Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Sheng Li Xue Bao ; 69(5): 703-714, 2017 Oct 25.
Artículo en Zh | MEDLINE | ID: mdl-29063118

RESUMEN

DREAM (downstream regulatory element antagonist modulator), Calsenilin and KChIP3 (potassium channel interacting protein 3) belong to the neuronal calcium sensor (NCS) superfamily, which transduces the intracellular calcium signaling into a variety of activities. They are encoded by the same gene locus, but have distinct subcellular locations. DREAM was first found to interact with DRE (downstream regulatory element) site in the vicinity of the promoter of prodynorphin gene to suppress gene transcription. Calcium can disassemble this interaction by binding reversibly to DREAM protein on its four EF-hand motifs. Apart from having calcium dependent DRE site binding, DREAM can also interact with other transcription factors, such as cAMP responsive element binding protein (CREB), CREB-binding protein (CBP) and cAMP responsive element modulator (CREM), by this concerted actions, DREAM extends the gene pool under its control. DREAM is predominantly expressed in central nervous system with its highest level in cerebellum, and accumulating evidence demonstrated that DREAM might play important roles in pain sensitivity. Novel findings have shown that DREAM is also involved in learning and memory processes, Alzheimer's disease and stroke. This mini-review provides a brief introduction of its discovery history and protein structure properties, focusing on the mechanism of DREAM nuclear translocation and gene transcription regulation functions.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Interacción con los Canales Kv/fisiología , Proteínas Represoras/fisiología , Animales , Señalización del Calcio/fisiología , Humanos , Proteínas de Interacción con los Canales Kv/genética , Umbral del Dolor , Proteínas Represoras/genética
2.
J Neurosci Res ; 93(2): 253-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25250856

RESUMEN

The superfamily of importin-ß-related proteins is the largest class of nuclear transport receptors and can be generally divided into importins and exportins according to their transport directions. Eleven importins and seven exportins have been identified, and the expression patterns of both classes are important for their functions in nucleocytoplasmic transport activities. This study demonstrates that all of the importins (importin-ß; transportin-1, -2, and -3; and importin-4, -5, -7, -8, -9, -11, and -13) and all the exportins (exportin-1, -2, -4, -5, -6, -7, and -t) are differentially expressed in the cerebral cortex, cerebellum, hippocampus, and brainstem and in primary cultures of cerebral cortical astrocytes and neurons. For astrocytes, we observed that different importins and exportins displayed different expression changes during 0-6 hr of ischemia treatment, especially an increase of both the mRNA and the protein of exportin-7. Immunostaining showed that exportin-7 accumulated inside the nucleus and around the nuclear envelope. In addition, we noticed an increased cytoplasmic distribution of one of the cargo proteins of exportin-7, LKB1, an important element in maintaining energy homeostasis. This increased cytoplasmic distribution was accompanied by an increased expression of exportin-7 under ischemia in astrocytes. We demonstrate that exportin-7 responds to ischemia in astrocytes and that this response involves translocation of LKB1, a protein that plays important roles during metabolic stress, from the nucleus to the cytoplasm.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/ultraestructura , Isquemia Encefálica/patología , Regulación de la Expresión Génica/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP ran/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Hipoxia de la Célula/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos , Regulación de la Expresión Génica/genética , Carioferinas/genética , Carioferinas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/citología , Neuronas/metabolismo , Fotoblanqueo , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , ARN Mensajero/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo , Proteína de Unión al GTP ran/genética
3.
Neurochem Res ; 40(9): 1929-44, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26248512

RESUMEN

Cadmium (Cd), a highly ubiquitous toxic heavy metal, can contaminate the environment, including agricultural soil, water and air, via industrial runoff and other sources of pollution. Cd accumulated in the body via direct exposure or through the food chain results in neurodegeneration and many other diseases. Previous studies on its toxicity in the central nervous system (CNS) focused mainly on neurons. To obtain a more comprehensive understanding of Cd toxicity for the CNS, we investigated how astrocytes respond to acute and chronic Cd exposure and its toxic molecular mechanisms. When primary cultures of cerebral cortical astrocytes incubated with 1-300 µM CdCl2, morphological changes, LDH release and cell death were observed in a time and dose-dependent manner. Further studies demonstrated that acute and chronic Cd treatment phosphorylated JNK, p38 and Akt to different degrees, while ERK1/2 was only phosphorylated under low doses of Cd (10 µM) exposure. Inhibition of JNK and PI3K/Akt, but not of p38, could partially protect astrocyte from cytotoxicity in chronic and acute Cd exposure. Moreover, Cd also induced a strong calcium signal, while BAPTA, a specific intracellular calcium (Ca(2+)) chelator, prevented Cd-induced intracellular increase of calcium levels in astrocytes; inhibited the Cd-induced activation of ERK1/2, JNK, p38 and Akt; and also significantly reduced astrocyte cell death. All of these results suggested that the Cd-Ca(2+)-MAPK and PI3K/Akt signaling pathways were involved in Cd-induced toxicity in astrocytes. This toxicity involvement indicates that these pathways may be exploited as a target for the prevention of Cd-induced neurodegenerative diseases.


Asunto(s)
Astrocitos/efectos de los fármacos , Cadmio/toxicidad , Señalización del Calcio , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Astrocitos/enzimología , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos ICR
4.
Glia ; 61(10): 1748-65, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23922257

RESUMEN

Water movement plays vital roles in both physiological and pathological conditions in the brain. Astrocytes are responsible for regulating this water movement and are the major contributors to brain edema in pathological conditions. Aquaporins (AQPs) in astrocytes play critical roles in the regulation of water movement in the brain. AQP1, 3, 4, 5, 8, and 9 have been reported in the brain. Compared with AQP1, 4, and 9, AQP3, 5, and 8 are less studied. Among the lesser known AQPs, AQP5, which has multiple functions identified outside the central nervous system, is also indicated to be involved in hypoxia injury in astrocytes. In our study, AQP5 expression could be detected both in primary cultures of astrocytes and neurons, and AQP5 expression in astrocytes was confirmed in 1- to 4-week old primary cultures of astrocytes. AQP5 was localized on the cytoplasmic membrane and in the cytoplasm of astrocytes. AQP5 expression was downregulated during ischemia treatment and upregulated after scratch-wound injury, which was also confirmed in a middle cerebral artery occlusion model and a stab-wound injury model in vivo. The AQP5 increased after scratch injury was polarized to the migrating processes and cytoplasmic membrane of astrocytes in the leading edge of the scratch-wound, and AQP5 over-expression facilitated astrocyte process elongation after scratch injury. Taken together, these results indicate that AQP5 might be an important water channel in astrocytes that is differentially expressed during various brain injuries.


Asunto(s)
Acuaporina 5/metabolismo , Astrocitos/metabolismo , Lesiones Encefálicas/patología , Encéfalo/metabolismo , Encéfalo/patología , Regulación de la Expresión Génica/fisiología , Animales , Animales Recién Nacidos , Hipoxia de la Célula/fisiología , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Factores de Tiempo , Transfección , Heridas y Lesiones/patología
5.
Glia ; 61(12): 2063-77, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24123203

RESUMEN

Astrocyte activation is a hallmark of central nervous system injuries resulting in glial scar formation (astrogliosis). The activation of astrocytes involves metabolic and morphological changes with complex underlying mechanisms, which should be defined to provide targets for astrogliosis intervention. Astrogliosis is usually accompanied by an upregulation of glial fibrillary acidic protein (GFAP). Using an in vitro scratch injury model, we scratched primary cultures of cerebral cortical astrocytes and observed an influx of calcium in the form of waves spreading away from the wound through gap junctions. Using the calcium blocker BAPTA-AM and the JNK inhibitor SP600125, we demonstrated that the calcium wave triggered the activation of JNK, which then phosphorylated the transcription factor c-Jun to facilitate the binding of AP-1 to the GFAP gene promoter to switch on GFAP upregulation. Blocking calcium mobilization with BAPTA-AM in an in vivo stab wound model reduced GFAP expression and glial scar formation, showing that the calcium signal, and the subsequent regulation of downstream signaling molecules, plays an essential role in brain injury response. Our findings demonstrated that traumatic scratch injury to astrocytes triggered a calcium influx from the extracellular compartment and activated the JNK/c-Jun/AP-1 pathway to switch on GFAP expression, identifying a previously unreported signaling cascade that is important in astrogliosis and the physiological response following brain injury.


Asunto(s)
Astrocitos/metabolismo , Calcio/metabolismo , Genes jun/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Transcripción AP-1/metabolismo , Animales , Astrocitos/citología , Señalización del Calcio/genética , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Gliosis/genética , Ratones , Ratones Endogámicos ICR , Factor de Transcripción AP-1/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA