4.7 Article

Distinct Modifications in Kv2.1 Channel via Chemokine Receptor CXCR4 Regulate Neuronal Survival-Death Dynamics

期刊

JOURNAL OF NEUROSCIENCE
卷 32, 期 49, 页码 17725-+

出版社

SOC NEUROSCIENCE
DOI: 10.1523/JNEUROSCI.3029-12.2012

关键词

-

资金

  1. University of Iowa Office of the Vice President of Research
  2. American Epilepsy Society [Pc-190423]
  3. Epilepsy Foundation
  4. National Institutes of Health/National Institute of Neurological Disorders and Stroke [NS069898, NS045549]

向作者/读者索取更多资源

The chemokine stromal cell-derived factor-1 alpha (SDF-1 alpha) has multiple effects on neuronal activity, survival, and death under conditions that generate a proinflammatory microenvironment within the brain, via signaling through C-X-C-type chemokine receptor 4 (CXCR4), although the underlying cellular/molecular mechanisms are unclear. Using rat hippocampal neurons, we investigated distinct modifications in the voltage-gated K+ (Kv) channel Kv2.1 in response to short- and long-term SDF-1 alpha/CXCR4-mediated signaling as an underlying mechanism for CXCR4-dependent regulation of neuronal survival and death. Acute exposure of neurons to SDF-1 alpha led to dynamic dephosphorylation and altered localization of Kv2.1 channel, resulting in enhanced voltage-dependent activation of Kv2.1-based delayed-rectifier Kv currents (I-DR). These changes were dependent on CXCR4- and/or NMDA receptor-mediated activation of calcineurin and provide neuroprotection. However, prolonged SDF-1 alpha treatment leads to CXCR4-mediated activation of p38 mitogen-activated protein kinase, resulting in phosphorylation of Kv2.1 at S800 and enhanced surface trafficking of the channel protein, resulting in increased I-DR/Kv2.1 current density. This, in combination with sustained dephosphorylation-induced enhancement of the voltage-dependent activation of I-DR/Kv2.1, predisposed neurons to excessive K+ efflux, a vital step for the neuronal apoptotic program. Such apoptotic death was dependent on CXCR4 and Kv2.1 function and was absent in cells expressing the Kv2.1-S800A mutant channel. Furthermore, similar modifications in Kv2.1 and CXCR4/Kv2.1-dependent apoptosis were observed following treatment of neurons with the human immunodeficiency virus-1 (HIV-1) glycoprotein gp120. Therefore, distinct modifications in Kv2.1 in response to short-and long-term CXCR4-mediated signaling could provide a basis for neuroprotection or apoptosis in neuropathologies, such as neuroinflammation, stroke, brain tumors, and HIV-associated neurodegeneration.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据