Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mov Disord ; 38(7): 1273-1281, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37166002

RESUMEN

BACKGROUND: The degeneration of nigral (A9) dopaminergic (DA) neurons results in cardinal motor symptoms that define Parkinson's disease (PD). Loss-of-function mutations in parkin are linked to a rare form of early-onset PD that is inherited recessively. OBJECTIVE: We generated isogenic human A9 DA neurons with or without parkin mutations to establish the causal relationship between parkin mutations and the dysfunction of human A9 DA neurons. METHODS: Using TALEN (transcription activator-like effector nuclease)- or CRISPR/Cas9-mediated gene targeting, we produced two isogenic pairs of naivetropic induced pluripotent stem cells (iPSCs) by repairing exon 3 deletions of parkin in iPSCs derived from a PD patient and by introducing the PD-linked A82E mutation into iPSCs from a healthy subject. The four lines of isogenic iPSCs were differentiated to A9 DA neurons, which fired spontaneous pacemaking action potentials (AP) dependent on L-type Ca2+ channels. RESULTS: The frequency of the pacemaking APs was significantly reduced by parkin mutations introduced to normal neurons. Consistent with this, isogenic repair of parkin mutations significantly increased the frequency from that observed in patient-derived neurons. CONCLUSIONS: The results show that parkin maintains robust pacemaking in human iPSC-derived A9 DA neurons. The function is critical to normal DA transmission required for controlling voluntary locomotor activities. © 2023 International Parkinson and Movement Disorder Society.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Neuronas Dopaminérgicas/metabolismo , Enfermedad de Parkinson/genética , Sustancia Negra/metabolismo , Ubiquitina-Proteína Ligasas/genética
2.
Mol Psychiatry ; 27(11): 4407-4418, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35610351

RESUMEN

The degeneration of nigral (A9) dopaminergic (DA) neurons causes motor symptoms in Parkinson's disease (PD). We use small-molecule compounds to direct the differentiation of human induced pluripotent stem cells (iPSCs) to A9 DA neurons that share many important properties with their in vivo counterparts. The method generates a large percentage of TH+ neurons that express appropriate A9 markers, such as GIRK2 and ALDH1A1, but mostly not the A10 marker CALBINDIN. Functionally, they exhibit autonomous pacemaking based on L-type voltage-dependent Ca2+ channels and show autoreceptor-dependent regulation of dopamine release. When transplanted in the striatum of 6-OHDA-lesioned athymic rats, the human A9 DA neurons manifest robust survival and axon outgrowth, and ameliorate motor deficits in the rat PD model. The ability to generate patient-specific A9 DA autonomous pacemakers will significantly improve PD research and facilitate the development of disease-modifying therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Humanos , Ratas , Animales , Dopamina , Neuronas Dopaminérgicas , Sustancia Negra , Oxidopamina , Enfermedad de Parkinson/terapia
3.
Mov Disord ; 37(1): 70-79, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34564901

RESUMEN

BACKGROUND: Despite intense efforts to develop an objective diagnostic test for Parkinson's disease, there is still no consensus on biomarkers that can accurately diagnose the disease. OBJECTIVE: Identification of biomarkers for idiopathic Parkinson's disease (PD) may enable accurate diagnosis of the disease. We tried to find molecular and cellular differences in dopaminergic (DA) neurons derived from healthy subjects and idiopathic PD patients with or without rest tremor at onset. METHODS: We measured the expression of genes controlling dopamine synthesis, sequestration, and catabolism as well as the levels of corresponding metabolites and reactive oxygen species in midbrain DA neurons differentiated from induced pluripotent stem cells (iPSCs) of healthy subjects and PD patients with or without rest tremor. RESULTS: Significant differences in DA-related gene expression, metabolites, and oxidative stress were found between midbrain DA neurons derived from healthy subjects and patients with PD. DA neurons derived from PD patients with or without rest tremor at onset exhibited significant differences in the levels of some of these transcripts, metabolites, and oxidative stress. CONCLUSION: The unique combination of these quantifiable molecular and cellular traits in iPSC-derived midbrain DA neurons can distinguish healthy subjects from idiopathic PD patients and segregate PD patients with or without rest tremor at onset. The strategy may be used to develop an objective diagnostic test for PD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Diferenciación Celular/genética , Neuronas Dopaminérgicas/metabolismo , Humanos , Mesencéfalo/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
4.
Nat Protoc ; 16(8): 3954-3980, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34215863

RESUMEN

Naive human pluripotent stem cells (hPSCs) can be used to generate mature human cells of all three germ layers in mouse-human chimeric embryos. Here, we describe a protocol for generating mouse-human chimeric embryos by injecting naive hPSCs converted from the primed state. Primed hPSCs are treated with a mammalian target of rapamycin inhibitor (Torin1) for 3 h and dissociated to single cells, which are plated on mouse embryonic fibroblasts in 2iLI medium, a condition essentially the same for culturing mouse embryonic stem cells. After 3-4 d, bright, dome-shaped colonies with mouse embryonic stem cell morphology are passaged in 2iLI medium. Established naive hPSCs are injected into mouse blastocysts, which produce E17.5 mouse embryos containing 0.1-4.0% human cells as quantified by next-generation sequencing of 18S ribosomal DNA amplicons. The protocol is suitable for studying the development of hPSCs in mouse embryos and may facilitate the generation of human cells, tissues and organs in animals.


Asunto(s)
Quimera/embriología , Embrión de Mamíferos/fisiología , Células Madre Embrionarias/fisiología , Fibroblastos/fisiología , Células Madre Pluripotentes/fisiología , Amidas/farmacología , Animales , Embrión de Mamíferos/citología , Células Madre Embrionarias/efectos de los fármacos , Femenino , Humanos , Ratones , Naftiridinas/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Piridinas/farmacología
5.
Exp Biol Med (Maywood) ; 246(2): 240-248, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33070653

RESUMEN

Degeneration of photoreceptors is a major cause of blindness. Identifying new methods for the generation of photoreceptors offers valuable options for a cell replacement therapy of blindness. Here, we show that primary adult human retinal pigmented epithelium (hRPE) cells were directly converted to postmitotic neurons with various properties of photoreceptors by the neurogenic transcription factor ASCL1 and microRNA124. At Day 8 after the induction of ASCL1 and miRNA124 expression in hRPE cells, 91% of all cells were Tuj1+, and 83% of all cells were MAP2+ neurons. The cone photoreceptor marker L/M-opsin, the rod photoreceptor marker rhodopsin, and the generic photoreceptor marker recoverin were expressed in 76%, 86%, and 92% of all cells, respectively. Real-time quantitative PCR measurements showed significant and continuous increases in the expression of photoreceptor markers phosducin and recoverin, rod cell markers phosphodiesterases 6 b and arrestin S-antigen, and cone cell markers L/M-opsin and S-opsin in three independent lines of primary hRPE cells at different days of transdifferentiation. Transmission electron microscopy of converted neurons showed disc-like structures similar to those found in photoreceptors. While the converted neurons had voltage-dependent Na+, K+, and Ca2+ currents, light-induced change in membrane potential was not detected. The study demonstrates the feasibility of rapid and efficient transdifferentiation of adult hPRE cells to neurons with many properties of photoreceptors. It opens up a new possibility in cell replacement therapy of blindness caused by photoreceptor degeneration.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Neuronas/citología , Células Fotorreceptoras de Vertebrados/citología , Epitelio Pigmentado de la Retina/citología , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Reprogramación Celular/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Mitosis , Epitelio Pigmentado de la Retina/ultraestructura , Factores de Tiempo
6.
Sci Rep ; 10(1): 10343, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587369

RESUMEN

The TET family of 5-methylcytosine (5mC) dioxygenases plays critical roles in development by modifying DNA methylation. Using CRISPR, we inactivated the TET1 gene in H9 human embryonic stem cells (hESCs). Mutant H9 hESCs remained pluripotent, even though the level of hydroxymethylcytosine (5hmC) decreased to 30% of that in wild-type cells. Neural differentiation induced by dual SMAD inhibitors was not significantly affected by loss of TET1 activity. However, in a morphogen-free condition, TET1 deficiency significantly reduced the generation of NESTIN+SOX1+ neuroectoderm cells from 70% in wild-type cells to 20% in mutant cells. This was accompanied by a 20-fold reduction in the expression level of PAX6 and a significant decrease in the amount of 5hmC on the PAX6 promoter. Overexpression of the TET1 catalytic domain in TET1-deficient hESCs significantly increased 5hmC levels and elevated PAX6 expression during differentiation. Consistent with these in vitro data, PAX6 expression was significantly decreased in teratomas formed by TET1-deficient hESCs. However, TET1 deficiency did not prevent the formation of neural tube-like structures in teratomas. Our results suggest that TET1 deficiency impairs the intrinsic ability of hESCs to differentiate to neuroectoderm, presumably by decreasing the expression of PAX6, a key regulator in the development of human neuroectoderm.


Asunto(s)
Células Madre Embrionarias Humanas/fisiología , Oxigenasas de Función Mixta/deficiencia , Placa Neural/crecimiento & desarrollo , Neurogénesis/genética , Factor de Transcripción PAX6/genética , Proteínas Proto-Oncogénicas/deficiencia , 5-Metilcitosina/metabolismo , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Línea Celular , Metilación de ADN/fisiología , Epigénesis Genética , Mutación del Sistema de Lectura , Regulación del Desarrollo de la Expresión Génica , Humanos , Oxigenasas de Función Mixta/genética , Neuronas/fisiología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción SOXB1/genética , Teratoma/genética , Teratoma/patología
7.
Sci Adv ; 6(20): eaaz0298, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32426495

RESUMEN

It has not been possible to generate naïve human pluripotent stem cells (hPSCs) that substantially contribute to mouse embryos. We found that a brief inhibition of mTOR with Torin1 converted hPSCs from primed to naïve pluripotency. The naïve hPSCs were maintained in the same condition as mouse embryonic stem cells and exhibited high clonogenicity, rapid proliferation, mitochondrial respiration, X chromosome reactivation, DNA hypomethylation, and transcriptomes sharing similarities to those of human blastocysts. When transferred to mouse blastocysts, naïve hPSCs generated 0.1 to 4% human cells, of all three germ layers, including large amounts of enucleated red blood cells, suggesting a marked acceleration of hPSC development in mouse embryos. Torin1 induced nuclear translocation of TFE3; TFE3 with mutated nuclear localization signal blocked the primed-to-naïve conversion. The generation of chimera-competent naïve hPSCs unifies some common features of naïve pluripotency in mammals and may enable applications such as human organ generation in animals.


Asunto(s)
Células Madre Pluripotentes , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Diferenciación Celular , Quimera , Humanos , Mamíferos , Ratones , Serina-Treonina Quinasas TOR
8.
Biochem Biophys Res Commun ; 516(3): 765-769, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31255287

RESUMEN

The direct conversion of accessible cells such as human fibroblasts to inaccessible cells, particularly neurons, opens up many opportunities for using the human model system to study diseases and discover therapies. Previous studies have indicated that the neuronal conversion of adult human skin fibroblasts is much harder than that for human lung fibroblasts, which are used in many experiments. Here we formally report this differential plasticity of human skin versus lung fibroblasts in their transdifferentiation to induced neurons. Using RNAseq of isogenic and non-isogenic pairs of human skin and lung fibroblasts at different days in their conversion to neurons, we found that several master regulators (TWIST1, TWIST2, PRRX1 and PRRX2) in the fibroblast Gene Regulatory Network were significantly downregulated in lung fibroblasts, but not in skin fibroblasts. By knocking down each of these genes and other genes that suppress the neural fate, such as REST, HES1 and HEY2, we found that the combined attenuation of HEY2 and PRRX2 significantly enhanced the transdifferentiation of human skin fibroblasts induced by ASCL1 and p53 shRNA. The new method, which overexpressed ASCL1 and knocked down p53, HEY2 and PRRX2 (ApH2P2), enabled the efficient transdifferentiation of adult human skin fibroblasts to MAP2+ neurons in 14 days. It would be useful for a variety of applications that require the efficient and speedy derivation of patient-specific neurons from skin fibroblasts.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas Represoras/genética , Piel/metabolismo , Proteína p53 Supresora de Tumor/genética , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Transdiferenciación Celular , Reprogramación Celular , Fibroblastos/citología , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/metabolismo , Humanos , Pulmón/citología , Pulmón/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Transducción de Señal , Piel/citología , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 Relacionada con Twist/antagonistas & inhibidores , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
9.
J Parkinsons Dis ; 8(4): 479-493, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30149462

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder. It is characterized by the degeneration of nigral dopaminergic (DA) neurons. While over 90% of cases are idiopathic, without a clear etiology, mutations in many genes have been linked to rare, familial forms of PD. It has been quite challenging to develop effective animal models of PD that capture salient features of PD. The discovery of induced pluripotent stem cells (iPSCs) makes it possible to generate patient-specific DA neurons to study PD. Here, we review the methods for the generation of iPSCs and discuss previous studies using iPSC-derived neurons from monogenic forms of PD. These investigations have revealed several converging pathways that intersect with the unique vulnerabilities of human nigral DA neurons. With the rapid development in stem cell biology, it is possible to generate patient-specific neurons that will be increasingly similar to those in the brain of the patient. Combined with the ability to edit the genome to generate isogenic iPSCs, the generation and analysis of patient-specific midbrain DA neurons will transform PD research by providing a valuable tool for mechanistic study and drug discovery.


Asunto(s)
Neuronas Dopaminérgicas/patología , Células Madre Pluripotentes Inducidas/patología , Enfermedad de Parkinson/patología , Sustancia Negra/patología , Neuronas Dopaminérgicas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/metabolismo
10.
Cell Rep ; 19(5): 1033-1044, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467897

RESUMEN

Locomotor symptoms in Parkinson's disease (PD) are accompanied by widespread oscillatory neuronal activities in basal ganglia. Here, we show that activation of dopamine D1-class receptors elicits a large rhythmic bursting of spontaneous excitatory postsynaptic currents (sEPSCs) in midbrain neurons differentiated from induced pluripotent stem cells (iPSCs) of PD patients with parkin mutations, but not normal subjects. Overexpression of wild-type parkin, but not its PD-causing mutant, abolishes the oscillatory activities in patient neurons. Dopamine induces a delayed enhancement in the amplitude of spontaneous, but not miniature, EPSCs, thus increasing quantal content. The results suggest that presynaptic regulation of glutamatergic transmission by dopamine D1-class receptors is significantly potentiated by parkin mutations. The aberrant dopaminergic regulation of presynaptic glutamatergic transmission in patient-specific iPSC-derived midbrain neurons provides a mechanistic clue to PD pathophysiology, and it demonstrates the usefulness of this model system in understanding how mutations of parkin cause movement symptoms in Parkinson's disease.


Asunto(s)
Dopamina/farmacología , Neuronas Dopaminérgicas/metabolismo , Potenciales Postsinápticos Excitadores , Mesencéfalo/citología , Mutación , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/genética , Anciano , Estudios de Casos y Controles , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/fisiopatología , Persona de Mediana Edad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología , Receptores de Dopamina D1/metabolismo
11.
Redox Biol ; 11: 606-612, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28110217

RESUMEN

Motor symptoms that define Parkinson's disease (PD) are caused by the selective loss of nigral dopaminergic (DA) neurons. Cell replacement therapy for PD has been focused on midbrain DA neurons derived from human fetal mesencephalic tissue, human embryonic stem cells (hESC) or human induced pluripotent stem cells (iPSC). Recent development in the direct conversion of human fibroblasts to induced dopaminergic (iDA) neurons offers new opportunities for transplantation study and disease modeling in PD. The iDA neurons are generated directly from human fibroblasts in a short period of time, bypassing lengthy differentiation process from human pluripotent stem cells and the concern for potentially tumorigenic mitotic cells. They exhibit functional dopaminergic neurotransmission and relieve locomotor symptoms in animal models of Parkinson's disease. In this review, we will discuss this recent development and its implications to Parkinson's disease research and therapy.


Asunto(s)
Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias Humanas/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Enfermedad de Parkinson/terapia , Diferenciación Celular/genética , Neuronas Dopaminérgicas/patología , Fibroblastos/metabolismo , Humanos , Mesencéfalo/patología , Mesencéfalo/trasplante , Enfermedad de Parkinson/patología
12.
Nat Commun ; 6: 10100, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26639555

RESUMEN

The direct conversion of fibroblasts to induced dopaminergic (iDA) neurons and other cell types demonstrates the plasticity of cell fate. The low efficiency of these relatively fast conversions suggests that kinetic barriers exist to safeguard cell-type identity. Here we show that suppression of p53, in conjunction with cell cycle arrest at G1 and appropriate extracellular environment, markedly increase the efficiency in the transdifferentiation of human fibroblasts to iDA neurons by Ascl1, Nurr1, Lmx1a and miR124. The conversion is dependent on Tet1, as G1 arrest, p53 knockdown or expression of the reprogramming factors induces Tet1 synergistically. Tet1 knockdown abolishes the transdifferentiation while its overexpression enhances the conversion. The iDA neurons express markers for midbrain DA neurons and have active dopaminergic transmission. Our results suggest that overcoming these kinetic barriers may enable highly efficient epigenetic reprogramming in general and will generate patient-specific midbrain DA neurons for Parkinson's disease research and therapy.


Asunto(s)
Transdiferenciación Celular/genética , Neuronas Dopaminérgicas/citología , Fibroblastos/citología , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Proteína p53 Supresora de Tumor/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular , Reprogramación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Mesencéfalo , MicroARNs/genética , Oxigenasas de Función Mixta , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Stem Cells Dev ; 24(21): 2591-604, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26218671

RESUMEN

The lack of robust Parkinson's disease (PD) phenotype in parkin knockout rodents and the identification of defective dopaminergic (DA) neurotransmission in midbrain DA neurons derived from induced pluripotent stem cells (iPSC) of PD patients with parkin mutations demonstrate the utility of patient-specific iPSCs as an effective system to model the unique vulnerabilities of midbrain DA neurons in PD. Significant efforts have been directed at developing efficient genomic engineering technologies in human iPSCs to study diseases such as PD. In the present study, we converted patient-specific iPSCs from the primed state to a naivetropic state by DOX-induced expression of transgenes (Oct4, Sox2, Klf4, c-Myc, and Nanog) and the use of 2iL (MEK inhibitor PD0325901, GSK3 inhibitor CHIR99021, and human LIF). These patient-specific naivetropic iPSCs were pluripotent in terms of marker expression, spontaneous differentiation in vitro, and teratoma formation in vivo. They exhibited morphological, proliferative, and clonogenic characteristics very similar to naive mouse embryonic stem cells (ESC). The high clonal efficiency and proliferation rate of naivetropic iPSCs enabled very efficient gene targeting of GFP to the PITX3 locus by transcription activator-like effector nuclease. The naivetropic iPSCs could be readily reverted to the primed state upon the withdrawal of DOX, 2iL, and the switch to primed-state hESC culture conditions. Midbrain DA neurons differentiated from the reverted iPSCs retained the original phenotypes caused by parkin mutations, attesting to the robustness of these phenotypes and the usefulness of genomic engineering in patient-specific naivetropic iPSCs for studying PD.


Asunto(s)
Diferenciación Celular/fisiología , Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Parkinson/genética , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Factor 4 Similar a Kruppel , Ratones
14.
Stem Cells ; 33(1): 68-78, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25332110

RESUMEN

Parkinson's disease (PD) is characterized by the degeneration of nigral dopaminergic (DA) neurons and non-DA neurons in many parts of the brain. Mutations of parkin, an E3 ubiquitin ligase that strongly binds to microtubules, are the most frequent cause of recessively inherited PD. The lack of robust PD phenotype in parkin knockout mice suggests a unique vulnerability of human neurons to parkin mutations. Here, we show that the complexity of neuronal processes as measured by total neurite length, number of terminals, number of branch points, and Sholl analysis was greatly reduced in induced pluripotent stem cell (iPSC)-derived TH(+) or TH(-) neurons from PD patients with parkin mutations. Consistent with these, microtubule stability was significantly decreased by parkin mutations in iPSC-derived neurons. Overexpression of parkin, but not its PD-linked mutant nor green fluorescent protein, restored the complexity of neuronal processes and the stability of microtubules. Consistent with these, the microtubule-depolymerizing agent colchicine mimicked the effect of parkin mutations by decreasing neurite length and complexity in control neurons while the microtubule-stabilizing drug taxol mimicked the effect of parkin overexpression by enhancing the morphology of parkin-deficient neurons. The results suggest that parkin maintains the morphological complexity of human neurons by stabilizing microtubules.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Mutación , Neuronas/fisiología , Enfermedad de Parkinson/genética , Células Madre Pluripotentes/fisiología , Ubiquitina-Proteína Ligasas/genética , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Neuronas/enzimología , Neuronas/ultraestructura , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Células Madre Pluripotentes/enzimología , Células Madre Pluripotentes/patología
15.
Anal Chem ; 85(7): 3545-52, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23458184

RESUMEN

Induced pluripotent stem cells (iPSC) are generated by reprogramming somatic cells to the pluripotent state. Identification and quantitative characterization of changes in the molecular organization of the cell during the process of cellular reprogramming is valuable for stem cell research and advancement of its therapeutic applications. Here we employ quantitative Raman microspectroscopy and biomolecular component analysis (BCA) for a comparative analysis of the molecular composition of nucleoli in skin fibroblasts and iPSC derived from them. We report that the cultured fibroblasts obtained from different human subjects, share comparable concentrations of proteins, RNA, DNA, and lipids in the molecular composition of nucleoli. The nucleolar molecular environment is drastically changed in the corresponding iPSC. We measured that the transition from skin fibroblasts to iPSC is accompanied by a statistically significant increase in protein concentrations ~1.3-fold, RNA concentrations ~1.3-fold, and DNA concentrations ~1.4-fold, while no statistically significant difference was found for the lipid concentrations. The analysis of molecular vibrations associated with diverse aminoacids and protein conformations indicates that nucleoli of skin fibroblasts contain similar subsets of proteins, with prevalence of tyrosine. In iPSC, we observed a higher signal from tryptophan with an increase in the random coil and α helix protein conformations, indicating changes in the subset of nucleolar proteins during cell reprogramming. At the same time, the concentrations of major types of macromolecules and protein conformations in the nucleoli of iPSC and human embryonic stem cells (hESC) were found to be similar. We discuss these results in the context of nucleolar function and conclude that the nucleolar molecular content is correlated with the cellular differentiation status. The approach described here shows the potential for spectroscopically monitoring changes in macromolecular organization of the cell at different stages of reprogramming.


Asunto(s)
Nucléolo Celular/química , Fibroblastos/química , Células Madre Pluripotentes Inducidas/química , Piel/citología , Nucléolo Celular/genética , Células Cultivadas , Reprogramación Celular , ADN/análisis , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Lípidos/análisis , Proteínas Nucleares/análisis , ARN/análisis , Espectrometría Raman/métodos
16.
Sci Transl Med ; 4(141): 141ra90, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764206

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder caused by genetic and environmental factors that results in degeneration of the nigrostriatal dopaminergic pathway in the brain. We analyzed neural cells generated from induced pluripotent stem cells (iPSCs) derived from PD patients and presymptomatic individuals carrying mutations in the PINK1 (PTEN-induced putative kinase 1) and LRRK2 (leucine-rich repeat kinase 2) genes, and compared them to those of healthy control subjects. We measured several aspects of mitochondrial responses in the iPSC-derived neural cells including production of reactive oxygen species, mitochondrial respiration, proton leakage, and intraneuronal movement of mitochondria. Cellular vulnerability associated with mitochondrial dysfunction in iPSC-derived neural cells from familial PD patients and at-risk individuals could be rescued with coenzyme Q(10), rapamycin, or the LRRK2 kinase inhibitor GW5074. Analysis of mitochondrial responses in iPSC-derived neural cells from PD patients carrying different mutations provides insight into convergence of cellular disease mechanisms between different familial forms of PD and highlights the importance of oxidative stress and mitochondrial dysfunction in this neurodegenerative disease.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Humanos , Indoles/uso terapéutico , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Neuronas/efectos de los fármacos , Fenoles/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Sirolimus/uso terapéutico , Ubiquinona/uso terapéutico
17.
Curr Neurol Neurosci Rep ; 12(4): 392-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22622410

RESUMEN

Parkinson's disease (PD) is a movement disorder associated with the degeneration of nigral dopaminergic (DA) neurons. One of the greatest obstacles for PD research is the lack of patient-specific nigral DA neurons for mechanistic studies and drug discovery. The advent of induced pluripotent stem cells (iPSCs) has overcome this seemingly intractable problem and changed PD research in many profound ways. In this review, we discuss recent development in the generation and analyses of patient-specific iPSC-derived midbrain DA neurons. Results from this novel platform of human cellular models of PD have offered a tantalizing glimpse of the promising future of PD research. With the development of the latest genomic modification technologies, dopaminergic neuron differentiation methodologies, and cell transplantation studies, PD research is poised to enter a new phase that utilizes the human model system to identify the unique vulnerabilities of human nigral DA neurons and disease-modifying therapies based on such mechanistic studies.


Asunto(s)
Neuronas Dopaminérgicas/citología , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Parkinson/terapia , Animales , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Mesencéfalo/citología , Enfermedad de Parkinson/patología
18.
Nat Commun ; 3: 668, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22314364

RESUMEN

Parkinson's disease (PD) is defined by the degeneration of nigral dopaminergic (DA) neurons and can be caused by monogenic mutations of genes such as parkin. The lack of phenotype in parkin knockout mice suggests that human nigral DA neurons have unique vulnerabilities. Here we generate induced pluripotent stem cells from normal subjects and PD patients with parkin mutations. We demonstrate that loss of parkin in human midbrain DA neurons greatly increases the transcription of monoamine oxidases and oxidative stress, significantly reduces DA uptake and increases spontaneous DA release. Lentiviral expression of parkin, but not its PD-linked mutant, rescues these phenotypes. The results suggest that parkin controls dopamine utilization in human midbrain DA neurons by enhancing the precision of DA neurotransmission and suppressing dopamine oxidation. Thus, the study provides novel targets and a physiologically relevant screening platform for disease-modifying therapies of PD.


Asunto(s)
Encéfalo/embriología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Sitios de Unión , Fibroblastos/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Monoaminooxidasa/biosíntesis , Mutación , Neuronas/metabolismo , Estrés Oxidativo , Oxígeno/química , Fenotipo , Piel/metabolismo , Factores de Tiempo
19.
Biometals ; 25(4): 787-93, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22310887

RESUMEN

Divalent metal ion transporter (DMT1) is the major transporter for iron entrance into mammalian cells and iron exit from endosomes during the transferrin cycle. Four major mRNA isoforms correspond to four protein isoforms, differing at 5'/3' and N-/C-termini, respectively. Isoforms are designated 1A versus 1B reflecting where transcription starts or +iron responsive element (+IRE) versus -IRE reflecting the presence/absence of an IRE in the 3' end of the mRNA. These differences imply regulation at transcriptional and posttranscriptional levels. Many proteins are degraded by a ubiquitination-dependent mechanism. Two different ubiquitin ligases (E3s) appear to be involved in DMT1 ubiquitination: Parkin or neuronal precursor cell-expressed developmentally downregulated 4 (Nedd4) family E3s which often utilize Nedd4 family interacting protein-1 and -2 (Ndfip1 and 2) to ubiquitinate their substrate proteins. Prior data suggest that Parkin ubiquitinates 1B DMT1 but not 1A DMT1 while Nedd4/Ndfips ligate ubiquitin to DMT1 in the duodenum where 1A/+IRE DMT1 predominates. Our assay for whether these systems target DMT1 depends on two HEK293 cell lines that express permanently transfected 1A/+IRE DMT1 or 1B/-IRE DMT1 after induction by doxycycline. Transient transfection with a Parkin construct before induction diminishes 1B/-IRE DMT1 detected by immune-blots but not 1A/+IRE DMT1. Mutant Parkin serves as a control that does not affect DMT1 levels. Thus DMT1 regulation in an isoform specific fashion can occur by ubiquitination and the events involved have implications for DMT1 function and disease processes.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Humanos , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas Nedd4 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
20.
Hum Mol Genet ; 20(6): 1074-83, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21177257

RESUMEN

Parkin, whose mutations cause Parkinson disease (PD), controls oxidative stress by limiting the expression of monoamine oxidases (MAO)--mitochondrial enzymes responsible for the oxidative de-amination of dopamine. Here, we show that parkin performed this function by increasing the ubiquitination and degradation of estrogen-related receptors (ERR), orphan nuclear receptors that play critical roles in the transcription regulation of many nuclear-encoded mitochondrial proteins. All three ERRs (α, ß and γ) increased the transcription of MAOs A and B; the effects were abolished by parkin, but not by its PD-linked mutants. Parkin bound to ERRs and increased their ubiquitination and degradation. In fibroblasts from PD patients with parkin mutations or brain slices from parkin knockout mice, degradation of ERRs was significantly attenuated. The results reveal the molecular mechanism by which parkin suppresses the transcription of MAOs to control oxidative stress induced by dopamine oxidation.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Monoaminooxidasa/genética , Enfermedad de Parkinson/metabolismo , Receptores de Estrógenos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Humanos , Ratones , Ratones Noqueados , Monoaminooxidasa/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Estrógenos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Receptor Relacionado con Estrógeno ERRalfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...