Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Protoc ; 1(6): e141, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34102035

RESUMEN

It is essential to generate isolated populations of human neuronal subtypes in order to understand cell-type-specific roles in brain function and susceptibility to disease pathology. Here we describe a protocol for in-parallel generation of cortical glutamatergic (excitatory) and GABAergic (inhibitory) neurons from human pluripotent stem cells (hPSCs) by using the neurogenic transcription factors Ngn2 and a combination of Ascl1 and Dlx2, respectively. In contrast to the majority of neural transdifferentiation protocols that use transient lentiviral infection, the use of stable hPSC lines carrying doxycycline-inducible transcription factors allows neuronal differentiation to be initiated by addition of doxycycline and neural medium. This article presents a method to generate lentivirus from cultured mammalian cells and establish stable transcription factor-expressing cell lines (Basic Protocol 1), followed by a method for monolayer excitatory and inhibitory neuronal differentiation from the established lines (Basic Protocol 2). The resulting neurons reproducibly exhibit properties consistent with human cortical neurons, including the expected morphologies, expression of glutamatergic and GABAergic genes, and functional properties. Our approach enables the scalable and rapid production of human neurons suitable for modeling human brain diseases in a subtype-specific manner and examination of differential cellular vulnerability. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Lentivirus production and generation of stable hPSC lines Support Protocol 1: Expansion and maintenance of hPSCs Basic Protocol 2: Differentiation of EX- and IN-neurons Support Protocol 2: Experimental methods for validation of EX- and IN-neurons.


Asunto(s)
Células Madre Pluripotentes , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Neurogénesis , Neuronas
2.
Science ; 359(6382): 1330-1331, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29567692

Asunto(s)
Encéfalo , Genoma , Humanos
3.
Science ; 356(6336)2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28450582

RESUMEN

Neuropsychiatric disorders have a complex genetic architecture. Human genetic population-based studies have identified numerous heritable sequence and structural genomic variants associated with susceptibility to neuropsychiatric disease. However, these germline variants do not fully account for disease risk. During brain development, progenitor cells undergo billions of cell divisions to generate the ~80 billion neurons in the brain. The failure to accurately repair DNA damage arising during replication, transcription, and cellular metabolism amid this dramatic cellular expansion can lead to somatic mutations. Somatic mutations that alter subsets of neuronal transcriptomes and proteomes can, in turn, affect cell proliferation and survival and lead to neurodevelopmental disorders. The long life span of individual neurons and the direct relationship between neural circuits and behavior suggest that somatic mutations in small populations of neurons can significantly affect individual neurodevelopment. The Brain Somatic Mosaicism Network has been founded to study somatic mosaicism both in neurotypical human brains and in the context of complex neuropsychiatric disorders.


Asunto(s)
Encéfalo/anomalías , Trastornos Mentales/genética , Mosaicismo , Enfermedades del Sistema Nervioso/genética , Células-Madre Neurales/fisiología , Neuronas/fisiología , Encéfalo/metabolismo , División Celular/genética , Daño del ADN , Análisis Mutacional de ADN/métodos , Reparación del ADN/genética , Replicación del ADN , Genoma Humano , Células Germinativas/metabolismo , Humanos , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo
4.
J Biol Chem ; 291(4): 1803-1816, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26631732

RESUMEN

Parkin is an E3 ligase that contains a ubiquitin-like (UBL) domain in the N terminus and an R1-in-between-ring-RING2 motif in the C terminus. We showed that the UBL domain specifically interacts with the R1 domain and negatively regulates Parkin E3 ligase activity, Parkin-dependent mitophagy, and Parkin translocation to the mitochondria. The binding between the UBL domain and the R1 domain was suppressed by carbonyl cyanide m-chlorophenyl hydrazone treatment or by expression of PTEN-induced putative kinase 1 (PINK1), an upstream kinase that phosphorylates Parkin at the Ser-65 residue of the UBL domain. Moreover, we demonstrated that phosphorylation of the UBL domain at Ser-65 prevents its binding to the R1 domain and promotes Parkin activities. We further showed that mitochondrial translocation of Parkin, which depends on phosphorylation at Ser-65, and interaction between the R1 domain and a mitochondrial outer membrane protein, VDAC1, are suppressed by binding of the UBL domain to the R1 domain. Interestingly, Parkin with missense mutations associated with Parkinson disease (PD) in the UBL domain, such as K27N, R33Q, and A46P, did not translocate to the mitochondria and induce E3 ligase activity by m-chlorophenyl hydrazone treatment, which correlated with the interaction between the R1 domain and the UBL domain with those PD mutations. These findings provide a molecular mechanism of how Parkin recruitment to the mitochondria and Parkin activation as an E3 ubiquitin ligase are regulated by PINK1 and explain the previously unknown mechanism of how Parkin mutations in the UBL domain cause PD pathogenesis.


Asunto(s)
Enfermedad de Parkinson/enzimología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Humanos , Mitocondrias/enzimología , Enfermedad de Parkinson/genética , Fosforilación , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Unión Proteica , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
5.
Nat Med ; 21(12): 1445-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26523971

RESUMEN

Focal malformations of cortical development (FMCDs) account for the majority of drug-resistant pediatric epilepsy. Postzygotic somatic mutations activating the phosphatidylinositol-4,5-bisphosphate-3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway are found in a wide range of brain diseases, including FMCDs. It remains unclear how a mutation in a small fraction of cells disrupts the architecture of the entire hemisphere. Within human FMCD-affected brain, we found that cells showing activation of the PI3K-AKT-mTOR pathway were enriched for the AKT3(E17K) mutation. Introducing the FMCD-causing mutation into mouse brain resulted in electrographic seizures and impaired hemispheric architecture. Mutation-expressing neural progenitors showed misexpression of reelin, which led to a non-cell autonomous migration defect in neighboring cells, due at least in part to derepression of reelin transcription in a manner dependent on the forkhead box (FOX) transcription factor FOXG1. Treatments aimed at either blocking downstream AKT signaling or inactivating reelin restored migration. These findings suggest a central AKT-FOXG1-reelin signaling pathway in FMCD and support pathway inhibitors as potential treatments or therapies for some forms of focal epilepsy.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Movimiento Celular/genética , Modelos Animales de Enfermedad , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Imagen por Resonancia Magnética , Malformaciones del Desarrollo Cortical/enzimología , Malformaciones del Desarrollo Cortical/cirugía , Ratones , Datos de Secuencia Molecular , Mosaicismo , Mutación/genética , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fenotipo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Recombinación Genética/genética , Proteína Reelina , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
7.
PLoS Genet ; 9(4): e1003412, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23593018

RESUMEN

DJ-1, a Parkinson's disease (PD)-associated gene, has been shown to protect against oxidative stress in Drosophila. However, the molecular mechanism underlying oxidative stress-induced phenotypes, including apoptosis, locomotive defects, and lethality, in DJ-1-deficient flies is not fully understood. Here we showed that Daxx-like protein (DLP), a Drosophila homologue of the mammalian Death domain-associated protein (Daxx), was upregulated under oxidative stress conditions in the loss-of-function mutants of Drosophila DJ-1ß, a Drosophila homologue of DJ-1. DLP overexpression induced apoptosis via the c-Jun N-terminal kinase (JNK)/Drosophila forkhead box subgroup O (dFOXO) pathway, whereas loss of DLP increased resistance to oxidative stress and UV irradiation. Moreover, the oxidative stress-induced phenotypes of DJ-1ß mutants were dramatically rescued by DLP deficiency, suggesting that enhanced expression of DLP contributes to the DJ-1ß mutant phenotypes. Interestingly, we found that dFOXO was required for the increase in DLP expression in DJ-1ß mutants and that dFOXO activity was increased in the heads of DJ-1ß mutants. In addition, subcellular localization of DLP appeared to be influenced by DJ-1 expression so that cytosolic DLP was increased in DJ-1ß mutants. Similarly, in mammalian cells, Daxx translocation from the nucleus to the cytosol was suppressed by overexpressed DJ-1ß under oxidative stress conditions; and, furthermore, targeted expression of DJ-1ß to mitochondria efficiently inhibited the Daxx translocation. Taken together, our findings demonstrate that DJ-1ß protects flies against oxidative stress- and UV-induced apoptosis by regulating the subcellular localization and gene expression of DLP, thus implying that Daxx-induced apoptosis is involved in the pathogenesis of DJ-1-associated PD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de Drosophila , Factores de Transcripción Forkhead , Proteínas del Tejido Nervioso , Proteínas Nucleares , Estrés Oxidativo , Enfermedad de Parkinson , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de la radiación , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mutación , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/efectos de la radiación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Tolerancia a Radiación/genética , Rayos Ultravioleta
8.
J Biol Chem ; 288(8): 5660-72, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23303188

RESUMEN

Mutations in PINK1 (PTEN-induced putative kinase 1) are tightly linked to autosomal recessive Parkinson disease (PD). Although more than 50 mutations in PINK1 have been discovered, the role of these mutations in PD pathogenesis remains poorly understood. Here, we characterized 17 representative PINK1 pathogenic mutations in both mammalian cells and Drosophila. These mutations did not affect the typical cleavage patterns and subcellular localization of PINK1 under both normal and damaged mitochondria conditions in mammalian cells. However, PINK1 mutations in the kinase domain failed to translocate Parkin to mitochondria and to induce mitochondrial aggregation. Consistent with the mammalian data, Drosophila PINK1 mutants with mutations in the kinase domain (G426D and L464P) did not genetically interact with Parkin. Furthermore, PINK1-null flies expressing the transgenic G426D mutant displayed defective phenotypes with increasing age, whereas L464P mutant-expressing flies exhibited the phenotypes at an earlier age. Collectively, these results strongly support the hypothesis that the kinase activity of PINK1 is essential for its function and for regulating downstream Parkin functions in mitochondria. We believe that this study provides the basis for understanding the molecular and physiological functions of various PINK1 mutations and provides insights into the pathogenic mechanisms of PINK1-linked PD.


Asunto(s)
Mutación , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/fisiología , Adenosina Trifosfato/metabolismo , Animales , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Drosophila melanogaster , Fibroblastos/citología , Células HEK293 , Células HeLa , Humanos , Inmunohistoquímica/métodos , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/metabolismo , Neuronas/metabolismo , Fenotipo , Proteínas Quinasas/metabolismo , Transgenes , Ubiquitina-Proteína Ligasas/metabolismo
9.
Biochem Biophys Res Commun ; 377(3): 975-80, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18957282

RESUMEN

PTEN-induced putative kinase 1 (PINK1) and Parkin, encoded by their respective genes associated with Parkinson's disease (PD), are linked in a common pathway involved in the protection of mitochondrial integrity and function. However, the mechanism of their interaction at the biochemical level has not been investigated yet. Using both mammalian and Drosophila systems, we here demonstrate that the PINK1 kinase activity is required for its function in mitochondria. PINK1 regulates the localization of Parkin to the mitochondria in its kinase activity-dependent manner. In detail, Parkin phosphorylation by PINK1 on its linker region promotes its mitochondrial translocation, and the RING1 domain of Parkin is critical for this occurrence. These results demonstrate the biochemical relationship between PINK1, Parkin, and the mitochondria and thereby suggest the possible mechanism of PINK-Parkin-associated PD pathogenesis.


Asunto(s)
Mitocondrias Musculares/enzimología , Enfermedad de Parkinson/enzimología , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Drosophila melanogaster/enzimología , Humanos , Fosforilación , Proteínas Quinasas/genética , Transfección
10.
Nature ; 441(7097): 1157-61, 2006 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-16672980

RESUMEN

Autosomal recessive juvenile parkinsonism (AR-JP) is an early-onset form of Parkinson's disease characterized by motor disturbances and dopaminergic neurodegeneration. To address its underlying molecular pathogenesis, we generated and characterized loss-of-function mutants of Drosophila PTEN-induced putative kinase 1 (PINK1), a novel AR-JP-linked gene. Here, we show that PINK1 mutants exhibit indirect flight muscle and dopaminergic neuronal degeneration accompanied by locomotive defects. Furthermore, transmission electron microscopy analysis and a rescue experiment with Drosophila Bcl-2 demonstrated that mitochondrial dysfunction accounts for the degenerative changes in all phenotypes of PINK1 mutants. Notably, we also found that PINK1 mutants share marked phenotypic similarities with parkin mutants. Transgenic expression of Parkin markedly ameliorated all PINK1 loss-of-function phenotypes, but not vice versa, suggesting that Parkin functions downstream of PINK1. Taken together, our genetic evidence clearly establishes that Parkin and PINK1 act in a common pathway in maintaining mitochondrial integrity and function in both muscles and dopaminergic neurons.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Dopamina/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Actividad Motora , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...