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











Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 3(1): 278, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483278

RESUMEN

Histone H3 lysine 4 methylation (H3K4me) is extensively regulated by numerous writer and eraser enzymes in mammals. Nine H3K4me enzymes are associated with neurodevelopmental disorders to date, indicating their important roles in the brain. However, interplay among H3K4me enzymes during brain development remains largely unknown. Here, we show functional interactions of a writer-eraser duo, KMT2A and KDM5C, which are responsible for Wiedemann-Steiner Syndrome (WDSTS), and mental retardation X-linked syndromic Claes-Jensen type (MRXSCJ), respectively. Despite opposite enzymatic activities, the two mouse models deficient for either Kmt2a or Kdm5c shared reduced dendritic spines and increased aggression. Double mutation of Kmt2a and Kdm5c clearly reversed dendritic morphology, key behavioral traits including aggression, and partially corrected altered transcriptomes and H3K4me landscapes. Thus, our study uncovers common yet mutually suppressive aspects of the WDSTS and MRXSCJ models and provides a proof of principle for balancing a single writer-eraser pair to ameliorate their associated disorders.


Asunto(s)
Anomalías Múltiples/genética , Agresión , Anomalías Craneofaciales/genética , Espinas Dendríticas/metabolismo , Trastornos del Crecimiento/genética , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Hipertricosis/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Animales , Modelos Animales de Enfermedad , Histona Demetilasas/deficiencia , N-Metiltransferasa de Histona-Lisina/deficiencia , Masculino , Metilación , Ratones , Proteína de la Leucemia Mieloide-Linfoide/deficiencia
3.
J Neurosci Res ; 95(1-2): 472-486, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870428

RESUMEN

The neuroimmune system is significantly sexually dimorphic, with sex differences evident in the number and activation states of microglia, in the activation of astrocytes, and in cytokine release and function. Neuroimmune cells and signaling are now recognized as critical for many neural functions throughout the life span, including synaptic plasticity and memory function. Here we address the question of how cytokines, astrocytes, and microglia contribute to memory, and specifically how neuroimmune modulation of memory differentially affects males and females. Understanding sex differences in both normal memory processes and dysregulation of memory in psychiatric and neurological disorders is critical for developing treatment and preventive strategies for memory disorders that are effective for both men and women. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Encéfalo , Citocinas/metabolismo , Sistema Inmunológico/fisiología , Memoria/fisiología , Neuroinmunomodulación/fisiología , Caracteres Sexuales , Animales , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/metabolismo , Humanos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Trastornos Mentales/complicaciones , Enfermedades del Sistema Nervioso/complicaciones
4.
Age (Dordr) ; 36(4): 9675, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24994537

RESUMEN

The α1-adrenergic receptor (α1AR) subtypes, α1AAR and α1BAR, have differential effects in the heart and central nervous system. Long-term stimulation of the α1AAR subtype prolongs lifespan and provides cardio- and neuro-protective effects. We examined the lifespan of constitutively active mutant (CAM)-α1BAR mice and the incidence of cancer in mice expressing the CAM form of either the α1AAR (CAM-α1AAR mice) or α1BAR. CAM-α1BAR mice have a significantly shortened lifespan when compared with wild-type (WT) animals; however, the effect was sex dependent. Female CAM-α1BAR mice lived significantly shorter lives, while the median lifespan of male CAM-α1BAR mice was not different when compared with that of WT animals. There was no difference in the incidence of cancer in either sex of CAM-α1BAR mice. The incidence of cancer was significantly decreased in CAM-α1AAR mice when compared with that in WT, and no sex-dependent effects were observed. Further study is warranted on cancer incidence after activation of each α1AR subtype and the effect of sex on lifespan following activation of the α1BAR. The implications of a decrease in cancer incidence following long-term α1AAR stimulation could lead to improved treatments for cancer.


Asunto(s)
Envejecimiento , Regulación Neoplásica de la Expresión Génica , Longevidad/fisiología , Neoplasias Experimentales/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Femenino , Estudios de Seguimiento , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/epidemiología , Transducción de Señal , Factores de Tiempo
5.
Mol Pharmacol ; 80(4): 747-58, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21791575

RESUMEN

The role of α(1)-adrenergic receptors (α(1)ARs) in cognition and mood is controversial, probably as a result of past use of nonselective agents. α(1A)AR activation was recently shown to increase neurogenesis, which is linked to cognition and mood. We studied the effects of long-term α(1A)AR stimulation using transgenic mice engineered to express a constitutively active mutant (CAM) form of the α(1A)AR. CAM-α(1A)AR mice showed enhancements in several behavioral models of learning and memory. In contrast, mice that have the α(1A)AR gene knocked out displayed poor cognitive function. Hippocampal brain slices from CAM-α(1A)AR mice demonstrated increased basal synaptic transmission, paired-pulse facilitation, and long-term potentiation compared with wild-type (WT) mice. WT mice treated with the α(1A)AR-selective agonist cirazoline also showed enhanced cognitive functions. In addition, CAM-α(1A)AR mice exhibited antidepressant and less anxious phenotypes in several behavioral tests compared with WT mice. Furthermore, the lifespan of CAM-α(1A)AR mice was 10% longer than that of WT mice. Our results suggest that long-term α(1A)AR stimulation improves synaptic plasticity, cognitive function, mood, and longevity. This may afford a potential therapeutic target for counteracting the decline in cognitive function and mood associated with aging and neurological disorders.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Afecto/fisiología , Cognición/fisiología , Longevidad/fisiología , Plasticidad Neuronal/fisiología , Receptores Adrenérgicos alfa 1/metabolismo , Afecto/efectos de los fármacos , Animales , Cognición/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Longevidad/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Cultivo de Órganos , Receptores Adrenérgicos alfa 1/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA