Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Genes Cells ; 21(11): 1176-1194, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27717094

RESUMEN

Terminal differentiation of neurons is accompanied by irreversible exit from the cell cycle and expression of neuronal phenotypes. The molecular mechanism whereby committed neuronal progenitors lose their ability to reenter the cell cycle is largely unknown. Here, we report that the nuclear transport system is rapidly remodeled in primary cortical progenitor cells (CPCs) at the very beginning of neuronal terminal differentiation. High levels of Ran GTPase-activating protein 1 (RanGAP), a key regulator of the Ran GTP-GDP cycle, in primary CPCs are drastically reduced upon neuronal induction. Small ubiquitin-like modifier (SUMO)-2/3-conjugated RanGAP undergoes desumoylation and degradation in neuronally committed CPCs, where reduced RanGAP levels impede the nuclear import of nucleocytoplasmic shuttling proteins including the DNA replication initiation factor Cdc6. Furthermore, RNAi-mediated down-regulation of RanGAP expression in undifferentiated CPCs induces neuronal phenotypes including cell cycle exit. Our data suggest that remodeling of the RanGAP-mediated nuclear transport system plays a key role in cell cycle exit for terminal differentiation of cortical neurons.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas Activadoras de GTPasa/metabolismo , Neurogénesis , Animales , Astrocitos/citología , Ciclo Celular , Corteza Cerebral/citología , Regulación hacia Abajo , Femenino , Ratones , Ratones Endogámicos ICR
2.
Nat Commun ; 7: 10943, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26971449

RESUMEN

Neurons rely heavily on mitochondria for their function and survival. Mitochondrial dysfunction contributes to the pathogenesis of neurodegenerative diseases such as Parkinson's disease. PGC-1α is a master regulator of mitochondrial biogenesis and function. Here we identify necdin as a potent PGC-1α stabilizer that promotes mitochondrial biogenesis via PGC-1α in mammalian neurons. Expression of genes encoding mitochondria-specific proteins decreases significantly in necdin-null cortical neurons, where mitochondrial function and expression of the PGC-1α protein are reduced. Necdin strongly stabilizes PGC-1α by inhibiting its ubiquitin-dependent degradation. Forced expression of necdin enhances mitochondrial function in primary cortical neurons and human SH-SY5Y neuroblastoma cells to prevent mitochondrial respiratory chain inhibitor-induced degeneration. Moreover, overexpression of necdin in the substantia nigra in vivo of adult mice protects dopaminergic neurons against degeneration in experimental Parkinson's disease. These data reveal that necdin promotes mitochondrial biogenesis through stabilization of endogenous PGC-1α to exert neuroprotection against mitochondrial insults.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Biogénesis de Organelos , Trastornos Parkinsonianos/genética , Sustancia Negra/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Corteza Cerebral/citología , ADN Mitocondrial , Modelos Animales de Enfermedad , Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Trastornos Parkinsonianos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Estabilidad Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Cell Signal ; 28(2): 94-107, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26655377

RESUMEN

Cellular signaling mediated by the EGF receptor (EGFR) plays a key role in controlling proliferation and differentiation of cortical progenitor cells (CPCs). However, regulatory mechanisms of EGFR signaling in CPCs remain largely unknown. Here we demonstrate that necdin, a MAGE (melanoma antigen) family protein, interacts with EGFR in primary CPCs and represses its downstream signaling linked to astrocyte differentiation. EGFR was autophosphorylated and interacted with necdin in EGF-stimulated CPCs. Necdin bound to autophosphorylated EGFR via its tyrosine kinase domain. EGF-induced phosphorylation of ERK was enhanced in necdin-null CPCs, where the interaction between EGFR and the adaptor protein Grb2 was strengthened, suggesting that endogenous necdin suppresses the EGFR/ERK signaling pathway in CPCs. In necdin-null CPCs, astrocyte differentiation induced by the gliogenic cytokine cardiotrophin-1 was significantly accelerated in the presence of EGF, and inhibition of EGFR/ERK signaling abolished the acceleration. Furthermore, necdin strongly suppressed astrocyte differentiation induced by overexpression of EGFR or its ligand binding-defective mutant equivalent to a glioblastoma-associated EGFR variant. These results suggest that necdin acts as an intrinsic suppressor of the EGFR/ERK signaling pathway in EGF-responsive CPCs to restrain astroglial development in a cell-autonomous manner.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/embriología , Receptores ErbB/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/metabolismo , Animales , Astrocitos/citología , Astrocitos/enzimología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Corteza Cerebral/metabolismo , Receptores ErbB/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteína Adaptadora GRB2/antagonistas & inhibidores , Proteína Adaptadora GRB2/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/enzimología , Dominios Proteicos
4.
PLoS One ; 9(6): e99503, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24911587

RESUMEN

Necdin, a pleiotropic protein that promotes differentiation and survival of mammalian neurons, is a member of MAGE (melanoma antigen) family proteins that share a highly conserved MAGE homology domain. Several MAGE proteins interact with ubiquitin E3 ligases and modulate their activities. However, it remains unknown whether MAGE family proteins interact with SUMO (small ubiquitin-like modifier) E3 ligases such as PIAS (protein inhibitor of activated STAT) family, Nsmce2/Mms21 and Cbx4/Pc2. In the present study, we examined whether necdin interacts with these SUMO E3 ligases. Co-immunoprecipitation analysis revealed that necdin, MAGED1, MAGEF1 and MAGEL2 bound to PIAS1 but not to Nsmce2 or Cbx4. These SUMO E3 ligases bound to MAGEA1 but failed to interact with necdin-like 2/MAGEG1. Necdin bound to PIAS1 central domains that are highly conserved among PIAS family proteins and suppressed PIAS1-dependent sumoylation of the substrates STAT1 and PML (promyelocytic leukemia protein). Remarkably, necdin promoted degradation of PIAS1 via the ubiquitin-proteasome pathway. In transfected HEK293A cells, amino- and carboxyl-terminally truncated mutants of PIAS1 bound to necdin but failed to undergo necdin-dependent ubiquitination. Both PIAS1 and necdin were associated with the nuclear matrix, where the PIAS1 terminal deletion mutants failed to localize, implying that the nuclear matrix is indispensable for necdin-dependent ubiquitination of PIAS1. Our data suggest that necdin suppresses PIAS1 both by inhibiting SUMO E3 ligase activity and by promoting ubiquitin-dependent degradation.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Humanos , Antígenos Específicos del Melanoma/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteínas Inhibidoras de STAT Activados/química , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Especificidad por Sustrato , Sumoilación , Ubiquitinación
5.
PLoS One ; 9(1): e84460, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24392139

RESUMEN

Neural precursor cells (NPCs) in the neocortex exhibit a high proliferation capacity during early embryonic development and give rise to cortical projection neurons after maturation. Necdin, a mammal-specific MAGE (melanoma antigen) family protein that possesses anti-mitotic and pro-survival activities, is expressed abundantly in postmitotic neurons and moderately in tissue-specific stem cells or progenitors. Necdin interacts with E2F transcription factors and suppresses E2F1-dependent transcriptional activation of the cyclin-dependent kinase Cdk1 gene. Here we show that necdin serves as a suppressor of NPC proliferation in the embryonic neocortex. Necdin is moderately expressed in the ventricular zone of mouse embryonic neocortex, in which proliferative cell populations are significantly increased in necdin-null mice. In the neocortex of necdin-null embryos, expression of Cdk1 and Sox2, a stem cell marker, is significantly increased, whereas expression of p16, a cyclin-dependent kinase inhibitor, is markedly diminished. Cdk1 and p16 expression levels are also significantly increased and decreased, respectively, in primary NPCs prepared from necdin-null embryos. Intriguingly, necdin interacts directly with Bmi1, a Polycomb group protein that suppresses p16 expression and promotes NPC proliferation. In HEK293A cells transfected with luciferase reporter constructs, necdin relieves Bmi1-dependent repression of p16 promoter activity, whereas Bmi1 counteracts necdin-mediated repression of E2F1-dependent Cdk1 promoter activity. In lentivirus-infected primary NPCs, necdin overexpression increases p16 expression, suppresses Cdk1 expression, and inhibits NPC proliferation, whereas Bmi1 overexpression suppresses p16 expression, increases Cdk1 expression, and promotes NPC proliferation. Our data suggest that embryonic NPC proliferation in the neocortex is regulated by the antagonistic interplay between necdin and Bmi1.


Asunto(s)
Neocórtex/citología , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Embrión de Mamíferos/metabolismo , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Neocórtex/embriología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Unión Proteica
6.
J Neurosci ; 33(25): 10362-73, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23785149

RESUMEN

Neural stem cells (NSCs) reside in vivo in hypoxic environments, and NSC proliferation is enhanced in vitro under hypoxic conditions. Various adaptive responses to hypoxia are mediated by hypoxia-inducible factors (HIFs), a family of basic helix-loop-helix Per-Arnt-Sim (PAS) transcription factors. Necdin, a MAGE (melanoma antigen) family protein, is expressed abundantly in postmitotic neurons and possesses potent antimitotic and antiapoptotic activities. We here report that hypoxia induces degradation of the necdin protein in primary NSCs by HIF-mediated ubiquitin-proteasome system. Necdin was expressed in primary NSCs prepared from the ganglionic eminences of mouse embryos. Hypoxia enhanced neurosphere formation of NSCs, in which the necdin protein level was significantly reduced. Primary NSCs prepared from necdin-deficient mice exhibited higher rates of proliferation and apoptosis than those from wild-type mice in normoxia, whereas there were no significant differences in the proliferation and apoptosis rates between necdin-deficient and wild-type NSCs in hypoxia. HIF-2α was predominantly expressed in hypoxic NSCs, where expression of HIF-responsive genes was upregulated. HIF-2α interacted with necdin via its PAS domain, which enhanced necdin ubiquitination. Lentivirus-mediated expression of the PAS domain in primary NSCs promoted necdin degradation and enhanced NSC proliferation in normoxia, whereas a small-molecule inhibitor of HIF-2α translation stabilized the necdin protein and reduced NSC proliferation in hypoxia. These results suggest that oxygen tension regulates the necdin protein level in NSCs through HIF-2α-mediated proteasomal degradation to modulate their proliferation and apoptosis.


Asunto(s)
Apoptosis/genética , Apoptosis/fisiología , Proliferación Celular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Células-Madre Neurales/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Oxígeno/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Western Blotting , Separación Celular , Fragmentación del ADN , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Lentivirus/genética , Ratones , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitina/metabolismo
7.
J Neurosci ; 32(16): 5562-72, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22514318

RESUMEN

The forkhead transcription factor Foxo1 regulates energy homeostasis by modulating gene expression in the hypothalamus. Foxo1 undergoes post-translational modifications such as phosphorylation and acetylation, which modulate its functional activities. Sirtuin1 (Sirt1), a nicotinamide adenine dinucleotide-dependent protein deacetylase, regulates the acetylation status of Foxo1 in mammalian cells. Necdin, a pleiotropic protein required for neuronal development and survival, interacts with both Sirt1 and p53 to facilitate p53 deacetylation. The necdin gene (Ndn), an imprinted gene transcribed only from the paternal allele, is strongly expressed in hypothalamic neurons. Here, we demonstrate that necdin controls the acetylation status of Foxo1 in vivo in hypothalamic arcuate neurons to modulate the thyroid function. Necdin forms a stable ternary complex with Sirt1 and Foxo1, diminishes Foxo1 acetylation, and suppresses the transcriptional activity of Foxo1 in vitro. Paternal Ndn mutant mice express high levels of acetylated Foxo1 and mRNAs encoding agouti-related protein and neuropeptide Y in the hypothalamus in vivo during the juvenile period. The mutant mice exhibit endocrine dysfunction characteristic of hypothalamic hypothyroidism. Chemically induced hyperthyroidism and hypothyroidism lead to hypothalamic responses similar to those under necdin-deficient and excessive conditions, respectively, suggesting that thyroid hormone serves as a negative regulator of this system. These results suggest that necdin regulates Foxo1 acetylation and neuropeptide gene expression in the arcuate neurons to modulate the hypothalamic-pituitary-thyroid axis during development.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Hipotálamo/citología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Glándula Tiroides/metabolismo , Acetilación , Factores de Edad , Proteína Relacionada con Agouti/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Dióxido de Carbono/metabolismo , Línea Celular Transformada , Inmunoprecipitación de Cromatina , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Neuropéptido Y/metabolismo , Proteínas Nucleares/deficiencia , Consumo de Oxígeno/genética , ARN Mensajero , Sirtuina 1/metabolismo , Tirotropina/sangre , Tirotropina/genética , Tiroxina/sangre , Transfección , Triyodotironina/sangre
8.
PLoS One ; 7(1): e30948, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22292082

RESUMEN

White adipose tissues are composed mainly of white fat cells (adipocytes), which play a key role in energy storage and metabolism. White adipocytes are terminally differentiated postmitotic cells and arise from their progenitor cells (preadipocytes) or mesenchymal stem cells residing in white adipose tissues. Thus, white adipocyte number is most likely controlled by the rate of preadipocyte proliferation, which may contribute to the etiology of obesity. However, little is known about the molecular mechanisms that regulate preadipocyte proliferation during adipose tissue development. Necdin, which is expressed predominantly in postmitotic neurons, is a pleiotropic protein that possesses anti-mitotic and pro-survival activities. Here we show that necdin functions as an intrinsic regulator of white preadipocyte proliferation in developing adipose tissues. Necdin is expressed in early preadipocytes or mesenchymal stem cells residing in the stromal compartment of white adipose tissues in juvenile mice. Lentivirus-mediated knockdown of endogenous necdin expression in vivo in adipose tissues markedly increases fat mass in juvenile mice fed a high-fat diet until adulthood. Furthermore, necdin-null mutant mice exhibit a greater expansion of adipose tissues due to adipocyte hyperplasia than wild-type mice when fed the high-fat diet during the juvenile and adult periods. Adipose stromal-vascular cells prepared from necdin-null mice differentiate in vitro into a significantly larger number of adipocytes in response to adipogenic inducers than those from wild-type mice. These results suggest that necdin prevents excessive preadipocyte proliferation induced by adipogenic stimulation to control white adipocyte number during adipose tissue development.


Asunto(s)
Adipocitos/fisiología , Proliferación Celular , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Células Madre/fisiología , Adipocitos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Células Madre/metabolismo , Células del Estroma/metabolismo , Células del Estroma/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA