Another important issue is that receptors can exist in a triheter

Another important issue is that receptors can exist in a triheteromeric form that contains both a GluN2A and a GluN2B subunit (Hatton and Paoletti, 2005 and Rauner and Köhr, 2011), where the role of each subunit cannot be established using currently available pharmacological tools. Additional problems in relating function to GluN2 subunit composition include their different spatiotemporal expression profiles. For example, in younger neurons, GluN2B is predominant and as such

may mediate excitotoxicity Selleck Sirolimus simply because most NMDARs are GluN2B-containing. Moreover, GluN2B- and GluN2A-containing NMDARs may be enriched at extrasynaptic and synaptic sites, respectively (Groc et al., 2006, Martel et al., 2009 and Tovar and Westbrook, 1999, but see Harris and Pettit, 2007 and Thomas et al., 2006). Since receptor location may be a determinant of excitotoxicity irrespective of subunit composition (Hardingham and Bading, 2010), a location-dependent effect may be misinterpreted as a subunit-specific effect. We have eschewed pharmacocentric approaches in favor of molecular genetics to determine whether equivalent levels of Ca2+ influx through GluN2A- and GluN2B-containing NMDARs differentially affect neuronal viability. We hypothesized that any differences would be due to their large CTDs because this is the primary area of sequence divergence, as well as being the part of GluN2 known to bind intracellular

signaling/scaffolding proteins (Ryan et al., 2008). By studying signaling from wild-type and chimeric GluN2A/2B subunits, using both acutely expressed subunits Afatinib as well as a mouse knockin model, we find that the presence of the CTD2B in an NMDAR renders Ca2+ influx through this receptor more toxic than the presence of CTD2A. This difference is observed in vivo as well as in vitro and is attributable in part to enhanced physical/functional coupling of CTD2B to the PSD-95/nNOS signaling cassette, which suppresses prosurvival CREB-mediated

gene expression, rendering neurons vulnerable to excitotoxic cell death. We wanted to investigate whether the subtype of GluN2 CTD influences the excitotoxicity of a given amount of NMDAR-mediated ion flux. We created constructs encoding and chimeric receptors based on GluN2B and GluN2A but with their respective CTDs replaced (denoted as CTR) with each other’s (GluN2B2A(CTR) and GluN2A2B(CTR), respectively, Figure 1A). In rat hippocampal neurons, we first expressed either wild-type GluN2BWT or GluN2B2A(CTR), at a developmental stage where endogenous NMDARs are overwhelmingly GluN2B-containing (Martel et al., 2009). Expression of GluN2BWT or GluN2B2A(CTR) both enhanced whole-cell currents to a similar level (Figure 1B) and did not differentially affect the proportion of extrasynaptic NMDARs (Figure 1C), as assessed by the “quantal block” method of irreversibly blocking synaptically located NMDARs (Papadia et al., 2008).

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>