101 results on '"Bernard L. Schneider"'
Search Results
2. Central anorexigenic actions of bile acids are mediated by TGR5
- Author
-
Sabrina Diano, Sungho Jin, Bernard L. Schneider, Antimo Gioiello, Nadia Bresciani, Laura A. Velázquez-Villegas, Yu Sun, Qingyao Huang, Philippe Zizzari, Aiste Baleisyte, Valérie S. Fénelon, Julijana Ivanisevic, Giuseppe Bruschetta, Kristina Schoonjans, Daniela Cota, Roberto Pellicciari, Ashley Castellanos-Jankiewicz, and Alessia Perino
- Subjects
Male ,receptor ,Endocrinology, Diabetes and Metabolism ,Receptors, G-Protein-Coupled ,Bile Acids ,Eating ,Mice ,0302 clinical medicine ,Receptors ,rat ,Receptor ,gaba ,Mice, Knockout ,Neurons ,2. Zero hunger ,0303 health sciences ,Drug discovery ,Chemistry ,Neuropeptide Y receptor ,G protein-coupled bile acid receptor ,Anorexia ,cortex ,medicine.anatomical_structure ,medicine.drug ,medicine.medical_specialty ,Central nervous system ,Hypothalamus ,Neuropeptide ,Mice, Transgenic ,leptin ,Cell Line ,Neuropeptides ,Bile Acids and Salts ,G-Protein-Coupled ,03 medical and health sciences ,Physiology (medical) ,Orexigenic ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Secretion ,030304 developmental biology ,G protein-coupled receptor ,Cell Biology ,agrp neurons ,Endocrinology ,glucagon-like peptide-1 ,Gene Expression Regulation ,area postrema ,activation ,030217 neurology & neurosurgery - Abstract
Bile acids (BAs) are signalling molecules that mediate various cellular responses in both physiological and pathological processes. Several studies report that BAs can be detected in the brain1, yet their physiological role in the central nervous system is still largely unknown. Here we show that postprandial BAs can reach the brain and activate a negative-feedback loop controlling satiety in response to physiological feeding via TGR5, a G-protein-coupled receptor activated by multiple conjugated and unconjugated BAs2 and an established regulator of peripheral metabolism3–8. Notably, peripheral or central administration of a BA mix or a TGR5-specific BA mimetic (INT-777) exerted an anorexigenic effect in wild-type mice, while whole-body, neuron-specific or agouti-related peptide neuronal TGR5 deletion caused a significant increase in food intake. Accordingly, orexigenic peptide expression and secretion were reduced after short-term TGR5 activation. In vitro studies demonstrated that activation of the Rho–ROCK–actin-remodelling pathway decreases orexigenic agouti-related peptide/neuropeptide Y (AgRP/NPY) release in a TGR5-dependent manner. Taken together, these data identify a signalling cascade by which BAs exert acute effects at the transition between fasting and feeding and prime the switch towards satiety, unveiling a previously unrecognized role of physiological feedback mediated by BAs in the central nervous system. Bile acids are shown to enter the brain and regulate short-term reductions in food intake after a meal by inhibiting neuropeptide release from agouti-related peptide/neuropeptide Y neurons.
- Published
- 2021
- Full Text
- View/download PDF
3. CSPα reduces aggregates and rescues striatal dopamine release in α-synuclein transgenic mice
- Author
-
Jeffrey W. Dalley, Oleg Anichtchik, David Klenerman, Bernard L. Schneider, Martyna Podgajna, Laura Calo, Eric Hidari, Emma Carlson, Maria Grazia Spillantini, and Michal Wegrzynowicz
- Subjects
0301 basic medicine ,Genetically modified mouse ,Parkinson's disease ,Dopamine ,animal diseases ,Mice, Transgenic ,Protein Aggregation, Pathological ,Synapse ,Neurotransmitter secretion ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,α-synuclein ,0302 clinical medicine ,synapse ,Report ,mental disorders ,medicine ,Animals ,Neurotransmitter ,AcademicSubjects/SCI01870 ,Chemistry ,Dopaminergic ,Neurodegeneration ,Membrane Proteins ,Parkinson Disease ,DNAJ chaperone family ,HSP40 Heat-Shock Proteins ,CSPα ,medicine.disease ,Corpus Striatum ,nervous system diseases ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,Synapses ,alpha-Synuclein ,Parkinson’s disease ,AcademicSubjects/MED00310 ,Neurology (clinical) ,030217 neurology & neurosurgery ,medicine.drug - Abstract
α-Synuclein aggregation at the synapse is an early event in Parkinson’s disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and α-synuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like α-synuclein, chaperones the SNARE complex assembly and controls neurotransmitter release. α-Synuclein can rescue neurodegeneration in CSPαKO mice. However, whether α-synuclein aggregation alters CSPα expression and function is unknown. Here we show that α-synuclein aggregation at the synapse is associated with a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with α-synuclein aggregates and in vivo reduces synaptic α-synuclein aggregates increasing monomeric α-synuclein and restoring normal dopamine release in 1-120hαSyn mice. These novel findings reveal a mechanism by which α-synuclein aggregation alters CSPα at the synapse, and show that CSPα rescues α-synuclein aggregation-related phenotype in 1-120hαSyn mice similar to the effect of α-synuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage Parkinson’s disease., CSPα and αsynuclein (αsyn) are involved in synaptic function and neurotransmitter release via parallel pathways. Caló et al. show that αsyn aggregation at the synapse is associated with a reduction in CSPα. An increase in CSPα reduces synaptic αsyn aggregates, rescuing impaired striatal dopamine release in αsyn transgenic mice.
- Published
- 2021
- Full Text
- View/download PDF
4. Single and Dual Vector Gene Therapy with AAV9-PHP.B Rescues Hearing in Tmc1 Mutant Mice
- Author
-
Olga Shubina-Oleinik, Bernard L. Schneider, Carl Nist-Lund, Jeffrey R. Holt, Jason Wu, Sofia Spataro, Irina Marcovich, Paola Solanes, and Hannah Goldberg
- Subjects
Male ,Hearing loss ,Genetic enhancement ,Genetic Vectors ,Mutant ,Biology ,Mice ,Drug Discovery ,otorhinolaryngologic diseases ,Genetics ,medicine ,Animals ,Inner ear ,Mechanotransduction ,Hearing Loss ,Molecular Biology ,Gene ,Mice, Knockout ,Pharmacology ,Membrane Proteins ,Genetic Therapy ,Dependovirus ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Knockout mouse ,Molecular Medicine ,Female ,Hair cell ,medicine.symptom ,RNA, Guide, Kinetoplastida - Abstract
AAV-mediated gene therapy is a promising approach for treating genetic hearing loss. Replacement or editing of the Tmc1 gene, encoding hair cell mechanosensory ion channels, is effective for hearing restoration in mice with some limitations. Efficient rescue of outer hair cell function and lack of hearing recovery with later-stage treatment remain issues to be solved. Exogenous genes delivered with the adeno-associated virus (AAV)9-PHP.B capsid via the utricle transduce both inner and outer hair cells of the mouse cochlea with high efficacy. Here, we demonstrate that AAV9-PHP.B gene therapy can promote hair cell survival and successfully rescues hearing in three distinct mouse models of hearing loss. Tmc1 replacement with AAV9-PHP.B in a Tmc1 knockout mouse rescues hearing and promotes hair cell survival with equal efficacy in inner and outer hair cells. The same treatment in a recessive Tmc1 hearing-loss model, Baringo, partially recovers hearing even with later-stage treatment. Finally, dual delivery of Streptococcus pyogenes Cas9 (SpCas9) and guide RNA (gRNA) in separate AAV9-PHP.B vectors selectively disrupts a dominant Tmc1 allele and preserves hearing in Beethoven mice, a model of dominant, progressive hearing loss. Tmc1-targeted gene therapies using single or dual AAV9-PHP.B vectors offer potent and versatile approaches for treating dominant and recessive deafness.
- Published
- 2021
- Full Text
- View/download PDF
5. SMN Depleted Mice Offer a Robust and Rapid Onset Model of Nonalcoholic Fatty Liver DiseaseSummary
- Author
-
Bernard L. Schneider, Rashmi Kothary, Maica Llavero-Hurtado, Thomas H. Gillingwater, Abdelmadjid Atrih, Alexandra Tierney, Jean Michaud, Ariane Beauvais, Thomas M. Wishart, Lucia Chehade, Marc-Olivier Deguise, Douglas J. Lamont, Simon H. Parson, Yves De Repentigny, Mary-Ellen Harper, and Chantal A. Pileggi
- Subjects
0301 basic medicine ,LC-MS, liquid chromatography-mass spectrometry ,DAVID, Database for Annotation, Visualization and Integrated Discovery ,IGF1, insulin-like growth factor 1 ,AST, aspartate aminotransferase ,RC799-869 ,muscular-atrophy phenotype ,CS, citric synthase ,animal-models ,Mice ,0302 clinical medicine ,iron ,Fibrosis ,Non-alcoholic Fatty Liver Disease ,LDL, low-density lipoprotein ,Nonalcoholic fatty liver disease ,MCD, methionine and choline deficient diet ,steatosis ,Medicine ,TNFR1, tumor necrosis factor receptor superfamily member 1A ,defects ,ANOVA, analysis of variance ,Original Research ,Mice, Knockout ,P, postnatal day ,FasR, Fas receptor ,NEFA, non-esterified fatty acid ,p53, tumor protein p53 ,TEAB, triethyl ammonium bicarbonate ,Fatty liver ,Gastroenterology ,NASH ,CPT1, carnitine palmitoyl transferase 1 ,p21, cyclin dependent kinase inhibitor 1A ,Diseases of the digestive system. Gastroenterology ,3. Good health ,ECM, extracellular matrix ,SMN ,TMT, tandem mass tagging ,mouse-model ,qPCR, quantitative polymerase chain reaction ,030211 gastroenterology & hepatology ,NASH, nonalcoholic steatohepatitis ,medicine.medical_specialty ,insulin ,HDL, high-density lipoprotein ,PBS, phosphate-buffered saline ,steatohepatitis ,Hypoglycemia ,SMN1, survival motor neuron 1 ,03 medical and health sciences ,Insulin resistance ,ROS, reactive oxygen species ,VLDL, very low density lipoprotein ,Internal medicine ,ALT, alanine aminotransferase ,NAFLD ,mitochondrial dysfunction ,Animals ,MCL, Markov Clustering Algorithm ,Hepatology ,ALP, alkaline phosphatase ,business.industry ,T2DM, type 2 diabetes mellitus ,IPA, ingenuity pathway analysis ,medicine.disease ,WT, wild-type ,Survival of Motor Neuron 1 Protein ,igfals, insulin-like growth factor binding protein acid labile subunit ,digestive system diseases ,Fatty Liver ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Metabolism ,AAV9, adeno-associated virus 9 ,SMA, spinal muscular atrophy ,network ,Akt, protein kinase B ,Hepatic stellate cell ,NAFLD, nonalcoholic fatty liver disease ,Steatosis ,Steatohepatitis ,business ,Bax, BCL2 associated X protein ,HFN4a, hepatic nuclear factor 4 alpha - Abstract
Background & Aims Nonalcoholic fatty liver disease (NAFLD) is considered a health epidemic with potential devastating effects on the patients and the healthcare systems. Current preclinical models of NAFLD are invariably imperfect and generally take a long time to develop. A mouse model of survival motor neuron (SMN) depletion (Smn2B/- mice) was recently shown to develop significant hepatic steatosis in less than 2 weeks from birth. The rapid onset of fatty liver in Smn2B/- mice provides an opportunity to identify molecular markers of NAFLD. Here, we investigated whether Smn2B/- mice display typical features of NAFLD/nonalcoholic steatohepatitis (NASH). Methods Biochemical, histologic, electron microscopy, proteomic, and high-resolution respirometry were used. Results The Smn2B/- mice develop microvesicular steatohepatitis within 2 weeks, a feature prevented by AAV9-SMN gene therapy. Although fibrosis is not overtly apparent in histologic sections of the liver, there is molecular evidence of fibrogenesis and presence of stellate cell activation. The consequent liver damage arises from mitochondrial reactive oxygen species production and results in hepatic dysfunction in protein output, complement, coagulation, iron homeostasis, and insulin-like growth factor-1 metabolism. The NAFLD phenotype is likely due to non-esterified fatty acid overload from peripheral lipolysis subsequent to hyperglucagonemia compounded by reduced muscle use and insulin resistance. Despite the low hepatic mitochondrial content, isolated mitochondria show enhanced β-oxidation, likely as a compensatory response, resulting in the production of reactive oxygen species. In contrast to typical NAFLD/NASH, the Smn2B/- mice lose weight because of their associated neurological condition (spinal muscular atrophy) and develop hypoglycemia. Conclusions The Smn2B/- mice represent a good model of microvesicular steatohepatitis. Like other models, it is not representative of the complete NAFLD/NASH spectrum. Nevertheless, it offers a reliable, low-cost, early-onset model that is not dependent on diet to identify molecular players in NAFLD pathogenesis and can serve as one of the very few models of microvesicular steatohepatitis for both adult and pediatric populations., Graphical abstract
- Published
- 2021
- Full Text
- View/download PDF
6. Central and peripheral delivery of AAV9-SMN target different pathomechanisms in a mouse model of spinal muscular atrophy
- Author
-
Rebecca A. Yaworski, Aoife Reilly, Vincent Tabard-Cossa, Ariane Beauvais, Bernard L. Schneider, Simon Thebault, Niko Hensel, Daniel R. Tessier, Rashmi Kothary, and Marc-Olivier Deguise
- Subjects
Pathology ,medicine.medical_specialty ,Neuromuscular disease ,business.industry ,Genetic enhancement ,Spinal muscular atrophy ,SMN1 ,Motor neuron ,medicine.disease ,SMA ,Peripheral ,medicine.anatomical_structure ,Toxicity ,medicine ,business - Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease caused by loss of the SMN1 gene. Although lower motor neurons are a primary target, there is evidence that peripheral organ defects contribute to SMA. Current SMA gene therapy uses a single, high titre intravenous bolus of AAV9-SMN resulting in impressive, yet limited amelioration of the clinical phenotype. However, risks of this treatment include liver toxicity. Intrathecal administration is under clinical trial but was interrupted due to safety concerns in a concomitant animal study. As there is no direct comparison between the different delivery strategies while avoiding high dose toxicity, we injected SMA mice with low dose scAAV9-cba-SMN either intravenously (IV) for peripheral SMN restoration or intracerebroventricularly (ICV) for CNS-focused SMN restoration. Here, IV injections restored SMN in peripheral tissues but not CNS, while ICV injections mildly increased SMN in the periphery and the CNS. Consequently, only ICV treatment rescued motor neuron degeneration. Surprisingly, both treatments resulted in an impressive rescue of survival, weight, motor function, and peripheral phenotypes including liver and pancreas pathology. Our work highlights independent contributions of peripheral organs to SMA pathology and suggests that treatments should not be restricted to the motor neuron.Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
7. The Links between ALS and NF-κB
- Author
-
Brian D. McCabe, Emma C. Källstig, and Bernard L. Schneider
- Subjects
Review ,Disease ,NF-κB ,neuroinflammation ,lcsh:Chemistry ,Pathogenesis ,chemistry.chemical_compound ,0302 clinical medicine ,NF-kB ,Amyotrophic lateral sclerosis ,lcsh:QH301-705.5 ,Spectroscopy ,Neurons ,0303 health sciences ,NF-kappa B ,General Medicine ,3. Good health ,Computer Science Applications ,Oligodendroglia ,Disease Susceptibility ,Microglia ,medicine.symptom ,Cell type ,Inflammation ,Environment ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Physical and Theoretical Chemistry ,Molecular Biology ,Neuroinflammation ,Alleles ,030304 developmental biology ,business.industry ,Organic Chemistry ,Amyotrophic Lateral Sclerosis ,Genetic Variation ,medicine.disease ,Enzyme Activation ,lcsh:Biology (General) ,lcsh:QD1-999 ,chemistry ,ALS ,business ,Neuroscience ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease wherein motor neuron degeneration leads to muscle weakness, progressive paralysis, and death within 3–5 years of diagnosis. Currently, the cause of ALS is unknown but, as with several neurodegenerative diseases, the potential role of neuroinflammation has become an increasingly popular hypothesis in ALS research. Indeed, upregulation of neuroinflammatory factors have been observed in both ALS patients and animal models. One such factor is the inflammatory inducer NF-κB. Besides its connection to inflammation, NF-κB activity can be linked to several genes associated to familial forms of ALS, and many of the environmental risk factors of the disease stimulate NF-κB activation. Collectively, this has led many to hypothesize that NF-κB proteins may play a role in ALS pathogenesis. In this review, we discuss the genetic and environmental connections between NF-κB and ALS, as well as how this pathway may affect different CNS cell types, and finally how this may lead to motor neuron degeneration.
- Published
- 2021
8. Expression of a miRNA targeting mutated SOD1 in astrocytes induces motoneuron plasticity and improves neuromuscular function in ALS mice
- Author
-
Bernard L. Schneider, Sylvain Pradervand, Cylia Rochat, Patrick Aebischer, Florence E. Perrin, Cédric Raoul, and Nathalie Bernard-Marissal
- Subjects
Denervation ,Gastrocnemius muscle ,medicine.anatomical_structure ,RNA interference ,SOD1 ,medicine ,Biology ,Gene mutation ,Amyotrophic lateral sclerosis ,medicine.disease ,Neuromuscular junction ,Cell biology ,Compound muscle action potential - Abstract
In amyotrophic lateral sclerosis (ALS) caused by SOD1 gene mutations, both cell-autonomous and non-cell-autonomous mechanisms lead to the selective degeneration of motoneurons. Here, we evaluate the therapeutic potential of gene therapy targeting mutated SOD1 in mature astrocytes using mice expressing the mutated SOD1G93A protein. An AAV-gfaABC1D vector encoding an artificial microRNA is used to deliver RNA interference against mutated SOD1 selectively in astrocytes. The treatment leads to the progressive rescue of neuromuscular junction occupancy, to the recovery of the compound muscle action potential in the gastrocnemius muscle, and significantly improves neuromuscular function. In the spinal cord, gene therapy targeting astrocytes protects a small pool of fast-fatigable motoneurons until disease end stage. In the gastrocnemius muscle of the treated SOD1G93A mice, the fast-twitch type IIb muscle fibers are preserved from atrophy. Axon collateral sprouting is observed together with muscle fiber type grouping indicative of denervation/re-innervation events. The transcriptome profiling of spinal cord motoneurons shows changes in the expression levels of factors regulating the dynamics of microtubules. Gene therapy delivering RNA interference against mutated SOD1 in astrocytes provides therapeutic effects enhancing motoneuron plasticity and improving neuromuscular function in ALS mice.
- Published
- 2021
- Full Text
- View/download PDF
9. PARKIN REGULATES DRUG TAKING-BEHAVIOR IN RAT MODEL OF METHAMPHETAMINE USE DISORDER
- Author
-
Akhil Sharma, Anna Moszczynska, Bernard L. Schneider, and Arman Harutyunyan
- Subjects
0301 basic medicine ,Male ,Knockout rat ,Drug taking ,Self Administration ,place preference ,Pharmacology ,Parkin ,Methamphetamine ,chemistry.chemical_compound ,0302 clinical medicine ,High doses ,Medicine ,dorsomedial striatum ,media_common ,Glutamate receptor ,Genomics ,Psychiatry and Mental health ,glutamate receptors ,basal ganglia ,RC321-571 ,medicine.drug ,media_common.quotation_subject ,Ubiquitin-Protein Ligases ,Rat model ,Addiction ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Nucleus accumbens ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,nucleus-accumbens ,expression ,Animals ,Rats, Long-Evans ,Biological Psychiatry ,business.industry ,Meth ,Rats ,nervous system diseases ,030104 developmental biology ,chemistry ,inflammation ,Methamphetamine use ,dopamine release ,activation ,Central Nervous System Stimulants ,business ,030217 neurology & neurosurgery - Abstract
There is no FDA-approved medication for methamphetamine (METH) use disorder. New therapeutic approaches are needed, especially for people who use METH heavily and are at high risk for overdose. This study used genetically engineered rats to evaluate PARKIN as a potential target for METH use disorder. PARKIN knockout, PARKIN-overexpressing, and wild-type young adult male Long Evans rats were trained to self-administer high doses of METH using an extended-access METH self-administration paradigm. Reinforcing/rewarding properties of METH were assessed by quantifying drug-taking behavior and time spent in a METH-paired environment. PARKIN knockout rats self-administered more METH and spent more time in the METH-paired environment than wild-type rats. Wild-type rats overexpressing PARKIN self-administered less METH and spent less time in the METH-paired environment. PARKIN knockout rats overexpressing PARKIN self-administered less METH during the first half of drug self-administration days than PARKIN-deficient rats. The results indicate that rats with PARKIN excess or PARKIN deficit are useful models for studying neural substrates underlying “resilience” or vulnerability to METH use disorder and identify PARKIN as a novel potential drug target to treat heavy use of METH.
- Published
- 2020
- Full Text
- View/download PDF
10. CSPα reduces aggregates and rescues striatal dopamine release in αsynuclein transgenic mice
- Author
-
Michal Wegrzynowicz, E Carlson, Bernard L. Schneider, David Klenerman, Laura Calo, Oleg Anichtchik, Jeffrey W. Dalley, Maria Grazia Spillantini, and Eric Hidari
- Subjects
Striatal dopamine ,Genetically modified mouse ,Chemistry ,Neurodegeneration ,Dopaminergic ,medicine.disease ,Phenotype ,Cell biology ,Synapse ,chemistry.chemical_compound ,Dopamine ,medicine ,Neurotransmitter ,medicine.drug - Abstract
αSynuclein aggregation at the synapse is an early event in Parkinson’s disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and αsynuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like αsynuclein, chaperones the SNARE complex assembly and neurotransmitter release. αSynuclein can rescue neurodegeneration in CSPαKO mice. However, whether αsynuclein aggregation alters CSPα expression and function is unknown. Here we show that αsynuclein aggregation at the synapse induces a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with αsynuclein aggregates and in vivo reduces synaptic αsynuclein aggregates restoring normal dopamine release in 1-120hαsyn mice. These novel findings reveal a mechanism by which αsynuclein aggregation alters CSPα at the synapse, and show that CSPα rescues αsynuclein aggregation-related phenotype in 1-120hαsyn mice similar to the effect of αsynuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage PD.
- Published
- 2020
- Full Text
- View/download PDF
11. Glutaredoxin 1 downregulation in the substantia nigra leads to dopaminergic degeneration in mice
- Author
-
Bernard L. Schneider, Etienne C. Hirsch, Reddy Peera Kommaddi, Ajit Ray, Aditi Verma, Deepti Bapat, Vijayalakshmi Ravindranath, Latha Diwakar, Indian Institute of Science [Bangalore] (IISc Bangalore), Ecole Polytechnique Fédérale de Lausanne (EPFL), Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Hirsch, Etienne, Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Parkinson's disease ,Tyrosine 3-Monooxygenase ,mouse model ,Dopamine ,translocation ,Down-Regulation ,Substantia nigra ,24 tyrosine hydroxylase 25 ,Striatum ,thioltransferase ,03 medical and health sciences ,Mice ,0302 clinical medicine ,glutaredoxin 1 ,tyrosine hydroxylase ,shRNA ,medicine ,oxidative stress ,Animals ,Humans ,glutathione ,Glutaredoxins ,dysfunction ,dopaminergic neurons ,Tyrosine hydroxylase ,Chemistry ,Pars compacta ,[SCCO.NEUR]Cognitive science/Neuroscience ,Dopaminergic ,Neurodegeneration ,[SCCO.NEUR] Cognitive science/Neuroscience ,neurodegeneration ,mitochondrial complex-i ,medicine.disease ,cell-death ,Cell biology ,Ventral tegmental area ,Substantia Nigra ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,nervous system ,parkinsons-disease ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
International audience; Background: Parkinson's disease (PD) is characterized by a severe loss of the dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). Perturbation of protein thiol redox homeostasis has been shown to play a role in the dysregulation of cell death and cell survival signaling pathways in these neurons. Glutaredoxin 1 (Grx1) is a thiol/disulfide oxidoreductase that catalyzes the deglutathionylation of proteins and is important for regulation of cellular protein thiol redox homeostasis.Objectives: We evaluated if the downregulation of Grx1 could lead to dopaminergic degeneration and PD-relevant motor deficits in mice.Methods: Grx1 was downregulated unilaterally through viral vector-mediated transduction of short hairpin RNA against Grx1 into the SNpc. Behavioral assessment was performed through rotarod and elevated body swing test. Stereological analysis of tyrosine hydroxylase-positive and Nissl-positive neurons was carried out to evaluate neurodegeneration.Results: Downregulation of Grx1 resulted in contralateral bias of elevated body swing and reduced latency to fall off, accelerating rotarod. This was accompanied by a loss of tyrosine hydroxylase-positive neurons in the SNpc and their DA projections in the striatum. Furthermore, there was a loss Nissl-positive neurons in the SNpc, indicating cell death. This was selective to the SNpc neurons because DA neurons in the ventral tegmental area were unaffected akin to that seen in human PD. Furthermore, Grx1 mRNA expression was substantially decreased in the SNpc from PD patients.Conclusions: Our study indicates that Grx1 is critical for the survival of SNpc DA neurons and that it is downregulated in human PD. © 2020 International Parkinson and Movement Disorder Society.
- Published
- 2020
- Full Text
- View/download PDF
12. A mouse model for spinal muscular atrophy provides insights into non-alcoholic fatty liver disease pathogenesis
- Author
-
Thomas H. Gillingwater, Simon H. Parson, Abdelmadjid Atrih, Douglas J. Lamont, Lucia Chehade, Jean Michaud, Rashmi Kothary, Alexandra Tierney, Maica Llavero-Hurtado, Yves De Repentigny, Bernard L. Schneider, Thomas M. Wishart, Chantal A. Pileggi, Marc-Olivier Deguise, Mary-Ellen Harper, and Ariane Beauvais
- Subjects
0303 health sciences ,medicine.medical_specialty ,business.industry ,Fatty liver ,nutritional and metabolic diseases ,Spinal muscular atrophy ,Hypoglycemia ,medicine.disease ,SMA ,3. Good health ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Steatohepatitis ,Hepatic fibrosis ,business ,030217 neurology & neurosurgery ,Dyslipidemia ,030304 developmental biology - Abstract
Background & aimsSpinal muscular atrophy (SMA) is an inherited neuromuscular disorder leading to paralysis and death in children. SMA patients are more susceptible to dyslipidemia as well as liver steatosis, features reproduced in SMA mouse models. As current pre-clinical models of NAFLD are invariably imperfect and generally take a long time to develop, the rapid development of liver steatosis in SMA mice provides a means to identify molecular markers of non-alcoholic fatty liver disease (NAFLD). Here, we investigated whetherSmn2B/-mice, a model of severe SMA, display typical features of NAFLD/non-alcoholic steatohepatitis (NASH).MethodsBiochemical, histological, electron microscopy, proteomic, and high-resolution respirometry were used.ResultsTheSmn2B/-mice develop steatohepatitis early in life. The consequent liver damage arises from mitochondrial reactive oxygen species production and results in impaired hepatic function including alterations in protein output, complement, coagulation, iron homeostasis, and IGF-1 metabolism. The steatohepatitis is reversible by AAV9-SMN gene therapy. The NAFLD phenotype is likely due to non-esterified fatty acid (NEFA) overload from peripheral lipolysis, subsequent to hyperglucagonemia compounded by reduced muscle use. Mitochondrial β-oxidation contributed to hepatic damage as we observed enhanced hepatic mitochondrial β-oxidation and reactive oxygen species production. Hepatic mitochondrial content, however, was decreased. In contrast to typical NAFLD/NASH, theSmn2B/-mice lose weight due to their neurological condition, develop hypoglycemia and do not develop hepatic fibrosis.ConclusionTheSmn2B/-mice represent a good model of microvesicular steatohepatitis. Like other models, it is not representative of the complete NAFLD/NASH spectrum. Nevertheless, it offers a reliable, low-cost, early onset model that is not dependent on diet to identify molecular players in NAFLD pathogenesis and can serve as one of the very few models of microvesicular steatohepatitis for both adult and pediatric populations.
- Published
- 2020
- Full Text
- View/download PDF
13. Blood Flow to the Spleen is Altered in a Mouse Model of Spinal Muscular Atrophy
- Author
-
Ariane Beauvais, Marc-Olivier Deguise, Rashmi Kothary, and Bernard L. Schneider
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Necrosis ,Genetic enhancement ,Genetic Vectors ,Spleen ,Mice, Transgenic ,Muscular Atrophy, Spinal ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,business.industry ,Ultrasonography, Doppler ,Spinal muscular atrophy ,Blood flow ,Genetic Therapy ,medicine.disease ,SMA ,Survival of Motor Neuron 2 Protein ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Regional Blood Flow ,Neurology (clinical) ,medicine.symptom ,business ,Perfusion ,030217 neurology & neurosurgery - Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder affecting young children. While pre-clinical models of SMA show small spleens, the same is not true in humans. Here, we show by doppler ultrasonography decreased splenic blood flow in Smn2B/- mice. Further, AAV9-SMN gene therapy does not rescue the distal ear and tail necrosis nor the spleen size in these mice, suggesting that the latter may be linked to a cardiovascular defect. Absence of smaller spleens in human patients is likely due to differences in presentation of defects in SMA between pre-clinical mouse models and human patients, particularly the susceptibility to cardiovascular issues.
- Published
- 2020
14. Scalable Production of AAV Vectors in Orbitally Shaken HEK293 Cells
- Author
-
Bernard L. Schneider, Daniel Blessing, Maria Rey, Catherine Pythoud, Vivianne Padrun, Nicole Déglon, Gabriel Vachey, and Florian M. Wurm
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,viral vectors ,purification ,Genetic enhancement ,viruses ,mammalian-cells ,serotype 8 ,high-titer ,adeno-associated virus ,medicine.disease_cause ,Virus ,Article ,03 medical and health sciences ,Transduction (genetics) ,gene-transfer ,0302 clinical medicine ,expression ,Genetics ,medicine ,orbitally shaken bioreactors ,lcsh:QH573-671 ,Molecular Biology ,Adeno-associated virus ,CNS transduction ,Chemistry ,lcsh:Cytology ,HEK 293 cells ,Transfection ,serum-free production ,suspension HEK293 cells ,Cell biology ,AAV8 ,AAV9 ,immune affinity chromatography ,transient transfection ,lcsh:Genetics ,030104 developmental biology ,recombinant-adenoassociated-virus ,Cell culture ,030220 oncology & carcinogenesis ,Molecular Medicine ,Nuclear localization sequence - Abstract
Adeno-associated virus (AAV) vectors are currently among the most commonly applied for in vivo gene therapy approaches. The evaluation of vectors during clinical development requires the production of considerable amounts of highly pure and potent vectors. Here, we set up a scalable process for AAV production, using orbitally shaken bioreactors and a fully characterized suspension-adapted cell line, HEKExpress. We conducted a proof-of-concept production of AAV2/8 and AAV2/9 vectors using HEKExpress cells. Furthermore, we compared the production of AAV2/9 vectors using this suspension cell line to classical protocols based on adherent HEK293 cells to demonstrate bioequivalence in vitro and in vivo. Following upstream processing, we purified vectors via gradient centrifugation and immunoaffinity chromatography. The in vitro characterization revealed differences due to the purification method, as well as the transfection protocol and the corresponding HEK293 cell line. The purification method and cell line used also affected in vivo transduction efficiency after bilateral injection of AAV2/9 vectors expressing a GFP reporter fused with a nuclear localization signal (AAV2/9-CBA-nlsGFP) into the striatum of adult mice. These results show that AAV vectors deriving from suspension HEKExpress cells are bioequivalent and may exhibit higher potency than vectors produced with adherent HEK293 cells. Keywords: adeno-associated virus, AAV8, AAV9, suspension HEK293 cells, transient transfection, orbitally shaken bioreactors, immune affinity chromatography, CNS transduction
- Published
- 2019
15. The RNA-Binding Protein PUM2 Impairs Mitochondrial Dynamics and Mitophagy During Aging
- Author
-
Davide D'Amico, Graham Knott, Mario Romani, Hao Li, Johan Auwerx, Kristina Schoonjans, Bernard L. Schneider, Vincenzo Sorrentino, Nicola Zamboni, Vera Lemos, Adrienne Mottis, and Francesca Potenza
- Subjects
Male ,Mitochondrial fission factor ,Aging ,Mitochondrion ,Mitochondrial Dynamics ,stress ,0302 clinical medicine ,Mitophagy ,Tissue homeostasis ,0303 health sciences ,Neurodegeneration ,Age Factors ,RNA-Binding Proteins ,Cell biology ,Mitochondria ,Up-Regulation ,c-elegans ,Mitochondrial fission ,Female ,Signal Transduction ,Repressor ,prion-like domains ,Biology ,Mitochondrial Proteins ,03 medical and health sciences ,medicine ,fission ,Animals ,Humans ,skeletal-muscle ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Muscle, Skeletal ,Molecular Biology ,030304 developmental biology ,life-span ,Membrane Proteins ,Cell Biology ,medicine.disease ,genomic stability ,gene-expression ,Mitochondria, Muscle ,Mice, Inbred C57BL ,Proteostasis ,HEK293 Cells ,age ,identification ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Summary Little information is available about how post-transcriptional mechanisms regulate the aging process. Here, we show that the RNA-binding protein Pumilio2 (PUM2), which is a translation repressor, is induced upon aging and acts as a negative regulator of lifespan and mitochondrial homeostasis. Multi-omics and cross-species analyses of PUM2 function show that it inhibits the translation of the mRNA encoding for the mitochondrial fission factor (Mff), thereby impairing mitochondrial fission and mitophagy. This mechanism is conserved in C. elegans by the PUM2 ortholog PUF-8. puf-8 knock-down in old nematodes and Pum2 CRISPR/Cas9-mediated knockout in the muscles of elderly mice enhances mitochondrial fission and mitophagy in both models, hence improving mitochondrial quality control and tissue homeostasis. Our data reveal how a PUM2-mediated layer of post-transcriptional regulation links altered Mff translation to mitochondrial dynamics and mitophagy, thereby mediating age-related mitochondrial dysfunctions.
- Published
- 2019
- Full Text
- View/download PDF
16. Amyloid-β plaque deposition measured using propagation-based X-ray phase contrast CT imaging
- Author
-
Alberto Astolfo, Aurélien Lathuilière, Bernard L. Schneider, Marco Stampanoni, and Vanessa Laversenne
- Subjects
Nuclear and High Energy Physics ,Materials science ,Amyloid beta ,amyloid plaques ,Phase contrast microscopy ,Nanotechnology ,Amyloid plaques ,Brain imaging ,brain imaging ,phase contrast ,030218 nuclear medicine & medical imaging ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Nuclear magnetic resonance ,Neuroimaging ,law ,Phase contrast ,Alzheimer Disease ,medicine ,Animals ,Instrumentation ,X-ray CT ,Alzheimer's disease ,Radiation ,Amyloid beta-Peptides ,biology ,medicine.diagnostic_test ,X-Rays ,X-ray ,Brain ,Magnetic resonance imaging ,medicine.disease ,Research Papers ,Peptide Fragments ,Biochemistry of Alzheimer's disease ,Positron emission tomography ,biology.protein ,Tomography, X-Ray Computed ,Alzheimer’s disease ,030217 neurology & neurosurgery - Abstract
Amyloid beta accumulation into insoluble plaques (Aβp) is known to play a significant role in the pathological process in Alzheimer's disease (AD). The presence of Aβp is also one of the neuropathological hallmarks for the disease. AD final diagnosis is generally acknowledged after the evaluation of Aβp deposition in the brain. Insoluble Aβp accumulation may also concur to cause AD as postulated in the so-called amyloid hypothesis. Therefore, the visualization, evaluation and quantification of Aβp are nowadays the keys for a better understanding of the disease, which may point to a possible cure for AD in the near future. Synchrotron-based X-ray phase contrast (XPC) has been demonstrated as the only imaging method that can retrieve the Aβp signal with high spatial resolution (up to 10 µm), high sensitivity and three-dimensional information at the same time. Although at the moment XPC is suitable for ex vivo samples only, it may develop into an alternative to positron emission tomography and magnetic resonance imaging in Aβp imaging. In this contribution the possibility of using synchrotron-based X-ray phase propagation computed tomography to visualize and measure Aβp on mouse brains is presented. A careful setup optimization for this application leads to a significant improvement of spatial resolution (∼1 µm), data acquisition speed (five times faster), X-ray dose (five times lower) and setup complexity, without a substantial loss in sensitivity when compared with the classic implementation of grating-based X-ray interferometry., Journal of Synchrotron Radiation, 23 (3), ISSN:0909-0495, ISSN:1600-5775
- Published
- 2016
17. Spinal cord stimulation improves forelimb use in an alpha-synuclein animal model of Parkinson's disease
- Author
-
Ivani Brys, Wojciech Bobela, Bernard L. Schneider, Romulo Fuentes, and Patrick Aebischer
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,Substantia nigra ,Spinal cord stimulation ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Alpha-synuclein ,Spinal Cord Stimulation ,Behavior, Animal ,Tyrosine hydroxylase ,business.industry ,General Neuroscience ,Parkinson Disease ,General Medicine ,medicine.disease ,Neuromodulation (medicine) ,Rats ,Surgery ,Substantia Nigra ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,chemistry ,Anesthesia ,alpha-Synuclein ,Forelimb ,business ,030217 neurology & neurosurgery ,Immunostaining - Abstract
Neuromodulation by spinal cord stimulation has been proposed as a symptomatic treatment for Parkinson's disease. We tested the chronic effects of spinal cord stimulation in a progressive model of Parkinson's based on overexpression of alpha-synuclein in the substantia nigra. Adult Sprague Dawley rats received unilateral injections of adeno-associated virus serotype 6 (AAV6) in the substantia nigra to express alpha-synuclein. Locomotion and forepaw use of the rats were evaluated during the next 10 weeks. Starting on week 6, a group of AAV6-injected rats received spinal cord stimulation once a week. At the end of the experiment, tyrosine hydroxylase and alpha-synuclein immunostaining were performed. Rats with unilateral alpha-synuclein expression showed a significant decrease in the use of the contralateral forepaw, which was mildly but significantly reverted by spinal cord stimulation applied once a week from the 6th to the 10th week after the AAV6 injection. Long-term spinal cord stimulation proved to be effective to suppress or delay motor symptoms in a sustained and progressive model of Parkinson's and might become an alternative, less invasive neuromodulation option to treat this disease.
- Published
- 2016
- Full Text
- View/download PDF
18. Allele-specific gene editing prevents deafness in a model of dominant progressive hearing loss
- Author
-
J. Keith Joung, Sofia Spataro, Bifeng Pan, Mikołaj Piotr Zaborowski, Gwenaëlle S. G. Géléoc, K. Domenica Karavitaki, Sara P. Garcia, Bernard L. Schneider, David P. Corey, Benjamin P. Kleinstiver, Paola Solanes, Bence György, Carl Nist-Lund, Yukako Asai, and Jeffrey R. Holt
- Subjects
0301 basic medicine ,Hearing loss ,Hearing Loss, Sensorineural ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Genome editing ,CRISPR-Associated Protein 9 ,medicine ,Animals ,Humans ,Guide RNA ,Allele ,Gene ,Alleles ,Cells, Cultured ,Genetics ,Gene Editing ,Cas9 ,Point mutation ,Membrane Proteins ,General Medicine ,Dependovirus ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,medicine.symptom ,DNA - Abstract
Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.
- Published
- 2018
19. Cortico–reticulo–spinal circuit reorganization enables functional recovery after severe spinal cord contusion
- Author
-
Laetitia Baud, Mark Anderson, Bernard L. Schneider, Elodie Rey, Galyna Pidpruzhnykova, Selin Anil, Quentin Barraud, Stéphane Pagès, Polina Shkorbatova, Julie Kreider, Cristina Martinez-Gonzalez, Grégoire Courtine, Janine Beauparlant, Lucia Friedli, Laura Batti, and Leonie Asboth
- Subjects
0301 basic medicine ,Rehabilitation ,business.industry ,General Neuroscience ,medicine.medical_treatment ,medicine.disease ,Spinal cord ,Neuromodulation (medicine) ,03 medical and health sciences ,Glutamatergic ,030104 developmental biology ,0302 clinical medicine ,Lumbar ,medicine.anatomical_structure ,Paralysis ,Medicine ,medicine.symptom ,business ,Neuroscience ,Spinal cord injury ,030217 neurology & neurosurgery ,Motor cortex - Abstract
Severe spinal cord contusions interrupt nearly all brain projections to lumbar circuits producing leg movement. Failure of these projections to reorganize leads to permanent paralysis. Here we modeled these injuries in rodents. A severe contusion abolished all motor cortex projections below injury. However, the motor cortex immediately regained adaptive control over the paralyzed legs during electrochemical neuromodulation of lumbar circuits. Glutamatergic reticulospinal neurons with residual projections below the injury relayed the cortical command downstream. Gravity-assisted rehabilitation enabled by the neuromodulation therapy reinforced these reticulospinal projections, rerouting cortical information through this pathway. This circuit reorganization mediated a motor cortex-dependent recovery of natural walking and swimming without requiring neuromodulation. Cortico-reticulo-spinal circuit reorganization may also improve recovery in humans.
- Published
- 2018
- Full Text
- View/download PDF
20. Adenoviral-mediated expression of G2019S LRRK2 induces striatal pathology in a kinase-dependent manner in a rat model of Parkinson's disease
- Author
-
Bernard L. Schneider, Elpida Tsika, Philippe Colin, Darren J. Moore, An Phu Tran Nguyen, and Julien Dusonchet
- Subjects
Pathology ,medicine.medical_specialty ,Kinase ,Time Factors ,Neurofilament ,Neurite ,Parkinson's disease ,Glycine ,Striatum ,Protein Serine-Threonine Kinases ,Biology ,Parkinsonism ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Adenoviridae ,lcsh:RC321-571 ,Transduction, Genetic ,Tubulin ,Forelimb ,Serine ,medicine ,Animals ,Humans ,Adenovirus ,Rats, Wistar ,Kinase activity ,Neurodegeneration ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,HEK 293 cells ,Parkinson Disease ,LRRK2 ,medicine.disease ,Corpus Striatum ,Rats ,nervous system diseases ,Disease Models, Animal ,HEK293 Cells ,Gene Expression Regulation ,Neurology ,Phosphopyruvate Hydratase ,Mutation ,alpha-Synuclein ,Female - Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset, autosomal dominant Parkinson's disease (PD). LRRK2 contains functional GTPase and kinase domains. The most common G2019S mutation enhances the kinase activity of LRRK2 in vitro whereas G2019S LRRK2 expression in cultured neurons induces toxicity in a kinase-dependent manner. These observations suggest a potential role for kinase activity in LRRK2-associated PD. We have recently developed a novel rodent model of PD with progressive neurodegeneration induced by the adenoviral-mediated expression of G2019S LRRK2. In the present study, we further characterize this LRRK2 model and determine the contribution of kinase activity to LRRK2-mediated neurodegeneration. Recombinant human adenoviral vectors were employed to deliver human wild-type, G2019S or kinase-inactive G2019S/D1994N LRRK2 to the rat striatum. LRRK2-dependent pathology was assessed in the striatum, a region where LRRK2 protein is normally enriched in the mammalian brain. Human LRRK2 variants are robustly expressed throughout the rat striatum. Expression of G2019S LRRK2 selectively induces the accumulation of neuronal ubiquitin-positive inclusions accompanied by neurite degeneration and the altered distribution of axonal phosphorylated neurofilaments. Importantly, the introduction of a kinase-inactive mutation (G2019S/D1994N) completely ameliorates the pathological effects of G2019S LRRK2 in the striatum supporting a kinase activity-dependent mechanism for this PD-associated mutation. Collectively, our study further elucidates the pathological effects of the G2019S mutation in the mammalian brain and supports the development of kinase inhibitors as a potential therapeutic approach for treating LRRK2-associated PD. This adenoviral rodent model provides an important tool for elucidating the molecular basis of LRRK2-mediated neurodegeneration.
- Published
- 2015
21. Motifs in the tau protein that control binding to microtubules and aggregation determine pathological effects
- Author
-
Paolo Paganetti, Pamela Valdés, Graham Knott, Bernard L. Schneider, Matthias Cacquevel, Catherine Maclachlan, Aurélien Lathuilière, Stéphanie Papin, and Andreas Muhs
- Subjects
0301 basic medicine ,lcsh:Medicine ,medicine.disease_cause ,Microtubules ,Mice ,0302 clinical medicine ,Phosphorylation ,Cognitive decline ,lcsh:Science ,Cytoskeleton ,Cerebral Cortex ,Neurons ,Tau Proteins / genetics ,Mutation ,Multidisciplinary ,biology ,Chemistry ,Neurons / metabolism ,Prosencephalon / pathology ,Cell biology ,Cerebral Cortex / pathology ,Protein Binding ,Frontotemporal dementia ,Microtubules / metabolism ,Genetic Vectors ,Tau protein ,Prosencephalon / metabolism ,Hyperphosphorylation ,tau Proteins ,Article ,03 medical and health sciences ,Prosencephalon ,Alzheimer Disease ,Microtubule ,mental disorders ,Genetic Vectors / metabolism ,medicine ,Animals ,Humans ,Alzheimer Disease / pathology ,lcsh:R ,medicine.disease ,Alzheimer Disease / metabolism ,Mice, Inbred C57BL ,030104 developmental biology ,Animals, Newborn ,Cerebral Cortex / ultrastructure ,Rotarod Performance Test ,Mutagenesis, Site-Directed ,biology.protein ,lcsh:Q ,Protein Conformation, beta-Strand ,Tau Proteins / metabolism ,030217 neurology & neurosurgery - Abstract
Tau pathology is associated with cognitive decline in Alzheimer’s disease, and missense tau mutations cause frontotemporal dementia. Hyperphosphorylation and misfolding of tau are considered critical steps leading to tauopathies. Here, we determine how motifs controlling conformational changes in the microtubule-binding domain determine tau pathology in vivo. Human tau was overexpressed in the adult mouse forebrain to compare variants carrying residues that modulate tau propensity to acquire a β-sheet conformation. The P301S mutation linked to frontotemporal dementia causes tau aggregation and rapidly progressing motor deficits. By comparison, wild-type tau becomes heavily hyperphosphorylated, and induces behavioral impairments that do not progress over time. However, the behavioral defects caused by wild-type tau can be suppressed when β-sheet breaking proline residues are introduced in the microtubule-binding domain of tau. This modification facilitates tau interaction with microtubules, as shown by lower levels of phosphorylation, and by the enhanced protective effects of mutated tau against the severing of the cytoskeleton in neurons exposed to vinblastine. Altogether, motifs that are critical for tau conformation determine interaction with microtubules and subsequent pathological modifications, including phosphorylation and aggregation.
- Published
- 2017
- Full Text
- View/download PDF
22. Loss of MITF expression during human embryonic stem cell differentiation disrupts retinal pigment epithelium development and optic vesicle cell proliferation
- Author
-
Kyle Wallace, Anna Petelinsek, Isabel Pinilla, Joseph M. Simonett, Bernard L. Schneider, Jason S. Meyer, M. Joseph Phillips, David M. Gamm, Sara E. Howden, Lynda S. Wright, Eric M. Clark, Elizabeth E. Capowski, and James A. Thomson
- Subjects
Cellular differentiation ,Retinal Pigment Epithelium ,Biology ,Gene Knockout Techniques ,Mice ,Neural Stem Cells ,Genetics ,medicine ,Animals ,Humans ,Protein Isoforms ,Progenitor cell ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Genetics (clinical) ,Embryonic Stem Cells ,Cell Proliferation ,Homeodomain Proteins ,Microphthalmia-Associated Transcription Factor ,Retinal pigment epithelium ,integumentary system ,Cell Differentiation ,General Medicine ,Optic vesicle ,Articles ,Microphthalmia-associated transcription factor ,Molecular biology ,Embryonic stem cell ,Neural stem cell ,body regions ,medicine.anatomical_structure ,sense organs ,Transcription Factors - Abstract
Microphthalmia-associated transcription factor (MITF) is a master regulator of pigmented cell survival and differentiation with direct transcriptional links to cell cycle, apoptosis and pigmentation. In mouse, Mitf is expressed early and uniformly in optic vesicle (OV) cells as they evaginate from the developing neural tube, and null Mitf mutations result in microphthalmia and pigmentation defects. However, homozygous mutations in MITF have not been identified in humans; therefore, little is known about its role in human retinogenesis. We used a human embryonic stem cell (hESC) model that recapitulates numerous aspects of retinal development, including OV specification and formation of retinal pigment epithelium (RPE) and neural retina progenitor cells (NRPCs), to investigate the earliest roles of MITF. During hESC differentiation toward a retinal lineage, a subset of MITF isoforms was expressed in a sequence and tissue distribution similar to that observed in mice. In addition, we found that promoters for the MITF-A, -D and -H isoforms were directly targeted by Visual Systems Homeobox 2 (VSX2), a transcription factor involved in patterning the OV toward a NRPC fate. We then manipulated MITF RNA and protein levels at early developmental stages and observed decreased expression of eye field transcription factors, reduced early OV cell proliferation and disrupted RPE maturation. This work provides a foundation for investigating MITF and other highly complex, multi-purposed transcription factors in a dynamic human developmental model system.
- Published
- 2017
23. G2019S LRRK2 enhances the neuronal transmission of tau in the mouse brain
- Author
-
Shariful Islam, An Phu Tran Nguyen, Pamela Valdés, Darren J. Moore, Guillaume Daniel, and Bernard L. Schneider
- Subjects
0301 basic medicine ,Genetically modified mouse ,Transgene ,Mice, Transgenic ,tau Proteins ,Neuropathology ,Biology ,Hippocampal formation ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Synaptic Transmission ,Neuronal Transmission ,03 medical and health sciences ,Mice ,0302 clinical medicine ,mental disorders ,Genetics ,medicine ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,CA1 Region, Hippocampal ,Genetics (clinical) ,LRRK2 Gene ,Mice, Knockout ,Neurons ,Brain ,Parkinson Disease ,General Medicine ,medicine.disease ,LRRK2 ,nervous system diseases ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Mutation ,Tauopathy ,030217 neurology & neurosurgery - Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset, autosomal dominant Parkinson's disease (PD). LRRK2 mutations typically give rise to Lewy pathology in the brains of PD subjects yet can induce tau-positive neuropathology in some cases. The pathological interaction between LRRK2 and tau remains poorly defined. To explore this interaction in vivo, we crossed a well-characterized human P301S-tau transgenic mouse model of tauopathy with human G2019S-LRRK2 transgenic mice or LRRK2 knockout (KO) mice. We find that endogenous or pathogenic LRRK2 expression has minimal effects on the steady-state levels, solubility and abnormal phosphorylation of human P301S-tau throughout the mouse brain. We next developed a new model of tauopathy by delivering AAV2/6 vectors expressing human P301S-tau to the hippocampal CA1 region of G2019S-LRRK2 transgenic or LRRK2 KO mice. P301S-tau expression induces hippocampal tau pathology and marked degeneration of CA1 pyramidal neurons in mice, however, this occurs independently of endogenous or pathogenic LRRK2 expression. We further developed new AAV2/6 vectors co-expressing human WT-tau and GFP to monitor the neuron-to-neuron transmission of tau within defined hippocampal neuronal circuits. While endogenous LRRK2 is not required for tau transmission, we find that G2019S-LRRK2 markedly enhances the neuron-to-neuron transmission of tau in mice. Our data suggest that mutant tau-induced neuropathology occurs independently of LRRK2 expression in two mouse models of tauopathy but identifies a novel pathogenic role for G2019S-LRRK2 in promoting the neuronal transmission of WT-tau protein. These findings may have important implications for understanding the development of tau neuropathology in LRRK2-linked PD brains.
- Published
- 2017
24. Endoplasmic reticulum and mitochondria in diseases of motor and sensory neurons: a broken relationship?
- Author
-
Roman Chrast, Nathalie Bernard-Marissal, and Bernard L. Schneider
- Subjects
0301 basic medicine ,Cancer Research ,Sensory Receptor Cells ,Immunology ,Review Article ,Mitochondrion ,Biology ,Endoplasmic Reticulum ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Organelle ,medicine ,Animals ,Humans ,Secretion ,lcsh:QH573-671 ,Amyotrophic lateral sclerosis ,Motor Neurons ,lcsh:Cytology ,Endoplasmic reticulum ,Autophagy ,Neurodegeneration ,Neurodegenerative Diseases ,Cell Biology ,medicine.disease ,Mitochondria ,030104 developmental biology ,Mitochondrial Membranes ,Neuroscience ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Recent progress in the understanding of neurodegenerative diseases revealed that multiple molecular mechanisms contribute to pathological changes in neurons. A large fraction of these alterations can be linked to dysfunction in the endoplasmic reticulum (ER) and mitochondria, affecting metabolism and secretion of lipids and proteins, calcium homeostasis, and energy production. Remarkably, these organelles are interacting with each other at specialized domains on the ER called mitochondria-associated membranes (MAMs). These membrane structures rely on the interaction of several complexes of proteins localized either at the mitochondria or at the ER interface and serve as an exchange platform of calcium, metabolites, and lipids, which are critical for the function of both organelles. In addition, recent evidence indicates that MAMs also play a role in the control of mitochondria dynamics and autophagy. MAMs thus start to emerge as a key element connecting many changes observed in neurodegenerative diseases. This review will focus on the role of MAMs in amyotrophic lateral sclerosis (ALS) and hereditary motor and sensory neuropathy, two neurodegenerative diseases particularly affecting neurons with long projecting axons. We will discuss how defects in MAM signaling may impair neuronal calcium homeostasis, mitochondrial dynamics, ER function, and autophagy, leading eventually to axonal degeneration. The possible impact of MAM dysfunction in glial cells, which may affect the capacity to support neurons and/or axons, will also be described. Finally, the possible role of MAMs as an interesting target for development of therapeutic interventions aiming at delaying or preventing neurodegeneration will be highlighted.
- Published
- 2017
25. An R-CaMP1.07 reporter mouse for cell-typespecific expression of a sensitive red fluorescent calcium indicator
- Author
-
Ladan Egolf, Bernard L. Schneider, Adriano Aguzzi, Junichi Nakai, Despoina Goniotaki, Hongkui Zeng, Philipp Bethge, Masamichi Ohkura, Stefano Carta, Jerry L. Chen, Fabian F. Voigt, Linda Madisen, Dayra A. Lorenzo, Fritjof Helmchen, University of Zurich, and Helmchen, Fritjof
- Subjects
0301 basic medicine ,Gene Expression ,lcsh:Medicine ,Neocortex ,Mice ,Cell Signaling ,Animal Cells ,Gene expression ,lcsh:Science ,Calcium signaling ,Neurons ,Multidisciplinary ,Genetically Modified Organisms ,Electroporation ,Animal Models ,Calcium Imaging ,Molecular Imaging ,3. Good health ,In Vivo Imaging ,medicine.anatomical_structure ,Experimental Organism Systems ,Cellular Types ,Genetic Engineering ,Preclinical imaging ,Research Article ,Signal Transduction ,Biotechnology ,Imaging Techniques ,Transgene ,Green Fluorescent Proteins ,10208 Institute of Neuropathology ,Mice, Transgenic ,Neuroimaging ,Mouse Models ,610 Medicine & health ,1100 General Agricultural and Biological Sciences ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Calcium imaging ,1300 General Biochemistry, Genetics and Molecular Biology ,In vivo ,medicine ,Animals ,Calcium Signaling ,Photons ,1000 Multidisciplinary ,Genetically Modified Animals ,10242 Brain Research Institute ,lcsh:R ,Biology and Life Sciences ,Cell Biology ,Neuronal Dendrites ,Molecular biology ,030104 developmental biology ,Cellular Neuroscience ,570 Life sciences ,biology ,Calcium ,lcsh:Q ,Neuroscience - Abstract
Genetically encoded calcium indicators (GECIs) enable imaging of in vivo brain cell activity with high sensitivity and specificity. In contrast to viral infection or in utero electroporation, indicator expression in transgenic reporter lines is induced noninvasively, reliably, and homogenously. Recently, Cre/tTA-dependent reporter mice were introduced, which provide high-level expression of green fluorescent GECIs in a cell-type-specific and inducible manner when crossed with Cre and tTA driver mice. Here, we generated and characterized the first red-shifted GECI reporter line of this type using R-CaMP1.07, a red fluorescent indicator that is efficiently two-photon excited above 1000 nm. By crossing the new R-CaMP1.07 reporter line to Cre lines driving layer-specific expression in neocortex we demonstrate its high fidelity for reporting action potential firing in vivo, long-term stability over months, and versatile use for functional imaging of excitatory neurons across all cortical layers, especially in the previously difficult to access layers 4 and 6. ISSN:1932-6203
- Published
- 2017
26. Un implant bioactif pour prévenir la maladie d'Alzheimer
- Author
-
Bernard L. Schneider and Aurélien Lathuilière
- Subjects
0301 basic medicine ,Web of science ,MEDLINE ,Library science ,Adoptive Transfer / instrumentation ,Alzheimer Disease / prevention & control ,General Biochemistry, Genetics and Molecular Biology ,Immunization / methods ,03 medical and health sciences ,0302 clinical medicine ,Biocompatible Materials / chemistry ,Biocompatible Materials / therapeutic use ,Antibodies / administration & dosage ,Medicine ,Humans ,Drug Compounding / methods ,Aged ,Aged, 80 and over ,business.industry ,General Medicine ,Prostheses and Implants ,ddc:616.8 ,030104 developmental biology ,Adoptive Transfer / methods ,business ,030217 neurology & neurosurgery - Abstract
Reference EPFL-ARTICLE-227589doi:10.1051/medsci/20173301013View record in Web of Science Record created on 2017-05-01, modified on 2017-05-01
- Published
- 2017
27. Overview of Mouse Models of Parkinson's Disease
- Author
-
Wojciech Bobela, Bernard L. Schneider, and Lu Zheng
- Subjects
Parkinson's disease ,Pars compacta ,Neurodegeneration ,Dopaminergic ,Substantia nigra ,General Medicine ,Biology ,medicine.disease ,Dopamine ,Basal ganglia ,Genetic model ,medicine ,Neuroscience ,medicine.drug - Abstract
Parkinson's disease is a neurodegenerative disorder characterized by the loss of neurons in specific regions of the nervous system, notably in the substantia nigra pars compacta and, in most cases, by the deposition of intraneuronal inclusions named Lewy bodies. These pathological alterations have profound effects on the brain function, leading to the progressive development of various symptoms, the most prominent being the impaired initiation of voluntary movements caused by the loss of dopamine signaling in the basal ganglia. Here, we provide an overview of the mouse models of Parkinson's disease, with the goal of guiding selection of the most appropriate model for studying the question at hand. Pharmacological approaches targeting dopamine signaling and toxins leading to selective degeneration of nigral neurons are used to validate symptomatic treatments that aim at restoring effective dopaminergic function for motor control. Alternative mouse models are based on genetic modifications that are meant to reproduce the inherited alterations associated with familial forms of Parkinson's disease. Although genetic models have most often failed to induce overt degeneration of nigral dopaminergic neurons, they provide essential tools to explore the multifactorial etiology of this complex neurodegenerative disorder. Curr. Protoc. Mouse Biol. 4:121-139 © 2014 by John Wiley & Sons, Inc. Keywords: Parkinson's disease; mouse models of neurodegeneration; genetic factors; toxins
- Published
- 2014
- Full Text
- View/download PDF
28. Large-scale chondroitin sulfate proteoglycan digestion with chondroitinase gene therapy leads to reduced pathology and modulates macrophage phenotype following spinal cord contusion injury
- Author
-
Bernard L. Schneider, Athanasios Didangelos, Elizabeth J. Bradbury, Karen D. Bosch, Rafael J. Yáñez-Muñoz, Nicholas D. James, Katalin Bartus, Elizabeth M. Muir, Joost Verhaagen, John H. Rogers, and Netherlands Institute for Neuroscience (NIN)
- Subjects
Pathology ,Time Factors ,Neural Conduction ,Chondroitin ABC Lyase ,BLADDER FUNCTION ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,0302 clinical medicine ,Macrophage ,PLASTICITY ,BRAIN ,contusion ,Spinal cord injury ,IN-VIVO ,Cells, Cultured ,Injections, Spinal ,0303 health sciences ,Cultured ,CD68 ,General Neuroscience ,Articles ,RECOVERY ,chondroitinase ,gene therapy ,HEMISECTION ,neuroprotection ,Female ,ABC ,medicine.medical_specialty ,Spinal ,Cells ,Chondroitin ABC lyase ,Nerve Tissue Proteins ,Biology ,Serotonergic ,Neuroprotection ,Injections ,03 medical and health sciences ,In vivo ,medicine ,Animals ,FUNCTIONAL AXONAL REGENERATION ,Spinal Cord Injuries ,030304 developmental biology ,REPAIR ,Animal ,Macrophages ,Genetic Therapy ,medicine.disease ,spinal cord injury ,Electric Stimulation ,Rats ,Disease Models, Animal ,chemistry ,Chondroitin Sulfate Proteoglycans ,Gene Expression Regulation ,Chondroitin sulfate proteoglycan ,CELLS ,Disease Models ,Sprague-Dawley ,030217 neurology & neurosurgery ,Psychomotor Performance - Abstract
Chondroitin sulfate proteoglycans (CSPGs) inhibit repair following spinal cord injury. Here we use mammalian-compatible engineered chondroitinase ABC (ChABC) delivered via lentiviral vector (LV-ChABC) to explore the consequences of large-scale CSPG digestion for spinal cord repair. We demonstrate significantly reduced secondary injury pathology in adult rats following spinal contusion injury and LV-ChABC treatment, with reduced cavitation and enhanced preservation of spinal neurons and axons at 12 weeks postinjury, compared with control (LV-GFP)-treated animals. To understand these neuroprotective effects, we investigated early inflammatory changes following LV-ChABC treatment. Increased expression of the phagocytic macrophage marker CD68 at 3 d postinjury was followed by increased CD206 expression at 2 weeks, indicating that large-scale CSPG digestion can alter macrophage phenotype to favor alternatively activated M2 macrophages. Accordingly, ChABC treatmentin vitroinduced a significant increase in CD206 expression in unpolarized monocytes stimulated with conditioned medium from spinal-injured tissue explants. LV-ChABC also promoted the remodelling of specific CSPGs as well as enhanced vascularity, which was closely associated with CD206-positive macrophages. Neuroprotective effects of LV-ChABC corresponded with improved sensorimotor function, evident as early as 1 week postinjury, a time point when increased neuronal survival correlated with reduced apoptosis. Improved function was maintained into chronic injury stages, where improved axonal conduction and increased serotonergic innervation were also observed. Thus, we demonstrate that ChABC gene therapy can modulate secondary injury processes, with neuroprotective effects that lead to long-term improved functional outcome and reveal novel mechanistic evidence that modulation of macrophage phenotype may underlie these effects.
- Published
- 2014
- Full Text
- View/download PDF
29. Intracerebroventricular Injection of Adeno-Associated Virus 6 and 9 Vectors for Cell Type–Specific Transgene Expression in the Spinal Cord
- Author
-
Elisabeth Dirren, Veronica Setola, Bernard L. Schneider, Patrick Aebischer, Chris Towne, and D.E. Redmond
- Subjects
Male ,Transgene ,Genetic Vectors ,Green Fluorescent Proteins ,Cell ,Central nervous system ,Gene Dosage ,Gene Expression ,Gene delivery ,Biology ,medicine.disease_cause ,Viral vector ,Mice ,Transduction (genetics) ,Transduction, Genetic ,Chlorocebus aethiops ,Genetics ,medicine ,Animals ,Transgenes ,Promoter Regions, Genetic ,Molecular Biology ,Adeno-associated virus ,Motor Neurons ,Dependovirus ,Spinal cord ,Virology ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Organ Specificity ,Astrocytes ,Molecular Medicine - Abstract
In the context of motoneuron diseases, gene delivery as an experimental or therapeutic approach is hindered by the challenge to specifically target cell populations that are widely distributed along the spinal cord. Further complicating the task, transgenes often need to be delivered to motoneurons and/or glial cells to address the non-cell-autonomous mechanisms involved in disease pathogenesis. Intracerebroventricular (ICV) injection of recombinant adeno-associated viruses (AAVs) in newborn mice allows distributing viral vectors throughout the central nervous system while limiting undesired transduction of peripheral organs. Here, we show that by combining the appropriate set of AAV serotype and promoter, specific transgene expression can be achieved in either motoneurons or astrocytes along the whole mouse spinal cord. ICV injection of recombinant AAV6 with the cytomegalovirus (cmv) promoter preferentially targets motoneurons, whereas AAV9 particles combined with the astrocyte-specific gfaABC(1)D promoter lead to significant transgene expression selectively targeted to astrocytes. Importantly, ICV coinjection of both AAV6-cmv and AAV9-gfaABC(1)D results in segregated expression of two different transgenes in motoneurons and astrocytes, respectively. Relevance of viral vector delivery via the cerebrospinal fluid was further investigated in young nonhuman primates. Intracisternal injection of recombinant AAV6-cmv led to robust cervical transduction of motoneurons, highlighting the potential of this approach for gene therapy and modeling of motoneuron diseases.
- Published
- 2014
- Full Text
- View/download PDF
30. Gene therapy: a promising approach for neuroprotection in Parkinson’s disease?
- Author
-
Pamela Valdés and Bernard L. Schneider
- Subjects
0301 basic medicine ,Parkinson's disease ,neurotrophic factors ,Genetic enhancement ,Neuroscience (miscellaneous) ,Disease ,Biology ,Gene delivery ,Neuroprotection ,lcsh:RC321-571 ,lcsh:QM1-695 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Neurotrophic factors ,medicine ,Autophagy ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,dopaminergic neurons ,General Commentary ,Neurodegeneration ,Dopaminergic ,aminochrome ,neurodegeneration ,astrocytes ,Gene Therapy ,lcsh:Human anatomy ,medicine.disease ,3. Good health ,Mitochondria ,030104 developmental biology ,Risk factors ,Parkinson’s disease ,Anatomy ,Neuroscience ,030217 neurology & neurosurgery ,dopamine oxidation - Abstract
With the development of effective systems for gene delivery to the central nervous system (CNS), gene therapy has become a therapeutic option for the treatment of Parkinson's disease (PD). Gene therapies that are the most advanced in the clinic have been designed to more effectively compensate for the lack of dopamine signaling in the basal ganglia and rescue the cardinal motor symptoms of PD. However, it remains essential to devise novel therapies to prevent neurodegeneration and disease progression. Since gene therapy has been initially proposed for the delivery of neurotrophins to support the survival and function of dopaminergic neurons, our understanding of PD etiology has changed dramatically. Genes implicated in familial forms of the disease and genetic risk factors associated with sporadic PD have been identified. The spreading of the α-synuclein pathology, as well as perturbations of the lysosomal and mitochondrial activities, appear to play critical roles in the pathogenesis. These findings provide novel targets for gene therapy against PD, but at the same time underline the complexity of this chronic disease. Here we review and discuss the successes and limitations of gene therapy approaches, which have been proposed to provide neuroprotection in PD.
- Published
- 2016
- Full Text
- View/download PDF
31. Overexpression of parkin in the rat nigrostriatal dopamine system protects against methamphetamine neurotoxicity
- Author
-
Bernard L. Schneider, Roberta Traini, Anna Moszczynska, Bin Liu, and Bryan A. Killinger
- Subjects
Male ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,Tyrosine 3-Monooxygenase ,Dopamine ,Ubiquitin-Protein Ligases ,Dopamine Agents ,Substantia nigra ,Striatum ,Biology ,Article ,Parkin ,Body Temperature ,Methamphetamine ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Developmental Neuroscience ,Internal medicine ,medicine ,Animals ,Chymotrypsin ,Analysis of Variance ,Toxicity ,Tyrosine hydroxylase ,Pars compacta ,Dopaminergic ,Meth ,Dependovirus ,Neuroprotection ,Corpus Striatum ,Rats ,nervous system diseases ,Substantia Nigra ,Disease Models, Animal ,Endocrinology ,nervous system ,Neurology ,chemistry ,Neurotoxicity Syndromes ,Neuroscience ,Synaptosomes ,medicine.drug - Abstract
Methamphetamine (METH) is a central nervous system psychostimulant with a high potential for abuse. At high doses, METH causes a selective degeneration of dopaminergic terminals in the striatum, sparing other striatal terminals and cell bodies. We previously detected a deficit in parkin after binge METH in rat striatal synaptosomes. Parkin is an ubiquitin-protein E3 ligase capable of protecting dopamine neurons from diverse cellular insults. Whether the deficit in parkin mediates the toxicity of METH and whether parkin can protect from toxicity of the drug is unknown. The present study investigated whether overexpression of parkin attenuates degeneration of striatal dopaminergic terminals exposed to binge METH. Parkin overexpression in rat nigrostriatal dopamine system was achieved by microinjection of adeno-associated viral transfer vector 2/6 encoding rat parkin (AAV2/6-parkin) into the substantia nigra pars compacta. The microinjections of AAV2/6-parkin dose-dependently increased parkin levels in both the substantia nigra pars compacta and striatum. The levels of dopamine synthesizing enzyme, tyrosine hydroxylase, remained at the control levels; therefore, tyrosine hydroxylase immunoreactivity was used as an index of dopaminergic terminal integrity. In METH-exposed rats, the increase in parkin levels attenuated METH-induced decreases in striatal tyrosine hydroxylase immunoreactivity in a dose-dependent manner, indicating that parkin can protect striatal dopaminergic terminals against METH neurotoxicity. (c) 2013 Elsevier Inc. All rights reserved.
- Published
- 2013
- Full Text
- View/download PDF
32. Focal expression of adeno-associated viral-mutant tau induces widespread impairment in an APP mouse model
- Author
-
Melissa R. Andrews, Jean-Charles Bensadoun, James W. Fawcett, Ishrut Hussain, Jill C. Richardson, Maria Grazia Spillantini, Fred S. Wouters, Matthias Cacquevel, Patrick Aebischer, Elisa Dassie, Bernard L. Schneider, and David R. Howlett
- Subjects
Viral vectors ,Aging ,Pathology ,medicine.medical_specialty ,Mutant ,Morris water navigation task ,Mice, Transgenic ,tau Proteins ,Biology ,Transfection ,medicine.disease_cause ,Adenoviridae ,Viral vector ,Amyloid beta-Protein Precursor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Adeno-associated virus ,Memory ,mental disorders ,medicine ,Animals ,Neurodegeneration ,Alzheimer's precursor protein ,Neuroinflammation ,030304 developmental biology ,Neurons ,Memory Disorders ,0303 health sciences ,General Neuroscience ,Alzheimer's disease ,Entorhinal cortex ,medicine.disease ,Molecular biology ,Mice, Inbred C57BL ,Tauopathy ,Tauopathies ,Neurology (clinical) ,Tau ,Geriatrics and Gerontology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Adeno-associated virus serotype 6 (AAV6) viral vectors encoding mutant and normal tau were used to produce focal tau pathology. Two mutant forms of tau were used; the P301S tau mutation is associated with neurofibrillary tangle formation in humans, and the 3PO mutation leads to rapid tau aggregation and is associated with pathogenic phosphorylation and cytotoxicity in vitro. We show that adeno-associated viral injection into entorhinal cortex of normal and tau knockout animals leads to local overexpression of tau, and the presence of human tau in axons projecting to and emanating from the entorhinal cortex. Starting at 2 months and increasing by 6 months post-injection neurons expressing mutant tau developed hyperphosphorylated tau pathology, in addition to dystrophic neurites. There was neuronal loss in tau-expressing regions, which was similar in normal and in TASTPM mice injected with mutant tau. There was neuroinflammation around plaques, and in regions expressing mutant tau. We saw no evidence that mutant tau had affected amyloid-beta pathology or vice versa. Morris water maze behavioral tests demonstrated mild memory impairment attributable to amyloid-beta pathology at 2 and 4 months, with severe impairment at 6 months in animals receiving adeno-associated viral-3PO. Therefore, TASTPM mice injected with mutant tau displayed many of the main features characteristic of human Alzheimer's disease patients and might be used as a model to test new drugs to ameliorate clinical features of Alzheimer's disease. (C) 2013 Elsevier Inc. All rights reserved.
- Published
- 2013
- Full Text
- View/download PDF
33. Direct and Retrograde Transduction of Nigral Neurons with AAV6, 8, and 9 and Intraneuronal Persistence of Viral Particles
- Author
-
Karin Löw, Patrick Aebischer, and Bernard L. Schneider
- Subjects
viruses ,Genetic Vectors ,Substantia nigra ,Striatum ,Genome, Viral ,Gene delivery ,Biology ,Injections ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Transduction (genetics) ,0302 clinical medicine ,Capsid ,Transduction, Genetic ,Genetics ,medicine ,Animals ,Deoxyribonuclease I ,Humans ,Axon ,Oxidopamine ,Molecular Biology ,Research Articles ,030304 developmental biology ,Cell Nucleus ,Neurons ,0303 health sciences ,Dopaminergic Neurons ,HEK 293 cells ,Dopaminergic ,Virion ,Biological Transport ,Dependovirus ,Virology ,Axons ,Rats ,Neostriatum ,Substantia Nigra ,medicine.anatomical_structure ,HEK293 Cells ,chemistry ,nervous system ,Nerve Degeneration ,Molecular Medicine ,Female ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Recombinant adeno-associated viral (AAV) vectors of serotypes 6, 8, and 9 were characterized as tools for gene delivery to dopaminergic neurons in the substantia nigra for future gene therapeutic applications in Parkinson's disease. While vectors of all three serotypes transduced nigral dopaminergic neurons with equal efficiency when directly injected to the substantia nigra, AAV6 was clearly superior to AAV8 and AAV9 for retrograde transduction of nigral neurons after striatal delivery. For sequential transduction of nigral dopaminergic neurons, the combination of AAV9 with AAV6 proved to be more powerful than AAV8 with AAV6 or repeated AAV6 administration. Surprisingly, single-stranded viral genomes persisted in nigral dopaminergic neurons within cell bodies and axon terminals in the striatum, and intact assembled AAV capsid was enriched in nuclei of nigral neurons, 4 weeks after virus injections to the substantia nigra. 6-Hydroxydopamine (6-OHDA)-induced degeneration of dopaminergic neurons in the substantia nigra reduced the number of viral genomes in the striatum, in line with viral genome persistence in axon terminals. However, 6-OHDA-induced axonal degeneration did not induce any transsynaptic spread of AAV infection in the striatum. Therefore, the potential presence of viral particles in axons may not represent an important safety issue for AAV gene therapy applications in neurodegenerative diseases.
- Published
- 2013
- Full Text
- View/download PDF
34. Behaviour-dependent recruitment of long-range projection neurons in somatosensory cortex
- Author
-
Stefano Carta, Joana Soldado-Magraner, Bernard L. Schneider, Jerry L. Chen, Fritjof Helmchen, University of Zurich, and Helmchen, Fritjof
- Subjects
610 Medicine & health ,Sensory system ,Biology ,Somatosensory system ,Mice ,03 medical and health sciences ,Discrimination, Psychological ,0302 clinical medicine ,Calcium imaging ,SX00 SystemsX.ch ,Cortex (anatomy) ,medicine ,Animals ,Premovement neuronal activity ,Projection (set theory) ,10194 Institute of Neuroinformatics ,030304 developmental biology ,Neurons ,1000 Multidisciplinary ,Afferent Pathways ,0303 health sciences ,Multidisciplinary ,Neocortex ,10242 Brain Research Institute ,Behavior, Animal ,Somatosensory Cortex ,Anatomy ,medicine.anatomical_structure ,Somatosensory evoked potential ,Vibrissae ,570 Life sciences ,biology ,Calcium ,SX11 Neurochoice ,Neuroscience ,030217 neurology & neurosurgery - Abstract
In the mammalian neocortex, segregated processing streams are thought to be important for forming sensory representations of the environment(1,2), but how local information in primary sensory cortex is transmitted to other distant cortical areas during behaviour is unclear. Here we show task-dependent activation of distinct, largely non-overlapping long-range projection neurons in the whisker region of primary somatosensory cortex (S1) in awake, behaving mice. Using two-photon calcium imaging, we monitored neuronal activity in anatomically identified S1 neurons projecting to secondary somatosensory (S2) or primary motor (M1) cortex in mice using their whiskers to perform a texture-discrimination task or a task that required them to detect the presence of an object at a certain location. Whisking-related cells were found among S2-projecting (S2P) but not M1-projecting (M1P) neurons. A higher fraction of S2P than M1P neurons showed touch-related responses during texture discrimination, whereas a higher fraction of M1P than S2P neurons showed touch-related responses during the detection task. In both tasks, S2P and M1P neurons could discriminate similarly between trials producing different behavioural decisions. However, in trials producing the same decision, S2P neurons performed better at discriminating texture, whereas M1P neurons were better at discriminating location. Sensory stimulus features alone were not sufficient to elicit these differences, suggesting that selective transmission of S1 information to S2 and M1 is driven by behaviour.
- Published
- 2013
- Full Text
- View/download PDF
35. Alpha-synuclein ferrireductase activity is detectible in vivo, is altered in Parkinson's disease and increases the neurotoxicity of DOPAL
- Author
-
John P. Hart, Jennifer S. McDowall, Kevin C. Honeychurch, Philippe Colin, Ioanna Ntai, David R. Brown, and Bernard L. Schneider
- Subjects
0301 basic medicine ,Male ,Parkinson's disease ,FMN Reductase ,Iron ,Biology ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,In vivo ,Dopamine ,Neurotoxicity ,medicine ,Animals ,Humans ,Molecular Biology ,Synuclein ,Alpha-synuclein ,Dopaminergic ,Brain ,Parkinson Disease ,Cell Biology ,medicine.disease ,nervous system diseases ,Cell biology ,Rats ,030104 developmental biology ,nervous system ,chemistry ,Biochemistry ,DOPAL ,Toxicity ,Ferrireductase ,alpha-Synuclein ,Female ,030217 neurology & neurosurgery ,medicine.drug - Abstract
The normal cellular role of α-synuclein is of potential importance in understanding diseases in which an aggregated form of the protein has been implicated. A potential loss or change in the normal function of α-synuclein could play a role in the aetiology of diseases such as Parkinson's disease. Recently, it has been suggested that α-synuclein could cause the enzymatic reduction of iron and a cellular increase in Fe(II) levels. Experiments were carried out to determine if such activity could be measured in vivo. Experiments with rats overexpressing human α-synuclein in nigral dopaminergic neurons demonstrated a correlation between α-synuclein expression and ferrireductase activity. Furthermore, studies on tissue from Parkinson's disease patient brains showed a significant decrease in ferrireductase activity, possibly due to deposition of large amounts of inactive protein. Cellular studies suggest that increase ferrireductase activity results in increased levels of dopamine metabolites and increased sensitivity to the toxicity of DOPAL. These findings demonstrate that α-synuclein ferrireductase activity is present in vivo and its alteration may play a role in neuron loss in disease.
- Published
- 2016
- Full Text
- View/download PDF
36. Axonal localization of integrins in the CNS is neuronal type and age dependent
- Author
-
Bernard L. Schneider, David A. Tumbarello, Joost Verhaagen, Menghon Cheah, Melissa R. Andrews, Elizabeth B. Moloney, Patrick Aebischer, Sara Soleman, Matthew R. J. Mason, Jean-Charles Bensadoun, James W. Fawcett, University of St Andrews. School of Medicine, Netherlands Institute for Neuroscience (NIN), Andrews, Melissa R [0000-0001-5960-5619], Cheah, Menghon [0000-0003-1659-3771], Tumbarello, David A [0000-0002-5169-0561], Moloney, Elizabeth [0000-0003-0746-5081], Schneider, Bernard [0000-0001-5485-8748], and Apollo - University of Cambridge Repository
- Subjects
Male ,Integrin ,Integrin alpha6 ,axon initial segment ,Rats, Sprague-Dawley ,Pioneer axon ,Adeno-associated virus ,Ganglia, Spinal ,Axon ,retinal ganglion cell ,Dorsal root ganglia ,sensorimotor cortex ,Integrin alpha Chains ,General Neuroscience ,Integrin beta1 ,dorsal root ganglia ,Brain ,Retinal ganglion-cells ,General Medicine ,New Research ,Axon initial segment ,Sciatic Nerve ,Sensorimotor cortex ,medicine.anatomical_structure ,Spinal Cord ,RC0321 Neuroscience. Biological psychiatry. Neuropsychiatry ,Neurite ,integrin ,Neuroscience(all) ,Genetic Vectors ,NDAS ,adeno-associated virus ,QH426 Genetics ,Biology ,Development ,Retinal ganglion ,medicine ,Animals ,QH426 ,Spinal Cord Injuries ,Lentivirus ,Optic Nerve ,Axons ,Animals, Newborn ,nervous system ,Rats, Inbred Lew ,Axoplasmic transport ,RC0321 ,Axon guidance ,Neuroscience - Abstract
The regenerative ability of CNS axons decreases with age however this ability remains largely intact in PNS axons throughout adulthood. These differences are likely to correspond with age-related silencing of proteins necessary for axon growth and elongation. In previous studies, it has been shown that reintroduction of the alpha9 integrin subunit (tenascin-C receptor, ?9) that is downregulated in adult CNS can improve neurite outgrowth and sensory axon regeneration after a dorsal rhizotomy or a dorsal column crush spinal cord lesion. In the current study, we demonstrate that virally-expressed integrins (?9, ?6, or ?1 integrin) in the adult rat sensorimotor cortex and adult red nucleus are excluded from axons following neuronal transduction. Attempts to stimulate transport by inclusion of a cervical spinal injury and thus an upregulation of extracellular matrix molecules at the lesion site, or co-transduction with its binding partner, ?1 integrin, did not induce integrin localization within axons. In contrast, virally-expressed ?9 integrin in developing rat cortex (postnatal day 5 or 10) demonstrated clear localization of integrins in cortical axons revealed by the presence of integrin in the axons of the corpus callosum and internal capsule as well as in the neuronal cell body. Furthermore, examination of dorsal root ganglia neurons and retinal ganglion cells demonstrated integrin localization both within peripheral nerve as well as dorsal root axons and within optic nerve axons, respectively. Together, our results suggest a differential ability for in vivo axonal transport of transmembrane proteins dependent on neuronal age and subtype.Significance Statement: Most CNS neurons have an intrinsically low ability to regenerate their axons. This study has asked whether the transport into axons of integrins, the receptors that mediate growth through extracellular matrix, might reveal reasons for the poor regenerative ability of CNS axons. Tagged integrins were expressed in sensory, retinal ganglion cell, cortical and red nucleus neurons. The integrins were transported down the axons of sensory and retinal ganglion cell axons, but not down the axons of adult cortical or red nucleus neurons. However, during the postnatal period of corticospinal axon growth, cortical neurons admitted integrins into their axons. The findings suggest that exclusion of integrins and other receptors from CNS axons may be a cause for their poor regenerative ability.
- Published
- 2016
37. O2‐13‐03: Efficacy of ACI‐35, A Liposomal Anti‐Phospho Tau Vaccine in Two Different Mouse Models of Alzheimer's Disease
- Author
-
Kasia Piorkowska, Valérie Giriens, Andreas Muhs, Philippe Colin, Maria Pihlgren, Nathalie Chuard, Andrea Pfeifer, Marija Vukicevic, Valerie Gafner, Bernard L. Schneider, Sameer Nazeeruddin, and Pamela Valdés
- Subjects
Liposome ,Epidemiology ,business.industry ,Health Policy ,Disease ,Pharmacology ,Phospho tau ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2016
- Full Text
- View/download PDF
38. A subcutaneous cellular implant for passive immunization against amyloid-β reduces brain amyloid and tau pathologies
- Author
-
Bernard L. Schneider, Patrick Aebischer, Vanessa Laversenne, Marco Stampanoni, Erhard Kopetzki, Helmut Jacobsen, Alberto Astolfo, Bernd Bohrmann, and Aurélien Lathuilière
- Subjects
0301 basic medicine ,Plaque, Amyloid / metabolism ,medicine.medical_treatment ,Immunization, Passive / methods ,Cell ,Plaque, Amyloid ,Antibodies, Monoclonal / administration & dosage ,Alzheimer Disease / prevention & control ,law.invention ,Mice ,0302 clinical medicine ,Subcutaneous Absorption ,law ,Cells, Cultured ,Drug Implants ,biology ,Antibodies, Monoclonal ,Brain ,Recombinant Proteins / administration & dosage ,Alzheimer's disease ,Cellular implant ,Recombinant Proteins ,Neuroprotection ,medicine.anatomical_structure ,Tauopathies ,Recombinant DNA ,Brain / metabolism ,Antibody ,medicine.symptom ,Recombinant Proteins / pharmacology ,Recombinant Proteins / pharmacokinetics ,Amyloid beta-Peptides / antagonists & inhibitors ,Amyloid ,Antibodies, Monoclonal / pharmacology ,medicine.drug_class ,Mice, Transgenic ,Brain damage ,Monoclonal antibody ,Tauopathies / prevention & control ,03 medical and health sciences ,Alzheimer Disease ,mental disorders ,medicine ,Animals ,Secretion ,Amyloid beta-Peptides ,business.industry ,Amyloid beta-Peptides / metabolism ,Immunization, Passive ,Immunotherapy ,Antibodies, Monoclonal / blood ,Antibodies, Monoclonal / pharmacokinetics ,Peptide Fragments ,030104 developmental biology ,Immunology ,biology.protein ,Encapsulation ,Immunization ,Neurology (clinical) ,Amyloid beta-Peptides / immunology ,business ,030217 neurology & neurosurgery ,Peptide Fragments / metabolism - Abstract
Passive immunization against misfolded toxic proteins is a promising approach to treat neurodegenerative disorders. For effective immunotherapy against Alzheimer's disease, recent clinical data indicate that monoclonal antibodies directed against the amyloid-β peptide should be administered before the onset of symptoms associated with irreversible brain damage. It is therefore critical to develop technologies for continuous antibody delivery applicable to disease prevention. Here, we addressed this question using a bioactive cellular implant to deliver recombinant anti-amyloid-β antibodies in the subcutaneous tissue. An encapsulating device permeable to macromolecules supports the long-term survival of myogenic cells over more than 10 months in immunocompetent allogeneic recipients. The encapsulated cells are genetically engineered to secrete high levels of anti-amyloid-β antibodies. Peripheral implantation leads to continuous antibody delivery to reach plasma levels that exceed 50 µg/ml. In a proof-of-concept study, we show that the recombinant antibodies produced by this system penetrate the brain and bind amyloid plaques in two mouse models of the Alzheimer's pathology. When encapsulated cells are implanted before the onset of amyloid plaque deposition in TauPS2APP mice, chronic exposure to anti-amyloid-β antibodies dramatically reduces amyloid-β40 and amyloid-β42 levels in the brain, decreases amyloid plaque burden, and most notably, prevents phospho-tau pathology in the hippocampus. These results support the use of encapsulated cell implants for passive immunotherapy against the misfolded proteins, which accumulate in Alzheimer's disease and other neurodegenerative disorders.
- Published
- 2016
39. Lentiviral Vectors for the Engineering of Implantable Cells Secreting Recombinant Antibodies
- Author
-
Aurélien Lathuilière and Bernard L. Schneider
- Subjects
0301 basic medicine ,Cell type ,medicine.drug_class ,Genetic enhancement ,Recombinant Proteins / genetics ,Genetic Vectors ,Transduction, Genetic / methods ,Biology ,Monoclonal antibody ,law.invention ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,law ,Ex vivo gene therapy ,Lentivirus / genetics ,medicine ,Humans ,Cell encapsulation ,Recombinant Proteins / immunology ,Recombinant antibodies ,Antibody-Producing Cells / immunology ,Gene Transfer Techniques ,Genetically modified organism ,Cell biology ,Transplantation ,Chronic delivery ,030104 developmental biology ,030220 oncology & carcinogenesis ,Recombinant DNA ,Cell transplantation ,Genetic Engineering / methods ,Antibody-Producing Cells / metabolism - Abstract
The implantation of genetically modified cells is considered for the chronic delivery of therapeutic recombinant proteins in vivo. In the context of gene therapy, the genetic engineering of cells faces two main challenges. First, it is critical to generate expandable cell sources, which can maintain stable high productivity of the recombinant protein of interest over time, both in culture and after transplantation. In addition, gene transfer techniques need to be developed to engineer cells synthetizing complex polypeptides, such as recombinant monoclonal antibodies, to broaden the range of potential therapeutic applications. Here, we provide a workflow for the use of lentiviral vectors as a flexible tool to generate antibody-producing cells. In particular, lentiviral vectors can be used to genetically engineer the cell types compatible with encapsulation devices protecting the implanted cells from the host immune system. Detailed methods are provided for the design and production of lentiviral vectors, optimization of cell transduction, as well as for the quantification and quality control of the produced recombinant antibody.
- Published
- 2016
40. α-Synuclein in Central Nervous System and from Erythrocytes, Mammalian Cells, and Escherichia coli Exists Predominantly as Disordered Monomer
- Author
-
Michel Prudent, Bernard L. Schneider, Mustafa T. Ardah, Patrick Aebischer, Elpida Tsika, David Eliezer, Omar M. A. El-Agnaf, Philippe Coune, Carole Desobry, Darren J. Moore, Hilal A. Lashuel, Martial K. Mbefo, Bruno Fauvet, Niels Lion, Mohamed Bilal Fares, Eliezer Masliah, and Sarah Michael
- Subjects
Central Nervous System ,Erythrocytes ,Protein Conformation ,animal diseases ,medicine.disease_cause ,Biochemistry ,Protein Structure, Secondary ,Rats, Sprague-Dawley ,Mice ,0302 clinical medicine ,Protein structure ,heterocyclic compounds ,Peptide sequence ,Gel electrophoresis ,0303 health sciences ,Brain ,Recombinant Proteins ,Protein Structure and Folding ,Chromatography, Gel ,alpha-Synuclein ,Protein folding ,Immunoblotting ,Molecular Sequence Data ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Tetramer ,Cell Line, Tumor ,Escherichia coli ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Protein Unfolding ,030304 developmental biology ,HEK 293 cells ,Cell Biology ,Molecular biology ,nervous system diseases ,Rats ,HEK293 Cells ,nervous system ,Mutation ,health occupations ,Protein quaternary structure ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Since the discovery and isolation of α-synuclein (α-syn) from human brains, it has been widely accepted that it exists as an intrinsically disordered monomeric protein. Two recent studies suggested that α-syn produced in Escherichia coli or isolated from mammalian cells and red blood cells exists predominantly as a tetramer that is rich in α-helical structure (Bartels, T., Choi, J. G., and Selkoe, D. J. (2011) Nature 477, 107-110; Wang, W., Perovic, I., Chittuluru, J., Kaganovich, A., Nguyen, L. T. T., Liao, J., Auclair, J. R., Johnson, D., Landeru, A., Simorellis, A. K., Ju, S., Cookson, M. R., Asturias, F. J., Agar, J. N., Webb, B. N., Kang, C., Ringe, D., Petsko, G. A., Pochapsky, T. C., and Hoang, Q. Q. (2011) Proc. Natl. Acad. Sci. 108, 17797-17802). However, it remains unknown whether or not this putative tetramer is the main physiological form of α-syn in the brain. In this study, we investigated the oligomeric state of α-syn in mouse, rat, and human brains. To assess the conformational and oligomeric state of native α-syn in complex mixtures, we generated α-syn standards of known quaternary structure and conformational properties and compared the behavior of endogenously expressed α-syn to these standards using native and denaturing gel electrophoresis techniques, size-exclusion chromatography, and an oligomer-specific ELISA. Our findings demonstrate that both human and rodent α-syn expressed in the central nervous system exist predominantly as an unfolded monomer. Similar results were observed when human α-syn was expressed in mouse and rat brains as well as mammalian cell lines (HEK293, HeLa, and SH-SY5Y). Furthermore, we show that α-syn expressed in E. coli and purified under denaturing or nondenaturing conditions, whether as a free protein or as a fusion construct with GST, is monomeric and adopts a disordered conformation after GST removal. These results do not rule out the possibility that α-syn becomes structured upon interaction with other proteins and/or biological membranes.
- Published
- 2012
- Full Text
- View/download PDF
41. Nigrostriatal overabundance of α-synuclein leads to decreased vesicle density and deficits in dopamine release that correlate with reduced motor activity
- Author
-
Wojciech Bobela, Safa Mohanna, Patrick Aebischer, Marco Cantoni, Bernard L. Schneider, Graham Knott, Meret Nora Gaugler, Ralf Schneggenburger, Mustafa T. Ardah, Özgür Genç, Omar El-Agnaf, and Jean-Charles Bensadoun
- Subjects
Time Factors ,Apomorphine ,Dopamine ,Degeneration (medical) ,Striatum ,Functional Laterality ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,0302 clinical medicine ,Dopamine Uptake Inhibitors ,Intermediate Filament Proteins ,Transduction, Genetic ,Motor behavior ,Electrochemistry ,Chromatography, High Pressure Liquid ,0303 health sciences ,Targeted Overexpression ,Neurodegeneration ,Dopaminergic ,Anatomy ,Cell biology ,Rat Substantia-Nigra ,Dopamine Agonists ,alpha-Synuclein ,Female ,Synaptic Vesicles ,medicine.drug ,Genetic Vectors ,Green Fluorescent Proteins ,Enzyme-Linked Immunosorbent Assay ,Substantia nigra ,In Vitro Techniques ,Motor Activity ,Neurotransmission ,Biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Microscopy, Electron, Transmission ,Mice Lacking ,Electron microscopy ,medicine ,Animals ,Humans ,030304 developmental biology ,Alpha-synuclein ,Analysis of Variance ,Dopaminergic Neurons ,Catecholamine Concentration ,Homovanillic Acid ,medicine.disease ,Corpus Striatum ,Rats ,Familial Parkinsons-Disease ,Dynamic Changes ,Amphetamine ,Gene Expression Regulation ,nervous system ,chemistry ,In-Vitro ,Vesicular Monoamine Transport Proteins ,Mutation ,Exploratory Behavior ,Potassium ,3,4-Dihydroxyphenylacetic Acid ,Lewy Bodies ,Neurology (clinical) ,Electron-Microscopy ,030217 neurology & neurosurgery - Abstract
alpha-Synuclein (alpha-syn) is a presynaptic protein present at most nerve terminals, but its function remains largely unknown. The familial forms of Parkinson's disease associated with multiplications of the alpha-syn gene locus indicate that overabundance of this protein might have a detrimental effect on dopaminergic transmission. To investigate this hypothesis, we use adeno-associated viral (AAV) vectors to overexpress human alpha-syn in the rat substantia nigra. Moderate overexpression of either wild-type (WT) or A30P alpha-syn differs in the motor phenotypes induced, with only the WT form generating hemiparkinsonian impairments. Wild-type alpha-syn causes a reduction of dopamine release in the striatum that exceeds the loss of dopaminergic neurons, axonal fibers, and the reduction in total dopamine. At the ultrastructural level, the reduced dopamine release corresponds to a decreased density of dopaminergic vesicles and synaptic contacts in striatal terminals. Interestingly, the membrane-binding-deficient A30P mutant does neither notably reduce dopamine release nor it cause ultrastructural changes in dopaminergic axons, showing that alpha-syn's membrane-binding properties are critically involved in the presynaptic defects. To further determine if the affinity of the protein for membranes determines the extent of motor defects, we compare three forms of alpha-syn in conditions leading to pronounced degeneration. While membrane-binding alpha-syns (wild-type and A53T) induce severe motor impairments, an N-terminal deleted form with attenuated affinity for membranes is inefficient in inducing motor defects. Overall, these results demonstrate that alpha-syn overabundance is detrimental to dopamine neurotransmission at early stages of the degeneration of nigrostriatal dopaminergic axons.
- Published
- 2012
- Full Text
- View/download PDF
42. Sustained expression of PGC-1 in the rat nigrostriatal system selectively impairs dopaminergic function
- Author
-
Carine Ciron, Patrick Aebischer, Julien Dusonchet, Bernard L. Schneider, and Sylvain Lengacher
- Subjects
Dopamine ,Mitochondrion ,medicine.disease_cause ,Oxidative Phosphorylation ,Rats, Sprague-Dawley ,Mice ,Adenosine Triphosphate ,0302 clinical medicine ,Cells, Cultured ,Genetics (clinical) ,Membrane Potential, Mitochondrial ,Neurons ,0303 health sciences ,Mitochondrial Biogenesis ,Complex-I ,Dopaminergic ,Amyotrophic-Lateral-Sclerosis ,Articles ,General Medicine ,Dependovirus ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Mitochondria ,3. Good health ,Substantia Nigra ,alpha-Synuclein ,Female ,medicine.drug ,medicine.medical_specialty ,Cell Survival ,Cellular respiration ,Cell Respiration ,Genetic Vectors ,Huntingtons-Disease ,Substantia nigra ,Oxidative phosphorylation ,Biology ,03 medical and health sciences ,Oxygen Consumption ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Cell-Death ,Molecular Biology ,030304 developmental biology ,Mouse Model ,Dopaminergic Neurons ,Parkinsons-Disease ,Corpus Striatum ,Rats ,Pgc-1-Alpha ,Endocrinology ,Gene Expression Regulation ,nervous system ,Mitochondrial biogenesis ,Gamma Agonist Pioglitazone ,Nerve Degeneration ,Trans-Activators ,030217 neurology & neurosurgery ,Oxidative stress ,Transcription Factors - Abstract
Mitochondrial dysfunction and oxidative stress have been implicated in the etiology of Parkinson's disease. Therefore, pathways controlling mitochondrial activity rapidly emerge as potential therapeutic targets. Here, we explore the neuronal response to prolonged overexpression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), a transcriptional regulator of mitochondrial function, both in vitro and in vivo. In neuronal primary cultures from the ventral midbrain, PGC-1α induces mitochondrial biogenesis and increases basal respiration. Over time, we observe an increasing proportion of the oxygen consumed by neurons which are dedicated to adenosine triphosphate production. In parallel to enhanced oxidative phosphorylation, PGC-1α progressively leads to a decrease in mitochondrial polarization. In the adult rat nigrostriatal system, adeno-associated virus (AAV)-mediated overexpression of PGC-1α induces the selective loss of dopaminergic markers and increases dopamine (DA) catabolism, leading to a reduction in striatal DA content. In addition, PGC-1α prevents the labeling of nigral neurons following striatal injection of the fluorogold retrograde tracer. When PGC-1α is expressed at higher levels following intranigral AAV injection, it leads to overt degeneration of dopaminergic neurons. Finally, PGC-1α overexpression does not prevent nigrostriatal degeneration in pathologic conditions induced by α-synuclein overexpression. Overall, we find that lasting overexpression of PGC-1α leads to major alterations in the metabolic activity of neuronal cells which dramatically impair dopaminergic function in vivo. These results highlight the central role of PGC-1α in the function and survival of dopaminergic neurons and the critical need for maintaining physiological levels of PGC-1α activity.
- Published
- 2012
- Full Text
- View/download PDF
43. Lentiviral vectors express chondroitinase ABC in cortical projections and promote sprouting of injured corticospinal axons
- Author
-
Bernard L. Schneider, Rafael J. Yáñez-Muñoz, Sherif G. Ahmed, Anna Y Allan, John H. Rogers, Elizabeth M. Muir, Rong-Rong Zhao, Joost Verhaagen, Jean-Charles Bensadoun, João Nuno Alves, James W. Fawcett, Kasper C. D. Roet, Hannah Rickman, Roger J. Keynes, Jessica C. F. Kwok, and Netherlands Institute for Neuroscience (NIN)
- Subjects
System ,Male ,Chondroitinase ABC ,Pyramidal Tracts ,CSPG, chondroitin sulfate proteoglycan ,Chondroitin ABC Lyase ,GAG, glycosaminoglycan ,LV-ChABC, lentiviral vector encoding chondroitinase ABC ,Mice ,0302 clinical medicine ,Functional Recovery ,Axon ,CST, corticospinal tract ,Spinal cord injury ,Gene-Transfer ,Cells, Cultured ,Cerebral Cortex ,0303 health sciences ,DMEM, Dulbecco's modified Eagle's medium ,General Neuroscience ,Brain ,3. Good health ,medicine.anatomical_structure ,medicine.symptom ,Delivery ,Plasticity ,Cells ,Neuroscience(all) ,Genetic Vectors ,Central nervous system ,Chondroitin ABC lyase ,Transgene Expression ,Biology ,CNS, central nervous system ,Gene Expression Regulation, Enzymologic ,Article ,Viral vector ,CS, chondroitin sulfate ,Lesion ,03 medical and health sciences ,PBS, phosphate buffered saline ,PGK, phosphoglycerate kinase ,medicine ,Animals ,Humans ,Axon regeneration ,Spinal Cord Injuries ,030304 developmental biology ,Sheep ,Pyramidal tracts ,Spinal-Cord-Injury ,Lentivirus ,CMV, cytomegalovirus ,Lentiviral vectors ,BDA, biotinylated dextran amine ,medicine.disease ,Corticospinal tract ,Nerve Regeneration ,Rats ,HEK293 Cells ,nervous system ,Therapy ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Highlights ► Lentiviral vectors can transduce neurons and glia to secrete chondroitinase. ► The active enzyme is secreted from long-distance axon projections from the cerebral cortex. ► Chondroitinase transduction promotes preservation and sprouting of damaged corticospinal axons., Several diseases and injuries of the central nervous system could potentially be treated by delivery of an enzyme, which might most effectively be achieved by gene therapy. In particular, the bacterial enzyme chondroitinase ABC is beneficial in animal models of spinal cord injury. We have adapted the chondroitinase gene so that it can direct secretion of active chondroitinase from mammalian cells, and inserted it into lentiviral vectors. When injected into adult rat brain, these vectors lead to extensive secretion of chondroitinase, both locally and from long-distance axon projections, with activity persisting for more than 4 weeks. In animals which received a simultaneous lesion of the corticospinal tract, the vector reduced axonal die-back and promoted sprouting and short-range regeneration of corticospinal axons. The same beneficial effects on damaged corticospinal axons were observed in animals which received the chondroitinase lentiviral vector directly into the vicinity of a spinal cord lesion.
- Published
- 2011
- Full Text
- View/download PDF
44. Neuroprotection by Gene Therapy Targeting Mutant SOD1 in Individual Pools of Motor Neurons Does not Translate Into Therapeutic Benefit in fALS Mice
- Author
-
Bernard L. Schneider, Patrick Aebischer, Veronica Setola, and Chris Towne
- Subjects
Transgene ,Genetic Vectors ,Mice, Transgenic ,Gene delivery ,Biology ,Adenoassociated Virus Vectors ,Injections, Intramuscular ,Neuroprotection ,Mice ,Superoxide Dismutase-1 ,Drug Discovery ,medicine ,Genetics ,Animals ,Humans ,Gene silencing ,RNA, Small Interfering ,Amyotrophic lateral sclerosis ,Transgenic Als Mice ,Molecular Biology ,Prolongs Survival ,Motor Neurons ,Rna Interference ,Pharmacology ,Gene knockdown ,Skeletal-Muscle ,Mouse Model ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Amyotrophic-Lateral-Sclerosis ,Genetic Therapy ,Dependovirus ,Motor neuron ,medicine.disease ,Molecular biology ,Muscle atrophy ,Cell biology ,Disease Models, Animal ,medicine.anatomical_structure ,Disease Progression ,Degeneration ,Molecular Medicine ,Original Article ,medicine.symptom ,Delivery - Abstract
A major challenge in neurological gene therapy is delivery of the transgene to sufficient cell numbers in an atraumatic manner. This is particularly difficult for motor neuron (MN) diseases that have cells located across the entire spinal cord, brain stem, and cortex. We have used the familial mouse model of amyotrophic lateral sclerosis (ALS) to examine the feasibility of body-wide intramuscular injections of adeno-associated virus serotype 6 (AAV6), a vector capable of axonal retrograde transport, to deliver therapeutic genetic information across the lower MN axis. Neonatal muscle delivery of AAV expressing small hairpin RNAs (shRNAs) against the toxic transgene in this model, human mutant superoxide dismutase 1 (mSOD1), led to significant mSOD1 knockdown in the muscle as well as innervating MNs. This knockdown conferred neuroprotection and halted muscle atrophy in individually targeted MN pools. However, despite the vector being targeted to MNs that innervate muscle groups controlling eating, breathing, and locomotion, this approach was unable to therapeutically impact on disease progression in the ALS mouse model. These results stress the complexity of gene delivery for mSOD1 silencing and suggest that critical thresholds of protein knockdown and transduction across various cell types are required to translate local neuroprotective effects into functional improvements.
- Published
- 2011
- Full Text
- View/download PDF
45. Rab1A Over-Expression Prevents Golgi Apparatus Fragmentation and Partially Corrects Motor Deficits in an Alpha-Synuclein Based Rat Model of Parkinson's Disease
- Author
-
Philippe Coune, Bernard L. Schneider, Patrick Aebischer, and Jean-Charles Bensadoun
- Subjects
Male ,Time Factors ,Parkinson's disease ,Apomorphine ,animal diseases ,Rats, Sprague-Dawley ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Forelimb ,0303 health sciences ,Dopaminergic ,Parkinson Disease ,substantia nigra ,symbols ,Female ,adeno-associated viral vector ,medicine.drug ,Tyrosine 3-Monooxygenase ,Genetic Vectors ,Substantia nigra ,Biology ,Alpha-synuclein ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,symbols.namesake ,medicine ,Animals ,Humans ,Fragmentation (cell biology) ,Secretory pathway ,030304 developmental biology ,Dopaminergic Neurons ,Rab1A ,Golgi apparatus ,motor behavior ,medicine.disease ,Rats ,nervous system diseases ,rab1 GTP-Binding Proteins ,Disease Models, Animal ,nervous system ,chemistry ,Mutation ,Exploratory Behavior ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Although the overabundance of human alpha-synuclein in nigral dopaminergic neurons is considered to play a pathogenic role in Parkinson's disease (PD), it remains unclear how alpha-synuclein leads to neuronal degeneration and motor symptoms. Here, we explored the effect of human alpha-synuclein in the rat substantia nigra following AAV-mediated gene delivery inducing a moderate loss of dopaminergic neurons together with motor impairments. A significant fraction of the surviving nigral neurons were found to express human alpha Syn and displayed a pathological fragmentation of the Golgi apparatus. This observation prompted further investigation on the role of the secretory pathway, in particular at the ER/Golgi level, in alpha-synuclein toxicity. To address this question, we co-expressed human alpha-synuclein with Rab1A, a regulator of ER-to-Golgi vesicular trafficking, and found a significant reduction of Golgi fragmentation. Rab1A did not protect the dopaminergic neurons from the alpha-synuclein-induced degeneration that occurred within several months following vector injection. However, we observed in animals co-expressing Rab1A an improvement of motor behavior that correlates with the rescue of normal Golgi morphology in alpha-synuclein-expressing dopaminergic neurons. The non-prenylable mutant Rab1A-DeltaCC did not produce any of the effects observed with the wild-type form of Rab1A, linking the protective role of Rab1A with its activity in ER-to-Golgi vesicular trafficking. In conclusion, Rab1A can rescue the Golgi fragmentation caused by the overabundance of alpha-synuclein in nigral dopaminergic neurons, improving the ability of the surviving neurons to control motor function in hemiparkinsonian animals.
- Published
- 2011
- Full Text
- View/download PDF
46. Long-range connectivity of mouse primary somatosensory barrel cortex
- Author
-
Bernard L. Schneider, Rachel Aronoff, Carine Ciron, Celine Mateo, Carl C.H. Petersen, and Ferenc Mátyás
- Subjects
Orientation column ,Neocortex ,Secondary somatosensory cortex ,General Neuroscience ,Thalamus ,Sensory system ,Anatomy ,Barrel cortex ,medicine.anatomical_structure ,Somatosensory evoked potential ,Perirhinal cortex ,medicine ,Psychology ,Neuroscience - Abstract
The primary somatosensory barrel cortex processes tactile vibrissae information, allowing rodents to actively perceive spatial and textural features of their immediate surroundings. Each whisker on the snout is individually represented in the neocortex by an anatomically identifiable 'barrel' specified by the segregated termination zones of thalamocortical axons of the ventroposterior medial nucleus, which provide the primary sensory input to the neocortex. The sensory information is subsequently processed within local synaptically connected neocortical microcircuits, which have begun to be investigated in quantitative detail. In addition to these local synaptic microcircuits, the excitatory pyramidal neurons of the barrel cortex send and receive long-range glutamatergic axonal projections to and from a wide variety of specific brain regions. Much less is known about these long-range connections and their contribution to sensory processing. Here, we review current knowledge of the long-range axonal input and output of the mouse primary somatosensory barrel cortex. Prominent reciprocal projections are found between primary somatosensory cortex and secondary somatosensory cortex, motor cortex, perirhinal cortex and thalamus. Primary somatosensory barrel cortex also projects strongly to striatum, thalamic reticular nucleus, zona incerta, anterior pretectal nucleus, superior colliculus, pons, red nucleus and spinal trigeminal brain stem nuclei. These long-range connections of the barrel cortex with other specific cortical and subcortical brain regions are likely to play a crucial role in sensorimotor integration, sensory perception and associative learning.
- Published
- 2010
- Full Text
- View/download PDF
47. Targeted overexpression of the parkin substrate Pael-R in the nigrostriatal system of adult rats to model Parkinson's disease
- Author
-
Julien Dusonchet, Patrick Aebischer, Jean-Charles Bensadoun, and Bernard L. Schneider
- Subjects
Time Factors ,Parkinson's disease ,Apomorphine ,Dopamine ,Gene Expression ,Parkin ,Rats, Sprague-Dawley ,0302 clinical medicine ,Chromatography, High Pressure Liquid ,0303 health sciences ,Behavior, Animal ,Neurodegeneration ,Dopaminergic ,Parkinson Disease ,Dependovirus ,Ubiquitin ligase ,Substantia Nigra ,Globus pallidus ,Neurology ,GABAergic ,Female ,Autosomal-recessive juvenile parkinsonism ,medicine.drug ,Insoluble accumulation ,Tyrosine 3-Monooxygenase ,Green Fluorescent Proteins ,Receptors, Cell Surface ,Biology ,lcsh:RC321-571 ,03 medical and health sciences ,Parkin-associated endothelin receptor-like receptor ,medicine ,Animals ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Adeno-associated viral vector ,030304 developmental biology ,Analysis of Variance ,medicine.disease ,Corpus Striatum ,Rats ,nervous system diseases ,Amphetamine ,Disease Models, Animal ,nervous system ,Vesicular Monoamine Transport Proteins ,biology.protein ,Stereotyped Behavior ,Neuroscience ,Psychomotor Performance ,030217 neurology & neurosurgery - Abstract
Loss of function of parkin, an ubiquitin ligase, is responsible for autosomal recessive juvenile parkinsonism (AR-JP). Parkin-associated endothelin receptor-like receptor (Pael-R) was identified as an authentic substrate of parkin and is thought to accumulate abnormally following loss of parkin activity, causing neurodegeneration of nigral dopaminergic neurons in AR-JP patients. Our aim is therefore to generate a model of AR-JP through overexpression of Pael-R in the nigrostriatal system of adult rats. Using recombinant adeno-associated virus pseudotyped with the serotype 6 capsid (rAAV2/6) as a gene delivery tool, we achieved targeted and robust overexpression of rat Pael-R in nigral neurons and their striatal terminals. Overexpressed Pael-R was shown to accumulate very rapidly in an insoluble form. Accumulation of the receptor triggered a rapid and severe loss of nigral neurons and nigrostriatal fibers and terminals. No cell recovery was observed for up to 6 months post-injection. GABAergic neurons of the globus pallidus were unaffected by Pael-R overexpression, highlighting the selective vulnerability of nigral dopaminergic neurons to abnormal levels of Pael-R. Pael-R-induced degeneration also led to a depletion of striatal dopamine resulting in spontaneous motor impairments, as measured in the cylinder and stepping tests for forelimb akinesia. Interestingly, behavioral deficits of individual animals were correlated with the extent of the nigrostriatal lesion. Insoluble accumulation of Pael-R in the nigrostriatal system of adult rats represents, therefore, a chronic, rapidly progressing and specific model of AR-JP, which recapitulates major pathological hallmarks of the disease.
- Published
- 2009
- Full Text
- View/download PDF
48. Systemic AAV6 Delivery Mediating RNA Interference Against SOD1: Neuromuscular Transduction Does Not Alter Disease Progression in fALS Mice
- Author
-
Bernard L. Schneider, Patrick Aebischer, Cédric Raoul, and Chris Towne
- Subjects
Liver/metabolism ,Myocardium/metabolism ,Molecular Sequence Data ,SOD1 ,RNA Interference ,Biology ,Models, Biological ,Lower motor neuron ,Superoxide dismutase ,Mice ,Transduction (genetics) ,Superoxide Dismutase-1 ,Models ,Spinal Cord/metabolism ,RNA interference ,Drug Discovery ,Genetics ,medicine ,Brain/metabolism ,Animals ,Gene silencing ,Dependovirus/*genetics ,Amyotrophic lateral sclerosis ,Molecular Biology ,Motor Neurons ,Pharmacology ,Base Sequence ,Superoxide Dismutase ,Myocardium ,Amyotrophic Lateral Sclerosis ,Brain ,Dependovirus ,Biological ,Spinal cord ,medicine.disease ,Molecular biology ,Amyotrophic Lateral Sclerosis/*genetics/*therapy ,medicine.anatomical_structure ,Liver ,Spinal Cord ,nervous system ,Disease Progression ,Cancer research ,biology.protein ,Superoxide Dismutase/*genetics ,Molecular Medicine ,Motor Neurons/metabolism - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder arising from the selective death of motor neurons. Approximately 20% of familial ALS (fALS) cases are caused by toxic gain-of-function mutations in the superoxide dismutase 1 (SOD1) gene. We as well as others have provided proof-of-principle for the use of RNA interference (RNAi) against mutant SOD1 as a potential therapy for fALS. With the aim of maximizing the delivery of these silencing instructions, we explored the efficacy of intravenous delivery of recombinant adeno-associated virus (rAAV) serotype 6 expressing small hairpin RNAs targeting mutant SOD1 in the G93A SOD1 fALS mouse model. This approach resulted in a systemic transduction profile, corresponding to transduction of the entire skeletal musculature as well as heart and liver. In addition, motor neurons at all levels of the spinal cord and brain stem were transduced, amounting to 3-5% of the lower motor neuron pool. SOD1 protein levels were reduced by >50% in all the muscles that were examined. Crucially, this silencing profile did not alter the course of the disease in this fALS model, thereby providing compelling evidence that SOD1-mediated damage within skeletal muscles does not contribute to death of motor neurons in ALS. Further, this study demonstrates that motor neurons can be transduced across the length of the spinal cord through a single noninvasive delivery of rAAV.
- Published
- 2008
- Full Text
- View/download PDF
49. Αlpha-Synuclein as a Mediator in the Interplay between Aging and Parkinson's Disease
- Author
-
Patrick Aebischer, Bernard L. Schneider, and Wojciech Bobela
- Subjects
Aging ,Parkinson's disease ,alpha-synuclein ,lcsh:QR1-502 ,Context (language use) ,Review ,Mitochondrion ,Biology ,Biochemistry ,lcsh:Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,nigral dopaminergic neurons ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Alpha-synuclein ,0303 health sciences ,proteostasis ,Dopaminergic Neurons ,Dopaminergic ,Parkinson Disease ,medicine.disease ,nervous system diseases ,Mitochondria ,Proteostasis ,chemistry ,Proteasome ,nervous system ,Synuclein ,Parkinson’s disease ,Neuroscience ,metabolism ,030217 neurology & neurosurgery - Abstract
Accumulation and misfolding of the alpha-synuclein protein are core mechanisms in the pathogenesis of Parkinson’s disease. While the normal function of alpha-synuclein is mainly related to the control of vesicular neurotransmission, its pathogenic effects are linked to various cellular functions, which include mitochondrial activity, as well as proteasome and autophagic degradation of proteins. Remarkably, these functions are also affected when the renewal of macromolecules and organelles becomes impaired during the normal aging process. As aging is considered a major risk factor for Parkinson’s disease, it is critical to explore its molecular and cellular implications in the context of the alpha-synuclein pathology. Here, we discuss similarities and differences between normal brain aging and Parkinson’s disease, with a particular emphasis on the nigral dopaminergic neurons, which appear to be selectively vulnerable to the combined effects of alpha-synuclein and aging.
- Published
- 2015
50. O1‐05‐03: Preventive anti‐Aß passive immunization using a subcutaneous cellular implant lowers brain amyloid burden and tau pathology
- Author
-
Marco Stampanoni, Bernd Bohrmann, Alberto Astolfo, Aurélien Lathuilière, Vanessa Laversenne, Bernard L. Schneider, and Patrick Aebischer
- Subjects
Pathology ,medicine.medical_specialty ,Tau pathology ,Epidemiology ,business.industry ,Health Policy ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Immunization ,Immunology ,Medicine ,Amyloid burden ,Neurology (clinical) ,Implant ,Geriatrics and Gerontology ,business - Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.