Abstract

Pharmacological blockade or genetic knockout of neuronal connexin 36 (Cx36)-containing gap junctions reduces neuronal death caused by ischemia, traumatic brain injury and NMDA receptor (NMDAR)-mediated excitotoxicity. However, whether Cx36 gap junctions contribute to neuronal death via channel-dependent or channel-independent mechanism remains an open question. To address this, we manipulated connexin protein expression via lentiviral transduction of mouse neuronal cortical cultures and analyzed neuronal death twenty-four hours following administration of NMDA (a model of NMDAR excitotoxicity) or oxygen-glucose deprivation (a model of ischemic injury). In cultures prepared from wild-type mice, over-expression and knockdown of Cx36-containing gap junctions augmented and prevented, respectively, neuronal death from NMDAR-mediated excitotoxicity and ischemia. In cultures obtained form from Cx36 knockout mice, re-expression of functional gap junction channels, containing either neuronal Cx36 or non-neuronal Cx43 or Cx31, resulted in increased neuronal death following insult. In contrast, the expression of communication-deficient gap junctions (containing mutated connexins) did not have this effect. Finally, the absence of ethidium bromide uptake in non-transduced wild-type neurons two hours following NMDAR excitotoxicity or ischemia suggested the absence of active endogenous hemichannels in those neurons. Taken together, these results suggest a role for neuronal gap junctions in cell death via a connexin type-independent mechanism that likely relies on channel activities of gap junctional complexes among neurons. A possible contribution of gap junction channel-permeable death signals in neuronal death is discussed.

Highlights

  • Gap junctions connect neighboring cells via intercellular channels that allow direct electrical communication and propagation of ions and small molecules between coupled cells [1]

  • We addressed whether: (1) changes in the amount of neuronal gap junctions directly affect neuronal death and survival and (2) neuronal death related to gap junctions depends upon their channel activity

  • Appropriate expression and formation of gap junctional complexes from WT and mutant connexin lentiviruses were characterized, as described below, in neuronal cortical cultures obtained from connexin 36 (Cx36) knockout mice and in stably-transduced HeLa cells

Read more

Summary

Introduction

Gap junctions connect neighboring cells via intercellular channels that allow direct electrical communication and propagation of ions and small molecules between coupled cells [1]. Group II metabotropic glutamate receptors (mGluR) control the injury-mediated increase in neuronal gap junction coupling and the blockade of these receptors both prevents the up-regulation of neuronal gap junctions and reduces injury-dependent neuronal death [6, 15] It has been suggested previously [20, 21] that overactivation of glutamate receptors (mainly NMDARs) is an underlying mechanism for the secondary neuronal death following neuronal injury. The increase in neuronal gap junction coupling that results from activation of group II mGluRs, is a significant, underlying driver of expansive neuronal death We proposed that this is a general mechanism for neuronal death during different types of neuronal injury [22, 23]. Stated another way: is the role of Cx36 in neuronal death limited to providing cell-cell fluxes of ions or metabolites through gap junction channels or are there activities specific to this connexin isoform beyond its channel functions? This was addressed in the present study

Materials and Methods
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call