Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Stem Cells ; 34(6): 1563-75, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26946488

RESUMO

Despite decades of research on amyotrophic lateral sclerosis (ALS), there is only one approved drug, which minimally extends patient survival. Here, we investigated pathophysiological mechanisms underlying ALS using motor neurons (MNs) differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying mutations in FUS or SOD1. Patient-derived MNs were less active and excitable compared to healthy controls, due to reduced Na(+) /K(+) ratios in both ALS groups accompanied by elevated potassium channel (FUS) and attenuated sodium channel expression levels (FUS, SOD1). ALS iPSC-derived MNs showed elevated endoplasmic reticulum stress (ER) levels and increased caspase activation. Treatment with the FDA approved drug 4-Aminopyridine (4AP) restored ion-channel imbalances, increased neuronal activity levels and decreased ER stress and caspase activation. This study provides novel pathophysiological data, including a mechanistic explanation for the observed hypoexcitability in patient-derived MNs and a new therapeutic strategy to provide neuroprotection in MNs affected by ALS. Stem Cells 2016;34:1563-1575.


Assuntos
4-Aminopiridina/farmacologia , Esclerose Lateral Amiotrófica/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios Motores/patologia , Esclerose Lateral Amiotrófica/genética , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Canais Iônicos/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação/genética , Neuroproteção/efeitos dos fármacos , Fenótipo , Proteína FUS de Ligação a RNA/genética , Superóxido Dismutase/genética , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
2.
Hum Mol Genet ; 23(8): 2005-22, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24271013

RESUMO

Neuronal ceroid lipofuscinosis (NCL) comprises ∼13 genetically distinct lysosomal disorders primarily affecting the central nervous system. Here we report successful reprograming of patient fibroblasts into induced pluripotent stem cells (iPSCs) for the two most common NCL subtypes: classic late-infantile NCL, caused by TPP1(CLN2) mutation, and juvenile NCL, caused by CLN3 mutation. CLN2/TPP1- and CLN3-iPSCs displayed overlapping but distinct biochemical and morphological abnormalities within the endosomal-lysosomal system. In neuronal derivatives, further abnormalities were observed in mitochondria, Golgi and endoplasmic reticulum. While lysosomal storage was undetectable in iPSCs, progressive disease subtype-specific storage material was evident upon neural differentiation and was rescued by reintroducing the non-mutated NCL proteins. In proof-of-concept studies, we further documented differential effects of potential small molecule TPP1 activity inducers. Fenofibrate and gemfibrozil, previously reported to induce TPP1 activity in control cells, failed to increase TPP1 activity in patient iPSC-derived neural progenitor cells. Conversely, nonsense suppression by PTC124 resulted in both an increase of TPP1 activity and attenuation of neuropathology in patient iPSC-derived neural progenitor cells. This study therefore documents the high value of this powerful new set of tools for improved drug screening and for investigating early mechanisms driving NCL pathogenesis.


Assuntos
Aminopeptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteínas de Membrana/genética , Modelos Neurológicos , Chaperonas Moleculares/genética , Mutação/genética , Lipofuscinoses Ceroides Neuronais/genética , Serina Proteases/genética , Aminopeptidases/metabolismo , Western Blotting , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Eletrofisiologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Fenofibrato/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Genfibrozila/farmacologia , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Técnicas Imunoenzimáticas , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Lipofuscinoses Ceroides Neuronais/metabolismo , Lipofuscinoses Ceroides Neuronais/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Serina Proteases/metabolismo , Tripeptidil-Peptidase 1
3.
Neurobiol Dis ; 82: 420-429, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26253605

RESUMO

Autosomal-dominant mutations within the gene FUS (fused in sarcoma) are responsible for 5% of familial cases of amyotrophic lateral sclerosis (ALS). The FUS protein is physiologically mainly located in the nucleus, while cytoplasmic FUS aggregates are pathological hallmarks of FUS-ALS. Data from non-neuronal cell models and/or models using heterologous expression of FUS mutants suggest cytoplasmic FUS translocation as a pivotal initial event which leads to neurodegeneration depending on a second hit. Here we present the first human model of FUS-ALS using patient-derived neurons carrying endogenous FUS mutations leading to a benign (R521C) or a more severe clinical phenotype (frameshift mutation R495QfsX527). We thereby showed that the severity of the underlying FUS mutation determines the amount of cytoplasmic FUS accumulation and cellular vulnerability to exogenous stress. Cytoplasmic FUS inclusions formed spontaneously depending on both, severity of FUS mutation and neuronal aging. These aggregates showed typical characteristics of FUS-ALS including methylated FUS. Finally, neurodegeneration was not specific to layer V cortical neurons perfectly in line with the current model of disease spreading in ALS. Our study highlights the value and usefulness of patient-derived cell models in FUS-ALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios/patologia , Proteína FUS de Ligação a RNA/genética , Adulto , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Progressão da Doença , Feminino , Humanos , Corpos de Inclusão/patologia , Corpos de Inclusão/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , Pessoa de Meia-Idade , Neurônios Motores/patologia , Neurônios Motores/fisiologia , Mutação , Neurônios/fisiologia , Fenótipo , Proteína FUS de Ligação a RNA/metabolismo , Índice de Gravidade de Doença , Medula Espinal/patologia , Medula Espinal/fisiopatologia
4.
Nat Commun ; 9(1): 335, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29362359

RESUMO

Amyotrophic lateral sclerosis (ALS) is the most frequent motor neuron disease. Cytoplasmic fused in sarcoma (FUS) aggregates are pathological hallmarks of FUS-ALS. Proper shuttling between the nucleus and cytoplasm is essential for physiological cell function. However, the initial event in the pathophysiology of FUS-ALS remains enigmatic. Using human induced pluripotent stem cell (hiPSCs)-derived motor neurons (MNs), we show that impairment of poly(ADP-ribose) polymerase (PARP)-dependent DNA damage response (DDR) signaling due to mutations in the FUS nuclear localization sequence (NLS) induces additional cytoplasmic FUS mislocalization which in turn results in neurodegeneration and FUS aggregate formation. Our work suggests that a key pathophysiologic event in ALS is upstream of aggregate formation. Targeting DDR signaling could lead to novel therapeutic routes for ameliorating ALS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Dano ao DNA , Neurônios Motores/metabolismo , Mutação , Agregação Patológica de Proteínas/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Transporte Ativo do Núcleo Celular/genética , Idoso , Idoso de 80 Anos ou mais , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Diferenciação Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Feminino , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Pessoa de Meia-Idade , Neurônios Motores/patologia , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Proteína FUS de Ligação a RNA/genética , Transdução de Sinais
5.
Nat Neurosci ; 19(12): 1610-1618, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27723745

RESUMO

Intronic hexanucleotide expansions in C9ORF72 are common in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, but it is unknown whether loss of function, toxicity by the expanded RNA or dipeptides from non-ATG-initiated translation are responsible for the pathophysiology. We determined the interactome of C9ORF72 in motor neurons and found that C9ORF72 was present in a complex with cofilin and other actin binding proteins. Phosphorylation of cofilin was enhanced in C9ORF72-depleted motor neurons, in patient-derived lymphoblastoid cells, induced pluripotent stem cell-derived motor neurons and post-mortem brain samples from ALS patients. C9ORF72 modulates the activity of the small GTPases Arf6 and Rac1, resulting in enhanced activity of LIM-kinases 1 and 2 (LIMK1/2). This results in reduced axonal actin dynamics in C9ORF72-depleted motor neurons. Dominant negative Arf6 rescues this defect, suggesting that C9ORF72 acts as a modulator of small GTPases in a pathway that regulates axonal actin dynamics.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/metabolismo , Proteínas/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Encéfalo/metabolismo , Proteína C9orf72 , Expansão das Repetições de DNA/genética , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo
6.
Stem Cells Transl Med ; 4(10): 1223-33, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26304036

RESUMO

UNLABELLED: Brain perivascular cells have recently been identified as a novel mesodermal cell type in the human brain. These cells reside in the perivascular niche and were shown to have mesodermal and, to a lesser extent, tissue-specific differentiation potential. Mesenchymal stem cells (MSCs) are widely proposed for use in cell therapy in many neurological disorders; therefore, it is of importance to better understand the "intrinsic" MSC population of the human brain. We systematically characterized adult human brain-derived pericytes during in vitro expansion and differentiation and compared these cells with fetal and adult human brain-derived neural stem cells (NSCs) and adult human bone marrow-derived MSCs. We found that adult human brain pericytes, which can be isolated from the hippocampus and from subcortical white matter, are-in contrast to adult human NSCs-easily expandable in monolayer cultures and show many similarities to human bone marrow-derived MSCs both regarding both surface marker expression and after whole transcriptome profile. Human brain pericytes showed a negligible propensity for neuroectodermal differentiation under various differentiation conditions but efficiently generated mesodermal progeny. Consequently, human brain pericytes resemble bone marrow-derived MSCs and might be very interesting for possible autologous and endogenous stem cell-based treatment strategies and cell therapeutic approaches for treating neurological diseases. SIGNIFICANCE: Perivascular mesenchymal stem cells (MSCs) recently gained significant interest because of their appearance in many tissues including the human brain. MSCs were often reported as being beneficial after transplantation in the central nervous system in different neurological diseases; therefore, adult brain perivascular cells derived from human neural tissue were systematically characterized concerning neural stem cell and MSC marker expression, transcriptomics, and mesodermal and inherent neuroectodermal differentiation potential in vitro and in vivo after in utero transplantation. This study showed the lack of an innate neuronal but high mesodermal differentiation potential. Because of their relationship to mesenchymal stem cells, these adult brain perivascular mesodermal cells are of great interest for possible autologous therapeutic use.


Assuntos
Células-Tronco Mesenquimais/citologia , Adulto , Animais , Encéfalo/embriologia , Diferenciação Celular , Linhagem da Célula , Criança , Feminino , Células-Tronco Fetais/citologia , Perfilação da Expressão Gênica , Sobrevivência de Enxerto , Xenoenxertos , Hipocampo/citologia , Humanos , Masculino , Mesoderma , Camundongos , Pessoa de Meia-Idade , Placa Neural , Especificidade de Órgãos , Pericitos/citologia , Transcriptoma , Substância Branca/citologia , Adulto Jovem
7.
Stem Cells Transl Med ; 3(4): 458-69, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24558163

RESUMO

Adult neural progenitor cells (aNPC) are a potential autologous cell source for cell replacement in neurologic diseases or for cell-based gene therapy of neurometabolic diseases. Easy accessibility, long-term expandability, and detailed characterization of neural progenitor cell (NPC) properties are important requisites for their future translational/clinical applications. aNPC can be isolated from different regions of the adult human brain, including the accessible subcortical white matter (aNPCWM), but systematic studies comparing long-term expanded aNPCWM with aNPC from neurogenic brain regions are not available. Freshly isolated cells from subcortical white matter and hippocampus expressed oligodendrocyte progenitor cell markers such as A2B5, neuron-glial antigen 2 (NG2), and oligodendrocyte transcription factor 2 (OLIG2) in ∼20% of cells but no neural stem cell (NSC) markers such as CD133 (Prominin1), Nestin, SOX2, or PAX6. The epidermal growth factor receptor protein was expressed in 18% of aNPCWM and 7% of hippocampal aNPC (aNPCHIP), but only a small fraction of cells, 1 of 694 cells from white matter and 1 of 1,331 hippocampal cells, was able to generate neurospheres. Studies comparing subcortical aNPCWM with their hippocampal counterparts showed that both NPC types expressed mainly markers of glial origin such as NG2, A2B5, and OLIG2, and the NSC/NPC marker Nestin, but no pericyte markers. Both NPC types were able to produce neurons, astrocytes, and oligodendrocytes in amounts comparable to fetal NSC. Whole transcriptome analyses confirmed the strong similarity of aNPCWM to aNPCHIP. Our data show that aNPCWM are multipotent NPC with long-term expandability similar to NPC from hippocampus, making them a more easily accessible source for possible autologous NPC-based treatment strategies.


Assuntos
Células-Tronco Adultas/metabolismo , Antígenos de Diferenciação/biossíntese , Hipocampo/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Células-Tronco Pluripotentes/metabolismo , Adulto , Células-Tronco Adultas/citologia , Células Cultivadas , Feminino , Hipocampo/citologia , Humanos , Masculino , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Células-Tronco Pluripotentes/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA