Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Commun ; 15(1): 2796, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38555298

RESUMO

The Y-linked SRY gene initiates mammalian testis-determination. However, how the expression of SRY is regulated remains elusive. Here, we demonstrate that a conserved steroidogenic factor-1 (SF-1)/NR5A1 binding enhancer is required for appropriate SRY expression to initiate testis-determination in humans. Comparative sequence analysis of SRY 5' regions in mammals identified an evolutionary conserved SF-1/NR5A1-binding motif within a 250 bp region of open chromatin located 5 kilobases upstream of the SRY transcription start site. Genomic analysis of 46,XY individuals with disrupted testis-determination, including a large multigenerational family, identified unique single-base substitutions of highly conserved residues within the SF-1/NR5A1-binding element. In silico modelling and in vitro assays demonstrate the enhancer properties of the NR5A1 motif. Deletion of this hemizygous element by genome-editing, in a novel in vitro cellular model recapitulating human Sertoli cell formation, resulted in a significant reduction in expression of SRY. Therefore, human NR5A1 acts as a regulatory switch between testis and ovary development by upregulating SRY expression, a role that may predate the eutherian radiation. We show that disruption of an enhancer can phenocopy variants in the coding regions of SRY that cause human testis dysgenesis. Since disease causing variants in enhancers are currently rare, the regulation of gene expression in testis-determination offers a paradigm to define enhancer activity in a key developmental process.


Assuntos
Disgenesia Gonadal , Testículo , Animais , Feminino , Humanos , Masculino , Linhagem Celular , Mamíferos/genética , Sequências Reguladoras de Ácido Nucleico , Células de Sertoli/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Testículo/metabolismo
2.
Brain ; 146(9): 3624-3633, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37410912

RESUMO

The centrosome, as the main microtubule organizing centre, plays key roles in cell polarity, genome stability and ciliogenesis. The recent identification of ribosomes, RNA-binding proteins and transcripts at the centrosome suggests local protein synthesis. In this context, we hypothesized that TDP-43, a highly conserved RNA binding protein involved in the pathophysiology of amyotrophic lateral sclerosis and frontotemporal lobar degeneration, could be enriched at this organelle. Using dedicated high magnification sub-diffraction microscopy on human cells, we discovered a novel localization of TDP-43 at the centrosome during all phases of the cell cycle. These results were confirmed on purified centrosomes by western blot and immunofluorescence microscopy. In addition, the co-localization of TDP-43 and pericentrin suggested a pericentriolar enrichment of the protein, leading us to hypothesize that TDP-43 might interact with local mRNAs and proteins. Supporting this hypothesis, we found four conserved centrosomal mRNAs and 16 centrosomal proteins identified as direct TDP-43 interactors. More strikingly, all the 16 proteins are implicated in the pathophysiology of TDP-43 proteinopathies, suggesting that TDP-43 dysfunction in this organelle contributes to neurodegeneration. This first description of TDP-43 centrosomal enrichment paves the way for a more comprehensive understanding of TDP-43 physiology and pathology.


Assuntos
Esclerose Lateral Amiotrófica , Degeneração Lobar Frontotemporal , Proteinopatias TDP-43 , Humanos , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteinopatias TDP-43/patologia , Degeneração Lobar Frontotemporal/patologia , Centrossomo/metabolismo , Centrossomo/patologia
3.
Sci Adv ; 9(24): eadd5002, 2023 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-37327344

RESUMO

Neurogenesis in the developing human cerebral cortex occurs at a particularly slow rate owing in part to cortical neural progenitors preserving their progenitor state for a relatively long time, while generating neurons. How this balance between the progenitor and neurogenic state is regulated, and whether it contributes to species-specific brain temporal patterning, is poorly understood. Here, we show that the characteristic potential of human neural progenitor cells (NPCs) to remain in a progenitor state as they generate neurons for a prolonged amount of time requires the amyloid precursor protein (APP). In contrast, APP is dispensable in mouse NPCs, which undergo neurogenesis at a much faster rate. Mechanistically, APP cell-autonomously contributes to protracted neurogenesis through suppression of the proneurogenic activator protein-1 transcription factor and facilitation of canonical WNT signaling. We propose that the fine balance between self-renewal and differentiation is homeostatically regulated by APP, which may contribute to human-specific temporal patterns of neurogenesis.


Assuntos
Precursor de Proteína beta-Amiloide , Células-Tronco Neurais , Humanos , Camundongos , Animais , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Diferenciação Celular , Neurônios/metabolismo , Neurogênese
4.
Cell Mol Life Sci ; 80(6): 150, 2023 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-37184603

RESUMO

Amyotrophic lateral sclerosis (ALS) is the most common motor neuron (MN) disease in adults with no curative treatment. Neurofilament (NF) level in patient' fluids have recently emerged as the prime biomarker of ALS disease progression, while NF accumulation in MNs of patients is the oldest and one of the best pathological hallmarks. However, the way NF accumulations could lead to MN degeneration remains unknown. To assess NF accumulations and study the impact on MNs, we compared MNs derived from induced pluripotent stem cells (iPSC) of patients carrying mutations in C9orf72, SOD1 and TARDBP genes, the three main ALS genetic causes. We show that in all mutant MNs, light NF (NF-L) chains rapidly accumulate in MN soma, while the phosphorylated heavy/medium NF (pNF-M/H) chains pile up in axonal proximal regions of only C9orf72 and SOD1 MNs. Excitability abnormalities were also only observed in these latter MNs. We demonstrate that the integrity of the MN axonal initial segment (AIS), the region of action potential initiation and responsible for maintaining axonal integrity, is impaired in the presence of pNF-M/H accumulations in C9orf72 and SOD1 MNs. We establish a strong correlation between these pNF-M/H accumulations, an AIS distal shift, increased axonal calibers and modified repartition of sodium channels. The results expand our understanding of how NF accumulation could dysregulate components of the axonal cytoskeleton and disrupt MN homeostasis. With recent cumulative evidence that AIS alterations are implicated in different brain diseases, preserving AIS integrity could have important therapeutic implications for ALS.


Assuntos
Esclerose Lateral Amiotrófica , Humanos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Filamentos Intermediários , Superóxido Dismutase-1/genética , Proteína C9orf72/genética , Neurônios Motores/patologia
5.
Front Mol Neurosci ; 14: 767041, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970118

RESUMO

Inflammation is a shared hallmark between amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). For long, studies were conducted on tissues of post-mortem patients and neuroinflammation was thought to be only bystander result of the disease with the immune system reacting to dying neurons. In the last two decades, thanks to improving technologies, the identification of causal genes and the development of new tools and models, the involvement of inflammation has emerged as a potential driver of the diseases and evolved as a new area of intense research. In this review, we present the current knowledge about neuroinflammation in ALS, ALS-FTD, and FTD patients and animal models and we discuss reasons of failures linked to therapeutic trials with immunomodulator drugs. Then we present the induced pluripotent stem cell (iPSC) technology and its interest as a new tool to have a better immunopathological comprehension of both diseases in a human context. The iPSC technology giving the unique opportunity to study cells across differentiation and maturation times, brings the hope to shed light on the different mechanisms linking neurodegeneration and activation of the immune system. Protocols available to differentiate iPSC into different immune cell types are presented. Finally, we discuss the interest in studying monocultures of iPS-derived immune cells, co-cultures with neurons and 3D cultures with different cell types, as more integrated cellular approaches. The hope is that the future work with human iPS-derived cells helps not only to identify disease-specific defects in the different cell types but also to decipher the synergistic effects between neurons and immune cells. These new cellular tools could help to find new therapeutic approaches for all patients with ALS, ALS-FTD, and FTD.

6.
J Neurol Neurosurg Psychiatry ; 92(9): 942-949, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33785574

RESUMO

OBJECTIVE: Mutations in superoxide dismutase 1 gene (SOD1), encoding copper/zinc superoxide dismutase protein, are the second most frequent high penetrant genetic cause for amyotrophic lateral sclerosis (ALS) motor neuron disease in populations of European descent. More than 200 missense variants are reported along the SOD1 protein. To limit the production of these aberrant and deleterious SOD1 species, antisense oligonucleotide approaches have recently emerged and showed promising effects in clinical trials. To offer the possibility to any patient with SOD1-ALS to benefit of such a gene therapy, it is necessary to ascertain whether any variant of unknown significance (VUS), detected for example in SOD1 non-coding sequences, is pathogenic. METHODS: We analysed SOD1 mutation distribution after SOD1 sequencing in a large cohort of 470 French familial ALS (fALS) index cases. RESULTS: We identified a total of 27 SOD1 variants in 38 families including two SOD1 variants located in nearsplice or intronic regions of the gene. The pathogenicity of the c.358-10T>G nearsplice SOD1 variant was corroborated based on its high frequency (as the second most frequent SOD1 variant) in French fALS, the segregation analysis confirmed in eight affected members of a large pedigree, the typical SOD1-related phenotype observed (with lower limb onset and prominent lower motor neuron involvement), and findings on postmortem tissues showing SOD1 misaccumulation. CONCLUSIONS: Our results highlighted nearsplice/intronic mutations in SOD1 are responsible for a significant portion of French fALS and suggested the systematic analysis of the SOD1 mRNA sequence could become the method of choice for SOD1 screening, not to miss these specific cases.


Assuntos
Esclerose Lateral Amiotrófica/genética , Mutação , Linhagem , Superóxido Dismutase-1/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise Mutacional de DNA , Feminino , Testes Genéticos , Terapia Genética , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo
7.
Neurobiol Aging ; 101: 181-186, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33626479

RESUMO

Neuroinflammation is a hallmark of Amyotrophic Lateral Sclerosis (ALS) in hSOD1G93A mouse models where microglial cells contribute to the progressive motor neuron degenerative process. S100-A8 and S100-A9 (also known as MRP8 and MRP14, respectively) are cytoplasmic proteins expressed by inflammatory myeloid cells, including microglia and macrophages. Mainly acting as a heterodimer, S100-A8/A9, when secreted, can activate Toll-like Receptor 4 on immune cells, leading to deleterious proinflammatory cytokine production. Deletion of S100a9 in Alzheimer's disease mouse models showed a positive outcome, reducing pathology. We now assessed its role in ALS. Unexpectedly, our results show that deleting S100a9 in hSOD1G93A ALS mice had no impact on mouse survival, but rather accelerated symptoms with no impact on microglial activation and motor neuron survival, suggesting that blocking S100-A9 would not be a valuable strategy for ALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/mortalidade , Calgranulina B/genética , Deleção de Genes , Histona-Lisina N-Metiltransferase , Superóxido Dismutase-1 , Animais , Calgranulina B/metabolismo , Modelos Animais de Doenças , Histona-Lisina N-Metiltransferase/metabolismo , Inflamação , Camundongos , Microglia/metabolismo , Superóxido Dismutase-1/metabolismo , Sobrevida
8.
Nat Neurosci ; 23(11): 1339-1351, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33077946

RESUMO

Microglia and peripheral macrophages have both been implicated in amyotrophic lateral sclerosis (ALS), although their respective roles have yet to be determined. We now show that macrophages along peripheral motor neuron axons in mouse models and patients with ALS react to neurodegeneration. In ALS mice, peripheral myeloid cell infiltration into the spinal cord was limited and depended on disease duration. Targeted gene modulation of the reactive oxygen species pathway in peripheral myeloid cells of ALS mice, using cell replacement, reduced both peripheral macrophage and microglial activation, delayed symptoms and increased survival. Transcriptomics revealed that sciatic nerve macrophages and microglia reacted differently to neurodegeneration, with abrupt temporal changes in macrophages and progressive, unidirectional activation in microglia. Modifying peripheral macrophages suppressed proinflammatory microglial responses, with a shift toward neuronal support. Thus, modifying macrophages at the periphery has the capacity to influence disease progression and may be of therapeutic value for ALS.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Axônios/imunologia , Macrófagos/imunologia , Microglia/imunologia , Neurônios Motores/imunologia , Nervo Isquiático/imunologia , Adulto , Idoso , Esclerose Lateral Amiotrófica/metabolismo , Animais , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Pessoa de Meia-Idade , Neurônios Motores/metabolismo , Nervo Isquiático/metabolismo , Medula Espinal/imunologia , Medula Espinal/metabolismo
9.
J Biol Chem ; 294(37): 13755-13768, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31346033

RESUMO

Protection of neuronal homeostasis is a major goal in the management of neurodegenerative diseases. Microtubule-associated Ser/Thr kinase 2 (MAST2) inhibits neurite outgrowth, and its inhibition therefore represents a potential therapeutic strategy. We previously reported that a viral protein (G-protein from rabies virus) capable of interfering with protein-protein interactions between the PDZ domain of MAST2 and the C-terminal moieties of its cellular partners counteracts MAST2-mediated suppression of neurite outgrowth. Here, we designed peptides derived from the native viral protein to increase the affinity of these peptides for the MAST2-PDZ domain. Our strategy involved modifying the length and flexibility of the noninteracting sequence linking the two subsites anchoring the peptide to the PDZ domain. Three peptides, Neurovita1 (NV1), NV2, and NV3, were selected, and we found that they all had increased affinities for the MAST2-PDZ domain, with Kd values decreasing from 1300 to 60 nm, while target selectivity was maintained. A parallel biological assay evaluating neurite extension and branching in cell cultures revealed that the NV peptides gradually improved neural activity, with the efficacies of these peptides for stimulating neurite outgrowth mirroring their affinities for MAST2-PDZ. We also show that NVs can be delivered into the cytoplasm of neurons as a gene or peptide. In summary, our findings indicate that virus-derived peptides targeted to MAST2-PDZ stimulate neurite outgrowth in several neuron types, opening up promising avenues for potentially using NVs in the management of neurodegenerative diseases.


Assuntos
Neuritos/metabolismo , Crescimento Neuronal/efeitos dos fármacos , Domínios PDZ/fisiologia , Estimulantes do Sistema Nervoso Central/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Microtúbulos/metabolismo , Neurônios/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacologia , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Vírus da Raiva , Relação Estrutura-Atividade , Proteínas Virais/metabolismo , Proteínas Virais/farmacologia
10.
Acta Neuropathol ; 138(1): 123-145, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30874923

RESUMO

Recently, we provided genetic basis showing that mitochondrial dysfunction can trigger motor neuron degeneration, through identification of CHCHD10 encoding a mitochondrial protein. We reported patients, carrying the p.Ser59Leu heterozygous mutation in CHCHD10, from a large family with a mitochondrial myopathy associated with motor neuron disease (MND). Rapidly, our group and others reported CHCHD10 mutations in amyotrophic lateral sclerosis (ALS), frontotemporal dementia-ALS and other neurodegenerative diseases. Here, we generated knock-in (KI) mice, carrying the p.Ser59Leu mutation, that mimic the mitochondrial myopathy with mtDNA instability displayed by the patients from our original family. Before 14 months of age, all KI mice developed a fatal mitochondrial cardiomyopathy associated with enhanced mitophagy. CHCHD10S59L/+ mice also displayed neuromuscular junction (NMJ) and motor neuron degeneration with hyper-fragmentation of the motor end plate and moderate but significant motor neuron loss in lumbar spinal cord at the end stage of the disease. At this stage, we observed TDP-43 cytoplasmic aggregates in spinal neurons. We also showed that motor neurons differentiated from human iPSC carrying the p.Ser59Leu mutation were much more sensitive to Staurosporine or glutamate-induced caspase activation than control cells. These data confirm that mitochondrial deficiency associated with CHCHD10 mutations can be at the origin of MND. CHCHD10 is highly expressed in the NMJ post-synaptic part. Importantly, the fragmentation of the motor end plate was associated with abnormal CHCHD10 expression that was also observed closed to NMJs which were morphologically normal. Furthermore, we found OXPHOS deficiency in muscle of CHCHD10S59L/+ mice at 3 months of age in the absence of neuron loss in spinal cord. Our data show that the pathological effects of the p.Ser59Leu mutation target muscle prior to NMJ and motor neurons. They likely lead to OXPHOS deficiency, loss of cristae junctions and destabilization of internal membrane structure within mitochondria at motor end plate of NMJ, impairing neurotransmission. These data are in favor with a key role for muscle in MND associated with CHCHD10 mutations.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Demência Frontotemporal/metabolismo , Mitocôndrias/patologia , Neurônios Motores/metabolismo , Junção Neuromuscular/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Morte Celular/genética , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/genética , Camundongos Transgênicos , Proteínas Mitocondriais/metabolismo , Degeneração Neural/genética , Degeneração Neural/patologia , Fenótipo
11.
Nat Commun ; 8(1): 861, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29021520

RESUMO

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder due to selective loss of motor neurons (MNs). Mutations in the fused in sarcoma (FUS) gene can cause both juvenile and late onset ALS. We generated and characterized induced pluripotent stem cells (iPSCs) from ALS patients with different FUS mutations, as well as from healthy controls. Patient-derived MNs show typical cytoplasmic FUS pathology, hypoexcitability, as well as progressive axonal transport defects. Axonal transport defects are rescued by CRISPR/Cas9-mediated genetic correction of the FUS mutation in patient-derived iPSCs. Moreover, these defects are reproduced by expressing mutant FUS in human embryonic stem cells (hESCs), whereas knockdown of endogenous FUS has no effect, confirming that these pathological changes are mutant FUS dependent. Pharmacological inhibition as well as genetic silencing of histone deacetylase 6 (HDAC6) increase α-tubulin acetylation, endoplasmic reticulum (ER)-mitochondrial overlay, and restore the axonal transport defects in patient-derived MNs.Amyotrophic lateral sclerosis (ALS) leads to selective loss of motor neurons. Using motor neurons derived from induced pluripotent stem cells from patients with ALS and FUS mutations, the authors demonstrate that axonal transport deficits that are observed in these cells can be rescued by HDAC6 inhibition.


Assuntos
Esclerose Lateral Amiotrófica/genética , Transporte Axonal , Desacetilase 6 de Histona/metabolismo , Neurônios Motores/metabolismo , Proteína FUS de Ligação a RNA/genética , Adolescente , Adulto , Idoso , Sistemas CRISPR-Cas , Feminino , Desacetilase 6 de Histona/antagonistas & inibidores , Humanos , Ácidos Hidroxâmicos , Indóis , Células-Tronco Pluripotentes Induzidas , Masculino , Mutação Puntual , Cultura Primária de Células , Pirimidinas
12.
Neurobiol Aging ; 58: 239.e11-239.e20, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28716533

RESUMO

Mutations in UBQLN2 have been associated with rare cases of X-linked juvenile and adult forms of amyotrophic lateral sclerosis (ALS) and ALS linked to frontotemporal dementia (FTD). Here, we report 1 known (c.1489C>T, p.Pro497Ser, P497S) and 3 novel (c.1481C>T, p.Pro494Leu, P494L; c.1498C>T, p.Pro500Ser, P500S; and c.1516C>G, p.Pro506Ala, P506A) missense mutations in the PXX domain of UBQLN2 in familial motor neuron diseases including ALS and spastic paraplegia (SP). A novel missense mutation (c.1462G>A, p.Ala488Thr, A488T) adjacent to this hotspot UBQLN2 domain was identified in a sporadic case of ALS. These mutations are conserved in mammals, are absent from ExAC and gnomAD browsers, and are predicted to be deleterious by SIFT in silico analysis. Patient lymphoblasts carrying a UBQLN2 mutation showed absence of ubiquilin-2 accumulation, disrupted binding with HSP70, and impaired autophagic pathway. Our results confirm the role of PXX repeat in ALS pathogenesis, show that UBQLN2-linked disease can manifest like a SP phenotype, evidence a highly reduced disease penetrance in females carrying UBQLN2 mutations, which is important information for genetic counseling, and underline the pivotal role of ubiquilin-2 in proteolysis regulation pathways.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ciclo Celular/genética , Demência Frontotemporal/genética , Estudos de Associação Genética , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Mutação de Sentido Incorreto/genética , Fenótipo , Proteólise , Paraplegia Espástica Hereditária/genética , Ubiquitinas/genética , Proteínas Adaptadoras de Transdução de Sinal , Idoso , Idoso de 80 Anos ou mais , Proteínas Relacionadas à Autofagia , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Dimerização , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Domínios Proteicos/genética , Ubiquitinas/química , Ubiquitinas/metabolismo , Inativação do Cromossomo X
13.
Proc Natl Acad Sci U S A ; 114(18): E3679-E3688, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28416701

RESUMO

α-Synuclein (αSyn) is the major gene linked to sporadic Parkinson's disease (PD), whereas the G209A (p.A53T) αSyn mutation causes a familial form of PD characterized by early onset and a generally severe phenotype, including nonmotor manifestations. Here we generated de novo induced pluripotent stem cells (iPSCs) from patients harboring the p.A53T mutation and developed a robust model that captures PD pathogenic processes under basal conditions. iPSC-derived mutant neurons displayed novel disease-relevant phenotypes, including protein aggregation, compromised neuritic outgrowth, and contorted or fragmented axons with swollen varicosities containing αSyn and Tau. The identified neuropathological features closely resembled those in brains of p.A53T patients. Small molecules targeting αSyn reverted the degenerative phenotype under both basal and induced stress conditions, indicating a treatment strategy for PD and other synucleinopathies. Furthermore, mutant neurons showed disrupted synaptic connectivity and widespread transcriptional alterations in genes involved in synaptic signaling, a number of which have been previously linked to mental disorders, raising intriguing implications for potentially converging disease mechanisms.


Assuntos
Axônios/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Mutação de Sentido Incorreto , Doença de Parkinson/metabolismo , Polineuropatias/metabolismo , Transmissão Sináptica , alfa-Sinucleína/metabolismo , Substituição de Aminoácidos , Axônios/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Polineuropatias/genética , Polineuropatias/patologia , alfa-Sinucleína/genética
14.
FASEB J ; 31(2): 828-839, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27856558

RESUMO

Tobacco smoking is a public health problem, with ∼5 million deaths per year, representing a heavy burden for many countries. No effective therapeutic strategies are currently available for nicotine addiction, and it is therefore crucial to understand the etiological and pathophysiological factors contributing to this addiction. The neuronal α5 nicotinic acetylcholine receptor (nAChR) subunit is critically involved in nicotine dependence. In particular, the human polymorphism α5D398N corresponds to the strongest correlation with nicotine dependence risk found to date in occidental populations, according to meta-analysis of genome-wide association studies. To understand the specific contribution of this subunit in the context of nicotine addiction, an efficient screening system for native human nAChRs is needed. We have differentiated human induced pluripotent stem (iPS) cells into midbrain dopaminergic (DA) neurons and obtained a comprehensive characterization of these neurons by quantitative RT-PCR. The functional properties of nAChRs expressed in these human DA neurons, with or without the polymorphism in the α5 subunit, were studied with the patch-clamp electrophysiological technique. Our results in human DA neurons carrying the polymorphism in the α5 subunit showed an increase in EC50, indicating that, in the presence of the polymorphism, more nicotine or acetylcholine chloride is necessary to obtain the same effect. This human cell culturing system can now be used in drug discovery approaches to screen for compounds that interact specifically with human native and polymorphic nAChRs.-Deflorio, C., Blanchard, S., Carisì, M. C., Bohl, D., Maskos, U. Human polymorphisms in nicotinic receptors: a functional analysis in iPS-derived dopaminergic neurons.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptores Nicotínicos/metabolismo , Encéfalo/citologia , Linhagem Celular , Humanos , Polimorfismo Genético , Receptores Nicotínicos/genética
15.
Biol Aujourdhui ; 210(1): 27-36, 2016.
Artigo em Francês | MEDLINE | ID: mdl-27286578

RESUMO

Among motor neuron diseases, spinal muscular atrophy type 1 and amyotrophic lateral sclerosis are very aggressive diseases with no cure. With the breakthrough of human induced pluripotent stem cells, iPS, researchers have now at their disposal a powerful tool to generate human motor neurons in culture and study the pathological defects in patient cells. In this review, we will see which tools for the study of patients motoneurons were developed from iPS cells and the different cellular models that were generated. We will also see how these models were validated and current research to identify new therapeutic leads.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/fisiologia , Doença dos Neurônios Motores/patologia , Neurônios Motores/fisiologia , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Humanos , Modelos Biológicos , Doença dos Neurônios Motores/terapia , Atrofia Muscular Espinal/patologia , Atrofia Muscular Espinal/terapia , Células-Tronco Neurais/fisiologia , Neuroglia/fisiologia
16.
Hum Mol Genet ; 25(14): 2972-2984, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27206984

RESUMO

Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.


Assuntos
Retículo Endoplasmático/metabolismo , GTP Fosfo-Hidrolases/genética , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , Animais , Sinalização do Cálcio/genética , Citosol/metabolismo , Retículo Endoplasmático/patologia , Fibroblastos , GTP Fosfo-Hidrolases/biossíntese , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Mitofagia/genética , Mutação , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
17.
Curr Stem Cell Res Ther ; 11(4): 301-12, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26018231

RESUMO

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease affecting primarily the population of motor neurons, even though a non-cell autonomous component, involving neighbouring non-neuronal cells, is more and more described. Despite 140 years of disease experience, still no efficient treatment exists against ALS. The inability to readily obtain the faulty cell types relevant to ALS has impeded progress in drug discovery for decades. However, the pioneer work of Shinya Yamanaka in 2007 in the stem cell field was a real breakthrough. Recent advances in cell reprogramming now grant access to significant quantities of CNS disease-affected cells. Induced pluripotent stem cells (iPSc) have been recently derived from patients carrying mutations linked to familial forms of ALS as well as from sporadic patients. Precise and mature protocols allow now their differentiation into ALS-relevant cell subtypes; sustainable and renewable sources of human motor neurons or glia are being available for ALS disease modelling, drug screening or for the development of cell therapies. In few years, the proof-of-concept was made that ALS disease-related phenotypes can be reproduced with iPSc and despite some remaining challenges, we are now not so far to provide platforms for the investigation of ALS therapeutics. This paper also reviews the pioneering studies regarding the applicability of iPSc technology in ALS animal models. From modest slowing down of ALS progression to no severe adverse effects, iPSc-based cell therapy resulted in promising premises in ALS preclinical paradigms, although long-term surveys are highly recommended.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco , Esclerose Lateral Amiotrófica/patologia , Animais , Diferenciação Celular/genética , Modelos Animais de Doenças , Humanos , Neurônios Motores/transplante
18.
Neurobiol Dis ; 82: 269-280, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26107889

RESUMO

Amyotrophic lateral sclerosis (ALS) is a severe and incurable neurodegenerative disease. Human motor neurons generated from induced pluripotent stem cells (iPSc) offer new perspectives for disease modeling and drug testing in ALS. In standard iPSc-derived cultures, however, the two major phenotypic alterations of ALS--degeneration of motor neuron cell bodies and axons--are often obscured by cell body clustering, extensive axon criss-crossing and presence of unwanted cell types. Here, we succeeded in isolating 100% pure and standardized human motor neurons by a novel FACS double selection based on a p75(NTR) surface epitope and an HB9::RFP lentivirus reporter. The p75(NTR)/HB9::RFP motor neurons survive and grow well without forming clusters or entangled axons, are electrically excitable, contain ALS-relevant motor neuron subtypes and form functional connections with co-cultured myotubes. Importantly, they undergo rapid and massive cell death and axon degeneration in response to mutant SOD1 astrocytes. These data demonstrate the potential of FACS-isolated human iPSc-derived motor neurons for improved disease modeling and drug testing in ALS and related motor neuron diseases.


Assuntos
Esclerose Lateral Amiotrófica , Citometria de Fluxo/métodos , Células-Tronco Pluripotentes Induzidas , Neurônios Motores , Adulto , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Astrócitos/patologia , Astrócitos/fisiologia , Axônios/patologia , Axônios/fisiologia , Sobrevivência Celular , Células Cultivadas , Criança , Técnicas de Cocultura , Genes Reporter , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Lentivirus , Neurônios Motores/patologia , Neurônios Motores/fisiologia , Mutação , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
19.
J Mol Biol ; 427(4): 775-791, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25268803

RESUMO

Mucopolysaccharidoses type III (MPSIII, Sanfilippo syndrome) are genetic diseases due to deficient heparan sulfate (HS) saccharide digestion by lysosomal exoglycanases. Progressive accumulation of undigested saccharides causes early-onset behavioural and cognitive symptoms. The precise role of these saccharides in the pathophysiological cascade is still unclear. We showed that exposure of wild-type neural cells to exogenous soluble HS fragments of at least eight saccharides activated integrin-based focal adhesions (FAs), which attach cells to the extracellular matrix. FAs were constitutively activated in MPSIII type B astrocytes or neural stem cells unless undigested saccharides were cleared by exogenous supply of the missing exoglycanase. Defective cell polarisation and oriented migration in response to focal extracellular stimuli in affected cells suggest improper sensing of the environment. We consistently observed abnormal organisation of the rostral migratory stream in the brain of adult mice with MPSIII type B. These results suggest that cell polarisation and oriented migration defects participate to the neurological disorders associated with Sanfilippo syndrome.


Assuntos
Astrócitos/metabolismo , Adesões Focais/metabolismo , Heparitina Sulfato/farmacologia , Mucopolissacaridose III/patologia , Células-Tronco Neurais/metabolismo , Animais , Astrócitos/citologia , Encéfalo/patologia , Movimento Celular/genética , Polaridade Celular/genética , Células Cultivadas , Ativação Enzimática , Quinase 1 de Adesão Focal/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/citologia , Paxilina/biossíntese , Paxilina/genética , Fosforilação , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Suínos
20.
Regen Med ; 9(4): 437-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25159062

RESUMO

AIM: Biomarker-based tracking of human stem cells xenotransplanted into animal models is crucial for studying their fate in the field of cell therapy or tumor xenografting. MATERIALS & METHODS: Using immunohistochemistry and in situ hybridization, we analyzed the expression of three human-specific biomarkers: Ku80, human mitochondria (hMito) and Alu. RESULTS: We showed that Ku80, hMito and Alu biomarkers are broadly expressed in human tissues with no or low cross-reactivity toward rat, mouse or pig tissues. In vitro, we demonstrated that their expression is stable over time and does not change along the differentiation of human-derived induced pluripotent stem cells or human glial-restricted precursors. We tracked in vivo these cell populations after transplantation in rodent spinal cords using aforementioned biomarkers and human-specific antibodies detecting apoptotic, proliferating or neural-committed cells. CONCLUSION: This study assesses the human-species specificity of Ku80, hMito and Alu, and proposes useful biomarkers for characterizing human stem cells in xenotransplantation paradigms.


Assuntos
Antígenos de Diferenciação/metabolismo , Rastreamento de Células/métodos , Células-Tronco Pluripotentes Induzidas , Transplante de Células-Tronco , Animais , Xenoenxertos , Humanos , Imuno-Histoquímica/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...