Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Stroke ; 55(6): 1562-1571, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38716662

RESUMEN

BACKGROUND: While stroke is a recognized short-term sequela of traumatic brain injury, evidence about long-term ischemic stroke risk after traumatic brain injury remains limited. METHODS: The Atherosclerosis Risk in Communities Study is an ongoing prospective cohort comprised of US community-dwelling adults enrolled in 1987 to 1989 followed through 2019. Head injury was defined using self-report and hospital-based diagnostic codes and was analyzed as a time-varying exposure. Incident ischemic stroke events were physician-adjudicated. We used Cox regression adjusted for sociodemographic and cardiovascular risk factors to estimate the hazard of ischemic stroke as a function of head injury. Secondary analyses explored the number and severity of head injuries; the mechanism and severity of incident ischemic stroke; and heterogeneity within subgroups defined by race, sex, and age. RESULTS: Our analysis included 12 813 participants with no prior head injury or stroke. The median follow-up age was 27.1 years (25th-75th percentile=21.1-30.5). Participants were of median age 54 years (25th-75th percentile=49-59) at baseline; 57.7% were female and 27.8% were Black. There were 2158 (16.8%) participants with at least 1 head injury and 1141 (8.9%) participants with an incident ischemic stroke during follow-up. For those with head injuries, the median age to ischemic stroke was 7.5 years (25th-75th percentile=2.2-14.0). In adjusted models, head injury was associated with an increased hazard of incident ischemic stroke (hazard ratio [HR], 1.34 [95% CI, 1.12-1.60]). We observed evidence of dose-response for the number of head injuries (1: HR, 1.16 [95% CI, 0.97-1.40]; ≥2: HR, 1.94 [95% CI, 1.39-2.71]) but not for injury severity. We observed evidence of stronger associations between head injury and more severe stroke (National Institutes of Health Stroke Scale score ≤5: HR, 1.31 [95% CI, 1.04-1.64]; National Institutes of Health Stroke Scale score 6-10: HR, 1.64 [95% CI, 1.06-2.52]; National Institutes of Health Stroke Scale score ≥11: HR, 1.80 [95% CI, 1.18-2.76]). Results were similar across stroke mechanism and within strata of race, sex, and age. CONCLUSIONS: In this community-based cohort, head injury was associated with subsequent ischemic stroke. These results suggest the importance of public health interventions aimed at preventing head injuries and primary stroke prevention among individuals with prior traumatic brain injuries.


Asunto(s)
Traumatismos Craneocerebrales , Vida Independiente , Accidente Cerebrovascular Isquémico , Humanos , Femenino , Masculino , Persona de Mediana Edad , Accidente Cerebrovascular Isquémico/epidemiología , Incidencia , Factores de Riesgo , Adulto , Traumatismos Craneocerebrales/epidemiología , Estudios Prospectivos , Anciano , Estudios de Cohortes
2.
Glia ; 66(10): 2174-2187, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30194875

RESUMEN

Astrogliosis is a hallmark of neuroinflammatory disorders such as multiple sclerosis (MS). A detailed understanding of the underlying molecular mechanisms governing astrogliosis might facilitate the development of therapeutic targets. We investigated whether Nav1.5 expression in astrocytes plays a role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine model of MS. We created a conditional knockout of Nav1.5 in astrocytes and determined whether this affects the clinical course of EAE, focal macrophage and T cell infiltration, and diffuse activation of astrocytes. We show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, unexpectedly, in a sex-specific manner. Removal of Nav1.5 in astrocytes leads to increased inflammation in female mice with EAE, including increased astroglial response and infiltration of T cells and phagocytic monocytes. These cellular changes are consistent with more severe EAE clinical scores. Additionally, we found evidence suggesting possible dysregulation of the immune response-particularly with regard to infiltrating macrophages and activated microglia-in female Nav1.5 KO mice compared with WT littermate controls. Together, our results show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, in a sex-specific manner.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Caracteres Sexuales , Animales , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Monocitos/metabolismo , Monocitos/patología , Esclerosis Múltiple/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Médula Espinal/metabolismo , Médula Espinal/patología , Linfocitos T/metabolismo , Linfocitos T/patología
3.
Mol Pain ; 13: 1744806916688016, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28326929

RESUMEN

Neuropathic pain is a major complication of spinal cord injury, and despite aggressive efforts, this type of pain is refractory to available clinical treatment. Our previous work has demonstrated a structure-function link between dendritic spine dysgenesis on nociceptive sensory neurons in the intermediate zone, laminae IV/V, and chronic pain in central nervous system and peripheral nervous system injury models of neuropathic pain. To extend these findings, we performed a follow-up structural analysis to assess whether dendritic spine remodeling occurs on superficial dorsal horn neurons located in lamina II after spinal cord injury. Lamina II neurons are responsible for relaying deep, delocalized, often thermally associated pain commonly experienced in spinal cord injury pathologies. We analyzed dendritic spine morphometry and localization in tissue obtained from adult rats exhibiting neuropathic pain one-month following spinal cord injury. Although the total density of dendritic spines on lamina II neurons did not change after spinal cord injury, we observed an inverse relationship between the densities of thin- and mushroom-shaped spines: thin-spine density decreased while mushroom-spine density increased. These structural changes were specifically noted along dendritic branches within 150 µm from the soma, suggesting a possible adverse contribution to nociceptive circuit function. Intrathecal treatment with NSC23766, a Rac1-GTPase inhibitor, significantly reduced spinal cord injury-induced changes in both thin- and mushroom-shaped dendritic spines. Overall, these observations demonstrate that dendritic spine remodeling occurs in lamina II, regulated in part by the Rac1-signaling pathway, and suggests that structural abnormalities in this spinal cord region may also contribute to abnormal nociception after spinal cord injury.


Asunto(s)
Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Células del Asta Posterior/ultraestructura , Traumatismos de la Médula Espinal/patología , Análisis de Varianza , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Nocodazol/farmacología , Nocodazol/uso terapéutico , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/patología , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata , Traumatismos de la Médula Espinal/tratamiento farmacológico
4.
Glia ; 64(10): 1628-45, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26919466

RESUMEN

Voltage-gated sodium channels are required for electrogenesis in excitable cells. Their activation, triggered by membrane depolarization, generates transient sodium currents that initiate action potentials in neurons, cardiac, and skeletal muscle cells. Cells that have not traditionally been considered to be excitable (nonexcitable cells), including glial cells, also express sodium channels in physiological conditions as well as in pathological conditions. These channels contribute to multiple functional roles that are seemingly unrelated to the generation of action potentials. Here, we discuss the dynamics of sodium channel expression in astrocytes and microglia, and review evidence for noncanonical roles in effector functions of these cells including phagocytosis, migration, proliferation, ionic homeostasis, and secretion of chemokines/cytokines. We also examine possible mechanisms by which sodium channels contribute to the activity of glial cells, with an eye toward therapeutic implications for central nervous system disease. GLIA 2016;64:1628-1645.


Asunto(s)
Astrocitos/metabolismo , Microglía/metabolismo , Canales de Sodio/metabolismo , Potenciales de Acción/fisiología , Animales , Humanos , Sistema Nervioso/citología
5.
Glia ; 62(7): 1162-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24740847

RESUMEN

Astrogliosis is a prominent feature of many, if not all, pathologies of the brain and spinal cord, yet a detailed understanding of the underlying molecular pathways involved in the transformation from quiescent to reactive astrocyte remains elusive. We investigated the contribution of voltage-gated sodium channels to astrogliosis in an in vitro model of mechanical injury to astrocytes. Previous studies have shown that a scratch injury to astrocytes invokes dual mechanisms of migration and proliferation in these cells. Our results demonstrate that wound closure after mechanical injury, involving both migration and proliferation, is attenuated by pharmacological treatment with tetrodotoxin (TTX) and KB-R7943, at a dose that blocks reverse mode of the Na(+) /Ca(2+) exchanger (NCX), and by knockdown of Nav 1.5 mRNA. We also show that astrocytes display a robust [Ca(2+) ]i transient after mechanical injury and demonstrate that this [Ca(2+) ]i response is also attenuated by TTX, KB-R7943, and Nav 1.5 mRNA knockdown. Our results suggest that Nav 1.5 and NCX are potential targets for modulation of astrogliosis after injury via their effect on [Ca(2+) ]i .


Asunto(s)
Astrocitos/fisiología , Gliosis/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Heridas y Lesiones/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Corteza Cerebral , Técnicas de Silenciamiento del Gen , Gliosis/tratamiento farmacológico , Canal de Sodio Activado por Voltaje NAV1.5/genética , Estimulación Física , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Tetrodotoxina/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Heridas y Lesiones/tratamiento farmacológico
6.
Neuroreport ; 25(15): 1208-15, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25144393

RESUMEN

Astrocytes actively participate in the response of the central nervous system to injury, including in multiple sclerosis. Astrocytes can play both beneficial and detrimental roles in response to neuroinflammation; however, in extreme cases, astrogliosis can result in the formation of a glial scar, which can impede the regeneration of injured neurons. Although astrocytes do not express the voltage-gated sodium channel Nav1.5 in the nonpathological human brain, they exhibit robust upregulation of Nav1.5 within acute and chronic multiple sclerosis lesions. Recent work has indicated that Nav1.5 contributes to the pathways that regulate glial scar formation in vitro through modulation of intracellular Ca levels. However, the temporal dynamics of astrocytic Nav1.5 channel expression in response to neuroinflammatory pathologies has not been investigated. We examined astrocytes from mice with monophasic and chronic-relapsing (CR) experimental autoimmune encephalomyelitis (EAE) by immunohistochemical analysis to determine whether Nav1.5 is expressed in these cells, and whether the expression correlates with the severity of disease and/or phases of relapse and remission. Our results demonstrate that Nav1.5 is upregulated in astrocytes in situ in a temporal manner that correlates with disease severity in both monophasic and CR EAE. Further, in CR EAE, Nav1.5 expression is upregulated during relapses and subsequently attenuated during periods of remission. These observations are consistent with the suggestion that Nav1.5 can play a role in the response of astrocytes to inflammatory pathologies in the central nervous system and suggest Nav1.5 may be a potential therapeutic target to modulate reactive astrogliosis in vivo.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple Crónica Progresiva/metabolismo , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Animales , Inmunohistoquímica , Vértebras Lumbares , Ratones Biozzi , Ratones Endogámicos C57BL , Corteza Motora/metabolismo , Índice de Severidad de la Enfermedad , Médula Espinal/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA