Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(10): e0069623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796129

RESUMO

IMPORTANCE: Human cytomegalovirus (HCMV) infection is the leading cause of non-heritable birth defects worldwide. HCMV readily infects the early progenitor cell population of the developing brain, and we have found that infection leads to significantly downregulated expression of key neurodevelopmental transcripts. Currently, there are no approved therapies to prevent or mitigate the effects of congenital HCMV infection. Therefore, we used human-induced pluripotent stem cell-derived organoids and neural progenitor cells to elucidate the glycoproteins and receptors used in the viral entry process and whether antibody neutralization was sufficient to block viral entry and prevent disruption of neurodevelopmental gene expression. We found that blocking viral entry alone was insufficient to maintain the expression of key neurodevelopmental genes, but neutralization combined with neurotrophic factor treatment provided robust protection. Together, these studies offer novel insight into mechanisms of HCMV infection in neural tissues, which may aid future therapeutic development.


Assuntos
Anticorpos Neutralizantes , Infecções por Citomegalovirus , Citomegalovirus , Expressão Gênica , Fatores de Crescimento Neural , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Organoides/citologia , Organoides/metabolismo , Organoides/virologia , Receptores Virais/antagonistas & inibidores , Receptores Virais/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos
2.
Hum Mol Genet ; 28(19): 3199-3210, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211843

RESUMO

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by deletions or mutations in survival motor neuron 1 (SMN1). The molecular mechanisms underlying motor neuron degeneration in SMA remain elusive, as global cellular dysfunction obscures the identification and characterization of disease-relevant pathways and potential therapeutic targets. Recent reports have implicated microRNA (miRNA) dysregulation as a potential contributor to the pathological mechanism in SMA. To characterize miRNAs that are differentially regulated in SMA, we profiled miRNA levels in SMA induced pluripotent stem cell (iPSC)-derived motor neurons. From this array, miR-23a downregulation was identified selectively in SMA motor neurons, consistent with previous reports where miR-23a functioned in neuroprotective and muscle atrophy-antagonizing roles. Reintroduction of miR-23a expression in SMA patient iPSC-derived motor neurons protected against degeneration, suggesting a potential miR-23a-specific disease-modifying effect. To assess this activity in vivo, miR-23a was expressed using a self-complementary adeno-associated virus serotype 9 (scAAV9) viral vector in the Smn2B/- SMA mouse model. scAAV9-miR-23a significantly reduced the pathology in SMA mice, including increased motor neuron size, reduced neuromuscular junction pathology, increased muscle fiber area, and extended survival. These experiments demonstrate that miR-23a is a novel protective modifier of SMA, warranting further characterization of miRNA dysfunction in SMA.


Assuntos
Vetores Genéticos/administração & dosagem , MicroRNAs/genética , Atrofia Muscular Espinal/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , MicroRNAs/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Índice de Gravidade de Doença , Proteína 2 de Sobrevivência do Neurônio Motor/genética
3.
Mol Cell ; 71(2): 216-228.e7, 2018 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-30029002

RESUMO

The polyglutamine (polyQ) diseases are a group of nine neurodegenerative diseases caused by the expansion of a polyQ tract that results in protein aggregation. Unlike other model organisms, Dictyostelium discoideum is a proteostatic outlier, naturally encoding long polyQ tracts yet resistant to polyQ aggregation. Here we identify serine-rich chaperone protein 1 (SRCP1) as a molecular chaperone that is necessary and sufficient to suppress polyQ aggregation. SRCP1 inhibits aggregation of polyQ-expanded proteins, allowing for their degradation via the proteasome, where SRCP1 is also degraded. SRCP1's C-terminal domain is essential for its activity in cells, and peptides that mimic this domain suppress polyQ aggregation in vitro. Together our results identify a novel type of molecular chaperone and reveal how nature has dealt with the problem of polyQ aggregation.


Assuntos
Chaperonas Moleculares/metabolismo , Peptídeos/metabolismo , Linhagem Celular , Dictyostelium/metabolismo , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Serina/metabolismo , Ubiquitina/metabolismo
4.
J Cardiovasc Pharmacol Ther ; 21(6): 549-562, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26940570

RESUMO

BACKGROUND: Dystrophin-deficient cardiomyopathy is a growing clinical problem without targeted treatments. We investigated whether nicorandil promotes cardioprotection in human dystrophin-deficient induced pluripotent stem cell (iPSC)-derived cardiomyocytes and the muscular dystrophy mdx mouse heart. METHODS AND RESULTS: Dystrophin-deficient iPSC-derived cardiomyocytes had decreased levels of endothelial nitric oxide synthase and neuronal nitric oxide synthase. The dystrophin-deficient cardiomyocytes had increased cell injury and death after 2 hours of stress and recovery. This was associated with increased levels of reactive oxygen species and dissipation of the mitochondrial membrane potential. Nicorandil pretreatment was able to abolish these stress-induced changes through a mechanism that involved the nitric oxide-cyclic guanosine monophosphate pathway and mitochondrial adenosine triphosphate-sensitive potassium channels. The increased reactive oxygen species levels in the dystrophin-deficient cardiomyocytes were associated with diminished expression of select antioxidant genes and increased activity of xanthine oxidase. Furthermore, nicorandil was found to improve the restoration of cardiac function after ischemia and reperfusion in the isolated mdx mouse heart. CONCLUSION: Nicorandil protects against stress-induced cell death in dystrophin-deficient cardiomyocytes and preserves cardiac function in the mdx mouse heart subjected to ischemia and reperfusion injury. This suggests a potential therapeutic role for nicorandil in dystrophin-deficient cardiomyopathy.


Assuntos
Cardiomiopatias/prevenção & controle , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Canais KATP/agonistas , Distrofia Muscular Animal/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Nicorandil/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Animais , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Canais KATP/metabolismo , Masculino , Camundongos Endogâmicos mdx , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Nicorandil/metabolismo , Doadores de Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Xantina Oxidase/metabolismo
5.
Hum Mol Genet ; 25(3): 514-23, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26643950

RESUMO

Spinal muscular atrophy (SMA) is a genetic disorder characterized by loss of motor neurons in the spinal cord leading to muscle atrophy and death. Although motor neurons (MNs) are the most obviously affected cells in SMA, recent evidence suggest dysfunction in multiple cell types. Astrocytes are a crucial component of the motor circuit and are intimately involved with MN health and maintenance. We have previously shown that SMA astrocytes are altered both morphologically and functionally early in disease progression, though it is unclear what causes astrocytes to become reactive. Oxidative stress is a common feature among neurodegenerative diseases. Oxidative stress can both induce apoptosis in neurons and can cause astrocytes to become reactive, which are features observed in the SMA induced pluripotent stem cell (iPSC) cultures. Therefore, we asked if oxidative stress contributes to SMA astrocyte pathology. We examined mitochondrial bioenergetics, transcript and protein levels of oxidative and anti-oxidant factors, and reactive oxygen species (ROS) production and found little evidence of oxidative stress. We did observe a significant increase in endogenous catalase expression in SMA iPSCs. While catalase knockdown in SMA iPSCs increased ROS production above basal levels, levels of ROS remained lower than in controls, further arguing against robust oxidative stress in this system. Viral delivery of survival motor neuron (SMN) reversed astrocyte activation and restored catalase levels to normal, without changing mitochondrial respiration or expression of oxidative stress markers. Taken together, these data indicate that SMN deficiency induces astrocyte reactivity, but does not do so through an oxidative stress-mediated process.


Assuntos
Astrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , Células-Tronco Neurais/metabolismo , Astrócitos/patologia , Catalase/antagonistas & inibidores , Catalase/genética , Catalase/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios Motores/patologia , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patologia , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Células-Tronco Neurais/patologia , Fosforilação Oxidativa , Estresse Oxidativo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Glutationa Peroxidase GPX1
6.
Hum Mol Genet ; 24(14): 4094-102, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25911676

RESUMO

Systemically low levels of survival motor neuron-1 (SMN1) protein cause spinal muscular atrophy (SMA). α-Motor neurons of the spinal cord are considered particularly vulnerable in this genetic disorder and their dysfunction and loss cause progressive muscle weakness, paralysis and eventually premature death of afflicted individuals. Historically, SMA was therefore considered a motor neuron-autonomous disease. However, depletion of SMN in motor neurons of normal mice elicited only a very mild phenotype. Conversely, restoration of SMN to motor neurons in an SMA mouse model had only modest effects on the SMA phenotype and survival. Collectively, these results suggested that additional cell types contribute to the pathogenesis of SMA, and understanding the non-autonomous requirements is crucial for developing effective therapies. Astrocytes are critical for regulating synapse formation and function as well as metabolic support for neurons. We hypothesized that astrocyte functions are disrupted in SMA, exacerbating disease progression. Using viral-based restoration of SMN specifically to astrocytes, survival in severe and intermediate SMA mice was observed. In addition, neuromuscular circuitry was improved. Astrogliosis was prominent in end-stage SMA mice and in post-mortem patient spinal cords. Increased expression of proinflammatory cytokines was partially normalized in treated mice, suggesting that astrocytes contribute to the pathogenesis of SMA.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Atrofia Muscular Espinal/patologia , Animais , Diferenciação Celular , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Fenótipo , Medula Espinal/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo
7.
Mol Pain ; 11: 5, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25888914

RESUMO

BACKGROUND: Cell-based therapy may hold promise for treatment of chronic pain. Mesenchymal stem cells (MSCs) are readily available and robust, and their secretion of therapeutic peptides can be enhanced by genetically engineering. We explored the analgesic potential of transplanting bone marrow-derived MSCs that have been transduced with lentivectors. To optimize efficacy and safety, primary sensory neurons were targeted by MSC injection into the dorsal root ganglia (DRGs). RESULTS: MSCs were transduced using lentivectors to express enhanced green fluorescent protein (EGFP) or to co-express the analgesic peptide glial cell line-derived neurotrophic factor (GDNF) and EGFP by a viral 2A bicistronic transgene cassette. Engineered MSCs were injected into the 4(th) lumbar (L4) and L5 DRGs of adult allogeneic rats to evaluate survival in the DRGs. MSCs were detected by immunofluorescence staining up to 2-3 weeks after injection, distributed in the extracellular matrix space without disrupting satellite glial cell apposition to sensory neurons, suggesting well-tolerated integration of engrafted MSCs into DRG tissue. To examine their potential for inhibiting development of neuropathic pain, MSCs were injected into the L4 and L5 DRGs ipsilateral to a spinal nerve ligation injury. Animals injected with GDNF-engineered MSCs showed moderate but significant reduction in mechanical allodynia and hyperalgesia compared to controls implanted with MSCs expressing EGFP alone. We also observed diminished long-term survival of allografted MSCs at 3 weeks, and the development of a highly-proliferating population of MSCs in 12% of DRGs after transplantation. CONCLUSIONS: These data indicate that genetically modified MSCs secreting analgesic peptides could potentially be developed as a novel DRG-targeted cell therapy for treating neuropathic pain. However, further work is needed to address the challenges of MSC survival and excess proliferation, possibly with trials of autologous MSCs, evaluation of clonally selected populations of MSCs, and investigation of regulation of MSC proliferation.


Assuntos
Analgesia , Gânglios Espinais/transplante , Células-Tronco Mesenquimais/citologia , Neuralgia/terapia , Neurônios Aferentes/citologia , Analgesia/métodos , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Gânglios Espinais/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais , Neuralgia/genética , Neuralgia/metabolismo , Manejo da Dor/métodos , Ratos Sprague-Dawley , Nervos Espinhais/metabolismo
8.
Stem Cells Transl Med ; 3(5): 564-74, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24657962

RESUMO

Using stem cells to replace degenerating muscle cells and restore lost skeletal muscle function is an attractive therapeutic strategy for treating neuromuscular diseases. Myogenic progenitors are a valuable cell type for cell-based therapy and also provide a platform for studying normal muscle development and disease mechanisms in vitro. Human pluripotent stem cells represent a valuable source of tissue for generating myogenic progenitors. Here, we present a novel protocol for deriving myogenic progenitors from human embryonic stem (hES) and induced pluripotent stem (iPS) cells using free-floating spherical culture (EZ spheres) in a defined culture medium. hES cell colonies and human iPS cell colonies were expanded in medium supplemented with high concentrations (100 ng/ml) of fibroblast growth factor-2 (FGF-2) and epidermal growth factor in which they formed EZ spheres and were passaged using a mechanical chopping method. We found myogenic progenitors in the spheres after 6 weeks of culture and multinucleated myotubes following sphere dissociation and 2 weeks of terminal differentiation. A high concentration of FGF-2 plays a critical role for myogenic differentiation and is necessary for generating myogenic progenitors from pluripotent cells cultured as EZ spheres. Importantly, EZ sphere culture produced myogenic progenitors from human iPS cells generated from both healthy donors and patients with neuromuscular disorders (including Becker's muscular dystrophy, spinal muscular atrophy, and familial amyotrophic lateral sclerosis). Taken together, this study demonstrates a simple method for generating myogenic cells from pluripotent sources under defined conditions for potential use in disease modeling or cell-based therapies targeting skeletal muscle.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias , Células-Tronco Pluripotentes Induzidas , Desenvolvimento Muscular , Fibras Musculares Esqueléticas , Esferoides Celulares , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/terapia , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transplante de Células-Tronco
9.
Glia ; 61(9): 1418-1428, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23839956

RESUMO

Spinal muscular atrophy (SMA) is a genetic disorder caused by the deletion of the survival motor neuron 1 (SMN1) gene that leads to loss of motor neurons in the spinal cord. Although motor neurons are selectively lost during SMA pathology, selective replacement of SMN in motor neurons does not lead to full rescue in mouse models. Due to the ubiquitous expression of SMN, it is likely that other cell types besides motor neurons are affected by its disruption and therefore may contribute to disease pathology. Here we show that astrocytes in SMAΔ7 mouse spinal cord and from SMA-induced pluripotent stem cells exhibit morphological and cellular changes indicative of activation before overt motor neuron loss. Furthermore, our in vitro studies show mis-regulation of basal calcium and decreased response to adenosine triphosphate stimulation indicating abnormal astrocyte function. Together, for the first time, these data show early disruptions in astrocytes that may contribute to SMA disease pathology.


Assuntos
Astrócitos/metabolismo , Cálcio/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Atrofia Muscular Espinal/patologia , Medula Espinal/citologia , Trifosfato de Adenosina/farmacologia , Fatores Etários , Aldeído Desidrogenase/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Linhagem Celular Transformada , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Atrofia Muscular Espinal/genética , Mutação/genética , Nestina/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH , Células-Tronco Pluripotentes/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Proteínas S100/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo
10.
Hum Mol Genet ; 22(4): 729-36, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23175440

RESUMO

Our fundamental understanding of how several thousand diverse RNAs are recognized in the soma, sorted, packaged, transported and localized within the cell is fragmentary. The COPa and COPb proteins of the coatomer protein I (COPI) vesicle complex were reported to interact with specific RNAs and represent a candidate RNA sorting and transport system. To determine the RNA-binding profile of Golgi-derived COPI in neuronal cells, we performed formaldehyde-linked RNA immunoprecipitation, followed by high-throughput sequencing, a process we term FLRIP-Seq (FLRIP, formaldehyde-cross-linked immunoprecipitation). We demonstrate that COPa co-immunoprecipitates a specific set of RNAs that are enriched in G-quadruplex motifs and fragile X mental retardation protein-associated RNAs and that encode factors that predominantly localize to the plasma membrane and cytoskeleton and function within signaling pathways. These data support the novel function of COPI in inter-compartmental trafficking of RNA.


Assuntos
Proteína Coatomer/metabolismo , Transporte de RNA , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Animais , Antígenos de Neoplasias/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Complexo I de Proteína do Envoltório/metabolismo , Sequência Consenso , Citoesqueleto/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Antígeno Neuro-Oncológico Ventral , Ligação Proteica , Subunidades Proteicas/metabolismo , Transporte Proteico , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Análise de Sequência de RNA , Transcriptoma
11.
PLoS One ; 7(6): e39113, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723941

RESUMO

Spinal muscular atrophy (SMA) is a genetic disorder caused by a deletion of the survival motor neuron 1 gene leading to motor neuron loss, muscle atrophy, paralysis, and death. We show here that induced pluripotent stem cell (iPSC) lines generated from two Type I SMA subjects-one produced with lentiviral constructs and the second using a virus-free plasmid-based approach-recapitulate the disease phenotype and generate significantly fewer motor neurons at later developmental time periods in culture compared to two separate control subject iPSC lines. During motor neuron development, both SMA lines showed an increase in Fas ligand-mediated apoptosis and increased caspase-8 and-3 activation. Importantly, this could be mitigated by addition of either a Fas blocking antibody or a caspase-3 inhibitor. Together, these data further validate this human stem cell model of SMA, suggesting that specific inhibitors of apoptotic pathways may be beneficial for patients.


Assuntos
Apoptose/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores , Caspase 8/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Ativação Enzimática , Proteína Ligante Fas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Fenótipo , Receptor fas/antagonistas & inibidores
12.
Exp Neurol ; 224(1): 155-62, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20227407

RESUMO

Huntington's disease (HD) is an autosomal dominant disorder caused by expansion of polyglutamine repeats in the huntingtin gene leading to loss of striatal and cortical neurons followed by deficits in cognition and choreic movements. Growth factor delivery to the brain has shown promise in various models of neurodegenerative diseases, including HD, by reducing neuronal death and thus limiting motor impairment. Here we used mouse neural progenitor cells (mNPCs) as growth factor delivery vehicles in the N171-82Q transgenic mouse model of HD. mNPCs derived from the developing mouse striatum were isolated and infected with lentivirus expressing either glial cell line-derived neurotrophic factor (GDNF) or green fluorescent protein (GFP). Next, mNPCs(GDNF) or mNPCs(GFP) were transplanted bilaterally into the striatum of pre-symptomatic N171-82Q mice. We found that mNPCs(GDNF), but not mNPCs(GFP), maintained rotarod function and increased striatal neuron survival out to 3months post-transplantation. Importantly, histological analysis showed GDNF expression through the duration of the experiment. Our data show that mNPCs(GDNF) can survive transplantation, secrete GDNF for several weeks and are able to maintain motor function in this model of HD.


Assuntos
Corpo Estriado/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Doença de Huntington/terapia , Degeneração Neural/terapia , Análise de Variância , Animais , Peso Corporal , Contagem de Células , Células Cultivadas , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Terapia Genética , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Imuno-Histoquímica , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Atividade Motora/genética , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Teste de Desempenho do Rota-Rod , Transplante de Células-Tronco
13.
Nat Rev Drug Discov ; 9(5): 367-72, 2010 05.
Artigo em Inglês | MEDLINE | ID: mdl-20339370

RESUMO

High-throughput screening technologies are widely used in the early stages of drug discovery to rapidly evaluate the properties of thousands of compounds. However, they generally rely on testing compound libraries on highly proliferative immortalized or cancerous cell lines, which do not necessarily provide an accurate indication of the effects of compounds in normal human cells or the specific cell type under study. Recent advances in stem cell technology have the potential to allow production of a virtually limitless supply of normal human cells that can be differentiated into any specific cell type. Moreover, using induced pluripotent stem cell technology, they can also be generated from patients with specific disease traits, enabling more relevant modelling and drug screens. This article discusses the opportunities and challenges for the use of stem cells in drug screening with a focus on induced pluripotent stem cells.


Assuntos
Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Pluripotentes/efeitos dos fármacos , Animais , Diferenciação Celular , Desenho de Fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Modelos Biológicos
14.
PLoS One ; 4(10): e7630, 2009 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-19898616

RESUMO

BACKGROUND: Stem cell expansion and differentiation is the foundation of emerging cell therapy technologies. The potential applications of human neural progenitor cells (hNPCs) are wide ranging, but a normal cytogenetic profile is important to avoid the risk of tumor formation in clinical trials. FDA approved clinical trials are being planned and conducted for hNPC transplantation into the brain or spinal cord for various neurodegenerative disorders. Although human embryonic stem cells (hESCs) are known to show recurrent chromosomal abnormalities involving 12 and 17, no studies have revealed chromosomal abnormalities in cultured hNPCs. Therefore, we investigated frequently occurring chromosomal abnormalities in 21 independent fetal-derived hNPC lines and the possible mechanisms triggering such aberrations. METHODS AND FINDINGS: While most hNPC lines were karyotypically normal, G-band karyotyping and fluorescent in situ hybridization (FISH) analyses revealed the emergence of trisomy 7 (hNPC(+7)) and trisomy 19 (hNPC(+19)), in 24% and 5% of the lines, respectively. Once detected, subsequent passaging revealed emerging dominance of trisomy hNPCs. DNA microarray and immunoblotting analyses demonstrate epidermal growth factor receptor (EGFR) overexpression in hNPC(+7) and hNPC(+19) cells. We observed greater levels of telomerase (hTERT), increased proliferation (Ki67), survival (TUNEL), and neurogenesis (beta(III)-tubulin) in hNPC(+7) and hNPC(+19), using respective immunocytochemical markers. However, the trisomy lines underwent replicative senescence after 50-60 population doublings and never showed neoplastic changes. Although hNPC(+7) and hNPC(+19) survived better after xenotransplantation into the rat striatum, they did not form malignant tumors. Finally, EGF deprivation triggered a selection of trisomy 7 cells in a diploid hNPC line. CONCLUSIONS: We report that hNPCs are susceptible to accumulation of chromosome 7 and 19 trisomy in long-term cell culture. These results suggest that micro-environmental cues are powerful factors in the selection of specific hNPC aneuploidies, with trisomy of chromosome 7 being the most common. Given that a number of stem cell based clinical trials are being conducted or planned in USA and a recent report in PLoS Medicine showing the dangers of grafting an inordinate number of cells, these data substantiate the need for careful cytogenetic evaluation of hNPCs (fetal or hESC-derived) before their use in clinical or basic science applications.


Assuntos
Técnicas de Cultura de Células/métodos , Cromossomos Humanos Par 19/ultraestrutura , Cromossomos Humanos Par 7/ultraestrutura , Neurônios/citologia , Células-Tronco/citologia , Trissomia , Encéfalo/embriologia , Aberrações Cromossômicas , Citogenética , Células-Tronco Embrionárias/citologia , Receptores ErbB/metabolismo , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Análise de Sequência com Séries de Oligonucleotídeos
15.
Cell Transplant ; 17(7): 753-62, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19044202

RESUMO

The use of human neural progenitor cells (hNPC) has been proposed to provide neuronal replacement or astrocytes delivering growth factors for brain disorders such as Parkinson's and Huntington's disease. Success in such studies likely requires migration from the site of transplantation and integration into host tissue in the face of ongoing damage. In the current study, hNPC modified to release glial cell line-derived neurotrophic factor (hNPCGDNF) were transplanted into either intact or lesioned animals. GDNF release itself had no effect on the survival, migration, or differentiation of the cells. The most robust migration and survival was found using a direct lesion of striatum (Huntington's model) with indirect lesions of the dopamine system (Parkinson's model) or intact animals showing successively less migration and survival. No lesion affected differentiation patterns. We conclude that the type of brain injury dictates migration and integration of hNPC, which has important consequences when considering transplantation of these cells as a therapy for neurodegenerative diseases.


Assuntos
Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Corpo Estriado/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Neurônios/citologia , Transplante de Células-Tronco , Células-Tronco/citologia
16.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2D.2, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18819085

RESUMO

Neural progenitor cells have tremendous utility for understanding basic developmental processes, disease modeling, and therapeutic intervention. The protocols described in this unit provide detailed information to isolate and expand human and rodent neural progenitor cells in culture for several months as floating aggregates (termed neurospheres) or plated cultures. Detailed protocols for cryopreservation, neural differentiation, exogenous gene expression using lentivirus, and transplantation into the rodent nervous system are also described.


Assuntos
Separação Celular/métodos , Feto/citologia , Neurônios/citologia , Neurônios/virologia , Roedores , Células-Tronco/citologia , Células-Tronco/virologia , Animais , Diferenciação Celular , Proliferação de Células , Células Clonais , Criopreservação , Enzimas/metabolismo , Humanos , Lentivirus , Infecções por Lentivirus , Neurônios/transplante , Esferoides Celulares/citologia , Transplante de Células-Tronco , Fatores de Tempo , Transgenes
17.
Cell Transplant ; 17(4): 383-95, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18522241

RESUMO

Human neural progenitor cells (hNPCs) have been proposed as a potential source of cells for ex vivo gene therapy. In this pilot study, three 5-year-old female cynomolgus monkeys received a single intracarotid infusion of MPTP, followed 1 week later by MRI-guided stereotaxic intrastriatal and intranigral injections of male hNPCs transgenic for GDNF. Immunosupression with oral cyclosporine (30-40 mg/kg) began 48 h before hNPC transplants and continued throughout the study. We monitored the animals using a clinical rating scale (CRS). Three months postsurgery, we euthanized the animals by transcardiac perfusion, then retrieved and processed their brains for morphological analysis. Our findings include the following. 1) hNPCs survived and produced GDNF in all animals 3 months postsurgery. 2) hNPCs remained in the areas of injection as observed by GDNF immunostaining and in situ hybridization for the human Y chromosome. 3) A "halo" of GDNF expression was observed diffusing from the center of the graft out into the surrounding area. 4) We observed increased TH- and VMAT2-positive fibers in areas of GDNF delivery in two of the three animals. The two animals with TH- and VMAT2-positive fibers also showed reductions in their CRS scores. 5) Some GFAP-positive perivascular cuffing was found in transplanted areas. 6) General blood chemistry and necropsies did not reveal any abnormalities. Therefore, we conclude that hNPCs releasing GDNF may be a possible alternative for intracerebral trophic factor delivery in Parkinson's disease.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Neurônios/efeitos dos fármacos , Transplante de Células-Tronco , Células-Tronco/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Animais , Dopaminérgicos/administração & dosagem , Dopaminérgicos/farmacologia , Feminino , Humanos , Macaca fascicularis , Masculino , Neurônios/citologia , Neurônios/metabolismo , Neurotoxinas/administração & dosagem , Neurotoxinas/farmacologia , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/terapia , Projetos Piloto , Células-Tronco/citologia , Transgenes
18.
Exp Neurol ; 209(1): 213-23, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18061591

RESUMO

Growth factors such as glial cell line-derived neurotrophic factor (GDNF) have been shown to prevent neurodegeneration and promote regeneration in many animal models of Parkinson's disease (PD). Insulin-like growth factor 1 (IGF-1) is also known to have neuroprotective effects in a number of disease models but has not been extensively studied in models of PD. We produced human neural progenitor cells (hNPC) releasing either GDNF or IGF-1 and transplanted them into a rat model of PD. hNPC secreting either GDNF or IGF-1 were shown to significantly reduce amphetamine-induced rotational asymmetry and dopamine neuron loss when transplanted 7 days after a 6-hydroxydopamine (6-OHDA) lesion. Neither untransduced hNPC nor a sham transplant had this effect suggesting GDNF and IGF-1 release was required. Interestingly, GDNF, but not IGF-1, was able to protect or regenerate tyrosine hydroxylase-positive fibers in the striatum. In contrast, IGF-1, but not GDNF, significantly increased the overall survival of hNPC both in vitro and following transplantation. This suggests a dual role of IGF-1 to both increase hNPC survival after transplantation and exert trophic effects on degenerating dopamine neurons in this rat model of PD.


Assuntos
Dopamina/fisiologia , Fator de Crescimento Insulin-Like I/biossíntese , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson Secundária/terapia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Anfetamina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Estimulantes do Sistema Nervoso Central/farmacologia , Interpretação Estatística de Dados , Ensaio de Imunoadsorção Enzimática , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/farmacologia , Lentivirus/genética , Masculino , Fibras Nervosas/enzimologia , Oxidopamina , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/patologia , Ratos , Ratos Endogâmicos Lew , Comportamento Estereotipado/efeitos dos fármacos , Simpatolíticos , Tirosina 3-Mono-Oxigenase/metabolismo
19.
J Neurosci Methods ; 163(2): 338-49, 2007 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397931

RESUMO

Human neural progenitor cells (hNPC) hold great potential as an ex vivo system for delivery of therapeutic proteins to the central nervous system. When cultured as aggregates, termed neurospheres, hNPC are capable of significant in vitro expansion. In the current study, we present a robust method for lentiviral vector-mediated gene delivery into hNPC that maintains the differentiation and proliferative properties of neurosphere cultures while minimizing the amount of viral vector used and controlling the number of insertion sites per population. This method results in long-term, stable expression even after differentiation of the hNPC to neurons and astrocytes and allows for generation of equivalent transgenic populations of hNPC. In addition, the in vitro analysis presented predicts the behavior of transgenic lines in vivo when transplanted into a rodent model of Parkinson's disease. The methods presented provide a powerful tool for assessing the impact of factors such as promoter systems or different transgenes on the therapeutic utility of these cells.


Assuntos
Técnicas de Transferência de Genes/normas , Terapia Genética/métodos , Vetores Genéticos/genética , Lentivirus/genética , Células-Tronco/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Biomarcadores/metabolismo , Transplante de Tecido Encefálico/métodos , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Feto , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Humanos , Neurônios/metabolismo , Neurônios/virologia , Doença de Parkinson/genética , Doença de Parkinson/terapia , Regiões Promotoras Genéticas/genética , Ratos , Ratos Endogâmicos Lew , Esferoides Celulares/citologia , Esferoides Celulares/fisiologia , Esferoides Celulares/virologia , Células-Tronco/virologia , Transgenes
20.
Exp Neurol ; 191 Suppl 1: S80-94, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15629764

RESUMO

Caspase-9 is a critical downstream effector molecule involved in apoptosis, a cell death process thought to be involved in the demise of dopamine (DA) neurons in the substantia nigra (SN) affected by Parkinson's disease (PD). In this study, we determined that a tetracycline-regulated adenovirus harboring a dominant-negative form of caspase-9 (Casp9DN) and the marker gene, enhanced green fluorescent protein (EGFP), under the control of a bidirectional promoter could each be regulated in vitro and in vivo by doxycycline. We next observed that Casp9DN gene delivery significantly protected against TNFalpha and cycloheximide-induced chromatin condensation in HeLa cells and prevented chromatin condensation and the appearance of the early apoptotic marker annexin V in 6-hydroxydopamine (6-OHDA) treated MN9D cells, a dopaminergic cell line. Effects of Casp9DN on DA neurons in vivo were also assessed. DA neurons were retrogradely labeled with fluorogold (FG) and transduced with Casp9DN and EGFP or EGFP alone. A progressive lesion of DA neurons was induced by striatal injection of 6-OHDA 1 week later. At 2 weeks post-lesion, a morphometric analysis of FG+ neurons in the SN revealed that the mean cell diameter of FG labeled neurons in the Casp9DN group was 8% and 21% larger than the EGFP and PBS groups, respectively (P <0.05). However, there was no difference among the treatment groups in the number of neurons remaining in the lesioned SN. These results suggest that while inhibiting apoptosis at the level of caspase-9 is protective in vitro, it is not protective against 6-OHDA-induced cell death in vivo.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Caspases/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Transtornos Parkinsonianos/metabolismo , Tetraciclina/farmacologia , Adenoviridae/efeitos dos fármacos , Animais , Apoptose , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Técnicas de Transferência de Genes , Genes Dominantes , Genes Reporter , Vetores Genéticos/efeitos dos fármacos , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Rim/citologia , Rim/metabolismo , Masculino , Neostriado/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Neurotoxinas , Oxidopamina , Transtornos Parkinsonianos/patologia , Transtornos Parkinsonianos/terapia , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA