with those in the control care group. Nome Contatti Istituto/Dipartimento Prof. Enrico Granieri [email protected] [email protected] [email protected] Sezione di Neurologia, Dipartimento di Discipline Medico-chirurgiche della Comunicazione e del Comportamento, Università di Ferrara Proposta di ricerca Area di interesse identificata E. Non-Pharmacological Therapies Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Promotion of Adapted Motor Activity among patients with Parkinson’s disease Fondazione Cassa di Risparmio di Cento, University of Ferrara 3 years An adapted motor activity could improve the pattern of body accelerometer and quality of life of patients with Parkinson’s disease. A pleasant, playful motor activity associated with music can give a motor performance improvement in terms of velocity and acceleration of the gait, stability in posture and better and rapid execution of movements. Objective of the study is to demonstrate with different technique and measures whether or not objective differences are detectable in subjects with Parkinson’s disease after an adapted motor activity based on emotional involvement through pleasant exercises, such as music and dance. For this study we developed a triaxial accelerometer that can be located in three different locations of the body that allow to measure the average accelerations in the body trunk. At least 100 patients suffering from Parkinson’s disease will be evaluated. The adapted motor activity session will be run twice a week, each of which lasted 60 minutes. They will be tested at entry of the study, after 4 months of continuative motor activity and after 3 months following the end of the sessions by accelerometer test, disability scale (UPDRS III), quality of life questionnaire (SF-36), MMSE, tapping test. Measures of different domains with tests and objective measures with accelerometric techniques could improve our capability for the clinical monitoring of patients with movement disorders as Parkinson disease. In the same contest we aim to demonstrate the efficacy, in clinical terms, of an adapted motor activity based on emotional involvement through pleasant exercises with the support of music as a powerful cognitive and affective stimuli. (preliminary results of pilot studies has been shown at the Congresses of Italian Society of Neurology and LIMPE in 2008, 2009 and 2010, at the Congresses of Neurology in Moskow [Russia] and Belgrade [Serbia] in 2010. Papers are submitted for publication in neurological journals). Nome Dott.ssa Mariachiara Sensi Contatti [email protected] Istituto/Dipartimento U.O.Neurologia ,Dpartimento di Neuroscienze Riabilitazione, Ospedale S.Anna Ferrara Proposta di ricerca Area di interesse identificata E. Non-Pharmacological Therapies Finanziamenti ricevuti Ente finanziatore New treatment strategies (non pharmacologic therapeutic strategies in neurodegenerative diseases) with Deep brain Stimulation in dystonic and Parkinson Disease patients. None Durata progetto 3 years Abstract del progetto To evaluate the long term response of Deep Brain Stimulations on motor and non motor symptoms in Parkinson disease and Dystonic Patients. The hypothesis is to test axial symptoms, dysartria, dysfagia, pain, both clinically and with non invasive imaging techniques . Titolo progetto 2. PHARMACOLOGY F. Drug Pharmacogenetics Nome Alberto Pilotto Contatti Phone: +39 0882 410271 Fax: +39 0882 410271 E-mail: [email protected] Unità Operativa di Geriatria & Laboratorio di Gerontologia-Geriatria Dipartimento di Scienze Mediche IRCCS Casa Sollievo della Sofferenza Viale Cappuccini, 1 – 71013 San Giovanni Rotondo (FG) Istituto/Dipartimento Proposta di ricerca Sporadic Alzheimer’s disease (AD) is a progressive neurodegenerative disorder occurring predominantly in older age. The prevalence of AD rise from 20% after 75 years to 30% after 85 years, and about 5% of people aged 65 years or older have AD. With about 3 to 4 million people affected in the United States and about 350,000 new cases per year, AD is the most frequent cause of dementia in U.S. and in Western countries. Among the main cause leading to AD, the deficit of cholinergic system plays a major role. Accordingly, one of the most common therapy for the symptomatic treatment of AD is the block of acetylcholinesterase (AChE) by means of acetylcholinesterase inhibitors. The inhibition of AChE increases the concentration of acetylcholine (ACh) in the synaptic cleft, thus restoring the physiological effects of ACh within the central nervous system. Recent studies reported a significant benefits of acetylcholinesterase inhibitors vs placebo on cognitive function, activities of daily living, and behavior. These improvements, however, are not always detectable in clinical practice. Thus, it has been recently suggested that the detection of improvements on cognitive function, activities of daily living, and behavior may be obtained by the concomitant use of drugs acting on other pathogenetic AD mechanism. In particular the European Medical Agency (EMEA) recommend the use of the AChE inhibitors donepezil and rivastigmine in mild-to-moderate AD, and the addition of memantine in moderate-to-severe AD. Memantine block the glutamate receptor, limiting the uncontrolled entrance of Ca2+ ion in the postsynaptic neuron, thus delaying the Ca2+-excess neurodegeneration. Most studies reported that interindividual differences in response to these drugs may be due to variability in drug metabolism related to behavioral, clinical, and genetic factors, mainly hereditary polymorphisms of drug-metabolizing and drug-transporting enzymes. Final objective of this proposal is to identify the genetic component underlying the response/non response to the most common drugs currently used in the treatment of AD, i.e. donepezil, rivastigmine and memantine, throughout the following specific objectives: 1) identification of significant difference in the distribution of the genotypes of the CYP2D6 gene polymorphisms among patients responders/non-responders to donepezil treatment; 2) identification of the specific DNA alteration producing modifications in the CYP2D6 enzyme activity; 3) confirmation of the modification of the enzyme activity by means of donepezil dosage in vivo; 4) identification of significant difference in the distribution of the genotypes of the acetylcholinesterase gene polymorphisms among patients responders/non-responders to rivastigmine treatment; 5) identification of the specific DNA alteration producing modifications in the acetylcholinesterase-mediated hydrolysis of rivastigmine; 6) confirmation of the modification of the enzyme activity by means of rivastigmine dosage in vivo; 7) identification of significant difference in the distribution of the genotypes of the OCT2 gene polymorphisms among patients responders/non-responders to memantine treatment; 8) identification of the specific DNA alteration producing modifications in the OCT2 transporter of memantine; 9) confirmation of the modification of the enzyme activity by means of memantine dosage in vivo. We expected that the identification of functional polymorphisms in the CYP2D6, AChE and OCT2 genes may influence the clinical efficacy of donepezil, rivastigmine and memantine in AD patients, and may be useful in identifying subgroups of AD patients with different clinical response to treatment. Criteria and indicators to verify results are: 1) the number of patients enrolled in the study in the first 6 months; 2) the number of genotypes investigated at the end of the 1th year of the project; 3) the number of specific gene alteration influencing the enzyme activity at the 2nd year of the project; 4) the quantitative data obtained from plasma drug dosage at the end of the 2nd year of the project. This project is coherent with the ministerial guidelines for the I.R.C.C.S. and well integrate the clinical practice in neurology. Area di interesse identificata Pharmacogenetics of drugs for Alzheimer’s Disease Finanziamenti ricevuti Ente finanziatore Effect of weight loss on metabolic, functional, cognitive status and biological markers of longevity in obese frail elderly Ministero della Salute Durata progetto Two years Abstract del progetto Obesity, and in particular abdominal obesity causes serious metabolic and medical complications and impairs quality of life. Moreover, in elderly persons, obesity can lead to frailty by promoting chronic inflammation and by exacerbating the decline in strength, endurance, balance and mobility associated with aging and physical inactivity. Weight loss, induced by calorie restriction and/or physical exercise, simultaneously improves multiple metabolic risk factors for cardiovascular disease and other medical abnormalities associated with obesity, and reduce morbidity and mortality. However, the appropriate treatment for obesity in elderly persons is controversial because of the potential harmful effects of weight loss on bone and muscle mass. It has been reported that weight loss can also modify biological pathways at the cellular level. Insulin and of insulin-like growth factor-1 (IGF-1) have been involved in cellular caloric equilibrium and other mechanisms, i.e. oxidative stress and inflammation. Thus genetic polymorphisms in genes involved in these metabolic pathways such as apolipoprotein E (APOE), sterol regulatory element-binding protein cleavage-activating protein (SCAP), peroxisome proliferators-activated receptor gamma-2 (PPRγ), α-2B-adrenergic receptor (ADRA2B), acyl-CoA synthetase 5 (ACSL5) and interleukin-6 (IL-6) may influence the effect of weight loss on biological mechanisms and possibly on functional and cognitive status in the frail elderly. It is possible that weight loss in obese elderly persons can be beneficial by improving metabolic, functional and cognitive status, or harmful by causing a decrease in muscle and bone mass. However, the clinical and physiological effects of weight loss in obese elderly subjects have never been carefully evaluated. Thus, appropriate treatment for this rapidly increasing segment of the elderly population remains controversial. The purpose of this project is to advance our scientific knowledge by determining the effects of weight loss on metabolic, functional and cognitive status in obese elderly subjects. Moreover, we will evaluate the role of weight loss on biological markers of metabolic health and longevity, i.e. serum level of Insulin, insulin-like growth factor-1 (IGF-1), transforming growth factor-β (TGF-β), tumor-necrosis factor-α (TNF-α), interleukin-1 (IL-1) and C-Reactive Protein (CRP), and their relationships with genetic polymorphisms in APOE, SCAP, PPR-γ, ADRA2B, ACSL5 and IL6 genes. As biomarker of longevity we will also evaluate the accumulation of mitochondrial (mt) DNA mutation through the analysis of the D310 mononucleotide repeat of mtDNA. Titolo progetto Titolo progetto Smart Home for Elderly People (HOPE) Ente finanziatore Ministero della salute/Ministero dell’Università e della Ricerca Scientifica Durata progetto Two years Abstract del progetto Since Europe’s and worldwide population grows older, the need for care is growing as well as people spend more money to get it. The solution Smart Home for Elderly People (HOPE) promises to reduce the need for carers, improve the life of older people and cut the cost of assistance. The main objective of the “Hope” project is to produce an Integrated Computer Technology (ICT) solution that will help the elderly people, specifically those that suffer with the Alzheimer s disease (AD), achieve a richer and more independent lifestyle. Dementia causes long and oppressive suffering to patients and their relatives and imposes enormous costs on society. About 25 million people suffered from dementia in recent years. As a 4-fold increase of this number is expected by 2050, dementia is one main health issue of the next decades. AD covers 50-70% of all dementia cases, no cure exists, and effective and reliable early diagnostic techniques are lacking. Early diagnosis and progress monitoring of AD is a central part of treatment until future drugs and prevention strategies become available. Elderly people with AD spend most of their time at home. The “HOPE” solution consists of an integrated, smart platform that will enable the elderly people with AD to use innovative technology for a more independent life, easy access to information, monitor their health, and serve as a source of inspiration for users as well as for people working with assistive devices. Moreover, it will enable them to perform by themselves activities they were not able to do before and which are important for their daily personal life. The main advantage of the proposed system is that it provides a basis for integrating services for the elderly population while they are at home. HOPE is a budgeted solution that will be installed at the elderly people’s homes, and will provide services for (a) life-long, self organized, appropriate educational environment and access to information, (b) care management and health support, (c) self monitoring and decision making. Optionally, the user will be able to activate software which automatically establishes the necessary interactive, triple-play connection for receiving tele-help and tele-assistance from specialized service providers, doctors or other medical personnel. The proposed application will be intelligent enough to offer safety in terms of controlling efficiently the home environment, economy in terms of controlling and decreasing the need for external help, and convenience in terms of adjusting the operation of connected sub-systems to achieve the best possible user experience. 2. PHARMACOLOGY G. Clinical studies Nome Contatti Tel. E mail Istituto/Dipartimento Prof. Carlo Caltagirone Via Ardeatina, 306 – 00179 Rome 06 51501409 [email protected] IRCCS Fondazione Santa Lucia – Laboratory of Clinical and Behavioral Neurology. Proposta di ricerca Area di interesse identificata Neurocognition in Alzheimer Disease Finanziamenti ricevuti Ente finanziatore Mortality of elderly using conventional and non conventional antipsychotic drugs for the behavioural and psychological symptoms of dementia and for other psychical disorders. AIFA Durata progetto 2007-2010 Titolo progetto Abstract del progetto Background. Antipsychotic drugs are used to control the behavioral and psychological disturbances that often occur in elderly, particularly in those who suffer from dementia. Nonconventional drugs are perceived by clinicians to be better tolerated than conventional ones and for this reason are more frequently adopted for frail and elderly patients. However, reports from ongoing randomized clinical trials on risperidone and olanzapine (two non conventional antipsychotic drugs) in elderly with dementia, signaled an increased mortality and incidence of cerebrovascular events in patients assigned to active drug with respect to placebo. After this initial report, a formal meta-analysis of fifteen randomized placebo-controlled trials, (3,353 patients randomized to nonconventional antipsychotic drugs and 1,757 randomized to placebo) confirmed the existence of a small increase in mortality in those patients who had been assigned to active drugs. For this reason, in European and extra European countries, the health authorities introduced some limitations to the use of these non conventional drugs. When a drug treatment for behavioral disturbances becomes necessary, these limitative measures favor the use of conventional drugs but it is not known whether conventional antipsychotics are safer than conventional ones. Objectives. To assess, in a large population of elderly residents in different Italian Regions (Piemonte, Emilia, Marche) and in the metropolitan area of Milan the overall and cause specific mortality associated with the use of conventional and non conventional antipsychotic drugs. Methods. Using the informative systems of the regional health authorities and of the health district of Milan we will compose a population cohort formed by all the persons of 60 years or older who were prescribed an antipsychotic drug for the first time from January 2001. It is expected that this cohort might be of over 30000 elderly persons. For each included person the last date of existence in life or the date of death will be retrieved. Mortality will be evaluated from first assumption of antipsychotic drug by type of drug used (conventional, non conventional) and by great groups of causes of death (tumors, cardio vascular diseases and other causes). Mortality will be separately assessed for elderly users of antipsychotic and antidementia drugs. Expected results. This retrospective analysis of survival on an extraordinarily large cohort will give further evidences about the potential role of treatment with antipsychotic drugs in survival of elderly with and without dementia. 3. HUMAN/CLINICAL RESEARCH A . Human –epidemiology/risk factors Country: Italy Contact person: Gabbianelli Rosita or Cinzia Nasuti School of Pharmacy, University of Camerino, Date: 29/01/2010 I) Strategic Issues The main focus of our research is the development of a progressive neurodegenerative PD rat model. The research is based upon a multidisciplinary and integrated consortium bringing together field based researchers (genetic, molecular biology, biochemistry, toxicology and chemistry), from 1 Asian and 7 European countries, and 1 SME (University Medical Center Groningen Netherlands; Institut für Organische Chemie der RWTH, Germany; Biological Research Center of the Hungarian Academy of Sciences, Hungary; Instituto de Tecnologia Química e Biológica, Portugal; Departamento de Engenharia Química ISEP, Portugal; Technological Educational Institute of Athens, Greece; Alagappa University, India; SME, Chema Diagnostica di Marco Fiore, Italy). Environmental contaminants accumulated through the food chain can perturb normal physiologic processes leading to the initiation and progression of the Parkinson’s disease (PD). The consequences deriving from early life exposure to these environmental factors can influence variability of life span and the health profile of the elderly in the human population. PD neuropathology is characterized by the degeneration of the nigrostriatal dopaminergic pathway. It is the second most common neurodegenerative disorder, affecting 1-2 % of the population over the age of 50. PD prevalence studies show that the number of individuals with PD over age 50 (4.1-4.6 millions) will double by 2030 in Western European nations (8.7-9.3 million). Moreover the burden of PD will also shift from more industrialized nations to developing Asian nations (India, China etc.) where survival of individuals is likely expected to grow with improving economic conditions and health care. Environmental factors such as lifestyle, diet, pesticides, metals and solvents, seem to be involved in the development of neurodegeneration by causing epigenetic modification in people with PD over 50 years old. Pesticides are one of the most important risk factors of epigenetic changes that affect the development of PD. Permethrin (PERM) is a pesticide mainly used in agriculture, in healthcare, in industrial and domestic settings, and in public health services. It is employed in tropical areas to prevent mosquito-borne disease. Several studies find that pyrethroids persist in house dust in significant concentrations for months after they are applied for pest control operation and specifically, their metabolites were found in greater than 50 % in the urine of the subjects tested (Riederer et al., 2008). The potential consequences of pesticide exposure are greater for children than for adults. Children have higher risk of pesticide exposure due to behaviours such as mouthing items and playing on floors. Nevertheless, the primary exposure pathway of pyrethroids in humans is thought to be through dietary intake of vegetables, fruits and cereals. It is a consequence of pesticide residues that has accumulated in soil and groundwater that is re-cycled for irrigation of crops. Given the complexity of the many factors to which the populations described in the PD epidemiological studies have been exposed, the criteria to identify candidate substances as causative factors of PD have to follow these points: 1) effects on the striatal dopaminergic system (decrease in dopamine level/increase of its metabolites, decreased in dopamine transporter and enzyme tyrosine hydroxylase activity; 2) damages on substantia nigra and striatum; 3) mechanistic effects (e.g. on oxidative stress, mitochondrial dysfunction/complex I inhibition, and alpha-synuclein aggregation). Our previous studies show that permethrin insecticide, belonging to the pyrethroids family, could be the ideal candidate to satisfy the main criteria suggested above. Firstly, mitochondria complex I activity was inhibited in striatum cells incubated with PERM (paper submitted to Neuroscience). Secondly, in vivo studies on pup rats treated with PERM during brain development (from 6th to 15th day of life) showed an imbalance in the dopaminergic system (Nasuti et al., 2007). High homovanillic acid levels and reduced dopamine concentrations were measured in the striatum of 35 day old rats treated. The period of treatment corresponding to neurodevelopment events (synaptogenesis) in rat brain, occurs in the third trimester of pregnancy in humans. Increased oxidative stress in striatum (glutathione depletion, increase of protein oxidation), blood cells (decrease of glutathione peroxidase, increase of lipid peroxidation, unbalance of monocytes and neuthrophils respiratory burst), and behavioural modifications were measured (Nasuti et al., 2007; Gabbianelli et al., 2009, a; Gabbianelli et al., 2009, b). Thirdly, markers related to oxidative stress, such as DNA damage, changes in plasma membrane fluidity and antioxidant enzyme activities were observed in both striatum and blood cells of adult rats treated with PERM for two months (Nasuti et al.,2003, 2008; Gabbianelli et al., 2002, 2004; paper submitted to Neuroscience). Based on assumptions described above we suggest the followed strategic issue: Pathogenic mechanisms, early diagnosis and prevention of Parkinson’s disease (PD) following pesticide intake. The research aims to capitalize on knowledge acquired in the field of pesticide effects on neurodegenerative disease starting from developmental to old age. Studies of the role of epigenetic mechanisms are a research priority. The studies take into account new approaches for early diagnosis of PD and strategies able to prevent the development of the neurodegeneration. Five major objectives to reach: 1) Define the role of the early-life pesticide intake as a causative environmental agent for the development of PD; 2) Define the correlation between pesticide and epigenetic mechanisms in regulating transcription of genes; 3) Identify the specific peripheral markers related to early diagnosis of PD; 4) Evaluate the risk following pesticide food intake and translate the outcome to human population; 5) Prevention of PD onset and/or progression by three strategies: reduction of pesticide residues in the environment through bioremediation, screening of compounds for neuroprotective therapy and development of diagnostic kits to monitor peripheral markers related to the early PD diagnosis. Country: Italy Contact person: Micaela Caserta (CNR) Date: January 28th, 2010 I) Strategic Issues Increase in longevity does not correlate with an increase in disease-free life expectancy. The molecular mechanisms at the basis of this lack of correlation are still matter of debate and require research investment. Cellular senescence has long been used as a cellular model for understanding mechanisms underlying the ageing process. Compelling evidence obtained in recent years demonstrate that DNA damage is a common mediator for both replicative senescence, which is triggered by telomere shortening, and premature cellular senescence induced by various stressors such as oncogenic stress and oxidative stress. Extensive observations suggest that DNA damage accumulates with age and that this may be due to an increase in production of reactive oxygen species (ROS) and a decline in DNA repair capacity. Genomic instability is therefore thought to play a causative role in the ageing process. Our working hypothesis is based on the possibility that chronic exposure to environmental factors causing oxidative stress during life, including pollutants but also noxious nutrients such as alcohol, could actually reduce the ability of relevant genes, implicated in the process of protecting the cell from protein and DNA damage, to respond properly. If, at the same time, inhibition of programmed cell death (apoptosis) occurs, cellular damage starts to accumulate. We will develop experimental strategies to analyse the mechanisms regulating the delicate balance between rescue and apoptotic pathways in two model systems, human monocytic cells and yeast S. cerevisiae, with the specific aim to define molecular targets of pharmacological intervention. II) Priority areas Human cells The major goal is to understand the molecular mechanisms by which environmental insults, causing oxidative stress in several cell types and tissues, alter the expression of genes involved in counterbalancing the damaging effects, like those coding for inducible heat shock proteins, detoxifying enzymes, proteins involved in DNA repair processes as well as factors controlling programmed cell death. One of the experimental systems that will be employed consist of human monocytic cell lines, that will be subjected to various stress agents (mutagenic stress, ethanol, heat shock, and oxidative stress) either as single treatment or as repeated cycles of treatment. The parameters that will be evaluated are: - mRNA level of selected genes (Hsp70, Hsp90, TRAP1, ERCC1, TDG, MCL1, DAD1); - chromatin remodelling and histone modifications occurring at their promoters; - induction of apoptosis/necrosis; - epigenetic modifications and activity of PARP-1 and SIRT1, two key regulators of the balance between life and death of stressed cells. The subsequent step will be to evaluate some of these parameters (those requiring a limited number of cells) on primary blood monocytic cells. S. cerevisiae Saccharomyces cerevisiae has played an important role as a model system to understand the biochemistry and molecular biology of mammalian cells. The genetic tools available and the short life span have also made S. cerevisiae a powerful system to study aging. The yeast chronological life span (CLS) is a measure of the survival of a non-dividing population of cells, and thus can model aging of mammalian non-dividing cells but also of higher eukaryotic organisms. The parallel description of the pro-aging role of homologs of Akt, S6 kinase, adenylate cyclase, and Tor in yeast and in higher eukaryotes, suggests that findings in S. cerevisiae will be valuable to understand human aging and diseases. Moreover, the similarities between mitochondria and age-dependent mitochondrial damage in yeast and mammalian cells indicate that S. cerevisiae is a valuable model to study mitochondrial dysfunction and diseases involving this organelle. Here, we propose to study the relevance of epigenetic modifications due to the yeast sirtuins (Sir2, HST1-4) at relevant aging loci such as telomeres and ribosomal DNA genes. Analyses will be carried out in condition of oxidative stress and at different times of chronological life. The results obtained will be then compared with parallel treatments in human cells were the sirtuin set (seven members in human) could act in a very similar way. III) Impact The comprehension of the basic mechanisms underlying cell response to environmental cues, alongside with the increase of knowledge on human biological variation in health and disease, will allow to identify specific pathways and genes whose activity needs to be strengthened or reduced by pharmacological intervention. Nome Valerio Carelli, MD, PhD Contatti [email protected] Istituto/Dipartimento Department of Neurological Sciences, University of Bologna Proposta di ricerca MELANOPSIN RETINAL GANGLION CELLS AND CIRCADIAN RHYTHMS IN PARKINSON AND ALZHEIMER DISEASE Area di interesse identificata Biomarkers (chronobiology and neurodegeneration) Finanziamenti ricevuti Titolo progetto MELANOPSIN RETINAL GANGLION CELLS AND CIRCADIAN RHYTHMS IN PARKINSON AND ALZHEIMER DISEASE Ente finanziatore none Durata progetto 2 years Abstract del progetto Background: Circadian rhythm abnormalities have been described in aging and neurodegenerative disorders such as Parkinson and Alzheimer disease (1,2). In particular, the “sundowning” (i.e. the appearance of psychomotor agitation during the evening-night) described in Alzheimer patients, has been related to an abnormal circadian rhythm of body temperature (3). In the last years many retinal abnormalities (mainly contrast sensitivity but also colour vision defects and others) and the presence of optic neuropathy have been documented in both Alzheimer and Parkinson disease (4-6). Moreover, in both diseases bright light therapy has been reported to be effective (7,8). Nevertheless, no data are available on the possible relationship between visual dysfunction and circadian rhythm abnormalities in these disorders. Their possible link comes from the recent discovery of a new class of photoreceptors, the melanopsin RGCs (mRGCs) (9). Melanopsin retinal ganglion cells (mRGCs) are a new class of photoreceptors playing a crucial role in non-image forming visual functions such as entrainment of circadian rhythms to light and dark cycle, pupillary light reflex and regulation of synthesis of melatonin during the night (10). These cells represent about 1% of RGCs and originate the retinohypothalamic tract (RHT) (11). Furthermore, In the retina of patients with Parkinson disease a depletion of dopamine levels has been documented. The amount of this reduction is strictly influenced by the time of last levodopa assumption (12). It is of note that dopaminergic amacrine cells interact with mRGCs and regulate melanopsin gene expression (13-15). In collaboration with Professor Alfredo Sadun (Doheny Eye Institute, USC, Los Angeles, California) we recently demonstrated that mRGCs are partially spared in mitochondrial optic neuropathies, characterized by selective loss of RGCs (i.e. LHON and DOA). Our study also showed that mRGCs are lost after age eighty in normal subjects (16,17). Rationale: Based on the current knowledge we hypothesize that the abnormalities of circadian rhythms documented in Alzheimer and Parkinson disease may be related to the presence of optic neuropathy and involvement of the mRGCs system. Aims: The aims of this project are: - to verify the presence of rest-activity rhythms by means of actigraphic recordings in Parkinson and Alzheimer patients - to verify the presence of optic neuropathy by measurement of nerve fiber layer thickness, as evaluated with Optical Coherence Tomography - to verify and quantify the presence of optic neuropathy and the involvement of mRGCs in post-mortem retinas of Alzheimer and Parkinson patients - to verify the presence of dopamine depletion in Parkinson retina - to correlate the presence of optic neuropathy and circadian rhythm abnormalities in patients with Alzheimer and Parkinson disease. References: 1. Wu YH, Swaab DF. Disturbance and strategies for reactivation of the circadian rhythm system in aging and Alzheimer's disease. Sleep Med. 2007 Sep;8(6):623-36. 2. Willis GL. Parkinson's disease as a neuroendocrine disorder of circadian function: dopamine-melatonin imbalance and the visual system in the genesis and progression of the degenerative process. Rev Neurosci. 2008;19(4-5):245-316. 3. Volicer L, et al. Sundowning and circadian rhythms in Alzheimer's disease. Am J Psychiatry. 2001 May;158(5):704-11. 4. Hinton DR, et al. Optic-nerve degeneration in Alzheimer's disease. N Engl J Med. 1986 Aug 21;315(8):485-7. 5. Sadun AA, Bassi CJ. Optic nerve damage in Alzheimer's disease. Ophthalmology. 1990 Jan;97(1):9-17. 6. Archibald NK, et al. The retina in Parkinson's disease. Brain. 2009 May;132(Pt 5):1128-45. 7. Paus S, et al. Bright light therapy in Parkinson's disease: a pilot study. Mov Disord. 2007; 22(10):1495-8. 8. Dowling GA, et al. Effect of morning bright light treatment for rest-activity disruption in institutionalized patients with severe Alzheimer's disease. Int Psychogeriatr. 2005; 17(2):221-36. 9. Berson DM, et al. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002 Feb 8;295(5557):1070-3. 10. Hattar S, et al. Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science. 2002 Feb 8;295(5557):1065-70. 11. Hannibal J, Fahrenkrug J. Neuronal input pathways to the brain's biological clock and their functional significance. Adv Anat Embryol Cell Biol. 2006;182:1-71. 12. Harnois C, Di Paolo T. Decreased Dopamine in the Retinas of Patients with Parkinson's Disease. Investigative Ophthalmology & Visual Science, 1990; 3 1(11) 13. Vugler AA, et al. Dopamine neurones form a discrete plexus with melanopsin cells in normal and degenerating retina. Exp Neurol. 2007 May; 205(1):26-35. 14. Sakamoto K, et al. Dopamine regulates melanopsin mRNA expression in intrinsically photosensitive retinal ganglion cells. Eur J Neurosci. 2005 Dec;22(12):3129-36. 15. Zhang DQ, et al. Wong KY, Sollars PJ, Berson DM, Pickard GE, McMahon DG. Intraretinal signaling by ganglion cell photoreceptors to dopaminergic amacrine neurons. Proc Natl Acad Sci U S A. 2008 Sep 16;105(37):14181-6. 16. La Morgia C et al. Melanopsin-containting retinal ganglion cells and circadian phototransduction are spared by Neurodegeneration in Mitochondrial Optic Neuropathies. Neurology March 17, 2009 Suppl 3 A183 (P04.075) 17. La Morgia C et al. Light-induced melatonin suppression in Mitochondrial Optic Neuropathies. Arvo Annual Meeting, May 3-7 2009 Fort Lauderdale, Florida N. 5663 Nome Prof. Roberto Lucchini Contatti Tel +39030 3996604 Fax +39030 3996080 Email: [email protected] Istituto/Dipartimento Dipartimento di Medicina Sperimentale e Applicata, Sezione di Medicina del Lavoro, Università di Brescia, P.le Spedali Civili 1, 25123 Brescia Proposta di ricerca Area di interesse identificata Gene-metal interaction as a determinant of neurodegenerative diseases Finanziamenti ricevuti 1. Titolo progetto PHIME - Public health impact of long-term, low-level mixed element exposure in susceptible population strata Ente finanziatore EU 6th Frame Program (www.phime.org) Durata progetto 2006-2011 Abstract del progetto 2.Titolo progetto PHIME: Gene-environment interaction on manganese induced parkinsonism are studied in children and elderly resident in areas with environmental exposure to heavy metals Epigenomics Studies of Toxic Hazards in Environmental health Research Ente finanziatore CARIPLO Foundation, Italy Durata progetto 2008-2011 Abstract del progetto Ente finanziatore This is an interdisciplinary program to conduct in-vitro and human investigations to evaluate the role of DNA methylation and histone modifications in mediating the health effects of metals and other environmental exposures PROFILO BIOLOGICO E GENETICO DELLA DISFUNZIONE DEI METALLI NELLA MALATTIA DI ALZHEIMER E NEL ‘MILD COGNITIVE IMPAIRMENT’ Ministero del Lavoro della Salute e delle Politiche Sociali Durata progetto 2009-2011 Abstract del progetto Several cross-sectional as well as cohort studies corroborate the concept of metal implication in Alzheimer’s disease (AD), including clinical observations suggesting that higher-than normal levels of copper (Squitti et al, Neurol 2002) and in particular ‘free’ copper – the quantity of serum copper which is not bound to ceruloplasmin (Squitti et al, Neurol 2005, 2006) – can distinguish AD from healthy controls. Here we tested the hypothesis that this is the case also for the classification of healthy elderly subjects vs. Mild Cognitive Impairment (MCI) which represents an intermediate cognitive state between ‘normalcy’ and full dementia. 3.Titolo progetto ABSTRACT Exposure to heavy metals can induce neurodegenerative effects in humans (Zatta et al., 2003). Various metals like aluminium, manganese, iron, copper, zinc are likely to play important roles in the pathogenesis of neurodegenerative illnesses such as Alzheimer’s Dementia (AD) and Parkinson’s Disease (PD). In particular for AD - the most common form of dementia in the elderly -a consensus has been recently established in the Metal Hypothesis of AD (Bush and Tanzi 2008). This hypothesis suggests that the interaction of Abeta – the major component of the senile plaques - with specific metals - iron, zinc and especially Cu - can drive Abeta pathogenicity and AD development and progression. Numerous clinical studies, including some from our group (Squitti et al, 2002, 2006, 2009) support this hypothesis. In particular, we have identified in AD patients a serum-level increase in the Cu quantity that does not bind to ceruloplasmin (free Cu) (Squitti et al, 2005), which correlates with the typical deficits, cerebrospinal markers (Squitti et al, 2006) and with a worse prognosis of the disease (Squitti et 2009a). The interaction between environmental exposure with genetic susceptibility is suspected as an important determinant of these chronic diseases as well (Wright and Baccarelli 2010; Baccarelli e Bollati, 2009). In fact, if on one hand metabolic dysfunction could have an effect on metal disbalance – especially at the liver lever, being the liver the controlling organ of metal homeostasis, on the other hand epigenetic factors have been highlighted in conjunction with oxidative stress mechanism to be triggered by exposure to neurotoxicants like heavy metals (Zawia et al., 2009). In particular, the effects of metal exposures in early life and throughout the developmental stages could account for clinical manifestations in older age. Our group has produced seminal contributions showing that exposure to metals (Wright and Baccarelli 2010; Tarantini and Baccarelli 2009) modifies global and gene-specific methylation states and that these changes can be detected in blood DNA. Epidemiological assessment needs accurate reconstruction of long-term and lifetime exposure, possibly with exposure biomarkers able to reflect long-term cumulative mechanism of neurotoxicity. Our group has developed various research projects in these areas, covering different aspects of metal exposure as determinant of AD (Papaleo et al., 2004) and PD (Lucchini et al., 2007; Squitti et al., 2009b). The understanding of exposure related determinants have important implication in terms of possible reduction of the epidemiological impacts of neurodegenerative diseases, through preventive intervention. The line of research we are interested in focuses on an integration of all the aspects – exposure-related as well as the metabolic ones - of the metal implication in neuro-degenerative disorders. In particular, we will explore the genetic and epigenetic influence on AD and PD patients exposed to heavy metals in the highly industrialized province of Brescia, and the hypothesis that gene controlling metal metabolism – such as the Wilson’s disease ATP7B gene - which controls the free copper levels in the body, the hemochromatosis HFE gene and Transferrin gene - involved in the regulation of body iron - arbour susceptibility loci for late-onset AD as well . Moreover, to provide proof-of-concept that a decoppering approach has a beneficial effect on AD progression, we are currently starting a large randomized controlled clinical trial based on the concept that metal complexing or ligand agents properly tune the redistribution of metals in the body. The influence of genes like parkin and PARK9 will also be assessed in relation to exposure to neurotoxic metals like manganese. Assessment of cumulative exposure (Sinha and Mark, 2005) to metals will be based on geospatial approach and will be applied to AD and PD patients and age-sex matched controls from geographical areas with different environmental exposures to heavy metals. REFERENCES Baccarelli A, Bollati V. Epigenetics and Environmental Chemicals. Curr Opin Pediatr 2009; 21: 243-51. Lucchini R, Albini E, Benedetti L, Borghesi S, Coccaglio R, Malara E, Parrinello G, Garattini S, Resola S, Alessio L, High Prevalence Of Parkinsonian Disorders Associated To Manganese Exposure In The Vicinities Of Ferroalloy Industries. Am J Ind Med 2007; 50: 11: 788-800 Papaleo B, Alessio, Apostoli P, Battista G, Benedetti F, Lucchini R, Pasqualetti P. Alzheimer disease: study of occupational risk factors [in Italian]. G Ital Med Lav Ergon 2004;26(4):310-312 Bush AI, Tanzi RE. Therapeutics for Alzheimer's disease based on the metal hypothesis. Neurotherapeutics. 2008 Jul;5(3):421-32. Review Squitti R, Lupoi D, Pasqualetti P, Dal Forno G, Vernieri F, Chiovenda P, Rossi L, Cortesi M, Cassetta E, Rossini PM. Elevation of serum copper levels in Alzheimer's disease. Neurology. 2002 Oct 22;59(8):1153-61.. Squitti R, Pasqualetti P, Dal Forno G, Moffa F, Cassetta E, Lupoi D, Vernieri F, Rossi L, Baldassini M, Rossini PM. Excess of serum copper not related to ceruloplasmin in Alzheimer disease. Neurology. 2005 Mar 22;64(6):1040-6. Squitti R, Barbati G, Rossi L, Ventriglia M, Dal Forno G, Cesaretti S, Moffa F, Caridi I, Cassetta E, Pasqualetti P, Calabrese L, Lupoi D, Rossini PM. Excess of nonceruloplasmin serum copper in AD correlates with MMSE, CSF [beta]-amyloid, and h-tau. Neurology. 2006 Jul 11;67(1):76-82. Squitti R, Bressi F, Pasqualetti P, Bonomini C, Ghidoni R, Binetti G, Cassetta E, Moffa F, Ventriglia M, Vernieri F, Rossini PM. Longitudinal prognostic value of serum "free" copper in patients with Alzheimer disease. Neurology. 2009a 6;72(1):50-5. Squitti R, Gorgone G, Panetta V, Lucchini R, Bucossi S, Albini E, Alessio L, Alberici A, Melgari JM, Benussi L, Binetti G, Rossini PM, Draicchio F. Implications of metal exposure and liver function in Parkinsonian patients resident in the vicinities of ferroalloy plants. J Neural Transm. 2009b Oct;116(10):1281-7 Tarantini L, Bonzini M, Apostoli P, Pegoraro V, Bollati V, Marinelli B, Cantone L, Rizzo G, Hou LF, Schwartz J, Bertazzi PA, Baccarelli A. Effects of Particulate Matter on Genomic DNA Methylation Content and iNOS Promoter Methylation. Environ Health Perspect 2009; 117: 217-22. Wright RO, Schwartz J, Wright RJ, Bollati V, Tarantini L, Park SK, Hu H, Sparrow D, Vokonas P, Baccarelli A. Biomarkers of Lead Exposure and DNA Methylation within Retrotransposons. Environ Health Perspect, online ahead of press Zatta P, Lucchini R, van Rensburg SJ, Taylor A.The role of metals in neurodegenerative processes: aluminum, manganese, and zinc. Brain Res Bull. 2003 Nov 15;62(1):15-28. Zawia NH, Lahiri DK, Cardozo-Pelaez F. Epigenetics, oxidative stress, and Alzheimer disease. Free Radic Biol Med. 2009 May 1;46(9):1241-9. Epub 2009 Feb 23. Nome Stefano Mattioli Contatti Occupational Medicine, S.Orsola-Malpighi University Hospital via Pelagio Palagi 9, I-40138 Bologna, Italy. Tel: +39-051 636 2761 Fax: +39-051 636 2609 E-mail: [email protected] Istituto/Dipartimento Section of Occupational Medicine, Department of Internal Medicine, Geriatrics and Nephrology, University of Bologna, Italy Proposta di ricerca Area di interesse identificata Using prevention to reduce the burden of Alzheimer’s disease (and other ageing diseases) Finanziamenti ricevuti Titolo progetto Occupational exposure to Valsalva manoeuvre and risk of Alzheimer disease Ente finanziatore None Durata progetto 2-3 Years (6 months for Study design and questionnaire preparation; at least 1 year for data collection; 6 month for data cleaning, quality controls and statistical analysis; 6 month for writing the manuscript and internal revision process) Abstract del progetto Introduction Chronic increased intracranial pressure or exposure to repetitive intermittent intracranial pressure elevations might contribute to Alzheimer’s disease (AD) via damage to the choroid plexus resulting from cumulative effects of repetitive intermittent intracranial pressure elevations, and leading to a reduction in cerebrospinal fluid (CSF) production, and hence to diminished CSF clearance of neurotoxins such as β-amyloid [Wostyn, Can chronic increased intracranial pressure or exposure to repetitive intermittent intracranial pressure elevations raise your risk for Alzheimer’s disease? Med Hypotheses 2004;62:925–30, 2004]. Weightlifters, glassblowers and wind instrument musicians can generate very high intracranial pressures due to repeated Valsalva manoeuvres (elevated subglottic pressures), which impair venous return from the head [Greenfield et al., Transient changes in cerebral vascular resistance during the Valsalva maneuver in man. Stroke 1984;15:76–9]. Herein, occupational exposure to Valsalva manoeuvres might be a risk factor for Alzheimer disease. Our aim will be the conduction of an etiological study to test the hypothesis of causal link between occupational history of frequent Valsalva manoeuvres and the onset of Alzheimer diseases in later ages. Methods We think that a case-control study design would be feasible to our purpose. Firstly, our hypothesis implies a long latency between exposure and disease onset. Moreover, regular lifting is a common task among manual workers. In a previous study conducted in Bologna, we estimated a lifetime prevalence of manual lifting works around 32% [Mattioli et al., Physical exertion (lifting) and retinal detachment among people with myopia. Epidemiology. 2008 Nov;19(6):868-71]. Considering the frequency of Alzheimer disease (10,000 new cases registered in Emilia-Romagna every year) we can recruit only incident cases, in order to minimize recall bias. Selection of cases will be conducted in cooperation with Neurological Clinic of Bologna; collaboration of relatives will be asked in order to properly collect information. Selection of controls (2 for each case) will be based on random sampling from the national health service registries. For data collection, we will use a standardized questionnaire designed for assessment of a series of potential occupational and non-occupational risk factors, based on those proposed in the literature. In particular, weight lifting will be assessed both by analysis of occupational history and by asking specific questions. Multivariate logistic regression analyses will be conducted to study the relationship between past occupational exposure to Valsalva manoeuvres and Alzheimer disease. Conclusions Chronic repetitive Valsalva manoeuvres have been proposed has a possible cause of Alzheimer disease. Weight lifting, a very common task for manual workers, might play an important role in Alzheimer diseases aetiology. If confirmed, this hypothesis could have important impact on the global burden of the disease. Indeed these findings could have significant implications in prevention, with important consequences on health and socio-economic fields. Epidemiological studies are needed to test this hypothesis and to asses other possible etiologic factors that could be implicated in the pathologic mechanism [Wostyn, 2004]. Nome Contatti Istituto/Dipartimento Giuseppe Mele (Professore Aggregato di Chimica) [email protected] Studio: 0832-297281 Cellulare: 333-9593228 Dipartimento di Ingegneria dell’Innovazione-Università del Salento Proposta di ricerca Area di interesse identificata Costruzione di un “Alzheimer Web-GIS” che consenta di georeferenziare casi di morbo di Alzheimer su aree campione associando ad ogni caso in esame una “base di dati” contenente parametri geografici e ambientali allo scopo di capire “se possibile” le correlazioni tra i parametri (geografici, scientifici, medici, sociali) ed i casi oggetto di studio. Nella costruzione del “Web_GIS”, sebbene in ambiti diversi, si potrebbe trasferire il know-how acquisito nei due progetti di seguito riportati di cui il proponente è stato Responsabile Scientifico. Finanziamenti ricevuti Titolo progetto (1) Acronimo ECODO-NET "Development of a model Web based virtual observatory of Acherontas, Kalamas and Torre Guaceto ecosystems and its application as a mobile exhibit and permanent environmental kiosk towards public awareness and sustainable development of coastal ecosystems (EcoDo-net)" focuses on the improvement of the management, the protection and the sustainable development of the Greek and Italian coastal ecosystems in the border area (Acherontas, Kalamas, their intermediary coastal zone and -Torre Guaceto) through the observation of the ecosystems' quality and the increase of public awareness about environmental issues. Programma Interreg IIIA Italia-Grecia 2000-2006 Asse III. Ambiente e Patrimonio Culturale Misura 3.1 Miglioramento della gestione degli ecosistemi Ente finanziatore FESR-Stato-Regione Puglia Durata progetto (1) 2 anni gennaio /2007-Dicembre/2008 Abstract del progetto (1) The program "Development of a model Web based virtual observatory of Acherontas, Kalamas and Torre Guaceto ecosystems and its application as a mobile exhibit and permanent environmental kiosk towards public awareness and sustainable development of coastal ecosystems (EcoDonet)" focuses on the improvement of the management, the protection and the sustainable development of the Greek and Italian coastal ecosystems in the border area (Acherontas, Kalamas, their intermediary coastal zone and -Torre Guaceto) through the observation of the ecosystems' quality and the increase of public awareness about environmental issues. Titolo progetto (2) Linee di intervento per la valorizzazione delle colture agricole locali della Valle della Cupa -Web GIS (Geographic Information System) as support for sustainable development local economy in the fields of agro-industry and fine chemicals Ente finanziatore Consorzio Universitario Interprovinciale Salentino Consorzio di Comuni della Valle della Cupa 1 anno 2006 Durata progetto(2) Abstract del progetto (2) There is nowadays an increasing interest in the development of natural products and new fine chemicals based on renewable organic materials using sustainable processes. At the same time innovative developments associated to main local productions (e.g. olive oil and wine industries) as well as to the extraction fine chemicals from less important local cultivar (e.g. mulberry, black-berry, bilberry etc.), are of topical importance especially in view of a sustainable “green chemistry”. Emerging technologies for simulating, controlling, and optimizing complex systems will be considered, crossing a long list of disciplines such as, botanical, chemical, computational, material science and process simulation. In this context we report our studies concerning a planned sampling of local agri-food and non-food species (traditional and/or minor cultivar), which could be interesting for this purpose. With this sampling we are able to trace the real distribution of local cultivations and to map these on a Geographic Information System (G.I.S.). Then, each sample will be listed depending on its own biological properties and physical condition of the selected cultivar. Nome Prof. Calogero Caruso MED04 Contatti Immunopatology (preclinical): Calogero Caruso, email: [email protected] Neurology: Roberto Monastero, email: [email protected] Oral Medicine: Giuseppina Campisi, email: [email protected] Istituto/Dipartimento Department of Pathobiology and Medical and Forensic Biotechnologies Proposta di ricerca Area di interesse identificata Experimental (preclinical) research The preclinical group (Immunopathology Unit) is evaluating the role of inflammaging in the expression/progression of the disease. So far the alterations of the inflammatory response and the pro-inflammatory profile of aged people have been studied as risk factors for AD development. This aspect has to be further characterized in terms of the comparison of the genes and proteins involved in determining the proinflammatory profile in healthy old and very old people and in demented (both Alzheimer and non Alzheimer). On the other hand, persistent peripheral multibacteria infection such as periodontitis (PD), associated with gram-anaerobic bacteria capable of exhibiting localized and systemic infections in the host, has been proposed as possible aggravating cofactor in subjects with vascular disease. In PD the balance between bacteria and host response is altered, resulting in a production of high levels of proinflammatory mediators and low levels of anti-inflammatory ones. Data regarding the PD association with neurodegenerative disorders are lacking; however, it seems plausible that, during PD, cytokines increase in AD brain due to stimulation of trigeminal nerve fibres, or directly from bacterial products. So, the Immunopathology group in association with the Oral Medicine group is going to gain insight into this relationship, using biological samples collected from the Neurological group. Many studies have reported different alterations of the immune system in AD, and the involvement of the acquired branches of the immune system. To investigate the systemic signs of immune responses in AD, the Immunopathology group is also analyzing the expression of activation markers on peripheral blood mononuclear cells from AD patients and age-matched healthy controls activated in vitro by recombinant amyloid peptide (rAβ42) and their cytokine production. This kind of study is also useful to obtain biomarkers of AD for monitoring the effectiveness of therapeutic interventions. Clinical research Risk factors for Mild Cognitive Impairment (MCI) - which may precede the clinical detection of AD and dementia - and predictors for the conversion from MCI to dementia are currently evaluated by the Neurological group. For this purpose, hospital-based data and crosssectional as well as prospective data from a population-based cohort are used. In particular, modifiable (non-genetic) and non-modifiable (genetic) risk factors are under evaluation. • Concerning modifiable risk factors, three etiologically-driven hypotheses are currently carried out: 1. The role of modifiable vascular risk factors (eg, diabetes, hypertension, dyslipidemia, obesity), alone or clustered, in the concept of metabolic syndrome are under investigation; 2. The role of behavioural symptoms, particularly depression, in determining the risk of MCI and its progression to dementia/AD are under evaluation; 3. The effects of oxidative stress and vitamin E on the development of MCI and dementia/AD, and their effect in the progression from MCI to dementia/AD are in assessment. • Concerning non-modifiable risk factors, we are going to evaluate the role of recently proposed and novel putative genes involved in AD. We hypothesize to include a large sample sample of AD and control subjects (case:control of 1,000:1,000); accordingly, this study would benefit from a valid statistical power in detecting novel potential genetic markers for AD. These genes will be evaluated alone, in clusters and in conjunction with the APOE4 allele. This study will be performed in association with Immunopathology Unit. Drugs currently in use to treat AD are effective only in 20% of patients; their therapeutic effect is predominantly under genetic control. Thus, these data would encourage new research in pharmacogenomics. Finanziamenti ricevuti Principal Investigator: Dr. Roberto Monastero ; Amount of fund: 590,000 euros Titolo progetto Ente finanziatore Epidemiology and risk factors for Mild Cognitive Impairment, Alzheimer's disease and dementia. The Zabut Aging Project: 5-year follow-up study on a rural Sicilian population Ministero della Salute, Italia Durata progetto 3 years Abstract del progetto We would extend recent research on risk factors for Mild Cognitive Impairment (MCI) and Alzheimer's disease (AD) and the progression of MCI to dementia in the elderly with a special focus regarding modifiable (non-genetic) and non-modifiable (genetic) risk factors. Baseline (n=1930 subjects) and 5-year population-based data (we are expecting to enrol approximately 1500 subjects) will, therefore, be used. Firstly, the role of vascular, modifiable vascular risk factors, alone or clustered in the concept of metabolic syndrome would be investigated. Secondly, the role of behavioural symptoms, particularly depression, in determining the risk of MCI and its progression to dementia/AD would be evaluated. Thirdly, the effects of oxidative stress and vitamin E on the development of MCI and dementia/AD, and their effect in the progression from MCI to dementia/AD would be assessed. Lastly, this project would evaluate the role of recently proposed and novel putative genes involved in AD. Roberto Monastero, MD, PhD Lab of Epidemiology and Psychology of Aging and Dementia Dept of Clinical Neuroscience University of Palermo Via La Loggia 1 90129 - Palermo, Italy Tel. +39-091-6555185 Fax. +39-091-6555113 email. [email protected] [email protected] Type: Progetto Giovani Ricercatori Ministero Salute, 2007 Title: Epidemiology and risk factors for Mild Cognitive Impairment, Alzheimer's disease and dementia. The Zabut Aging Project: 5-year follow-up study on a rural Sicilian population Principal Investigator: Dr. Roberto Monastero Amount of fund: 590,000 euros Lenght: 3-year project Abstract We would extend recent research on risk factors for Mild Cognitive Impairment (MCI) and Alzheimer's disease (AD) and the progression of MCI to dementia in the elderly with a special focus regarding modifiable (non-genetic) and non-modifiable (genetic) risk factors. Baseline (n=1930 subjects) and 5-year population-based data (we are expecting to enrol approximately 1500 subjects) will, therefore, be used. Firstly, the role of modifiable vascular risk factors, alone or clustered, in the concept of metabolic syndrome would be investigated. Secondly, the role of behavioural symptoms, particularly depression, in determining the risk of MCI and its progression to dementia/AD would be evaluated. Thirdly, the effects of oxidative stress and vitamin E on the development of MCI and dementia/AD, and their effect in the progression from MCI to dementia/AD would be assessed. Lastly, this project would evaluate the role of recently proposed and novel putative genes involved in AD. Due to the large, hypothesized sample of AD and control subjects (about 1,000 for each group), this study would benefit from a valid statistical power in detecting novel potential genetic markers for AD. These genes would be evaluated alone, in clusters and in conjunction with the APOE4 allele. Nome Emilio Di Maria, MD PhD Contatti [email protected]; +39 0105634368 Istituto/Dipartimento Dept. of Neuroscience, Ophthalmology and Genetics - University of Genova and Division of Medical Genetics, Galliera Hospital, Genova, Italy Proposta di ricerca Area di interesse identificata Problem statement: The clinical manifestations of Alzheimer’s disease (AD) often include behavioural disturbances, such as apathy and mood disorders, as well as psychotic symptoms (delusions and hallucinations). These features, which significantly affect the clinical outcome, are globally referred to as behavioural and psychological symptoms in dementia (BPSD). BPSD are recognised to cause a poorer course of disease, contributing to increase the social, pharmacological, and managing costs of AD. Family studies have reinforced the hypothesis that the occurrence of psychosis in AD patients is partially determined by genetic factors. The present proposal is in accordance to the priorities listed in the letter of intent for the joint initiative, namely: Genetic susceptibility to Alzheimer’s disease Biomarkers Studying early onset forms of Alzheimer’s disease to follow-up disease progression. Objective: to identify genetic risk factors responsible for the occurrence of behavioural and neuropsychiatric symptoms in patients with Alzheimer’s disease. We intend to pursue this objective by using both main approaches: i) genome-wide association studies; ii) candidate gene approach, in that the analysis will be performed only on genetic markers which were demonstrated to be associated with neurodegeneration or psychiatric disorders. To accomplish this plan, a network of Italian clinical centres and laboratory of genetics with distinguished expertise in the field has already been built. Preliminary results: A large cohort of patients with AD has been recruited through a multicentre effort. Patients were assessed at their admittance with a complete sociodemographic and clinical data collection. The study protocol was approved by the Institutional Review Boards. Patients enrolment is still ongoing. By applying the candidate gene approach outlined above, we have already demonstrated that the DAOA gene is a genetic risk factor for the occurrence of psychotic symptoms in AD [Di Maria et al, J Alzh Dis 2009]. Relevance: The identification of genetic factors influencing the occurrence of BPSD in AD will shed new light on the disease pathophysiology and allow a better prediction of the illness. On the basis of new lines of evidence, earlier and tailored treatments could be appropriately administered. Finanziamenti ricevuti Note: the Italian network on behavioural and psychological symptoms in dementia, quoted in the proposal section, has not been formally assembled. Therefore, the funded project granted to other researchers involved could not be reported. Titolo progetto MacAge – MCI-AD Conversion: Analysis of Genetic Effects. A cohort Ente finanziatore association study on the role of tau gene haplotypes and ApoE in determining the evolution from mild cognitive impairment to Alzheimer’s disease. CARIGE Foundation, Italy Durata progetto 1 year Abstract del progetto Background: The association of microtubule-associated tau protein gene (MAPT) with Alzheimer’s disease (AD) had been controversial, but recent literature demonstrated that the MAPT H1c haplotype is a genetic risk factor for AD [Myers AJ, 2005]. H1c haplotype is associated with increased tau expression and predisposes, in late-onset AD, to the unbalance between the 4-repeat- and the 3-repeat-containing isoforms (4rep/3rep), thus leading to tau dysfunction and deposition [Myers AJ, 2007]. A distortion of the 4rep/3rep expression pattern was also found in cholinergic neurons from mild cognitive impairment (MCI), a clinical condition which is prodromal to AD [Ginsberg SD, 2006]. Reducing tau levels in mice protects against amyloid-β-induced neuronal dysfunction [Roberson ED, 2007]. Preliminary results: We previously demonstrated that the tau H1 haplotype is a genetic risk factor for corticobasal degeneration, a sporadic tauopathy [Di Maria E, 2000]. More recently, a longitudinal study (PI: Prof. M. Tabaton) characterised individuals with amnestic MCI (aMCI), in order to find biological markers associated with the risk of AD [Assini A, 2004; Odetti P, 2005]. To date, 224 individuals with aMCI were recruited. We foresee to collect within the first phase of the project a cohort of at least 240 cases: all followed-up for 1 year, about ⅓ for ≥2 years. Objective: To evaluate the hypotheses that the evolution from MCI to AD is influenced by the tau locus, and that its effect is mediated by a qualitative and quantitative unbalance in the expression of the tau gene isoforms. The specific aims will be accomplished according to the following experimental plan (total duration: 1 year). - Patients evaluation: i) patients follow-up, blood sampling (according to the current protocol for the longitudinal study); ii) DNA, plasma, serum, cell lines banking (Galliera Genetic Bank). - Cohort association study: i) the tau tagging polymorphisms will be genotyped in the case series; ii) the association study will investigate the effect of the tau haplotypes on the MCI-AD transition, taking into account ApoE genotypes, as well as age, age at onset, gender and possible interacting variables. - Gene expression study: expression level of the tau isoforms will be investigated in lymphoblasts from patients belonging to the two subsets (MCI and MCI-AD converted) and carrying different tau genotypes. Relevance and deliverables: The identification of genetic factors influencing the evolution from MCI to AD will shed new light on the disease pathophysiology and allow a better prediction of the disease progression. On the basis of new lines of evidence, earlier and tailored therapeutic approaches can be envisaged. Nome Contatti Tel. E mail Istituto/Dipartimento Prof. Carlo Caltagirone Via Aretina, 306 – 00179 Rome 05 51501409 [email protected] IRCCS Fondazione Santa Lucia – Laboratory of Clinical and Behavioral Neurology. Proposta di ricerca Area di interesse identificata Preclinical Diagnosis Finanziamenti ricevuti Titolo progetto Ente finanziatore Markers to diagnose neurodegenerative diseases and of new targets to test the pharmacological treatments. Italian Ministry of Health Durata progetto 2006-2008 Abstract del progetto The present investigation involved 200 individual with AD, whose cognitive functioning was monitored along a 18-months period by means of administration of the Mini Mental State Examination. The results document, on one side, a progressive decline of cognitive performance and, on the other side, a significant relationship between the occurrence of some specific genome variants and both the severity and the earlier onset of disease. In particular, the presence of i) allele e4 of ApoE polymorphism, of ii) allele T of the polymorphism of the serotonin receptor 5HT2A (102T/C), of iii) some variants of GST genes as well as of the iv) polymorphism -137C/C of the promoter region for IL-18, significantly influence both the onset and the clinical disease course. Moreover, in line with the above data, the analysis of the cellular components of innate immunity confirmed that the AD patients’ immune system cells are more likely to produce phlogistic factors such as IL-6 and IL-18. Furthermore, it has also been showed that the previously reported decrease of proteinic level of ADAM10 within the AD patients’ platelets, is not owed to reduced levels of ADAM10 mRNA but, rather, to others events that involve the translation processes or the proteolysis of the molecule itself. Finally, groundbreaking in vivo and in vitro animals studies indicate that the Tat-Pro ADAM peptide can produce alterations of the molecular composition of the glutamatergic synapses, affecting the synaptic functioning. In conclusion, the present study adds some valuable information about the role played by some disease mediators, such as early inflammatory episodes and synaptic changes, thus, helping to identify new molecular cascades and cellular processes involved in the pathogenesis of neurodegeneration and, likely, to be used as markers of the disease. Indeed, by means of project here presented, we were allowed to implement an interdisciplinary work to explore the physio-pathogenetic pathways in AD, involving a wide number of individuals with AD that belong to several Italian centres. As a matter of fact, in the view of the multidisciplinary network, relying upon the synergistic coordination between basic and clinical researchers, the involvement of the most representative ITINAD units allowed us to reach results that significantly advance our understanding of AD pathogenesis. Moreover, the same results might represent relevant clues to individuate an advanced strategy in order both to early diagnose AD and to formulate an accurate prognosis. Nome Manservigi Roberto Contatti Tel. +39 0532 455401/455412 Fax +39 0532 974470 Mail: [email protected] Istituto/Dipartimento Department of Experimental and Diagnostic Medicine Proposta di ricerca Area di interesse identificata Neurodegenerative Diseases Finanziamenti ricevuti Titolo progetto Search of herpetic proteins involved in the pathogenesis of Alzheimer. Ente finanziatore MiUR Durata progetto 2006-2008 (two years) Abstract del progetto The overall objective of this proposal is to study the correlation between HSV-1 infection and Alzheimer’s disease (AD) and to understand those HSV-1 functions that are involved in the pathogenesis of AD. : Finanziamenti ricevuti Titolo progetto Ente finanziatore Search of herpetic proteins involved in the pathogenesis of Alzheimer's disease for the development of new therapeutic strategies MiUR Durata progetto 2008-2010 (two years) Abstract del progetto Despite the very numerous studies on Alzheimer's disease (AD), especially on amyloid plaques and neurofibrillary tangles, little information has been obtained thus on the cause of the disease. Moreover, different experimental evidences define a role for Herpes simplex virus type 1 (HSV1) infection as a risk factor for AD development. The goal of this project is to study and characterize the different interactions between viral proteins and the disease development, in order to obtain a better understanding of AD origins and, above all, to develop innovative therapies. 3. HUMAN/CLINICAL RESEARCH B. Epidemiology genetics Nome Contatti Istituto/Dipartimento Luisa Benussi, Giuliano Binetti 0339-030-3501725 [email protected] IRCCS “Centro S. Giovanni di Dio-Fatebenefratelli” NeuroBioGen LabMemory Clinic, Brescia Italy Proposta di ricerca Alzheimer disease (AD) is a genetically complex and heterogeneous disorder. Less than 10% of AD patients are familial with an autosomal dominant inheritance of the disease (FAD). In autosomal dominant AD families, rare mutations in the amyloid precursor protein gene, and the presenilin 1 and 2 genes have been identified. In sporadic patients the disease is expected to result from the interaction of multiple susceptibility genes and unknown environmental factors. So far, the e4 allele of the apolipoprotein E (APOE) gene has been the only consistently replicated genetic risk factor for late-onset AD. Through genome wide association studies (GWAS), a novel opportunity of identifying dementia risk genes and associated disease pathways has emerged with several potential AD risk genes already reported. Further validation and replication in other patients cohort combined with functional characterization of these genes could lead to a better understanding of the pathophysiology underlying dementia and open new therapeutic possibilities for AD. In the last years our group has contributed to: 1) the identification of genetic factors that influence the disease risk and the pharmacological response in AD patients (Nicosia et al., Dement Geriatr Cogn Disord. 2001;Ventriglia et al, Mol Psychiatry 2002; Scassellati et al, Neurosci Lett 2004; Ventriglia et al, Aging Clin Exp Res 2005; Emanuele et al, NBA 2009; Di Maria et al, JAD 2010; Santoro et al, CNS Drugs 2010); 2) the identification of mutations that are associated with familial forms of AD (Finckh et al, Am J Hum Genet 2000; Finckh et al, Neurology 2000; Binetti et al, Neurosci Lett 2003;Binetti et al, Ann Neurol 2003; Signorini et al, Curr Alzheimer Res 2004; Alberici et al, Eur J Neurol 2007;Ghidoni et al, JAD 2009); 3) the assessment of areas of intervention for genetic counseling of dementia by a crosscultural comparison between Italians and Americans (Binetti et al., PEC 2006). The specific objectives of this proposal are: 1) Identification of genetic factors that influence the disease susceptibility and the pharmacological response in AD patients (genome wide association studies); 2) Identification of new mutations and novel AD-associated loci (genetic screening; linkage analysis; quantitative trait - proteomic profiles- linkage analysis); 3) Evaluating the psychosocial impact of genetic testing in families with hereditary forms of AD. To carry out this project our group will collaborate with: Dr Roberta Ghidoni (Proteomics Unit, IRCCS Fatebenefratelli, Brescia) on genetic and proteomic studies; Dr F Tagliavini (IRCCS Fondazione Istituto Carlo Besta, Milano), Dr D Galimberti and Dr E Scarpini (University of Milan) on genetics and genetic counseling; Prof C Franceschi (Università di Bologna), Dr G Forloni (Istituto Mario Negri, Milano) on genetics and pharmacogenetics. Our Group, along with the mentioned National collaborators, can offer to the proposed project the following resources: 1) Clinical expertise in Alzheimer disease; 2) A large collection of biological samples from sporadic and familial AD patients (more than 1000 index patients) and a similar number of age-matched controls; 3) A large collection of families with autosomal dominant inheritance trait for AD (more than 90 families); 4) Genetic testing as well as a genetic counselling program for families with hereditary forms of dementia. Area di interesse identificata Genetic susceptibility to Alzheimer’s disease and genome wide association studies Finanziamenti ricevuti Titolo progetto Project1: Validation of genetic and biochemical markers for early diagnosis of AD and the prediction of conversion of MCI into AD, and Ente finanziatore Durata progetto Abstract del progetto design of a multivariate molecular protocol having high diagnostic and prognostic accuracy. Project2: Marker biologici di neurodegenerazione utilizzabili nella pratica clinica ai fini della diagnosi precoce e differenziale, della individuazione delle forme con risposta ottimale alle attuali terapie Project3:GenEtica- Profili bioetici e biogiuridici della genetica tra ricerca sperimentale, consulenza e prospettive terapeutiche. Project1:Italian Ministry of Health Progr Strategico PS39, prog 1 Project2:Italian Ministry of Health Progr Strategico conv.71 prog 6 Project3: Italian Ministry of University and Research, FIRB2006 Project1:01-01-2009/31-12-2010 Project2:7-01-2008/7-04-2010 Project3:15-10-2008/15/10/2011 Project 1 and Project 2 aims: 1) The development and validation of a multi-factorial protocol that integrates molecular, imaging, neurophysiological, neuropsychological and behavioral data for early diagnosis of AD, in particular during the preclinical phase and the prodromal stage of "mild cognitive impairment" when the symptoms are not severe enough to fulfill the current diagnostic criteria for AD. 2) Validation of this new diagnostic protocol within a regional public health network, and evaluation of the health care, organization and economic implications of its transfer to the National Health Service. Project3 aims: 1) to evaluate the psychosocial impact of genetic testing in families with hereditary forms of dementia; 2) to evaluate the efficacy of the pre-test educational session and the information comprehension during genetic counselling 3) analysis of the ethical issues arising from a genetic counselling service for dementia Nome Contatti Istituto/Dipartimento Maria Del Zompo Phone: +39 070 6092438 Fax: +39 070 653584 Email: [email protected] Department of Neurosciences “B.B. Brodie”, University of Cagliari, Cagliari, Italy Proposta di ricerca Area di interesse identificata - Genetic susceptibility to Alzheimer’s disease and genome wide association studies - Biobanking (blood sample, CSF, brain repository…) (In the Biobank of the Section of Clinical Pharmacolgy of the Department of Neuroscience, University of Cagliary, are cryopreserved the lymphoblastoid cell lines of 147 patients with Alzheimer's disease and 116 controls. AD patients of proven Sardinian ancestry (parents, grandparents and great-grandparents) were diagnosed according to DSM-IV, and National Institute of Neurologic and Communicative Disorders and Stroke–AD and Related Disorders Association (NINCDS-ADRDA) criteria for possible or probable AD (McKhann et al, 1984 ). Cognitive screening was performed by means of Mini-Mental State Examination (MMSE) (Folstein et al, 1975). All control subjects were cognitively intact volunteers with an MMSE score ≥ 26 and with negative family history for dementia.) 3. HUMAN/CLINICAL RESEARCH C. Biomarkers and early diagnosis and imaging Nome Contatti Istituto/Dipartimento Proposta di ricerca Area di interesse identificata Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Finanziamenti ricevuti Titolo progetto Patrizia Mecocci [email protected] +39 075 5783270 Sezione di Gerontologia e Geriatria Dipartimento di Medicina Clinica e Sperimentale Università degli Studi di Perugia 1)Studies on plasma biomarkers of oxidative/nitrosative stress, inflammation, lipid and glucose metabolism, in older adults, both in population based and clinical based longitudinal studies. Samples (already collected or to be collected) derived from Italy and other European countries, with which we are collaborating, to develop easily available biomarkers that can be used in the elderly to: a) study age-related changes in metabolism, also in relation to lifestyle (i.e. diet, physical activity); b) obtain diagnostic and prognostic tools, which can be used in different age-related disorders (cognitive decline/dementia; multimorbidity; disability). The main goal is to obtain noninvasive tools that can be used in age-related disorders for i) investigating pathogenetic mechanisms; ii) diagnosis, prognosis and monitoring of preventative and therapeutic interventions. Age-related cognitive decline and dementia are one of our main research areas, and our studies integrated plasma biomarkers and neuroimaging data. 2) Studie on cellular and animal models of aging, using markers of oxidative/nitrosative stress and mitochondrial structure and activity to evaluate the effects of novel therapeutics (chemically modified antioxidants, vitamins, nutriceuticals) that can be used in humans to prevent age-related cognitive disorders. ZINCAGE Contract n° 506850 European Union in the 6th Framework Program (FP6): "Food Quality and Safety", Priority 5, Thematic Area T6 3 years This is an epidemiological study on the influence of diet and lifestyle on healthy ageing, aimed at preventing adult degenerative disease, particularly focusing on cardiovascular and neurodegenerative diseases and also addressing malnutrition of the elderly. InnoMed (Innovative Medicines for Europe) Project. Contract n° 518170. European Union in the 6th Framework Program (FP6): Life Science Health, Priority 5. 3 years The project, led by the European Federation of Pharmaceutical Industry and Associations (EFPIA), addresses the complex issues associated with the future of biomedical research in the EU,and addresses ways of achieving accelerated development of, safe and more effective medicines, aiming to revitalize the European biopharmaceutical research environment. The project is mainly devoted to study cognitive impairment in the elderly and Alzheimer’s disease. K4Care Project (Knowledge-Based Homecare eServices for an Ageing Europe) Ente finanziatore Durata progetto Abstract del progetto Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto Finanziamenti ricevuti Titolo progetto Contract n° 026968 European Union in the 6th Framework Program (FP6): Information, Society, Technology, Priority 2 3 years The main objective of the K4CARE project is to improve the capabilities of the new EU society to manage and respond to the increasing number of senior population requiring a personalized healthcare assistance. The project will capture and integrate the information, skills, expertises, and experiences of specialised centres and professionals of several old and new EU countries, and will incorporate them in an intelligent web platform in order to give service to health professionals, patients, and citizens in general. Chronic conditions, such as dementia, are the main focus of the project. PRIN Oxidative and nitrosative damage of mitochondrial proteins in models of cell senescence and in subjects with Alzheimer's disease: a pre-clinical study on natural forms and new synthetic analogues of vitamin E MIUR 2 years Mitochondria are key targets of oxidative stress with relevance in cell senescence (mitochondrial theory of aging) and possibly in the pathogenetic mechanisms of age-related diseases such as Mild Cognitive Impairment (MCI)and Alzheimer's disease (AD). In this context emerging aspects deal with the study of post-transductional modifications (PTMs) in proteins caused by reactive oxygen or nitrogen species (ROS and RNS, respectively). The aim of this project is to evaluate PTMs in mitochondrial proteins in "in vitro" models of cellular senescence as well as in peripheral cells from subjects with MCI and AD also evaluating the role as protecting agents of natural forms and newly synthesized analogues of vitamin E. Ente finanziatore Young researchers 2007 Epidemiology and risk factors for Mild Cognitive Impairment, Alzheimer’s disease and dementia. The Zabut Aging Project: 5-year follow-up study on a rural Sicilian population. Ministero della Salute Durata progetto 3 years Abstract del progetto The study will evaluate the risk factors for MCI and AD and the progression of MCI to dementia in the elderly with a special focus regarding modifiable (non-genetic) and non-modifiable (genetic) risk factors. Baseline and 5-year population-based data will, therefore, be used. Main topics are i) the role of modifiable vascular risk factors; ii) the role of behavioural symptoms, particularly depression, in determining the risk of MCI and its progression to dementia/AD; iii) the effects of oxidative stress and vitamin E on the development of MCI and dementia/AD; iv) the role of recently proposed and novel putative genes involved in would encourage new research in pharmacogenomics. Nome Prof. Salvatore Monaco Contatti [email protected] 045-8124285/4461 Neuropatologia/Scienze Neurologiche e della Visione Istituto/Dipartimento Proposta di ricerca Dementia is a clinical syndrome characterised by a progressive loss of cognitive abilities and executive functions, with a significant compromise of patient’s social, relational and working functions. Recent data suggest that pathological and biochemical deteriorations take place years before the clinical emergence of the different dementia conditions. The search for novel markers with a high diagnostic sensitivity is therefore one of the major and high-priority objective of dementia research. There are several methodologies that are presently used or under development, including proteomics which has been shown as one of the most promising and useful for the identification of novel markers. Aim of the present program is therefore to identify clinical and biological markers of dementia according to the following experimental strategy: A). To study mild cognitive impairment and Alzheimer’s disease patients by assessing a specific clinical, neuropsychological, genetic and biochemical profile at the individual level. B). To identify novel genotypic markers associated to the specific clinical profile from all individual patients. C). To identify novel CSF and plasmatic phenotypic markers from all patients, by using a proteomic approach. Area di interesse identificata Biomarkers Biobanking Translational research Finanziamenti ricevuti Titolo progetto Disabilità cognitiva e comportamentale nelle demenze e nelle psicosi Ente finanziatore Fondazione CARIVERONA Durata progetto 3 anni Abstract del progetto Nome Rita Casadio, Pier Luigi Martelli Contatti [email protected] [email protected] Istituto/Dipartimento Dipartimento di Biologia Evoluzionistica Sperimentale Proposta di ricerca Genetic association studies have contributed lists of genetic variations (mainly SNPs) that are putatively related with the insurgence and/or the prognosis of the Alzheimer disease (AD). Next generation sequencing techniques will enormously increase the amount of data on genome wide variability associated with the AD. These data will be useful for discovering new genetic markers of the risk and for better understanding the molecular mechanisms underlying the aetiology of the AD. To benefit from these data, an important analysis effort has to be done in two different and complementary directions: 1) prioritisation, for highlighting among all the reported variations those that are more likely to be related to the AD; 2) annotation, for understanding if the variations occur in coding or in regulatory regions of the genome and for discovering their likely molecular effects. Computational methods that are able to integrate information from different sources in a Systems Biology perspective are then essential. In particular we propose to implement a pipeline able to: [DA 1] search if the variations occur in a protein coding region, in a RNA-coding region or in a regulatory element; [DA 2] predict the effect of protein mutations on localization, structure, stability, and function; [DA 3] predict the correlation between protein mutations and diseases; [DA 4] predict the effect of protein mutations on the protein-protein and protein-DNA interactions; [DA 5] identify the mRNAs targeted by miRNAs and to predict the effect of variations in miRNAs; [DA 6] predict the effect of variations in regulatory regions on the binding with transcription factors; [DA 7] analyse the effect of cariations on regulatory and interaction networks and on metabolic and signalling pathways; [DA 8] collect, compare and store the results of previous analyses for different variations. The Biocomputing Group of the University of Bologna developed several methods that can be integrated in the pipeline (see www.biocomp.unibo.it). Among the others, efficient tools of interest in this particular case are devoted to: 1) the structural and functional annotation of protein sequences (BAR: Bartoli et al. 2009); 2) the prediction of membrane protein topology (ENSEMBLE: Martelli et al. 2003); 3) the prediction of perturbations in protein stability upon mutations (I-Mutant: Capriotti et al. 2008); 4) the prediction of the correlation between protein mutations and diseases (SNPs&GO: Calabrese et al. 2009); 5) the protein localization (BaCelLo: Pierleoni et al. 2009) 6) the prediction of protein interaction sites (ISpred: Fariselli et al. 2002) and their analysis in the context of genome-wide interaction networks (Bartoli et al., in press) 7) the prediction of silencing effect of miRNAs (Montanucci et al. 2008). References Bartoli L, Montanucci L, Fronza R, Martelli PL, Fariselli P, Carota L, Donvito G, Maggi G, Casadio R -The Bologna Annotation Resource: a non-hierarchical method for the functional and structural annotation of protein sequences relying on a comparative large-scale genome analysis- J Proteome Res 8:4362-4371 (2009) Bartoli L, Martelli PL, Rossi I, Fariselli P, Casadio R -The prediction of protein-protein interaction sites in genome-wide protein interaction networks: the test case of the human cell cycle- Curr Prot Pept Sci (in press) Calabrese R, Capriotti E, Fariselli P, Martelli PL, Casadio R -Functional annotations improve the predictive score of human disease-related mutations in proteins- Hum Mutat 30:1237-1244 (2009) Capriotti E, Fariselli P, Rossi I, Casadio R -A three-state prediction of single point mutations on protein stability changes- BMC Bioinformatics 9 Suppl 2:S6 (2008) Fariselli P, Pazos F, Valencia A, Casadio R -Prediction of protein-protein interaction sites in heterocomplexes with neural networks- Eur J Biochem 269:1356-1361 (2002) Martelli PL, Fariselli P, Casadio R -An ENSEMBLE machine learning approach for the prediction of all-alpha membrane proteins- Bioinformatics 19:I205-I211 (2003) Montanucci L, Fariselli P, Martelli PL, Rossi I, Casadio R -In Silico Evidence of the Relationship Between miRNAs and siRNAs- Open Appl Inf Journal 2:9-13 (2008) Pierleoni A, Martelli PL, Fariselli P, Casadio R -BaCelLo: a balanced subcellular localization predictor- Bioinformatics 22:e408-e416 (2006) Area di interesse identificata Genome wide association studies Nome Contatti Istituto/Dipartimento Elio Scarpini, Daniela Galimberti 0255033847 [email protected] Università di Milano, Dipartimento di Scienze Neurologiche Proposta di ricerca Given the rationale that AD is a multifactorial disease and that inflammation plays a role in its pathogenesis, this proposal aims to study in detail genes encoding for inflammatory molecules, including progranulin and chemokines, all of which located in chromosome 17q.21, both from a genetic and functional point of view. This research will take advantage from the availability of a biobank including 500 patients with AD diagnosed according to current criteria, and a similar number of age-matched controls. Genomic DNA have been collected for each subject. In addition, in a group of about 200 patients and controls, serum, plasma, CSF and RNA from PBMC and CSF cells have been collected as well. In particular, the specific objectives of this research are aimed to: 1. Carry out a thorough association analysis of the region of chromosome 17 containing PGRN and the selected chemokine cluster (CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL14, CCL15, CCL16, CCL18, CCL23, CCR7, CCR10). Optimal tagging SNPs covering all the linkage blocks will be chosen. To this aim, we will perform a power calculation using the Genetic Power Calculator program (http://pngu.mgh.harvard.edu/~purcell/gpc/) based on sample size, the average observed MAF, and assuming a multiplicative model and an estimated prevalence of AD in Italy of 1 case per 1000 inhabitants after 65 years of age. 2. Evaluate expression levels of the above mentioned genes in cells isolated from CSF and PBMC by Real-Time PCR. Prioritized hits selected at aim 1) will be correlated with expression levels to test whether these SNPs influence the level of transcription. 3. Analyze the pattern of miRNA which act as silencer of mRNA studied in aim 2). miRNA possibly influencing the translation of the above mentioned genes will be selected by submitting queries at the website http://microrna.sanger.ac.uk/, which contains sequences of all published mature miRNA, together with their predicted source hairpin precursors. 4. Evalutate, by ELISA, CSF and serum levels of PGRN and chemokines. 5. Combine genetic, expression and functional data with the clinical phenotype of patients, including longitudinal full neuropsychological testing, treatment efficacy and brain/hippocampal atrophy at MRI, in order to identify a genetic pattern and/or novel pathogenetic mechanisms suitable to predict which patients will likely decline more rapidly or will respond to therapy. Centers collaborating to this project: University of Brescia (A. Padovani, D. Finazzi), IRCCS Fatebenefratelli Brescia (G. Binetti, R. Ghidoni), IRCCS Castellanza (M. Franceschi), IRCCS S. Raffaele, Milan (S. Cappa), University of Turin (I. Rainero). From the University of Milan: C. Mariani, R. Dominici, Area di interesse identificata Genetic susceptibility to Alzheimer’s disease and genome wide association studies Finanziamenti ricevuti Titolo progetto - Genetic Risk factors and peripheral biological markers of conversion from Mild Cognitive Impairment to Alzheimer's Disease (to DG) - Introducing new bio-markers in clinical practice for the early diagnosis of Alzheimer disease: methodological, clinical and organisational aspects for Ente finanziatore Durata progetto Abstract del progetto the National Health System (to ES) Progetti ( 1 and 4) parte di Programma Strategico (PS39), Italian Ministry of Health 01-01-2009/31-12-2010 This program has two primary aims: A. The development and validation of a multi-factorial protocol that integrates molecular, imaging, neurophysiological, neu- ropsychological and behavioral data for early diagnosis of Alzheimer's disease (AD), in particular during the preclinical pha se and the prodromal stage of "mild cognitive impairment" (MCI) when the symptoms are not severe enough to fulfill the current diagnostic criteria for AD. The achievement of this objective is one of the priorities of the international research com munity, in the perspective of specific, disease-modifying therapies that are under development. B. Validation of this new diagnostic protocol within a regional public health network, and evaluation of the health care, orga nization and economic implications of its transfer to the National Health Service (NHS). The program will be articulated into four complementary and strictly integrated projects with the following specific aims: PROJECT 1. Validation of genetic and biochemical markers for early diagnosis of AD and the prediction of conversion of MCI into AD, and design of a multivariate molecular protocol having high diagnostic and prognostic accuracy. PROJECT 2. Development of standard operating procedures (SOPs) and quality control (QC) for neuroimaging (MRI, 18F-FDG PET and 18F-DDNP PET) and clinical neurophysiological analysis (including electroencephalography, magnetoencephalography and transcranial magnetic stimulation) for the diagnosis of prodromic or incipient AD. PROJECT 3. Definition of cognitive and behavioral indicators that differentiate various forms of MCI and enable the diagno-sis of incipient AD. PROJECT 4. Validation of a comprehensive diagnostic protocol based on the results obtained in the above studies, in a series of Alzheimer Evaluation Units of Lombardia Region, and estimation of costs and benefits of its transfer to the NHS. Nome Prof. Paolo Maria Rossini Contatti Email: [email protected] Istituto/Dipartimento Neurology, University “Campus Biomedico”, Rome, Italy Proposta di ricerca Individual evaluation of cognitive decline: definition of a low-cost non-invasive examination battery and implementation of a predictive algorithm to identify Mild cognitive impairment. Area di interesse identificata Mild cognitive impairment (MCI) is a state of the elderly brain affecting a large number of individuals mainly characterized by memory impairment not yet encompassing the definition of dementia and, in the majority of cases, represents a precursor of Alzheimers’ Disease (AD). Because of MCI’s prodromal qualities do not greatly impair daily functioning, MCI can be identified only by specific clinical and neuropsychological evaluation. Within such a theoretical frame a reliable marker of MCI-to-AD progression is not yet available, even if it would be highly desirable both for research and health system purposes. Ideally, these markers should be non-invasive, widely available and of low-cost in order to screen out large population samples. Along this line a combination of neurophysiological (electroencephalography, transcranial magnetic stimulation), blood chemistries (including metals, pro- and antioxidant compounds) and genetics are a potential candidate. . In this way we have collected numerous studies (1-10). The line of research we are interested in focuses on the study of healthy subjects and patients (suffering from MCI, AD and vascular dementia – VaD-) by means of this multidimensional investigation and will try to identify prognostic indexes able to predict the risk of conversion from MCI to AD. References: 1.Babiloni C, Frisoni GB, Vecchio F, Pievani M, Geroldi C, De Carli C, Ferri R, Vernieri F, Lizio R, Rossini PM Global functional coupling of resting EEG rhythms is related to white-matter lesions along the cholinergic tracts in subjects with amnesic mild cognitive impairment. J Alzheimers Dis. 2010 Jan;19(3):859-71. 2.Moretti DV, Frisoni GB, Fracassi C, Pievani M, Geroldi C, Binetti G, Rossini PM, Zanetti O. MCI patients' EEGs show group differences between those who progress and those who do not progress to AD.Neurobiol Aging. 2009 Dec 16. [Epub ahead of print] 3. Babiloni C, Frisoni GB, Vecchio F, Pievani M, Geroldi C, De Carli C, Ferri R, Vernieri F, Lizio R, Rossini PM. Global Functional Coupling of Resting EEG Rhythms is Related to White-Matter Lesions Along the Cholinergic Tracts in Subjects with Amnesic Mild Cognitive Impairment. J Alzheimers Dis. 2009 Nov 17. [Epub ahead of print] 4. Moretti DV, Pievani M, Geroldi C, Binetti G, Zanetti O, Cotelli M, Rossini PM, Frisoni GB. Increasing hippocampal atrophy and cerebrovascular damage is differently associated with functional cortical coupling in MCI patients. Alzheimer Dis Assoc Disord. 2009 Oct-Dec;23(4):323-32. 5. Moretti DV, Pievani M, Geroldi C, Binetti G, Zanetti O, Rossini PM, Frisoni GB. EEG markers discriminate among different subgroup of patients with mild cognitive impairment. Am J Alzheimers Dis Other Demen. 2010 Feb;25(1):58-73. Epub 2009 Feb 9. 6. Moretti DV, Fracassi C, Pievani M, Geroldi C, Binetti G, Zanetti O, Sosta K, Rossini PM, Frisoni GB. Increase of theta/gamma ratio is associated with memory impairment. Clin Neurophysiol. 2009 Feb;120(2):295-303. Epub 2009 Jan 1. 7. Babiloni C, Pievani M, Vecchio F, Geroldi C, Eusebi F, Fracassi C, Fletcher E, De Carli C, Boccardi M, Rossini PM, Frisoni GB. White-matter lesions along the cholinergic tracts are related to cortical sources of EEG rhythms in amnesic mild cognitive impairment. Hum Brain Mapp. 2009 May;30(5):1431-43. 8. Babiloni C, Visser PJ, Frisoni G, De Deyn PP, Bresciani L, Jelic V, Nagels G, Rodriguez G, Rossini PM, Vecchio F, Colombo D, Verhey F, Wahlund LO, Nobili F. Cortical sources of resting EEG rhythms in mild cognitive impairment and subjective memory complaint. Neurobiol Aging. 2008 Nov 20. [Epub ahead of print] 9 Moretti DV, Pievani M, Fracassi C, Geroldi C, Calabria M, De Carli CS, Rossini PM, Frisoni GB. Brain vascular damage of cholinergic pathways and EEG markers in mild cognitive impairment. J Alzheimers Dis. 2008 Nov;15(3):357-72. 10. Rossini PM, Buscema M, Capriotti M, Grossi E, Rodriguez G, Del Percio C, Babiloni C. Is it possible to automatically distinguish resting EEG data of normal elderly vs. mild cognitive impairment subjects with high degree of accuracy? Clin Neurophysiol. 2008 Jul;119(7):1534-45. Epub 2008 May 15. Finanziamenti ricevuti Titolo progetto Ente finanziatore Is it possible to automatically distinguish resting EEG data of normal elderly vs. mild cognitive impairment subjects with high degree of accuracy? AFaR, Associazione Fatebenefratelli per Ricerca Biomedica e Sanitaria Durata progetto 2005-2006; 2006-2007; 2007-2008; 2008-2009; 2009-2010 Abstract del progetto To explore if Implicit function as squashing time (IFAST) based electroencephalographic (EEG) can reliably distinguish of mild cognitive impairment (MCI) and Alzheimer's disease (AD) subjects with Nome ORAZIO ZANETTI Contatti TEL 030 3501 358 Email: [email protected] U.O.Alzheimer-Centro per la Memoria IRCCS Centro S.Giovanni di Dio-Fatebenefratelli, Brescia Istituto/Dipartimento Proposta di ricerca Nell’agosto 2007 un gruppo di studiosi ha proposto nuovi criteri diagnostici per la diagnosi di malattia di Alzheimer (AD) prodromica o preclinica (Dubois et al.:Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007 Aug;6(8):734-46.) incorporando, oltre ai dati anamnestici, clinici e neurospicologici l’utilità di marker neuroradiologici (atrofia ippocampale o ipoperfusione temporo-parietale alla PET), e liquorali (riduzione della proteina ABeta 42 ed aumento di TAU). Dal luglio 2006 è attivo presso l’IRCCS S.Giovanni di Dio Fatebenefratelli un Ambulatorio Traslazionale per la Memoria (ATM) che coinvolge tutte le Unità Operative cliniche e di ricerca dell’Istituto al fine di offrire, soprattutto ai pazienti lievi un percorso diagnostico in grado di rispondere all’interrogativo ciclopico, con risvolti laceranti, che ci veniva posto: “vorrei sapere se ho o non ho la malattia di Alzheimer”. Nell’arco di 12 mesi sono stati valutati 144 pazienti consecutivamente afferiti all’ ATM, divisi in quattro gruppi sulla base della diagnosi clinica (utilizzando i noti criteri diagnostici NINCDSADRDA per la AD): 12 pazienti sono affetti da disturbo soggettivo di memoria (SMC, MMSE 28.0±1.1); 37 da “mild cognitive impairment” (MCI, MMSE 25.1±3.6), 55 da malattia di Alzheimer (MMSE 21.1±3.5), e 40 da demenze non Alzheimer (MMSE 21.6±5.5)(Vedi tabella). A tutti i pazienti sono state proposte le seguenti indagini: 1) la volumetria ippocampale alla MR; 2) l’analisi visiva della PET con fluorodesossiglucosio; 3) la concentrazione di Tau totale e di Abeta1-42 sul liquor cerebrospinale. La sensibilità di ogni marker è più elevata (43 to 71%) per la malattia di Alzheimer rispetto all’ MCI (18 to 31%). La specificità dei marker rispetto a SMC e demenze non Alzheimer è risultata buona, essendo superiore al’ 83% ed al 69% rispettivamente. La positività di almeno un marker presenta un’elevata sensibilità per la AD ed un’elevata specificità per il SMC. L’utilizzo di un numero maggiore di marker (due o tre) contemporaneamente aumenta notevolmente la specificità diagnostica a scapito però della sensibilità. In conclusione i marker recentemente proposti per la diagnosi precoce di malattia di Alzheimer possono essere di utilità nella pratica clinica. La valutazione longitudinale consentirà di valutare meglio il loro “valore aggiunto” rispetto alla tradizionale diagnosi che si fonda prevalentemente sull’indagine clinica corroborata da una semplice TAC encefalica. Per un oculato impiego delle risorse e delle nuove tecnologie è infatti importante stabilire con precisione quali siano i pazienti (verosimilmente coloro con un disturbo cognitivo molto lieve) che maggiormente possono trarre vantaggi da una procedura diagnostica complessa e costosa. Area di interesse identificata Impiego dei makers biologici e neuro radiologici nella diagnosi differenziale precoce Finanziamenti ricevuti – € 20.000 (Programma strategico 2006) Titolo progetto Ente finanziatore Strumenti e procedure diagnostiche per le demenze utilizzabili nella clinica ai fini della diagnosi differenziale precoce Ministero della Salute – Programma strategico 2006- Durata progetto triennale Abstract del progetto Nome Sandro Sorbi Contatti [email protected] tel +390554298465 Istituto/Dipartimento Dept. of Neurological and Psychiatric Sciences, University of Florence, Italy Proposta di ricerca Area di interesse identificata Research in the field of aging and dementia is focusing on the characterization of the early stages of cognitive decline. It has been suggested that many patients with Mild cognitive impairment (MCI) represents already not recognized AD patients. The purpose of the research is to identify tools and diagnostic procedures that can identify early stages of the disease. We will study the various domains in order to: a) Diagnose and characterize demented patients for a proper classification of the forms of dementia (Alzheimer's disease, fronto temporal dementia, FTD, dementia with motor signs such as Lewy body disease and others) b) Diagnose and characterize patients with Mild Cognitive Impairement c) To study the neuropsychological aspects to identified early MCI patients, both multi domain and amnesic MCI. d) Analyze the genetic patient profile to identify the genetic risk factors involved in the development and progression of AD. The study of genetic factors for AD will be performed in patients with familial forms of dementia and in patients with MCI. In addition neuropsychological and neuroimaging evaluations will be performed for each patients. The study will be divided into the following phases: analysis of autosomal dominant genetic mutations: new families affected by AD will be identified and characterized clinically and genetically. The goal will be to identify new causative mutations. the Initial screening of molecular diagnosis will be a detailed genetic screening of the genes associated with familial form of the disease, presenilins (PS-1 and PS-2) and the amyloid precursor protein (APP) - Search for other possible factors of genetic susceptibility will be performed on both familial and sporadic AD patients. It will be studied allelic variations of genes potentially involved in chromosomal regions of interest. It will also be carried out the study of Apolipoprotein E genotype (ApoE), considered today the only susceptibility factor associated with the disease. Finanziamenti ricevuti Titolo progetto Ente finanziatore “Strumenti e procedure diagnostiche per le demenze utilizzabili nella clinica ai fini della diagnosi precoce e differenziale, della individuazione delle forme a rapida o lenta progressione e delle forme con risposta ottimale alle attuali terapie” MINISTERO DELLA SALUTE , RICERCA FINALIZZATA 2006 Durata progetto 6/1/2008-6/4/2010 Abstract del progetto The main aim of the project is to integrate the results of neurocognitive, neuropsychological, neuroimaging and genetic studies as markers for the differential diagnosis of early dementia. Titolo progetto GENETIC STUDY ON ALZHEIMER'S DISEASE AND THE INVOLVEMENT OF OTHER NON-GENETIC FACTORS THAT MAY INFLUENCE THE CLINICAL EXPRESSION OF THE DISEASE Ente finanziatore Progetto di ricerca scientifica d'Ateneo (ex quota 60%) Durata progetto 2009-2010 Abstract del progetto GENETIC STUDY ON ALZHEIMER'S DISEASE Titolo progetto "Analisi dei fattori genetici coinvolti nelle malattie neurodegenerative: Utilizzo e applicazione di tecnologie di avanguardia per lo studio della malattia di Alzheimer, della demenza frontotemporale e della malattia di Parkinson" /* Ente finanziatore COMPAGNIA SAN PAOLO Durata progetto 2008-2010 Abstract del progetto GENETIC STUDY ON ALZHEIMER'S DISEASE Proposta di ricerca Area di interesse identificata Aim of the study is to determine the clinical and genetic features of movement disorders in patient affected by dementia and the clinical features of cognitive decline in patients affected by movement disorders. All the patients recruited will be assessed at baseline and during the follow-up with selected clinical scales and neuropsychological tests. Beyond movement disorder and cognitive deficit, attention will be paid to behavioural disturbances, mood disorders and impulse control disorders in these patients. Patients with a familial history of movement disorders or dementia will undergo genetic analysis on the genes implicated in autosomal dominant forms of Parkinson disease including PARK1, PARK4, SNCA and LRRK2. All the patients recruited will be submitted to FP-CIT-SPECT in order to verify and to measure the presence of nigrostriatal pathway C. Nome Flavio Nobili, Guido Rodriguez degeneration. We plan to enrol 50-100 patients affected by those neurodegenerative disorders that may be characterized by movement disorders and cognitive decline: Parkinson’s disease, Alzheimer’s disease with parkinsonism, Frontotemporal dementia, Lewy body disease, vascular dementia, corticobasal degeneration and progressive supranuclear palsy. Finanziamenti ricevuti Titolo progetto “ Epidemiologia e sviluppo di strumenti di integrazione socio-sanitaria nella gestione dei pazienti affetti da Malattia di Parkinson e da altre malattie degenerative con concomitante disturbo motorio e declino cognitivo” Ente finanziatore MINISTERO DELLA SALUTE, RICERCA FINALIZZATA 2007 Durata progetto 12/2009-12/2010 Abstract del progetto Main aim of the project is to analyze and develop in detail the natural history of Parkinson's disease and Parkinsonism due to cognitive decline and behavioural disturbances. Epidemiological study is of fundamental importance in developing diagnostic and therapeutic strategies and new methods of integrating social and health care. In particular we will identify and recruit individuals with Parkinson's disease and classical subjects with parkinsonism linked to cognitive decline, it will estimate the incidence of dementia in the two types of disturbances. Will be performed genetic analysis of genes related to Parkinson's disease in people with familiarity with the disease. All patients will undergo a battery of standardized tests (Unified Parkinson's Disease Rating Scale and Hoehn and Yahr and neuropsychological tests) and FP-CIT-SPECT imaging. Contatti Istituto/Dipartimento [email protected]; [email protected] Tel: 010 3537568; Fax: 010 5556893; cell: 333 8331613 Neurofisiologia Clinica, Dip. di Neuroscienze, Oftalmologia e Genetica, Università di Genova Proposta di ricerca A. ‘A multimodality index for the early diagnosis of Alzheimer’s disease’ Area di interesse identificata Early diagnosis Finanziamenti ricevuti Titolo progetto ‘Descripa’ (http://www.descripa.eu/) Ente finanziatore EU FP-5 Durata progetto 4 years Abstract del progetto Titolo progetto The DESCRIPA study aims to develop screening guidelines and clinical criteria for Alzheimer’s disease in non-demented subjects. The clinical criteria will be based on a prospective cohort study of non-demented subjects from a memory clinic. The screening guidelines will be based on a meta-analysis of prospective population-based cohort studies in Europe. ‘ICTUS’ (Neuroepidemiology. 2007;29(1-2):29-38.) Ente finanziatore EU FP-5 Durata progetto 3 years Abstract del progetto Observational study of cognitive and behavioural effects of acetylcholinesterase inhibitors in patients with mild to moderate Alzheimer’s disease Finanziamenti ricevuti Titolo progetto ‘Descripa’ (http://www.descripa.eu/) Ente finanziatore EU FP-5 Durata progetto 4 years Abstract del progetto The DESCRIPA study aims to develop screening guidelines and clinical criteria for Alzheimer’s disease in non-demented subjects. The clinical criteria will be based on a prospective cohort study of non-demented subjects from a memory clinic. The screening guidelines will be based on a meta-analysis of prospective population-based cohort studies in Europe. ‘ICTUS’ (Neuroepidemiology. 2007;29(1-2):29-38.) Titolo progetto Ente finanziatore EU FP-5 Durata progetto 3 years Abstract del progetto Observational study of cognitive and behavioural effects of acetylcholinesterase inhibitors in patients with mild to moderate Alzheimer’s disease BACKGROUND. During the last decade, there has been a great effort to improve diagnostic accuracy of Alzheimer’s disease (AD) since the earliest stages. The main advances have been observed with morphological Magnetic Resonance (MR) and functional (FDG-PET) brain imaging, and with Cereborspinal fluid (CSF) assays of Tau protein and β amyloid fragments. The accuracy of these tools is rather good when comparing patients with overt AD dementia versus healthy controls, but decreases in the very early stages of the disease. In these stages, the subjects still maintain full autonomy in everyday life, but subtle deficit in memory and/or other cognitive domains can already be shown by cognitive tests (Mild Cognitive Impairment, MCI). Moreover, some patients complain of memory disturbances that cannot be confirmed by cognitive assessments (subjective memory complaints, SMC) but that may sometimes be prodromic of AD. Pharmacological research is currently very active in trying to identify drugs that can modify the natural course of the disease (‘disease-modifying’). While they are tested mainly in overt AD dementia at present, it is obvious that effective drugs will be used as soon as possible in the course of the disease, i.e., at the MCI stage, or even earlier. Thus, an early diagnosis is of paramount importance to start effective treatment as soon as these drugs will be available. Yet, individual diagnostic accuracy of functional and morphological neuroimaging and CSF assays in these early stages is not fully satisfactory versus healthy controls but even more versus the other commonest forms of dementia, such as dementia with Lewy bodies (DLB), frontotemporal dementia (FTD) and vascular dementia (VaD). The more recent amyloid PET imaging has added value to neuroimaging, showing very high sensitivity but limited specificity versus healthy controls, DLB and Parkinson’s disease dementia. The reasons of incomplete accuracy are complex and partially understood. Genetic heterogeneity, environmental factors, and phenotipic variants of AD may explain incomplete sensitivity on the one hand. On the other hand, overlapping atrophy, hypometabolic and biochemical patterns with FTD, DLB and VaD may account for incomplete specificity of MR, PET, and CSF assays, respectively. The idea that the ‘shadow’ area of each diagnostic tool could be 'unmasked' by the other tools has moved researchers to join together the data deriving from more than one modality. Some work has already been done, showing an increase accuracy when two examinations are employed together. In this context, the ‘core’ cognitive deficit of AD, i.e., verbal episodic memory delayed recall, has also been included. Previous attempts to obtain combination indexes include neuropsychology and PET (1-2), neuropsychology, PET and Apolipoprotein E (ApoE) genotype (3), neuropsychology and MR imaging (4), Single Photon Emission Computed Tomography (SPECT) and MR imaging (5), CSF markers and SPECT (6). However, the data are still largely incomplete, especially regarding the very early stages of the disease. AIM. To test the hypothesis that joining together the information deriving from MR and FDG-PET imaging, memory tests, CSF assays and genetic predisposing factors (such as the ApoE genotype) can improve diagnostic accuracy in patients with AD dementia at the MCI stage. A single index could be achieved with integrated accuracy deriving from each tool. METHODS. Patients. A multicentric approach is needed to reach large patient series in a limited time. Patients with MCI of the amnestic type (aMCI), as well as age-matched healthy controls, should be recruited by Centres with proven expertise in dementia diagnosis and management. These subjects should undergo full neuropsychological examination (common minimum battery to be agreed), MR imaging, and FDG-PET. CSF assays are performed in MCI patients and in that part of healthy controls giving the informed consent for this invasive procedure, providing Ethic Committees approve this manoeuvre in healthy subjects. Administration of radiopharmaceuticals to healthy subjects must also be allowed by Ethic Committees and by the National Authority governing radiation exposure to healthy subjects for research purposes. Then, both patients and controls are regularly followed by means of clinical and neuropsychological examinations on yearly basis for at least three years to pick up those MCI patients matching the current criteria for the diagnosis of AD or other dementias. Other study groups are represented by patients affected with FTD, VaD or LBD in a mild stage (i.e., MMSE score >20), recruited by the same centres, following the internationally accepted guidelines. Image analysis. Several software are available to automatically segment the whole brain or specific brain regions, such as the hippocampus and the inferior parietal lobule, that better distinguish AD patients and controls. These software can either run both MR and PET images (such as Statistical parametric Mapping, SPM) or specifically one of the two (such as the boundary shift integral for MR and ‘Neurostat’ for PET). As for MR images, the output could be a volumetric index. The same applies to FDG-PET where a metabolic index can be automatically computed. For both MR and PET, other indexes expressing the probability to have maximum or minimum atrophy/hypometabolism versus a control group can be computed. Statistical analysis. Statistics should rely on a multidimensional approach able to identify the best combination of indexes that distinguish between AD patients (at MCI stage) and either healthy controls or DLB, FTD, and VaD, respectively. Stepwise and discriminant approaches are favourites. EXPECTED RESULTS. An index able to discriminate AD from healthy controls and from the other commonest foms of dementia with very high accuracy, i.e., >95%. This index should work, even if with a more limited accuracy, in those cases where just some of the requested information is available, for instance just with neuropsychology, ApoE, MR and PET images, without CSF biomarkers. Once established, such an index may be applied also to subjects with SMC to unveil AD pathology, as well as to screen population at high risk of AD, such as direct relatives of AD patients. REFERENCES. (1) Anchisi D, et al. Heterogeneity of brain glucose metabolism in mild cognitive impairment and clinical progression to Alzheimer disease. Arch Neurol 2005;62:1728-33. (2) Nobili F, et al. Principal component analysis of FDG PET in amnestic MCI. Eur J Nucl Med Mol Imaging 2008;35:2191-2202. (3) Mosconi L, et al. MCI conversion to dementia and the APOE genotype. A prediction study with FDG-PET. Neurology 2004;63:2332-40. (4) Visser PJ, et al. Medial temporal lobe atrophy and memory dysfunction as predictors for dementia in subjects with mild cognitive impairment. J Neurol 1999;246:477-85. (5) El Fakhri G, et al. MRI-guided SPECT perfusion measures and volumetric MRI in prodromal Alzheimer disease. Arch Neurol. 2003;60:1066-72. (6) Okamura N et al. Combined Analysis of CSF Tau Levels and [(123)I]Iodoamphetamine SPECT in Mild Cognitive Impairment: Implications for a Novel Predictor of Alzheimer's Disease. Am J Psychiatry. 2002;159:474-6. (7) Barnes J, et al. Automatic calculation of hippocampal atrophy rates using a hippocampal template and the boundary shift integral. Neurobiol Aging. 2007;28:1657-63. (8) Ishii K, et al. Statistical brain mapping of 18F-FDG PET in Alzheimer's disease: validation of anatomic standardization for atrophied brains. J Nucl Med. 2001;42:548-57. Nome Professor Paolo Caffarra, M.D. Contatti e-mail: [email protected] phone/fax: 0039-0521-704116 Department of Neuroscience University of Parma Via Gramsci 14 43100 Parma Italy Istituto/Dipartimento Proposta di ricerca Area di interesse identificata The need of an early diagnosis is essential for promoting a strategic pharmacological and non-pharmacological intervention on Alzheimer’s disease (AD). The identification of cognitive and neurobiological markers which might represent salient pre-clinical predictors of the insurgence of AD is essential nowadays. Our research focuses on the discovery of cognitive instruments useful for a differential diagnosis of AD, such as memory and executive tasks, and neuroimaging markers using different techniques, such as magnetic resonance imaging (MRI) and positron emission tomography (PET). Recently, we have set up an innovative and computerized experimental procedure to score semantic abilities in patients suffering from Mild Cognitive Impairment (MCI) and mild AD (Arango-Lasprilla et al., 2007; Ahmed et al., 2008) , investigating also the functional and metabolic neural substrates using MRI and F18FDG-PET. Different aspects of structural MRI will be observed, such as volumetric analysis, diffusion tensor imaging (DTI), imaging of default mode at rest and imaging of the deposition of iron in memory-related critical brain regions, i.e. hippocampus. In particular, the observation of iron deposition in the brain of patients at risk for AD, represents an innovative approach in the neuroimaging field of dementia (Ding et al., 2009). F18-FDG-PET constitutes an effective technique in the monitoring of early Alzheimer’s disease progression (Fouquet et al., 2009). Cognitive and neuroimaging follow-up data will be informative on the identification of specific possible markers of conversion (Devanand et al., 2007; McGeown et al., 2009). The role of the polymorphism of APOE is observed and correlated with clinical progression of the disease, cognitive performance and neuroimaging pattern. References: Ahmed S, Arnold R, Thompson SA, Graham KS, Hodges JR. (2008). Naming of objects, faces and buildings in mild cognitive impairment. Cortex, 44, 746-52. Arango-Lasprilla JC, Cuetos F, Valencia C, Uribe C, Lopera F. (2007). Cognitive changes in the preclinical phase of familial Alzheimer's disease. J Clin Exp Neuropsychol. 29, 892-900. Devanand DP, Pradhaban G, Liu X, Khandji A, De Santi S, Segal S, Rusinek H, Pelton GH, Honig LS, Mayeux R, Stern Y, Tabert MH, de Leon MJ. (2007). Hippocampal and entorhinal atrophy in mild cognitive impairment: prediction of Alzheimer disease. Neurology, 68, 828-36. Ding B, Chen KM, Ling HW, Sun F, Li X, Wan T, Chai WM, Zhang H, Zhan Y, Guan YJ. (2009). Correlation of iron in the hippocampus with MMSE in patients with Alzheimer's disease. J Magn Reson Imaging, 29, 793-8. McGeown, W.J., Shanks, M.F., Forbes-McKay, K.E., Venneri, A. (2009). Patterns of brain activity during a semantic task differentiate normal aging from early Alzheimer's disease. Psychiatry Res., 173, 218-27. Finanziamenti ricevuti Titolo progetto “CORRELATI FUNZIONALI E STRUTTURALI DELLE FUNZIONI COGNITIVE NELLA FASE PRE-CLINICA E CONCLAMATA DELLE DEMENZE DEGENERATIVE” Ente finanziatore Cassa di Risparmio di Parma e Piacenza Durata progetto 24 mesi Abstract del progetto The present project aims to investigate the neurobiological substrates associated with the insurgence and progression of Alzheimer’s disease (AD). In particular, the attention will be focused on the study of brain regions more vulnerable and affected by AD pathology and responsible for AD symptoms, such as hippocampal, posterior cingulate and parietal circuits involved in memory function. The project will concentrate on the identification of distinct neurofunctional and structural pattern which characterize the different forms of dementia, in order to establish the effectiveness of neuroimaging techniques in the diagnostic process of dementia. Particular attention will be give on the study of neuroimaging correlates of Mild Cognitive Impairment as this is considered a critical phase in the progression to dementia: the identification of typical preclinical neuroimaging markers of AD will provide the possibility to act an early and effective intervention. Nome Contatti Tel. E mail Istituto/Dipartimento Stefano F. Cappa DIBIT via Olgettina 58 20132 Milano tel 0226434887 [email protected] Vita Salute University and San Raffaele Scientific Institute, Division of Neuroscience Proposta di ricerca. Our multi-disciplinary reaserach unit, combines expertises from different backgrounds (neurology, neuropsychology, linguistics, cognitive psychology, epistemology), with the general aim to investigate the neural mechanisms of language and high-order cognition. The experimental approaches include behavioural studies in normal subjects and in neurological patients, brain imaging using magnetic resonance techniques and positron emission tomography, and neurophysiological investigations based on evoked responses and transcranial magnetic stimulation. In the field of dementia we are leading a number of investigation in the areas of linguistic function, memory and social neuroscience. Besides the theoretical interest , these studies are of relevance for issues such as early diagnosis (in particular distinction from healthy aging), differential diagnosis among different causes of degenerative dementia, and assessment of treatment effect. The studies are typically based on the development of innovative behavioural testing procedures, combined with multimodal imaging, biomarkers and genetic investigations, and are largely based on collaboration amiong multiple clinical and research units, both within the San Raffaele Foundation and with other national and international institution. Area di interesse identificata Cognitive neuroscience of dementia: clinical and theoretical aspects Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract del progetto The reseach has been supported by grants from the Ministry of Health, the MIUR, the EEC and Human Frontiers organization Nome Contatti Tel. E mail Istituto/Dipartimento Daniela Perani 02-26432224 -2223 [email protected] V-S San Raffaele University, Division of Neuroscienze and Nuclear Medicine Dpt. San Raffaele Scientific Institute Proposta di ricerca Area di interesse identificata Studies are aimed at investigating with PET the brain functional parameters and neurotransmission systems in neurological and psychiatric diseases : 18 F-FDG is used for the evaluation of brain glucose metabolism and 11CPIB for amyloid deposition. Present protocols include dementias (AD and FTLD, LBD, PDD) in early and pre-clinical phase, including in Mild Cognitive Impairment. 11 C-PK and its developed forms are used in the study of neuroinflammation. Present protocols include Parkinson’s disease and parkinsonisms such as Lewy Body Dementia, prion diseases, and Amiotrophic Lateral Sclerosis. To this aim mathematical models for quantification of activity are under development. 11 C-Raclopride and 11C-Fe CIT for the study of dopaminergic system. Present protocols include Parkinson’s disease and parkinsonisms, particularly in early phase. Voxel-based methods for automatic quantification are under study validation. 11 C MP4 for measuring the AchE activity, in AD and MCI subjects in order to obtain correlations between reduction of enzymatic activity and cognitive status. To this aim mathematical models for quantification of activity are under development. 11 C-MDL for 5HT2 serotonin receptors and 11C-raclopride. Present protocols include psychiatric conditions such as obsessive compulsive disorders and depression 11 C-FMZ for the evaluation of the gabaergic system. Present protocols include movement disorders such as dystonias (genetic and sporadic cases) Finanziamenti ricevuti Titolo progetto 1. Diagnosis by Statistical and Intelligent Systems (ICT 2009, 5.3). 2. Molecular imaging for the early diagnosis and monitoring of Alzheimer’s disease in old individuals with cognitive disturbances: Ente finanziatore Durata progetto Abstract del progetto an ADNI-compatible prospective study. 3. Identification of diagnostic biomarkers in Amyotrophic Lateral Sclerosis patients, and development of related computational methods. 1. Comunità Europea 2. Ministero della Sanità 3. Ministero della Sanità 1. 2010-2013 2. 2010-2012 3. 2008-2010 1. Validazione di nuovi metodi voxel-based e pattern recognition per il neuroimaging funzionale PET, SPECT e strutturale MRI nella diagnosi precoce e nella diagnosi differenziale delle malattie neuroddegenerative associate a demenza. 2. Studio dei correlati neurofunzionali, anatomici e molecolari nel disturbi cognitivi lievi. 3. Studio di biomarker in vitro e in vivo nella Sclerosi Laterale Amiotrofica. Nome Contatti Tel. E mail Istituto/Dipartimento Maurizio Popoli Tel: +390250318361 E-mail: [email protected] Center of Neuropharmacology, Department of Pharmacological Sciences, CEND, University of Milan Proposta di ricerca Area di interesse identificata 3C. Global proteomics of human fibroblasts for the identification of early biomarkers for dementia and Alzheimer’s Disease Finanziamenti ricevuti Titolo progetto Ente finanziatore Identification of biomarkers for Alzheimer’s dementia: genomics and proteomics of neurodegeneration Ministero della Salute Durata progetto 2004-2006 Abstract del progetto Ente finanziatore Main object of the present project was the identification of genetic and biological markers for Mild Cognitive Impairment (MCI) in order to identify a strategy useful for an early diagnosis and for the individuation of subjects at risk for the development of Alzheimer’s disease. Neurodegeneration, physiological cerebral aging and dementia: an integrated approach for a early diagnosis, evolution predictors, and for an advanced care organization. Ministero della Sanità Durata progetto 2000-2003 Abstract del progetto Aim of this project was the investigation of biological, cellular, genetic and neurotoxic processes underling dementia for the identification and definition of predictive markers of pathology in the general population and in presence of risk factors. Titolo progetto Nome Contatti Tel. E mail Istituto/Dipartimento Gianfranco Spalletta Via Ardeatina, 306 -00179 Rome 06-51501575 [email protected] IRCCS Santa Lucia Foundation/Department of Clinical and behavioral neurology - Neuropsychiatry laboratory Proposta di ricerca Area di interesse identificata Parkinson disease and parkinsonisms Finanziamenti ricevuti Titolo progetto Analysis of cognitive and neuropsychiatric disturbances in Parkinson disease and parkinsonisms: implications for early diagnosis Ente finanziatore Italian Ministry of Health Durata progetto 22/12/2008 - 22/12/2011 Abstract del progetto The prevalence rate for neuropsychiatric symptoms of depression, anxiety, and hallucinations in subjects with Parkinson Disease (PD) has a very wide range. This suggests how undefined the results of studies done up till now are. Moreover, longitudinal studies suggest that the majority of subjects with PD could develop dementia during the course of the disease. This study aims at evaluating the differential features of cognitive and neuropsychiatric disturbances in subjects with PD and other Parkinsonisms at the onset of the disease vs. advanced phases. A crosssectional model could help to understand the phenomena changing along time in a time-limited project. If psychopathological disturbances associated with Parkinson disease are difficult to be studied, those that accompany other motor disturbances are almost completely unknown. Therefore, it is urgent to fill this gap in knowledge with studies aimed at a better diagnostic typing between PD, LBD and other degenerative and vascular parkinsonisms. Another important issue concerns the validity of diagnostic criteria of DSM-IV for mood disturbances and anxiety disturbances in subjects with motor disturbances. Thus, in this crosssectional study, 300 patients affected by motor disturbances will be recruited. In particular, we will include 150 subjects with idiopatic Parkinson Disease (PD) of which 50 at onset and 100 in the advanced phase, 50 subjects with Dementia with Lewy Bodies (DLB); 50 subjects with vascular PD; 25 subjects with Progressive Supranuclear Palsy (PSP); 20 subjects with corticobasal degeneration (CBD) and 10 subjects with multisystemic atrophy (MSA). Patients will be assessed using a clinical battery comprising: a neurological battery including socio-demographic and clinical history, Unified Parkinson’s Disease Rating Scale (UPDRS), and Hoehn and Yahr scale. The neuropsychological battery will include: Mini Mental State Examination, Rey 15-word test, reduced Wisconsin Card Sorting, reduced Stroop Color-Word test, Verbal and visual-spatial Nback, Verbal semantic fluency, Verbal phonological fluency, Sentence construction, Immediate and deferred copy of Rey figure. The neuropschiatric battery will include: Psychiatric diagnosis according with the DSMIV-TR (in this case we will use both the standard diagnostic criteria and modified diagnostic criteria proposed by the Starkstein group in order to improve the neuropsychiatric diagnosis in patients with motor disturbances), Beck Depression Inventory, Toronto Alexithymia Scale, Penn Emotional Recognition Test, Snaith-Hamilton Pleasure Scale. In another part of the study we will include 20 idiopatic PD patients treated with Deep Brain Stimulation (DBS). They will be clinically evaluated with UPDRS Part III and with the neuropsychological and neuropsychiatric battery above-mentioned. The evaluation of subjects with stimulator implants will be carried out without any treatment and in 3 different phases: 1) during proper and stable antiparkinsonian therapy and Stimulator on, 2) during pharmacological therapy and DBS stimulator off since 90 minutes, 3) absence of pharmacological therapy with DBS stimulator on. The neuropsychological and behavioural tests will be carried out in a random way on different days. In the subgroup of patients with DBS the evaluation for depression will be carried out with a visual-analogical scale to instantly capture the severity of the depression in each of the 4 experimental conditions. Results of studies in this field will allow to propose new diagnostic criteria for neuropsychiatric and behavioural disturbances which will help clinicians in the early and differential diagnosis of movement disorders allowing also a better treatment. The research on patients in treatment with DBS will allow a better understanding of the features of non motor symptoms and directions towards treatments. The study will also provide a picture of the differential expression of cognitive versus neuropsychiatric symptoms within each pathological entity. Such achievements will be possibly imply a reduction of the burden caused by these diseases on the patient’s family, as well as a decrease of the social and heath costs. Nome Contatti Tel. E mail Istituto/Dipartimento Gianfranco Spalletta Via Ardeatina, 306 - 00179 Rome O6-51501575 [email protected] IRCCS Santa Lucia Foundation/Department of Clinical and behavioral neurology - Neuropsychiatry laboratory Proposta di ricerca Area di interesse identificata Mild Cognitive Impairment and Dementias Finanziamenti ricevuti Titolo progetto Ente finanziatore Cognitive and behavioural indicators of conversion from Mild Cognitive Impairment to neurodegenerative dementia and development of cognitive rehabilitation protocols Italian Ministry of Health Durata progetto 01/01/2009 - 31/12/2011 Abstract del progetto According to a four-group classification, the MCI amnestic single domain is characterized by a selective deficit of amnesic performances, multidomain amnestic MCI presents memory impairment and at least one other cognitive domain impairment, nonamnestic single domain MCI is associated with the compromise of one nonamnestic domain, and nonamnestic multi-domain MCI is associated with the compromise of two or more nonamnestic cognitive domains. The hypothetic usefulness of subdividing MCI to identify specifically the type of dementia to which the patient will convert has partially failed. However, there is solid evidence that multi-domain MCI has the highest rate of conversion in dementia. Although there is convergence on the description of the high risk of developing AD in subjects with chronic depression, often associated with hippocampal atrophy, there are also studies which clarify with sufficient reliability that other neuropsychiatric disturbances may predict conversion to AD. Regarding the fact that therapeutic strategies for the pathogenesis of AD have failed up till now, and considering the high risk of patients with MCI to develop AD, a revision of the clinical-diagnostic criteria of AD has been recently proposed. This revision makes AD diagnosis similar to those of amnesic MCI with the exception of biological and neuroradiological markers recognized as specific for AD diagnostic purposes. Unfortunately, the specific neuropsychiatric symptoms of AD were not included even in this recent diagnostic revision proposal. This research project will attempt to identify clinical characteristics useful for predicting the risk of developing neurodegenerative dementia from the 4 different forms of MCI, the type of dementia more frequently associated with each clinical characteristics, the progression severity of the illness; d) the effect of caregiver support on the illness progression, and the efficacy of cognitive rehabilitation on every subtype of clinical characteristics. The method of open study on cognitive rehabilitation will give an early clinical indication of the potential of this treatment. One hundred patients diagnosed with MCI will be included in the study. The patients included will carry out an in depth anamnestic evaluation and a diagnostic\categorical evaluation for MCI subtype. In addition, categorical neuropsychiatric disorders of depression, psychosis, and apathy will be assessed. Dimensional neuropsychiatric symptom severity will be measured with a battery of tests including: Neuropsychiatric Inventory-12 items, CERAD Disforia, and Dementia Apathy Interview and Rating. The age at onset of each behavioural disturbance and the first memory deficit will be accurately evaluated. Moreover, To obtain a global index of cognitive impairment, we will administer the MMSE. The Mental Deterioration Battery and other cognitive task will be used to assess individual cognitive domains. A subsample of 30 MCI patients will be treated with 12 weeks cognitive rehabilitation. Also, a subsample of caregivers of 30 MCI patients will undergo psychometric evaluation at baseline and at the three and six-month follow-up. All subjects will be included during the first 6 months period of the project. All diagnostic, cognitive and neuropsychiatric tests will be administered every six months until 18 months from the first evaluation. This project will allow a more correct clinical-diagnostic subclassification of MCI subgroups in order to make the clinical subtypes of MCI more homogeneous both on the diagnostic level and the biological correlates. In particular, this most reliable subclassification should help to fasten the recognition of the disorders and the differential diagnosis, thus facilitating a more precocious and specific treatment of patients who will develop the different types of dementia. This project will also suggest the potential role of cognitive rehabilitation therapy and caregivers support on the cognitive and behavioural symptom progression of MCI. To obtain this result potentially means a reduced cost of the management of dementia for the national health service and a better outcome of the illness for patients. Nome Paolo Calabresi Contatti Clinica Neurologica, Università degli Studi di Perugia, Ospedale S. Maria della Misericordia, 06132 Perugia, Italy e-mail: [email protected] Istituto/Dipartimento Clinica Neurologica, Laboratori di Neurologia sperimentale, Dipartimento di Specialitá Medico Chirurgiche e Sanitá Pubblica, Università degli Studi di Perugia Proposta di ricerca Area di interesse identificata Electrophysiological study of striatal physiology in normal conditions and in experimental parkinsonism: use of genetic and pathogenetic animal models. Pre-clinical research is conducted at the Laboratory of Experimental Neurology of the Clinica Neurologica at the Hospital S. Maria della Misericordia in Perugia. The activities focus on the study of important neurological pathologies such Parkinson’s disease (PD) and other neurological and neurodegenerative conditions (Huntington disease, Alzheimer disease, Multiple sclerosis, stroke, epilepsy) by means of in vitro animal models. The main topic of the laboratory is the study of the mechanisms involved in the physiopathology of such diseases by means of up to date electrophysiology and behavioural techniques in tight collaboration with the Laboratories of Neurophysiology located at the Fondazione S. Lucia in Rome. Motor and non-motor symptoms occurrence in PD represent the downstream effect of a pathological cascade resulting in the degeneration of dopaminergic neurons of the substantia nigra pars compacta (SNpc) projecting to the nucleus striatum. In this nucleus physiological activation of neuronal pathways are able to produce longterm depression (LTD), long-term potentiation (LTP) and “depotentiation” of synaptic transmission that are all disrupted by the PD-related pathological process. While Levodopa (L-DOPA) remains the gold standard of symptomatic therapy of PD it also causes severe longterm side effects (L-DOPA-induced dyskinesia, LID). The cellular and molecular mechanisms of LID formation and maintenance have started to be elucidated. For the study of these phenomena we take advantage of several animal models of PD. A well accepted model is obtained with the injection of 6hydroxydopamine (6-OHDA) into the SN causing permanent DAdenervation of the ipsilateral striatum mimicking the symptoms of the human PD. The in vitro model of PD by acute application of the neurotoxin rotenone that affects the complex I of mitochondrial chain is an important tool to explore mitochondrial function linked to PD. In order to assess specific aspects of the pathogenesis of PD, we employ transgenic animal lines expressing different mutant forms of αsynucleins such as A53T-α-synuclein mice and mice expressing human α-synuclein lacking the C-terminal 20 amino acids (α -Syn120). Furthermore, in order to study and to prevent LID development (priming) and maintenance in PD models we focussed on the modulation of the Ras-ERK pathway by the use of lentiviral vector (LV)mediated expression of either small harpin RNAs (shRNAs) specific for Ras-GRF1 and DARPP-32. Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Durata progetto Nuovi approcci terapeutici per le discinesie indotte dal trattamento con L-DOPA in un modello sperimentale di Parkinson: ruolo delle subunità del recettore NMDA e della via molecolare Ras-ERK Prin 2008 Two years LIDYAS - An innovative approach to treat levodopa-induced dyskinesia based on targeting striatal intracellular signalling Progetto San Paolo di Torino - Coordinatore: Istituto Nazionale di Neuroscienze (INN) Three years A multidisciplinary approach to test the therapeutic potential of neural stem cell transplantation in preclinical mouse models of Parkinson’s disease Progetto Strategico ex art56 Centro San Raffaele del Monte Tabor Two years Meccanismi di danno neuronale alla base della neurodegenerazione nelle patologie dei gangli della base Progetto Finalizzato 2005 Two years SYNSCAFF - Synaptic scaffolding proteins orchestrating cortical synapse organisation during development. SIXTH FRAMEWORK PROGRAMME - PRIORITY [LSH-2003-2.1.3-5] [Cortical development] Proposal/Contract no.:511995 Four years Mechanisms of L-DOPA-induced dyskinesia in an experimental model of Parkinson's Disease: focus on NMDA receptors Ministero dell’Istruzione dell’Università e della Ricerca (Prin 2005) Two years Ruolo dell’adenosina nella trasmissione sinaptica eccitatoria striatale: interazione con il sistema dopaminergico Sigma Tau One years Titolo progetto Ente finanziatore Durata progetto Titolo progetto Ente finanziatore Mechanisms underlying cell-type specific vulnerability of striatal neurons: implications for huntington's disease Telethon GGP02035 Two years Durata progetto REPLACES - Restorative Plasticity At Corticostriatal Excitatory Synapses Comunità Europea - HEALTH-2007-2.2.1-7 - Grant agreement no.: 222918 Four years Titolo progetto Ente finanziatore Durata progetto Imaging and biological markers of disease progression Progetto STRATEGICO Ministero della Sanità - Sottoprogetto 5 Two years Nome Contatti Tel. E mail Istituto/Dipartimento Lucilla Parnetti Clinica Neurologica, Università degli Studi di Perugia, Ospedale S. Maria della Misericordia, 06132 Perugia, Italy Tel: +39 075 578 3545; Fax: +39 075 578 3621. [email protected] Clinica Neurologica, Centro Disturbi della Memoria – Unità Valutativa Alzheimer - Laboratorio di Neurochimica Clinica, Dipartimento di Specialitá Medico Chirurgiche e Sanitá Pubblica, Università degli Studi di Perugia Proposta di ricerca Area di interesse identificata Cerebrospinal fluid biomarkers in Alzheimer’s disease and other neurodegenerative diseases Clinical research in the Laboratory of Neurochemistry is performed in close contact with the Center for Memory disturbancies, the Center for Movement disorders and the Center for Demyelinating disease of the Hospital S. Maria della Misericordia in Perugia. In the Laboratory of Neurochemistry, a biobank of cerebrospinal fluid (CSF) and plasma samples from patients with neurological diseases is available, making feasible retrospective studies on several pathologies such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and other neurodegenerative diseases. The main topic of the laboratory is the study of CSF biomarkers for the early and/or differential diagnosis of AD. The classical biomarkers Aβ1-42, tau protein (t-tau), and phosphorylated tau 181 (p-tau) are routinely assayed in CSF of patients with suspected dementia and/or patients with mild cognitive impairment (MCI) for which is also available a clinical follow up by means of neuropsychological tests.Techniques available in the Laboratory of Neurochemistry include mainly immunoassays such as ELISA and xMAP technique. The latter allows us to multiplex several analytes increasing the number of molecules that can be measured starting from the same volume of sample. CSF biomarkers show promise as a diagnostic tool in patients with AD, but recent multicenter studies showed that the levels of these biomarkers vary between different research centers. To study the source of variation our laboratory is actually involved in a world quality control program run by the Alzheimer Association in conjunction with Clinical Neurochemistry Laboratory in Gothenburg, Sweden, with the final goal of standardize these assays for clinical routine. Beside the classical CSF biomarkers we are actually studying other proteins as possible biomarkers for neurodegenerative diseases. We have recently shown that heart-fatty acid binding protein (H-FABP) is significantly increased in MCI patients which converted to AD and it may represent a new marker of neurodegeneration. Other projects are related to progranulin measurement in CSF, a protein which has been related to frontotemporal dementia and amyotrophic lateral sclerosis and to the evaluation of Aβ1-40 as a useful marker to differentiate various dementia conditions. Finally we are also evaluating the usefulness of the measurement of α-synuclein and classical CSF biomarkers to distinguish synucleinopathies (PD and dementia with Lewy Bodies) from other neurodegenerative diseases. Finanziamenti ricevuti Titolo progetto cNEUPRO (contract no. 199 LSHM-CT-2007-037950) Ente finanziatore EU – FP6 Durata progetto Three years (2007-2010) Abstract del progetto Ente finanziatore Recent research has clearly demonstrated that multiparametric neurochemical dementia diagnostics (NDD) in cerebrospinal fluid (CSF) does improve the early and differential diagnosis of dementias. cNEUPRO will apply advanced proteomic tools to discover novel neurochemical dementia markers in blood and CSF for the improved early and possibly predictive diagnosis of AD. A predictive dementia diagnosis will support the most effective use of forthcoming preventive therapeutic strategies. Our initiative will establish European SOPs for current neurochemical dementia diagnostics (NDD). A strong methodological impact is put on the quality of pre-analytical sample handling and clinical phenotyping, which has been neglected in industry-driven discovery studies. cNEUPRO integrates innovative biotech and bioinformatic companies with leading clinical and proteomic dementia research centres. cNEUPRO will also support the discovery of new diagnostic targets and is also promising to identify novel scaffolds for advanced molecular neuroimaging. CSF lysosomal hydrolases’ activity as possible marker of Parkinson’s disease MJFF Durata progetto 1 year Abstract del progetto Objective/Rationale: Recent studies have linked lysosomal dysfunction to the accumulation of α-synuclein oligomers and α-synuclein-mediated cell death. Clinical, neuropathological and genetic associations between Gaucher’s disease, a lysosomal storage disease, and Parkison’s disease (PD) have been reported. Also, a reduction of the activities of β-glucocerebrosidase, α and β-mannosidase has been reported in CSF of PD patients. To date there is no accepted diagnostic test for Parkinson’s disease based on biochemical analysis of blood or cerebrospinal fluid (CSF). The potential use of CSF lysosomal enzyme activities as a diagnostic indicator, or as a marker of disease progression in PD, will be investigated. Moreover the CSF levels of alpha-synuclein – a candidate diagnostic biomarker of PD – will be combined with the lysosomal enzyme activities in order to verify if there is any association between these biochemical parameters.Project Description: De-novo and treated patients referring to the Section of Neurology, University of Perugia, Italy, will be included in the study. CSF samples will be collected from patients with PD (n=80) and healthy subjects (n=50). Lysosomal enzyme activities (betaglucocerebrosidase, alpha-mannosidase, beta-mannosidase, beta- Titolo progetto hexosaminidase, beta-galactosidase, alpha-fucosidase, arylsulfatase A, arylsulfatase B, cathepsin D, and cathepsin S) will be determined in CSF samples using specific fluorimetric substrates. The levels of monomeric and oligomeric alpha-synuclein, using a sensitive and specific ELISA method able to reveal levels as low as 1 pg/mL in human biological fluids, including plasma and CSF, will be also evaluated. 3. HUMAN/CLINICAL RESEARCH D. Diagnosis: biomarkers Nome Contatti Istituto/Dipartimento Roberta Ghidoni, Giuliano Binetti +39-030-3501725 [email protected] IRCCS “Centro San Giovanni di Dio-Fatebenefratelli” Proteomics Unit, NeuroioGenLab Memory Clinic, Brescia, Italy Proposta di ricerca Over the past 10 years, frontotemporal lobar degeneration (FTLD) has emerged as the most common cause of dementia under the age of 60 years, outstripping even Alzheimer disease in prevalence. The increasingly common occurrence of FTLD, and its devasting impact on families and patients within their most productive years, urges novel approaches to cure or to prevent it. To date, there are no Food and Drug Administration approved medications for FTLD. FTLD has a strong genetic component, with up to 50% of cases being caused by mutation. Recently the progranulin gene (PGRN) was shown to be the most frequent genetic determinant: 68 mutations have been described in 212 families which account for 5–10% of FTLD cases worldwide (http://www.molgen.ua.ac.be/FTDmutations/). At present, the most common PGRN mutations worldwide are the Arg493X mutation, which has been identified in 37 patients belonging to 30 genealogically unrelated families with FTLD (the “Mayo Clinic” cohort) (Rademakers R et al,Lancet Neurol. 2007) and the PGRN Leu271LeufsX10 mutation, that we identified in 49 patients belonging to 38 unrelated Italian families (the “Brescia” cohort) (Benussi et al. NBA 2008;Benussi L and Ghidoni R et al, NBD 2009; Benussi L et al. ADAD in press).We recently reported that low plasma progranulin levels predict progranulin mutations in frontotemporal lobar degeneration. (Ghidoni R et al, Neurology 2008). Progranulin therefore has become a promising target for new therapeutic FTLD approaches. Our resources are: 1) Clinical expertise in FTLD: the IRCCS in Brescia has become a reference center for FTLD and related disorders serving a large geographical area in Lombardia. 2) The largest (along with the “Mayo Clinic cohort”) biological sample collection of PGRN mutation carriers (the “Brescia” cohort). Our scientific work in the last years has generated one of the largest cohort of FTLD families and biological sample collection of PGRN mutation carriers (affected and presymptomatic), that is an invaluable resource for research studies 3) Cellular and animal disease models. Our specific aims are: 1) Elucidating the role of progranulin in the disease onset and progression: we will study the molecular mechanisms underlying neurodegeneration in PGRN mutations carriers (plasma/serum) as well as in cellular (human fibroblasts and lymphocytes ) and animal models. 2) Biomarkers discovery for the differential diagnosis of FTLD vs Alzheimer’s disease (AD). FTLD is often misdiagnosed as another type of dementia (AD) and hence biochemical diagnostic markers would aid in differential diagnosis. Analysis of CSF is so far the most convenient method for studying the biology of neurodegenerative diseases in living patients. For a global proteomic study, surface-enhanced laser desorption/ionization mass spectrometry (SELDI-TOF MS) as well as two-dimensional gel electrophoresis (2-DE) will be used to study differential CSF protein expression. 3) Searching for new genes: Identification of novel disease-associated loci by linkage and gene expression analysis. Centers collaborating to this project: IRCCS Centro S.Giovanni di Dio-Fatebenefratelli, Brescia;. Istituto di Ricerche Farmacologiche "Mario Negri" , Milan (G. Forloni); Università degli Studi di Brescia (PF Spano); University of Milan (D. Galimberti, E. Scarpini); IRCCS Besta, Milan (F. Tagliavini) Area di interesse identificata Translational research Finanziamenti ricevuti Titolo progetto Proteomics of cognitive and movement disorders: Identification of molecular mechanisms associated with disease onset and phenotypic variability of frontotemporal lobar degeneration- (2009-2633) Ente finanziatore FONDAZIONE CARIPLO, Bando Ricerca Biomedica 2009 Durata progetto 01-01-2010/01-07-2012 Abstract del progetto The main objective of the present study is to unravel the pathophysiological bases of progranulopathies by 1) proteomic approach- For a global proteomic study, SELDI-TOF as well as two-dimensional gel electrophoresis (2-DE) will be used to study differential protein expression in subjects carrying or not PGRN mutations 2) neuroimaging study- The aim of the neuroimaging study is to describe the MR features of PGRN mutations with conventional, non-conventional, and functional neuroimaging. 3) personality and behavioral assessment- We will assess the personality and behavioral trait of asymptomatic at risk family members carrying PGRN mutations 4) cellular and animal models studies- Titolo progetto Project 1: “Validation of genetic and biochemical markers for early diagnosis of AD and the prediction of conversion of MCI into AD, and design of a multivariate molecular protocol having high diagnostic and prognostic accuracy”. Project 2: “Marker biologici di neurodegenerazione utilizzabili nella pratica clinica ai fini della diagnosi precoce e differenziale, della individuazione delle forme con risposta ottimale alle attuali terapie” Project 1: Italian Ministry of Health, Progr. Strategico PS39, prog. 1 Project 2: Italian Ministry of Health, Progr. Strategico conv.71, prog. 6 Project 1: 01-01-2009/31-12-2010 Project 2: 07-01-2008/07-04-2010 These programs have two primary aims: 1) The development and validation of a multi-factorial protocol that integrates molecular, imaging, neurophysiological, neuropsychological and behavioral data for early diagnosis of AD, in particular during the preclinical phase and the prodromal stage of "mild cognitive impairment" when the symptoms are not severe enough to fulfill the current diagnostic criteria for AD. 2) Validation of this new diagnostic protocol within a regional public health network, and evaluation of the health care, organization and economic implications of its transfer to the National Health Service. Ente finanziatore Durata progetto Abstract del progetto Referente: Prof. Carlo Ferrarese Phone. 039.233.3595 Fax. 039.233.2449 Email: [email protected] Clinica Neurologica, Ospedale San Gerardo Via Pergolesi 33 – 20052 Monza (MB) Laboratorio di Neurobiologia Dipartimento di Neuroscienze e Tecnologie Biomediche – Università di Milano-Bicocca Via Cadore 48 – 20052 Monza (MB) The main interest of our group consists in the integration of clinical and basic scientific research to understand the pathogenic mechanisms of neurodegenerative disorders and to identify new strategies for diagnosing, treating and preventing such untreatable diseases. Our center is composed by a clinical core (Department of Neurology, 42 beds plus Laboratory of Neurophysiology and Laboratory of Neuropsychology) at the san Gerardo Hospital (Monza, Italy) and a biological facility (Laboratory of Neurobiology, Department of Neuroscience and Biomedical Technologies, University of Milano-Bicocca). Regarding dementias and cognitive dysfunction, the main body of our clinical work is centered at the Memory Clinic (U.V.A. outpatient facility) that sees more than 300 patients among Alzheimer’s disease (AD) and other types of dementia. Clinical activity is deeply involved with research epidemiological, clinical, and neurobiological. Novel experimental therapies are offered; for example, a phase 3 trial testing the efficacy of an anti-beta amyloid antibody in AD is currently recruiting patients. The strict connection existing between the Memory Clinic and the Laboratory of Neurobiology allows studying novel peripheral markers in AD (mainly biochemical alterations in blood elements and skin fibroblasts) that might in the future be used for accelerating diagnostic processes, helping during follow-up, targeting personalized therapies, and refining prognostic previsions. Bioresearch methods currently employed include: cell cultures, immunochemistry, molecular biology, binding and uptake techniques, HPLC, confocal microscopy. For example, our group recently demonstrated an increase of sAPPαplasma levels with respect to healthy controls, while no differences were found regarding beta-amyloid (Abeta) levels. Moreover, we set up an ELISA assay for the determination of plasma anti-Abeta 1-42 levels, although no differences were found between AD patients and controls. However, we observed that AD patients not receiving acetylcholinesterase medications (AChEI) displayed anti-Abeta 1-42 antibodies plasma levels significantly lower with respect both, healthy controls, and AD patients receiving AChEI. Hence, we are currently analyzing the effect of the AChEI therapy on the immune response in AD patients. A putative immunomodulatory effect of these drugs might eventually further support immunoglobulin-based therapies for treating dementia. Furthermore, our group extensively studies the interactions existing between glutamatergic system, APP processing and signal transduction mechanisms (kinases) in peripheral (platelets, fibroblasts, lymphomonocytes) and cell models (neuroblastoma SHSY5Y). Another neurobiological interest of our group is related to Abeta catabolism. We focused on neprilisin, a major Abeta catabolic enzyme, studying its activity and expression in human fibroblasts. Neprilisin activity is reduced of ~50% in AD versus controls, while the expression in unchanged. In vitro “aged” Abeta treatment reduces neprilisin expression in both groups, while the activity decreases in controls and increases in AD patients, possibly due to a compensatory mechanism; this preliminary data suggests that Abeta is able to influence its own catabolism. A more innovative approach for reducing the toxic effect of Abeta consists in administering nanoparticles, molecules of nanometric dimensions able to bind Abeta limiting its aggregation and resulting toxicity. Within an FP7 European project we are testing the biocompatibility of various nanoparticles, and their ability to bind Abeta in vitro (cultured fibroblasts) and in biological fluids (plasma and serum) in order to identify the best candidates for future in vivo administrations. In the neuropsychological field, our current interests include: (a) development and validation of multiple ultrafast tests (5 minutes duration max) of the global cognitive status in order to propose them as rapid screening tools for general practitioners; (b) validation of a test of divided attention for the differential diagnosis of degenerative dementias; (c) development and validation of a novel test of denomination for the differential diagnosis between mild cognitive impairment (MCI) and dementia; (d) extensive study of visuo-spatial functions by a broad test battery, in order to define the differential cognitive profile between Lewy body dementia, corticobasal degeneration and posterior cortical atrophy; (e) study of the experimental neuropsychological (decision making, moral judgment, multi-tasking, humour appreciation) and neurobiological correlates of BDNF in AD, frontotemporal dementia and Lewy body or Parkinson dementia; (f) multicentric longitudinal cohort study recruiting patients affected by degenerative and vascular dementias or MCI to be followed for five years by repeatedly administering a neuropsychological test battery. Finally, our clinical department collaborates with the Department of Nuclear Medicine - Molecular Bioimaging Centre for both clinical and research purposes. Currently we are studying the existence of correlations between biochemical markers and functional neuroimaging data (PET-SPET). Tracers able to mark beta-amyloid or microglial activation, among others, might eventually integrate and confirm the value of the above described peripheral markers. Nome Piero Parchi M.D., Ph.D. Contatti [email protected] phone: 051-2092740 fax: 051-2092751 Istituto/Dipartimento Dipartimento di Scienze Neurologiche Università di Bologna, Italy Proposta di ricerca Aree di interesse identificate - Biobanking (blood sample, CSF, brain repository…) Standardization of diagnostic criteria and diagnostic instruments, harmonization and assessment tools Early diagnosis; Biomarkers Basic research: Biochemical characterization of abnormal protein aggregates to understand phenotypic diversity in neurodegenerative diseases Finanziamenti ricevuti Titolo progetto Ente finanziatore Durata progetto Abstract (estratti) del progetto 1) Molecular Pathology of the Human Brain; 2) Development of new diagnostic approaches for prion diseases 3) Validation and search of CSF biomarkers for the prediction of the clinical conversion from mild cognitive impairment (MCI) to dementia 1) EU, 2) Italian Ministry of Health, 3) Galletti Foundation 1) 5 years (Brain-Net II, project ended December 2009), COST application currently under evaluation (at final step). 2) 3 years 3) 3 years 1) Studies on human brain tissue have enabled the discovery of key factors of the most common brain diseases. Aβ, tau, synuclein and TDP-43 are outstanding examples of proteins identified in diseases such as Alzheimer (AD), Parkinson (PD) and Frontotemporal Lobar Degeneration (FTLD). The advent of molecular genetic techniques enabling insights into temporal or permanent changes of genetic activity (epigenetics) and the identification of novel regulators of translation (miRNA) have ushered in a new era that makes investigations of human brain tissue still indispensable. In this project ) we will provide highquality human brain tissue as an essential research resource by applying and further developing a shared database, common diagnostic criteria and ethical standards at European level. Furthermore, new technologies will be employed to investigate the causes of neurodegenerative disorders using such tissues. This collaborative group of experts (see http://www.brainneteurope.org) competent in all aspects of work with human brain tissue will foster European research on these devastating brain diseases. 2) The overall objective of this project is to improve the diagnosis and the prognosis of human TSE diseases through the development of new biochemical tests and the optimization of already available diagnostic tools. To this aims we will apply new proteomic approaches to identify TSE-specific patterns in the CSF and in plasma (or serum) of patients with sporadic CJD and we will characterize novel biochemical properties of the abnormal PrPTSE aggregates that may further improve the strain-specific diagnosis of the disease subtype. 3) As one of the Units involved in a larger project (see Form sent by Dr. Gallassi from our Department) we will validate and search for molecular CSF markers that might be of value in predicting the conversion from MCI to full-blown dementia and critical for the monitoring of patients once new therapies become available. We will focus on already established markers such as A-beta, total tau and fosfo-tau and more recently discovered molecules such as TDP-43 and progranulin. Different techniques such as Elisa and western blotting will be applied. The data will be correlated with those obtained from the neuroimaging and neuropsychological serial evaluation. SICILIANO Analisi biochimica di parametri di stress ossidativo. Uno dei problemi clinici più attuali riguardala difficoltà a comprendere se lievi disturbi della memoria in età senile siano oppure no segni iniziali di Malattia di Alzheimer. A tal proposito va ricordato come attualmente si faccia riferimento a un quadro clinico-neurologico quale il decadimento cognitivo lieve (“Mild Cognitive Impairment o MCI”) ,quale condizione caratterizzata, come appunto il termine indica, da una lieve compromissione di funzioni mentali, in primo luogo la memoria, ma potenzialmente a rischio di sviluppare nel tempo una malattia di Alzheimer e pertanto da seguire attentamente nel suo ulteriore decorso. Un altro aspetto fondamentale è inoltre rappresentato dal fatto che, in assenza di un marcatore biologico in vivo che permetta di fare diagnosi di certezza della malattia in vita, assume particolare importanza la ricerca di parametri di laboratorio che siano in grado, in aggiunta a fornire elementi indicativi sulle cause di malattia, di essere indicatori di eventuale rischio, evoluzione e diagnosi precoce di malattia, oltre che predittivi di risposta al trattamento. Lo stress ossidativo è stato chiamato in causa come fattore implicato nella patogenesi delle malattie neurodegenerative, inclusa la malattia di Alzheimer, in base all’osservazione che si ha evidenza precoce di danno mediato da specie reattive dell’ossigeno, non soltanto a livello del sistema nervoso centrale ma anche a livello periferico. Poiché le specie reattive dell’ossigeno sono molecole altamente instabili, la loro misura diretta in campioni di plasma o siero può non riflettere la loro reale concentrazione in vivo; per tale motivo vengono dosati solo i prodotti finali di stress ossidativo. Distinguiamo i biomarker possono essere distinti in due grandi gruppi: 1. Marker di danno ossidativo alle proteine, ai lipidi e al DNA, che rivelano lo stato di ossidazione di tali macromolecole e che danno un’idea del grado di gravità del processo degenerativo in atto all’interno della cellula; 2. Marker dello stato antiossidante, che riflettono la capacità totale dei sistemi cellulari, enzimatici e non enzimatici, di difesa contro l’attacco delle specie chimiche reattive. Presso il nostro laboratorio sono valutati markers di danno ossidativo alle proteine (AOPP), markers dello stato antiossidante quali la capacità ferro-riducente del plasma (FRAP) e la concentrazione del glutatione totale, ridotto e ossidato. Gli AOPP consistono in un insieme di proteine tra cui la tiroglobulina, la γ-globulina, l’albumina e la mioglobulina (Witko-Sarsat et al., 1996). L’elettroforesi delle proteine mostra che il picco di AOPP ad alto peso molecolare è prevalentemente dovuto all’albumina che appare sotto forma di aggregati che probabilmente derivano da ponti disolfuro e/o da "cross-linking" della tirosina; al contrario il picco di AOPP a basso peso molecolare contiene albumina nella sua forma monomerica (Witko-Sarsat et al., 1996). In "vivo" i livelli plasmatici di AOPP correlano con i livelli di dimeri di tirosina, un marcatore di danno ossidativo delle proteine, e con la pentossidina, un prodotto di glicosilazione strettamente associato al danno ossidativo delle proteine (Selmeci et al., 2005). La relazione tra gli AOPP e i prodotti di perossidazione lipidica appare invece poco chiara. Le attuali conoscenze sembrano indicare che i lipidi non sono necessari per la formazione degli AOPP, ma che in “vivo” possono aumentare tale processo (Witko-Sarsat et al., 1996). Al valore della FRAP contribuiscono, per circa il 60% l’acido urico, per il 15% l’acido ascorbico, per il 5% l’α-tocoferolo, per il 10% le proteine e per il 5% la bilirubina (Benzie et al., 1996). La metodica che permette di determinare questo parametro sfrutta la capacità di questi antiossidanti di ridurre il ferro, sotto forma di ione ferrico presente nel reattivo FRAP, - a ione ferroso. La principale funzione del glutatione è quella di operare come antiossidante proteggendo le cellule dall'azione di specie proossidanti (Dringer et al., 2000). Le proprietà antiossidanti del glutatione sono dovute alla presenza del gruppo sulfidrilico della cisteina che gli permette di interagire sia con specie reattive dell’ossigeno, sia con altre sostanze elettrofile nell’ambito di numerosi sistemi antiossidanti e non (Pompella et al., 2003). Il glutatione allo stato ridotto (GSH) espleta la sua funzione riducendo i radicali liberi e convertendosi in glutatione ossidato (GSSG), composto da due molecole di glutatione unite da un ponte disolfuro. In questo senso, il GSH interviene nel metabolismo dell’acqua ossigenata, degli idroperossidi e di altri perossidi organici, è capace di inattivare il radicale idrossile, radicali perossilici, il perossinitrito, l’acido ipocloroso, radicali alcossilici e l’ossigeno singoletto. Può anche ripristinare la forma ridotta di altri antiossidanti come la vitamina E e la vitamina C. Valutazione clinica, test da sforzo e prelievi ematici per la valutazione dello stato ossidativo I parametri di stress ossidativo sopra descritti possono essere studiati sia in condizioni basali che dopo protocolli di esercizio. Le principali sedi di produzione di radicali liberi sono i mitocondri, gli organelli intracellulari deputati al metabolismo ossidativo; i mitocondri svolgono altresì un ruolo di primo piano nel mantenere l’omeostasi del calcio e nell’innescare la cascata di segnali che sottendono la degenerazione cellulare attraverso il meccanismo della apoptosi. Lo studio della funzione mitocondriale e dello stress ossidativo ad essa connesso può essere valutato attraverso l’andamento dei parametri ematici di stress ossidativo in relazione all’esercizio muscolare. Esempio di test da sforzo per la valutazione di parametri di stress ossidativo: I pazienti sono sottoposti a un test di sforzo incrementale sui muscoli dell’avambraccio, eseguito con l’ausilio di un miometro connesso ad un “hand-grip” (Digital Multi-Myometer, MIE Medical Research Ltd., Leeds, UK). Il protocollo di esercizio incrementale consiste nell’impugnare l’hand-grip del miometro e contrarre massimamente la mano contro la resistenza offerta dallo stesso, determinando, in Newton, il livello di contrazione volontaria massimale (CVM). Saranno eseguite, da tutti i pazienti, una serie di fasi contrattili o “steps”, condotte in maniera intermittente per un periodo di un minuto, con intervalli di riposo di 2 minuti tra uno “step” e l’altro. In ciascuno “step” le contrazioni intermittenti sono eseguite con frequenza di una al secondo contro la resistenza richiesta offerta dal miometro. L’esercizio inizia con un primo “step” al 10% della CVM, un secondo “step” al 40% della CVM ed un ultimo “step” al 70% della CVM. Il paziente potrà seguire su un display luminoso il livello di forza generato in ogni contrazione. Questo tipo di esercizio è principalmente aerobico all’inizio del test e diviene progressivamente anaerobico man mano che aumenta la forza esercitata per progressivo reclutamento delle unità motorie rapide (Milner-Brown et al., 1973). I prelievi ematici venosi per la determinazione dei marcatori biochimici di stress ossidativo sono eseguiti a livello basale, dopo il secondo “step”, dopo il terzo “step” e dopo 15 minuti dalla fine dell’esercizio. Nome Laura Calzà Contatti [email protected] Tel. +39 051 2097947 Cell. +39 335 310979 Istituto/Dipartimento DIMORFIPA Proposta di ricerca Area di interesse identificata Multidisciplinary projects Cognitive decline in multiple sclerosis. Cognitive defects are recognized as early events in MS, not correlated with white matter lesion. We are interested in studying the pathogenesis of early neural distress and lesion in MS animal models and neuroprotective strategies Standardization of diagnostic criteria and diagnostic instruments, harmonization and assessment tools Quality control programs for Abeta fragments in CSF and plasma and tau/Ptau in CSF using xMAP platform. Focus on clinical trials standardization Finanziamenti ricevuti Titolo progetto Translational medicine for novel diagnostic and therapeutic tools for neurodegenerative diseases Ente finanziatore Joint Regione Emilia Romagna-University of Bologna Durata progetto 3 years, under negotiation Abstract del progetto Development of a GLP-grade service for drug testing in animal models of Alzheimer disease, based on computerized analysis of video-tracking for learning and memory performance. Development Titolo progetto of cell-based platform based on neural stem cells derived from Alzheimer mice for the screening of chemical libraries of antiAlzheimer drugs. Effect of low-dose of ….. in chronic administration on cognitive behavior and morphological parameters in Tg2576 mouse Ente finanziatore Drug Company (covered by disclosure agreement) Durata progetto 12 months Abstract del progetto The project is aimed to evaluate efficacy of a novel gammasecretase inhibitor on behavioral performance, histopathology, biomarkers and synapse morphology and molecular markers in mice curring the swedish mutation of APP. This esperiment will test efficacy of chronic treatment from 5 to 19 months of age. Two doses of the test compund and one dose of a reference will be tested in transgenic vs wild tipe, age matching mice Plasma assay of Ab40/Ab42 (multiplex X-MAP-based immuno assay technology) in phase 1 clinical trial Titolo progetto Ente finanziatore Drug Company (covered by disclosure agreement) Durata progetto 20 months Abstract del progetto Placebo-Controlled, Ascending Single-Dose Study to Evaluate the Safety, Pharmacokinetics and Pharmacodynamics of …. in Healthy Young Male Subjects. The safety and tolerability of five ascending oral doses of … will be evaluated in 6 separate study sessions. In each of the study sessions, subjects will be randomized to … or placebo treatment. Overall, 60 subjects will participate in the study, 48 treated with … and 12 treated with placebo. Our lab in committed in efficacy variables blind analysis (Plasma -amyloid concentrations, A40/A42 fragments). Joint QC initiative “round Robin” study, an international interlaboratory evaluation of AB 140/1-42 levels from a common plasma sample set, toward standardization of multiplex/multiparametric methods for AB 140/1-42 quantification in plasma. Participating centers, 15; number of samples to analyze, 19. Remyelination failure in multiple sclerosis: a case of inflammationinduced tissue hypothyroidism? Titolo progetto Ente finanziatore AISM/FISM Principla investigator: Luciana Giardino Durata progetto 2 years Abstract del progetto Nome This project is aimed to investigate a possible mechanims for remyelination failure in multiple sclerosis. This project is aimed to verify if the inflammation associated to demyelination causes a defect of T4 to T3 conversion or a decreased expression of thyroid hormone receptors (nuclear receptors and membrane transporters) in the nervous tissue, thus leading to a local/cellular hypothyroidism and, thus, to the defect of white matter repair. This project could also provide insides for other neurodegenerative disease including white matter alterations, like Alzheimer disease Laura Calzà Contatti [email protected] Tel. +39 051 2097947 Cell. +39 335 310979 Istituto/Dipartimento DIMORFIPA Proposta di ricerca Area di interesse identificata Development of competitive animal models for AD GLP grade animal facility and behavioural testing for cognitive performance in AD animal models using video tracking and computer analysis; screening of new molecules after acute administration Basic research Brain metabolism of thyroid hormone: a risk factor for AD? We are interested in exploring possible connections between thyroid hormone content, receptor and activating enzyme expression, APP metabolism and Ab toxicity in the brain in normal and AD mice. Recent advances in knowledge on thyroid hormone brain metabolism and molecular biology of nuclear and membrane thyroid hormone receptors and transporters offer new possible targets to explore this topic. New treatment strategies Nome Contatti Istituto/Dipartimento Fabrizio Tagliavini Tel +39 02 2394 2260; Fax +39 02 2394 2101 Email: [email protected] Sito web: http://www.istituto-besta.it UO Neuropatologia – Neurologia 5 Fondazione IRCCS Istituto Neurologico “Carlo Besta” Via Celoria ,11 - 20133 Milano- ITALY Proposta di ricerca The research group: The Division comprises (i) a Clinical Unit (Dementia Center) devoted to the diagnosis and treatment of patients with degenerative dementias (over 2000 out-patients and 150 in-patients per year) and (ii) a Laboratory Unit (certified ISO 9001:2000 - Registration Number 26080, dedicated to the analysis of genes and biomarkers associated with degenerative dementias, post-mortem characterization of the disease process and disease-specific protein, and experimental studies on disease pathogenesis and the development of therapeutic strategies. The course of action of this activity can be expressed as a “Bed to Bench to Bed” cycle, with the ultimate goal to identify disease-modifying drugs in preclinical settings and apply them to patients. In this regard, the Clinical Unit is currently coordinating a multicentre phase II clinical trial supported by AIFA, to test the effectiveness of an anti-amyloidogenic molecule identified in experimental models. This comprehensive approach has also the great advantage to collect biological samples (plasma, DNA, CSF and brain tissue) from fully characterized patients. In this regard the Laboratory Unit is part of the Network of Excellence “BrainNet” Europe devoted to biobanking, harmonization of assessment tools and standardization of diagnostic criteria of neurodegenerative diseases. The permanent staff of the Division is composed by 8 MD and two technicians. In addition, the staff comprises 13 post-doctoral fellows (7 PhD, 3 MDV, 1 MD, 1 psychologist), 2 PhD students and 1 technician who are committed to the research activities on degenerative dementias with different expertise and roles. On the overall, the team has large experience in a variety of techniques spanning from neuropathology (including immunohistochemistry, morphometry, electron microscopy and atomic force microscopy), to molecular genetics, biochemistry (purification and characterization of disease-specific proteins), cellular and molecular biology, and animal models. The Division of Neuropathology is provided with fully equipped laboratories for histology, immunohistochemistry, electron microscopy, atomic force microscopy, biochemistry, and molecular and cellular biology, and a 60 sqm BL3 facility. Moreover an animal facility composed by a 140 sqm environmental controlled area, provided with a HVAC system and heap-filtered ventilated racks, with separate rooms for maintaining and breeding mouse lines, and carrying out surgical procedures, collection of samples and post-mortem examination is available. Research lines on Alzheimer’s disease A. Clinical research • Early diagnosis of Alzheimer’s disease (AD) We will characterize the phenotypic expression of AD and other degenerative dementias through multiplex analysis of a number of variables including neuropsychological and behavioural profile, neuroimaging, neurophysiological changes with special attention to sleep abnormalities, genetics, and plasma and CSF biomarkers. Furthermore, we will carry out longitudinal studies of presymptomatic carriers of gene mutations to detect early markers of conversion to the disease state. • Clinical trials We will carry out phase II and phase III clinical trials to assess the effectiveness of potentially diseasemodifying drugs, including molecules targeted to Aβ and protein tau. • Identification of disease-specific biomarkers We are currently working on a peripheral marker of AD having high specificity and sensitivity, based on cyclic amplification of misfolded Aβ and Aβ oligomers from biological fluids, similar to the PMCA technique successfully developed by Claudio Soto for prion diseases. The same approach will be used to amplify and detect misfolded tau. • Biobanking and neuropathological and molecular characterization of patients We will collect systematically plasma, DNA and CSF from patients with different types of dementia and non-demented individuals, and perform autopsies for neuropathological characterization and biochemical and molecular studies. B. Preclinical research • Recessive A673V APP mutation: molecular mechanisms and development of a new therapeutic strategy for sporadic AD We have recently identified an APP mutation (A673V) that causes early-onset AD only in the homozygous state while the heterozygous carriers are not affected. This mutation strongly boosts the production and amyloidogenic properties of Aβ. However, the interaction of A673V-mutated and wild-type peptides inhibits amyloidogenesis and A-mediated neurotoxicity (Di Fede et al., Science 2009, 323:1473-7). These findings are consistent with the observation that the A673V heterozygous carriers do not develop disease and offer grounds for a novel therapeutic strategy based on modified Aβ peptides. A research priority of our lab is to unravel the molecular mechanisms of the opposite effects of the A673V APP mutation in homo- or heterozygous state on amyloidogenesis and to develop a lead compound for AD therapy based on A673V-modified Aβ peptides or peptido-mimetic molecules. To accomplish these objectives we have generated a panel of transfected cells and transgenic C. elegans expressing human APP or Aβ with the A673V mutation, respectively, and transgenic mouse lines expressing A673V-mutated APP in the homozygous or heterozygous state. Furthermore, we have identified a prototypic lead compound corresponding to a six-mer Aβ peptide with the A673V substitution and are currently working on brain delivery systems. • Pathways leading to tau pathology A key event in AD pathogenesis is the hyperphosphorylation, misfolding and aggregation of the microtubule-associated protein tau leading to formation of neurofibrillary tangles, disruption of the neuronal cytoskeleton and neurodegeneration. Following the “Aβ cascade hypothesis” this event is triggered by aggregated Aβ species, particularly oligomeric assembly intermediates. However, the pathways linking Aβ and tau pathology are unknown. This is largely due to lack of animal models able to reproduce these two central aspects of AD. We have developed a mouse model of prion disease that shows a secondary tauopathy following deposition of PrP amyloid (Giaccone et al., Neurobiol. Aging 2008, 29:1864-73). This model will be used to investigate the molecular basis of tau pathology induced by deposition of an amyloid protein. • Role of nuclear tau in neurodegeneration in fronto-temporal dementia Tau is the major microtubule-associated protein of neurons and its primary role is to promote assembly and stabilization of microtubules required for morphogenesis and axonal transport. We have recently found that tau plays an important role also in chromosome stability, and mutations in the tau gene cause chromosome aberrations in peripheral cells in addition to formation of neurofibrillary tangles in neurons and glial cells. Our objective is to elucidate the molecular mechanisms underlying the genomic instability due to tau mutations using cellular models, and to investigate the contribution of the aneuploidy caused by mutated tau to neurodegeneration using a transgenic mouse model of tauopathy. Area di interesse identificata - Genetic susceptibility to Alzheimer’s disease (AD) CLINICA/PRECLINICA - Early diagnosis - Biomarkers - Developing competitive animal models to study AD - Studying early onset forms of AD to follow-up disease progression - Cinical trials with or without involvement of pharmaceutical companies - Basic research - Biobanking (blood sample, CSF, brain repository…) - Standardization of diagnostic criteria - New treatment strategies - Translational research Finanziamenti ricevuti 1. Titolo progetto Genoproteomics of Age Related Disorders (GuARD). Ente finanziatore Durata progetto Abstract del progetto 2. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto 3. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto 4. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto 5. Titolo progetto Ente finanziatore Durata progetto Abstract del progetto PI: Roberto Sitia Subproject leader: Fabrizio Tagliavini Cariplo Foundation 01/01/07-30/06/10 Project 3.2: Neurodegeneration. The main goals of this project are: (1) biochemical analysis of misfolded protein aggregates from brains of patients with sporadic and genetic forms of Alzheimer disease and prion diseases; (2) proteome analysis on brain tissue from the same patients and from gender- and age-matched non-demented control individuals; (3) proteome analysis on brain tissue from a mouse model of variant Creutzfeldt-Jakob disease. Alzheimer's disease: development and validation of a multi-factorial protocol for the diagnosis and follow-up of disease in the prodromal and incipient phase. PI: Fabrizio Tagliavini Italian Ministry of Health 01/01/09-30/06/11 This program has two primary aims: (1) the development and validation of a multi-factorial protocol that integrates molecular, imaging, neurophysiological, neuropsychological and behavioral data for early diagnosis of AD, in particular during the stage of MCI; (2) the validation of this diagnostic protocol within a public health network, and evaluation of the health-care, organization and economic implications of its transfer to the NHS. Molecular mechanisms underlying synaptic dysfunction in prototypic neurodegenerative diseases related to protein misfolding (nEUROsyn). PI: Fabrizio Tagliavini European Union/Italian Ministry of Health 01/02/09-31/01/12 This project is designed to investigate the mechanisms that lead to synaptic impairment and demise of neurons in AD and Huntington diseases using cellular and animal models, and patient material. The main goals are to: (1) study synaptic dysfunction; (2) identify alterations of synaptic and dendritic spine remodelling; (3) monitor changes in receptor trafficking at the synapse and disturbances in trafficking of different cellular components. A randomized, double-blind pilot study versus placebo for the evaluation of the efficacy of doxycycline administered by oral route in patients affected by Creutzfeld-Jakob disease. PI: Fabrizio Tagliavini Italian Italian Agency of Drug (AIFA) 01/09/06-31/08/10 This project is a multicentre phase II clinical trial to test the effectiveness of an anti-amyloidogenic molecule (doxycline) identified in experimental models. Biological markers of neurodegeneration to be used in clinical practice for early and differential diagnosis of degenerative dementias, the identification of fast and slow progressing forms, and the evaluation of response to treatment. PI: Gianfranco Spalletta, Project Leader: Fabrizio Tagliavini Italian Ministry of Health 01/07/08 to 01/06/10 The main goal of the project is the identification of peripheral markers of neurodegeneration in Alzheimer disease, Levy body dementia, frontotemporal dementia and prion diseases to allow (1) early differential diagnosis, (2) identification of fast and slow progressing forms, (3) evaluation of response to therapy. Nome Contatti Tel. E mail Istituto/Dipartimento Gianfranco Spalletta Via Ardeatina, 306 – 00179 Rome 06-51501575 [email protected] IRCCS Santa Lucia Foundation/Department of Clinical and behavioral neurology - Neuropsychiatry laboratory Proposta di ricerca Area di interesse identificata Mild Cognitive Impairment Finanziamenti ricevuti Titolo progetto Ente finanziatore Behavioral and Psychological Predictors of Cognitive Outcome in Subjects with Mild Cognitive Impairment: the Role of Non-conventional Neuroimaging and Serotonergic Genes Italian Ministry of Health Durata progetto 01/09/2010 – 31/08/2012 Abstract del progetto The present project will identify complex behavioural-endophenotypes, of behavioral-neuroimaging-genetics nature, useful for predicting the cognitive course of neurodegenerative dementias starting from their preclinical phase or MCI. In particular results will help to predict the outcome of the cognitive impairment, the relationship between cortical and subcortical brain atrophy and neuropsychiatric-neuropsychological longitudinal clinical manifestations, and a composite neuroimagingserotonin genetics index for better prediction of behavioral expression during the 18 months longitudinal course of the project. In order to do this an effort will be made to try to make the predictive capability of each clinical marker reliable for every single patient. Two-hundred people with a diagnosis of MCI will be recruited in this study. These patients will be drug free from psychopharmacological and acetilcholinesterase inhibitor drugs. One-hundred healthy controls (HC) will be also recruited. The subjects included will carry out an in depth anamnesis evaluation, a diagnostic evaluation for depression, psychosis, and apathy associated with dementia, and a psycho-behavioural dimensional assessment using a battery of tests including: Neuropsychiatric Inventory; CERAD Disforia; Dementia Apathy Interview Rating, Apathy Scale and Pittsburgh Sleep Quality Index. Moreover, the subjects will be evaluated through the MMSE, the Mental Deterioration Battery (MDB) and tests for basic and instrumental functional activities. This is a mixed case-control study (MCI at the baseline vs. HC) and longitudinal study (18-month follow-up only for MCI subjects). The first six months of research will be used for the inclusion of HC and MCI subjects in the study. Each cognitive and behavioural battery tests and diagnostic criteria will be administered in the MCI subjects every six months until 18 months from the first evaluation. Caregiver stress will be monitored at the baseline and the end of the follow-up. Image acquisition will be done at the baseline: all subjects will be examined using a 3 Tesla Allegra MR Imager with a standard quadrature head coil. Participants will undergo the same MR imaging protocol including whole-brain T2*weighted, T1-weighted and DTI scanning. Standard clinical sequences (FLAIR and T2-weighted images) will be acquired on the same subjects in order to exclude different pathologies. Image processing will be performed using FreeSurfer, FSL 4.1 and in-house developed software in Matlab vers. 6.5. The FreeSurfer toolkit will be employed to perform automatic cortical thickness reconstruction. The high resolution anatomical T1-weighted images will be also used to extract several region of interest corresponding to subcortical grey matter nuclei. The automatic segmentation of these nuclei will be obtained with the software FIRST, part of the FSL toolkit. Moreover, the DTI data will be coregistered to the T1 images in order to extract regional values of diffusion parameters (MD, FA) known to be indicative of tissue microstructure disruption, from the same nuclei identified in the previous step. The DTI data will be also employed to identify the main fiber tracts within the white matter. These tracts that will represent additional regions of interest from where extracting diffusion parameters. Genomic DNA will be purified from 200 microl of human whole blood. Polymorphisms and haplotypes of several genes will be analyzed in this study: the Apolipoprotein epsilon 4 (ApoE4) variant will be investigated in HC and MCI subjects in order to verify its influence on behavioural manifestations and symptom progression; the 102T/C polymorphisms of the 5HT2A receptor gene will be investigated for psychosis behavioural phenotype; to test the hypothesis that allelic variation in 5HTT gene-linked polymorphic region (5-HTTLPR) genotype is associated with behavioural syndromes and symptoms and cognitive outcome, a common 44-base pair deletion (s allele) polymorphism in the 5-HTTLPR which associated with reduced 5HTT transcription efficiency and 5HT uptake in vitro will be studied. The main focus of the research in the field of complex disorders, such as dementias, is to found new complex predictors which are valid and reliable in the prediction of clinical outcome starting from the early preclinical phases. In particular, mixed clinical (behavioural) and biological (neuroimaging, genetics) predictors may help to precociously identify those subjects who have a rapid decline of cognitive performances and those who will respond positively to the treatment with a stable course of the cognitive performances and who also improve. Another interesting possibility is that exist behavioural variables which may predict the course of the illness in the case they will not improve at the end of the follow-up. The longitudinal branch of this project is devoted to identify if this possibility exists and which are the behavioural predictors in terms of frequency of behavioural disorders at the end of the follow-ups. 3. HUMAN/CLINICAL RESEARCH E. Diagnosis: neuropsychology Nome Contatti Istituto/Dipartimento Maurizio Balestrino 010-3537078/7085/7066; [email protected]; FAX 0103538631; mobile phone 348-7814154 Department of Neuroscience, Ophtalmology and Genetics, University of Genova, Italy Proposta di ricerca Dementia of Alzheimer's type represents about 40% of the cases where a last will is challenged based on real or supposed mental incapacity of the testator (1). Usually, elder people find in their last will the reassurance that after their death their loved ones will have the resources to carry on their lives in agreement with the old person’s desires. However, when a person suffers (or is suspected to suffer) from Alzheimer’s disease, their last will is often disputed, on the ground that he/she may not have been mentally competent to make decisions. Thus, the peace of mind that comes with having written a thoughtful last will is considerably decreased. This is a growing problem, that is already painful from the social point of view (see for example http://elder-abuse-cyberray.blogspot.com/2007/06/where-there-iswill.html). Moreover, somebody may really take advantage of the declining cognitive status of an Alzheimer’s patient to have him/her write a will in his/her favor (2). The dispossessed heirs then claim that the will is invalid because the deceased was, at the time of writing it, incapacitated due to mental deterioration (3). In such cases, a judiciary trial follows, however the judgement is often difficult, because the writer of the will can no longer be examined, and the judgement must rely solely on the retrospective evaluation of medical records and of witnesses’ statements. Since both types of evidence are often incomplete or even discordant, the expert’s judgement is often difficult and flawed by obvious limitations. However, the last will has often been handwritten. If so, the quality of the handwriting may add very important elements to the expert’s evaluation, because it shifts the investigation from the retrospective medical records or witnesses’ claims to the direct observation of the deceased subject’s performance. The act of writing is by no means a mere movement of the hand and fingers, rather it is a cognitive performance from several viewpoints. The writer must (1) correctly build in his/her mind a suitable sentence, (2) translate words into graphic symbols, and (3) correctly arrange the writing in rows and columns, in such a way that it can be decoded. The first two tasks relate to speech abilities, and are usually negatively affected by aphasia. The third task implies a correct visuospatial ability. Both aphasia and visuospatial disturbances are prominent symptoms of Alzheimer’s type dementia (4). Accordingly, it has been shown that handwriting is compromised in demented patients (5,6,7,8). Even more important, a correlation exists between severity of handwriting disruption and severity of cognitive deficiency (for example, see Table 3 of reference (7) and Table 7 of reference (9)). While a wrong or confused handwriting is an obvious sign of mental impairment, the evaluation of this item in forensic psychiatry is currently hampered by the lack of the objective and quantitative tools that are required for a forensic examination. Although it has been demonstrated by several authors that writing is altered in demented patients (7,5,8,6), these data are difficult to apply to the evaluation of a single case, as it is required for judiciary purposes. By contrast, a quantitative score of handwriting that was able to identify severe mental deterioration with a high probability level would be an invaluable tool in the forensic evaluation. In preliminary research we addressed this issue by designing a semiquantitative standardized score system to evaluate handwriting, and investigated both its inter-rater reliability and its correlation with standard measures of cognitive performance (10). In that preliminary research we showed (a) that our semiquantitative handwriting scoring method has good interrater agreement and (b) that there is a statistically significant correlation between the degree of cognitive deterioration (as gauged by the score of both Mini Mental State Examination – MMSE - and of Milan Overall Dementia Assessment – MODA scale) and the handwriting score (10). We now propose to continue that research by testing a larger number of patients, to more definitely quantify the correlation between severity of dementia and handwriting score. Specifically, we plan to (1) Repeat our investigation on a larger number of patients to hopefully better validate it. (2) Administer to our patients not only the MMSE and MODA tests, but also the Alzheimer´s disease assessment scale ( ADAS-COG), an internationally accepted tool for the assessment of Alzheimer’s disease. (3) Validate the test by identifying cutoff values of our handwriting score that reliably identify persons with normal mental capacity and persons with mild, moderate or severe dementia. We will test the hypothesis that (a) a writing score lower than a certain value (to be indentified in ou proposed research) reliably identifies patients whose mental deterioration is minimal or inexistent and (b) a writing score higher than a certain value (writing score=5 according to our preliminary research, see ref. 10) reliably identifies persons with severe mental deterioration. We will carry out our research using a protocol similar to our published work (10). We will exclude patients having dementia not of Alzheimer's type, patients having severe motor deficit in the dominant hand, and patients having aphasia so severe as to prevent communication altogether. We believe that our reseach will help to bring justice and peace of mind to Alzheimer's disease victims and to their families, by providing a method, scientifically rooted in clinical neuropsychology, that will help preventing abuse and exploitation of Alzheimer’s disease patients through their last will. While this is in itself an important goal of our research, we also believe that the quantification of the relationship between Alzheimer’s disease and handwriting will open a new way of testing cognitive impairment in Alzheimer’s disease, with important fallout to the entire field of neuropsychological testing of Alzheimer’s disease patients. References 8) Shulman, K. I., Cohen, C. A., and Hull, I., (2005) Psychiatric issues in retrospective challenges of testamentary capacity. Int.J Geriatr.Psychiatry 20:63-69. 9) Peer, I. N., (1981) Wills, testamentary capacity and undue influence. Bull.Am Acad Psychiatry Law 9:15-22. 10) Redmond, F. C., (1987) Testamentary capacity. Bull.Am Acad Psychiatry Law 15:247-256. 11) Victor, M. and Ropper, A. H., (2005) Adams and Victor's Principles of Neurology. 8th Ed.: 12) Werner, Perla, Rosenblum, Sara, Bar-On, Gady, Heinik, Jeremia, and Korczyn, Amos, (1-7-2006) Handwriting Process Variables Discriminating Mild Alzheimer's Disease and Mild Cognitive Impairment. Journals of Gerontology Series B: Psychological Sciences and Social Sciences 61:228236. 13) Croisile, B., (2005) [Writing, aging and Alzheimer's disease]. Psychol Neuropsychiatr.Vieil. 3:183197. 14) Silveri, Maria Caterina, Corda, Francesca, and Di Nardo, Miriam, (2007) Central and peripheral aspects of writing disorders in Alzheimer's disease. J Clin Exp Neuropsychol. 29:179-186. 15) Forbes, Katrina E., Shanks, Michael F., and Venneri, Annalena, (1-3-2004) The evolution of dysgraphia in Alzheimer's disease. Brain Research Bulletin 63:19-24. 16) Luzzatti, Claudio, Laiacona, Marcella, and Agazzi, Daniela, (2003) Multiple patterns of writing disorders in dementia of the Alzheimer type and their evolution. Neuropsychologia 41:759-772. 17) Fontana P, Dagnino F, Cocito L, Balestrino M. (2008) Handwriting as a gauge of cognitive status: a novel forensic tool for posthumous evaluation of testamentary capacity. Neurol Sci. 29(4):257-61. 18) Brazzelli, M., Capitani, E., Della Sala, S., Spinnler, H., and Zuffi, M., (1994) A neuropsychological instrument adding to the description of patients with suspected cortical dementia: the Milan overall dementia assessment. J Neurol Neurosurg.Psychiatry 57:1510-1517. 19) Folstein, M. F., Robins, L. N., and Helzer, J. E., (1983) The Mini-Mental State Examination. Archives of General Psychiatry 40:812. Area di interesse identificata - Standardization of diagnostic criteria and diagnostic Finanziamenti ricevuti Titolo progetto // Ente finanziatore // Durata progetto // instruments, harmonization and assessment tools Multidisciplinary projects Responsabile: Angiola Maria Fasanaro (Neurologo) Collaboratori: Raffaele Rea (Volontario Neuropsicologo) Anna Carotenuto ( Volontario Neuropsicologo) Maila D’Anonio (Volontario Neuropsicologo) Luisa Colucci ( Tirocinante Neuropsicologo) Sabrina Carpi ( Tirocinante Neuropsicologo) Massimiliano Bosco ( Tirocinante Neuropsicologo) Ivana Molino ( Tirocinante Neuropsicologo) Gabriella Finizio (Tirocinante Neuropsicologo) Antonio Ziello ( Tirocinante Neuropsicologo) La Unità Valutativa Alzheimer, da me diretta è attiva dal 2000, e fa parte della Struttura Semplice di Malattie Involutive Cerebrali, Dipartimento di Neuroscienze, AORN "A. Cardarelli". Le attività dell’ ambulatorio sono prevalentemente svolte su soggetti geriatrici. L’esperienza maturata nel corso di 10 anni di attività ha permesso di valutare oltre 900 pazienti , ed attualmente circa 400 sono in terapia regolare. I dati sono raccolti in cartelle cliniche ad hoc ed archiviati elettronicamente. I protocolli seguono le Linee Guida Internazionali e si avvalgano dell’ applicazione di test neuropsicologici specifici di approfondimento. Particolare attenzione viene dedicata all’ amnesic/non amnesic Mild Cognitive Impairment e ai fattori vascolari associati alle demenze. I pazienti sono controllati a distanza di uno, tre e sei mesi dall’inizio della terapia, ed ogni sei mesi successivamente. Oltre alle attività diagnostiche e di monitoraggio l’Unità svolge un servizio di counseling diretto ai caregiver e programmi di riabilitazione diretti a preservare capacità residue. Attualmente il centro si avvale della collaborazione di 9 neuropsicologi tra volontari e tirocinanti che costantemente si occupano delle attività ambulatoriali e di ricerca. In quest’ ambito è stata svolta una collaborazione regolare nel triennio 2005-2007 con L’ Università di Tunisi per un progetto di implementazione delle metodiche neuropsicologiche.(progetto finanziato dal Ministero degli Affari Esteri.) Abbiamo una collaborazione stabile con la Cattedra di Neuropsicologia Clinica della Seconda Università degli Studi di Napoli. e con il Dipartimento di Medicina Sperimentale e Sanità Pubblica dell’Università di Camerino. E in corso di pubblicazione una ricerca realizzata insieme all’Università Orientale di Napoli e all’ Università La Sapienza di Roma. I principali progetti di ricerca attualmente attivi 1) Caratteristiche neuropsicologiche differenziali tra pazienti con Demenza fronto-temporale e con variante frontale della Malattia di Alzheimer. 2) Il contributo del network frontale alla genesi delle allucinazioni in pazienti con malattia di Alzheimer e con malattia di Parkinson. 3) I fenomeni attrattivi spaziali ed il closing-in 4) Caratteristiche fMRI del Mild Cognitive Impairment Amnesico. 5) Caratteristiche neuropsicologiche delle varianti atipiche della Malattia di Alzheimer. 6) Valutazione cognitiva e monitoraggio degli effetti terapeutici dei trattamenti farmacologici (Rivastigmina patch, Precursore Colinergico Colina alfoscerato, RO5313534 ) sui sintomi cognitivi e comportamentali della malattia di Alzheimer e nelle forme con danno vascolare associato. 7) Analisi dei costi sociali ed economici delle demenze. 8) Arte visiva come proposta di riabilitazione cognitiva e motivazionale 9) Depressione e Malattia di Alzheimer studio sui correlati gene-ambiente e risposta al trattamento farmacologico. D. Nome Roberto Gallassi Contatti [email protected] Istituto/Dipartimento Department of Neurological Sciences, University of Bologna 1) Centre for the Neurological Study of Brain Aging – Gallassi Roberto 2) Neurogenetic Unit – Carelli Valerio 3) Neuropathology Unit – Parchi Piero Department of Internal Medicine, Aging and Nephrology, University of Bologna 1) MR Spectroscopy Unit – Lodi Raffaele Proposta di ricerca Area di interesse identificata Earli diagnosis Finanziamenti ricevuti Titolo progetto Predictors of conversion from MCI to dementia Ente finanziatore Fondazione Gino Galletti Durata progetto 3 years Abstract del progetto Background MCI is a well known clinical condition intermediate between normal ageing and dementia. It is not completely ascertained if this entity is a beginning of a dementia or a clinical syndrome which can have different outcomes. Anyway, the annual conversion rate of MCI to dementia is variable in different previous studies, ranging from about 4% to 12% or more. The investigated factors of conversion taken into account are clinical aspects, neuropsychological tasks, conventional and functional neuroimaging measures, biological markers and genetic factors. Previous neuropsychological studies indicates that MCI may be characterized by subtle impairment in learning and delayed recall, particularly in tasks of episodic memory and visuospatial memory, in executive functions, phonemic and semantic verbal fluency, etc. In our recent study, we found that a behavioural memory battery, visual attention, long-term verbal memory tasks and Neuropsychiatric Inventory caregiver distress and apathy scores, resulted in independent predictors of conversion to dementia. We also found that single domain impairment, including memory, has a minor probability to develop dementia than multiple domain impairment. Rationale We hypothesize that some cognitive functions and neuropsychological tasks are more sensitive in predicting the conversion of dementia in relationship with modifications in other clinical, biochemical, genetic and neuroimaging findings. Aim The aims of this project are: - - - - to evaluate at baseline demographic features, clinical characteristics, cognitive and behavioural profile, biochemical and genetic findings in a sample of MCI patients of amnesic or not amnesic type and with single or multiple domain impairment. to perform clinical and MR neuroimaging longitudinal observation during three years of the MCI sample, evaluating the evolution of the single patients, in particular the conversion to dementia to correlate with a multi-dimensional approach the relationship between clinical, cognitive and behavioural findings and the other methods of investigation such as quantitative MR neuroimaging measures, haematic and cerebrospinal fluid biological data and the genetic profile of patients to evaluated the independent predictors of conversion from MCI to the various type of dementia F. Biobanking Bioinformatics 3. HUMAN/CLINICAL RESEARCH G. Ambient assisted living and socioeconomics Roberto Monastero, MD, PhD Lab of Epidemiology and Psychology of Aging and Dementia Dept of Clinical Neuroscience University of Palermo Via La Loggia 1 90129 - Palermo, Italy Tel. +39-091-6555185 Fax. +39-091-6555113 email. [email protected] [email protected] PROGETTO2 Type: Ambient Assisted Living (AAL) Joint Program, 2008 Title: Home-based Empowered Living for Parkinson’s disease Patients (HELP) Responsibles: Prof. Giuseppina Campisi, Dr. Roberto Monastero Amount of fund: ......................... Lenght: 3-year project Abstract Medical and social development have produced remarkable changes in the population distribution at worldwide level, leading to a dramatical increase in the life expectancy of the human population. the higher life expectancy is usually associated with the appearance of chronic conditions, such as Parkinson's disease (PD), and associated comorbidites, which burdens and have a great impact in the quality of life of both the persons suffering the illness and his/her relatives and caregivers. Besides, chronic conditions also derive in higher costs for the care delivery process, becoming a problem for the whole society. In this sense, the technological advances of the last decades can, and should, provide solutions to better manage the treatment for the patients, reducing the social and economic impact of the chronic conditions. The Homebased Empowered Living for Parkinson’s disease Patients project will include a system able to administer drug therapy in a controlled and either continuous or on-demand basis, to control disease progression and to mitigate PD symptoms. Accordingly, it will give a comprehensive service to help Parkinson's patients to better cope with their illness and reduce comorbidity. Country: ITALY Contact person: Alessandra Pedrocchi (Politecnico di Milano) Date: 14th Jan 2010 I) Strategic Issues TOPIC 1 Neuro-rehabilitation and brain functional imaging: rehabilitation customization driven by best exploitation of cerebral plasticity and health system sustainability The spreading of neuro-motor pathologies has a great impact on the healthcare system European Countries, being among the first causes of death (stroke is the third) but utmost being the first case of high severity long-term disabilities. The recovery and the progress of neuromotor diseases is strongly affected by the pathogenesis but also by the rehabilitation training, which has the purpose to best exploit the cerebral plasticity governing neural motor control reorganization. Beside the traditional therapist assisted training, different novel solutions has spread in the latest years based on robotic systems, functional electrical stimulation (neuroprostheses) and hybrid systems where robots and electrical stimulation are integrated. Nonetheless, the international scientific community still lack of a complete well supported comparative validation of these approaches and consequently a specific customization of the best training cocktail for each patient is still not documented by scientific results and it is mainly devoted to clinician personal convictions and experience. Especially, we claim that in term of motor recovery and motor relearning, i.e. cerebral re-organization, there is no clear scientific substrate to support the use of robotic devices and/or neuroprostheses aside or in alternative to standard rehabilitation. However, in the latest decade the possibility to investigate in-vivo cerebral activations associated to movement execution has become accessible , thanks to fMRI, EEG and fNIRS,… The challenge to be tackle in the next future is to integrate these two aspects aiming at studying the impact of rehabilitation selections on the cerebral plasticity reorganization in neuromotor pathologies. Beside the efficacy of those methods and their best exploitation, it also needs to be tested the sustainability of these solutions for the long term economical, environmental and social impact of biomedical technologies in the rehabilitation of neuromotor pathologies. Nome Alberto Pilotto Contatti Phone: +39 0882 410271 Fax: +39 0882 410271 E-mail: [email protected] Unità Operativa di Geriatria & Laboratorio di Gerontologia-Geriatria Dipartimento di Scienze Mediche IRCCS Casa Sollievo della Sofferenza Viale Cappuccini, 1 – 71013 San Giovanni Rotondo (FG) Istituto/Dipartimento Proposta di ricerca Sporadic Alzheimer’s disease (AD) is a progressive neurodegenerative disorder occurring predominantly in older age. The prevalence of AD rise from 20% after 75 years to 30% after 85 years, and about 5% of people aged 65 years or older have AD. With about 3 to 4 million people affected in the United States and about 350,000 new cases per year, AD is the most frequent cause of dementia in U.S. and in Western countries. Among the main cause leading to AD, the deficit of cholinergic system plays a major role. Accordingly, one of the most common therapy for the symptomatic treatment of AD is the block of acetylcholinesterase (AChE) by means of acetylcholinesterase inhibitors. The inhibition of AChE increases the concentration of acetylcholine (ACh) in the synaptic cleft, thus restoring the physiological effects of ACh within the central nervous system. Recent studies reported a significant benefits of acetylcholinesterase inhibitors vs placebo on cognitive function, activities of daily living, and behavior. These improvements, however, are not always detectable in clinical practice. Thus, it has been recently suggested that the detection of improvements on cognitive function, activities of daily living, and behavior may be obtained by the concomitant use of drugs acting on other pathogenetic AD mechanism. In particular the European Medical Agency (EMEA) recommend the use of the AChE inhibitors donepezil and rivastigmine in mild-to-moderate AD, and the addition of memantine in moderate-to-severe AD. Memantine block the glutamate receptor, limiting the uncontrolled entrance of Ca2+ ion in the postsynaptic neuron, thus delaying the Ca2+-excess neurodegeneration. Most studies reported that interindividual differences in response to these drugs may be due to variability in drug metabolism related to behavioral, clinical, and genetic factors, mainly hereditary polymorphisms of drug-metabolizing and drug-transporting enzymes. Final objective of this proposal is to identify the genetic component underlying the response/non response to the most common drugs currently used in the treatment of AD, i.e. donepezil, rivastigmine and memantine, throughout the following specific objectives: 1) identification of significant difference in the distribution of the genotypes of the CYP2D6 gene polymorphisms among patients responders/non-responders to donepezil treatment; 2) identification of the specific DNA alteration producing modifications in the CYP2D6 enzyme activity; 3) confirmation of the modification of the enzyme activity by means of donepezil dosage in vivo; 4) identification of significant difference in the distribution of the genotypes of the acetylcholinesterase gene polymorphisms among patients responders/non-responders to rivastigmine treatment; 5) identification of the specific DNA alteration producing modifications in the acetylcholinesterase-mediated hydrolysis of rivastigmine; 6) confirmation of the modification of the enzyme activity by means of rivastigmine dosage in vivo; 7) identification of significant difference in the distribution of the genotypes of the OCT2 gene polymorphisms among patients responders/non-responders to memantine treatment; 8) identification of the specific DNA alteration producing modifications in the OCT2 transporter of memantine; 9) confirmation of the modification of the enzyme activity by means of memantine dosage in vivo. We expected that the identification of functional polymorphisms in the CYP2D6, AChE and OCT2 genes may influence the clinical efficacy of donepezil, rivastigmine and memantine in AD patients, and may be useful in identifying subgroups of AD patients with different clinical response to treatment. Criteria and indicators to verify results are: 1) the number of patients enrolled in the study in the first 6 months; 2) the number of genotypes investigated at the end of the 1th year of the project; 3) the number of specific gene alteration influencing the enzyme activity at the 2nd year of the project; 4) the quantitative data obtained from plasma drug dosage at the end of the 2nd year of the project. This project is coherent with the ministerial guidelines for the I.R.C.C.S. and well integrate the clinical practice in neurology. Area di interesse identificata Pharmacogenetics of drugs for Alzheimer’s Disease Finanziamenti ricevuti Titolo progetto Ente finanziatore Effect of weight loss on metabolic, functional, cognitive status and biological markers of longevity in obese frail elderly Ministero della Salute Durata progetto Two years Abstract del progetto Obesity, and in particular abdominal obesity causes serious metabolic and medical complications and impairs quality of life. Moreover, in elderly persons, obesity can lead to frailty by promoting chronic inflammation and by exacerbating the decline in strength, endurance, balance and mobility associated with aging and physical inactivity. Weight loss, induced by calorie restriction and/or physical exercise, simultaneously improves multiple metabolic risk factors for cardiovascular disease and other medical abnormalities associated with obesity, and reduce morbidity and mortality. However, the appropriate treatment for obesity in elderly persons is controversial because of the potential harmful effects of weight loss on bone and muscle mass. It has been reported that weight loss can also modify biological pathways at the cellular level. Insulin and of insulin-like growth factor-1 (IGF-1) have been involved in cellular caloric equilibrium and other mechanisms, i.e. oxidative stress and inflammation. Thus genetic polymorphisms in genes involved in these metabolic pathways such as apolipoprotein E (APOE), sterol regulatory element-binding protein cleavage-activating protein (SCAP), peroxisome proliferators-activated receptor gamma-2 (PPR), -2B-adrenergic receptor (ADRA2B), acyl-CoA synthetase 5 (ACSL5) and interleukin-6 (IL-6) may influence the effect of weight loss on biological mechanisms and possibly on functional and cognitive status in the frail elderly. It is possible that weight loss in obese elderly persons can be beneficial by improving metabolic, functional and cognitive status, or harmful by causing a decrease in muscle and bone mass. However, the clinical and physiological effects of weight loss in obese elderly subjects have never been carefully evaluated. Thus, appropriate treatment for this rapidly increasing segment of the elderly population remains controversial. The purpose of this project is to advance our scientific knowledge by determining the effects of weight loss on metabolic, functional and cognitive status in obese elderly subjects. Moreover, we will evaluate the role of weight loss on biological markers of metabolic health and longevity, i.e. serum level of Insulin, insulin-like growth factor-1 (IGF-1), transforming growth factor- (TGF-), tumor-necrosis factor- (TNF-), interleukin-1 (IL-1) and C-Reactive Protein (CRP), and their relationships with genetic polymorphisms in APOE, SCAP, PPR-, ADRA2B, ACSL5 and Titolo progetto IL-6 genes. As biomarker of longevity we will also evaluate the accumulation of mitochondrial (mt) DNA mutation through the analysis of the D310 mononucleotide repeat of mtDNA. Smart Home for Elderly People (HOPE) Ente finanziatore Ministero della salute/Ministero dell’Università e della Ricerca Scientifica Durata progetto Two years Abstract del progetto Since Europe’s and worldwide population grows older, the need for care is growing as well as people spend more money to get it. The solution Smart Home for Elderly People (HOPE) promises to reduce the need for carers, improve the life of older people and cut the cost of assistance. The main objective of the “Hope” project is to produce an Integrated Computer Technology (ICT) solution that will help the elderly people, specifically those that suffer with the Alzheimer s disease (AD), achieve a richer and more independent lifestyle. Dementia causes long and oppressive suffering to patients and their relatives and imposes enormous costs on society. About 25 million people suffered from dementia in recent years. As a 4-fold increase of this number is expected by 2050, dementia is one main health issue of the next decades. AD covers 50-70% of all dementia cases, no cure exists, and effective and reliable early diagnostic techniques are lacking. Early diagnosis and progress monitoring of AD is a central part of treatment until future drugs and prevention strategies become available. Elderly people with AD spend most of their time at home. The “HOPE” solution consists of an integrated, smart platform that will enable the elderly people with AD to use innovative technology for a more independent life, easy access to information, monitor their health, and serve as a source of inspiration for users as well as for people working with assistive devices. Moreover, it will enable them to perform by themselves activities they were not able to do before and which are important for their daily personal life. The main advantage of the proposed system is that it provides a basis for integrating services for the elderly population while they are at home. HOPE is a budgeted solution that will be installed at the elderly people’s homes, and will provide services for (a) life-long, self organized, appropriate educational environment and access to information, (b) care management and health support, (c) self monitoring and decision making. Optionally, the user will be able to activate software which automatically establishes the necessary interactive, triple-play connection for receiving tele-help and tele-assistance from specialized service providers, doctors or other medical personnel. The proposed application will be intelligent enough to offer safety in terms of controlling efficiently the home environment, economy in terms of controlling and decreasing the need for external help, and convenience in terms of adjusting the operation of connected sub-systems to achieve the best possible user experience. Nome Vincenzo Mario Bruno GIORGINO Contatti TEL. (WK) (0039) (0)11 6706093 e-mail [email protected] Cell. 3280380926 Istituto/Dipartimento Department of Social Sciences - Faculty of Economics - University of Torino Proposta di ricerca A transdisciplinary oriented proposal from a social sciences perspective From the bedside to the bench and return The major challenge in the field of senile dementia is represented by the existent gap between different disciplinary approaches, namely between bio-medical and social/human sciences. The project aims to answer to this gap within a transdisciplinary perspective, mainly based on the effort to integrate experiential data coming from social sciences methods with biomedical ones. The former are considered a possible source of knowledge that, under certain conditions, can be meshed with the latter. The research strategy is based on the recruitment of around 50 patients with mild cognitive impairment or symptoms of early Alzheimer’s disease and their caregivers, both formal and informal. Selection criteria of participants within a grounded theory methodology will be based on the greater similarity, i.e. the most similar personal/social characteristics as well as of the context. The research design is a mixed method approach oriented to collect data from the experiential level principally through methods such as journals, systematic observation, in-depth interviews and focus groups with the caregivers, both formal and informal. The same could be said for Alzheimer’s patients as far as possible and, in this regard, first-person observation is of the utmost importance as it allows a knowledge that is unavailable with usual third person methods; the former can be assessed with adequate criteria as recent literature is showing. One major assumption is that the loss of cognitive functions could not mean the loss of the awareness of this painful condition; moreover, if the patient is usually unable to express his/her feelings, it does not mean that he/she does not have feelings at all. So far, we can develop adequate tools of systematic observation to explore these more subtle processes. The project may include a two month period of an experimental training for caregivers in which they could learn to practice some contemplative methods in their everyday life. This is a goal of translational relevance that can include the patients, depending by their specific condition, and calling for a specific kind of exercises. Following this perspective, we can set up two groups of around 25 caregivers each. To the first group is offered a two month contemplative practice program, while the second will follow the current standard service provision. If in the literature social ties are recognized of major importance both at health promotion and prevention level than during the support for the sick, they do not exist in a vacuum: they call for personal commitment, i.e. emotional, cognitive and economic resources. Social support and self healing are here intended as nurtured by a disciplined learned skills of contemplative techniques: see for example the literature about TM and mindfulness approaches which provide a cultural maintenance to our biological self. As a further development, a contribution by an economist could develop a specific research strategy for a study about the costs of this kind of intervention compared with standard ones. A valuable consequence of this approach could also be appreciated at cognitive level: a fresh understanding of difficult situations could emerge in everyday life by the caregivers or in the professional realm, suggesting new opportunities of managing the interactional lived situation and opening the way to new possible treatments. The hypothesis is that contemplative methods could enhance not only the coping function but also the general interactional process related to the management of this illness, including a better interaction and understanding with/of the patient’s experience, currently shadowed in clinical research. One can envisages also the consequences that this kind of approach could have at the governance level, in which caregivers and, when possible, patients can be co-producers of the healing process, as recent NHS policy programs show in the UK. A broader encompassing framework based on an experiential-empirical perspective could enhance the scientific development and open to more effective intervention. Vincenzo Mario Bruno Giorgino Area di interesse identificata Experimental methods oriented to personal experience Transdisciplinary research Sociology of health, of health profession and informal care