Elsevier

Experimental Neurology

Volume 238, Issue 2, December 2012, Pages 93-102
Experimental Neurology

TNF-α triggers rapid membrane insertion of Ca2 + permeable AMPA receptors into adult motor neurons and enhances their susceptibility to slow excitotoxic injury

https://doi.org/10.1016/j.expneurol.2012.08.004Get rights and content

Abstract

Excitotoxicity (caused by over-activation of glutamate receptors) and inflammation both contribute to motor neuron (MN) damage in amyotrophic lateral sclerosis (ALS) and other diseases of the spinal cord. Microglial and astrocytic activation in these conditions results in release of inflammatory mediators, including the cytokine, tumor necrosis factor‐alpha (TNF-α). TNF-α has complex effects on neurons, one of which is to trigger rapid membrane insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type glutamate receptors, and in some cases, specific insertion of GluA2 lacking, Ca2 + permeable AMPA receptors (Ca-perm AMPAr). In the present study, we use a histochemical stain based upon kainate stimulated uptake of cobalt ions (“Co2 + labeling”) to provide the first direct demonstration of the presence of substantial numbers of Ca-perm AMPAr in ventral horn MNs of adult rats under basal conditions. We further find that TNF-α exposure causes a rapid increase in the numbers of these receptors, via a phosphatidylinositol 3 kinase (PI3K) and protein kinase A (PKA) dependent mechanism. Finally, to assess the relevance of TNF-α to slow excitotoxic MN injury, we made use of organotypic spinal cord slice cultures. Co2 + labeling revealed that MNs in these cultures possess Ca-perm AMPAr. Addition of either a low level of TNF-α, or of the glutamate uptake blocker, trans-pyrrolidine-2,4-dicarboxylic acid (PDC) to the cultures for 48 h resulted in little MN injury. However, when combined, TNF-α + PDC caused considerable MN degeneration, which was blocked by the AMPA/kainate receptor blocker, 2,3-Dihydroxy-6-nitro-7-sulfamoylbenzo (F) quinoxaline (NBQX), or the Ca-perm AMPAr selective blocker, 1-naphthyl acetylspermine (NASPM). Thus, these data support the idea that prolonged TNF-α elevation, as may be induced by glial activation, acts in part by increasing the numbers of Ca-perm AMPAr on MNs to enhance injurious excitotoxic effects of deficient astrocytic glutamate transport.

Highlights

► First direct demonstration of functional Ca2 + permeable AMPA receptors in adult MNs. ► TNF-α rapidly increases numbers of these receptors in the plasma membrane. ► This TNF-α effect occurs via a PI3K and PKA dependent mechanism. ► TNF-α also increases MN Ca2 + permeable AMPA receptors in slice cultures. ► Low level TNF-α exposure enhances slow excitotoxic MN injury in slice cultures.

Introduction

Amyotrophic lateral sclerosis (ALS) is a catastrophic disease characterized by relatively selective degeneration of upper and lower (spinal) motor neurons (MNs), for which there is as yet no good treatment. Observations of deficiencies in spinal cord glutamate uptake in human ALS patients, resulting from selective loss of the astrocytic glutamate transporter, GLT-1, suggested an excitotoxic contribution (Rothstein et al., 1992, Rothstein et al., 1995). It is progressively evident that cell death in ALS depends critically upon interactions between MNs and neighboring cells. Activation of both microglia and astrocytes occurs prominently in both human disease and animal models of ALS (Philips and Robberecht, 2011, Wegorzewska et al., 2009), likely contributing to MN injury via a number of mechanisms, a key one of which may be release of factors including the cytokine tumor necrosis factor-alpha (TNF-α) (Mhatre et al., 2004, Tweedie et al., 2007).

Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are tetramers, composed of combinations of subunits (GluA1-4). Most AMPA receptors are Ca2 + impermeable due to the presence of a GluA2 subunit; GluA2 lacking receptors are Ca2 + permeable. TNF-α has complex effects, which could be either beneficial or injury promoting (Cheng et al., 1994, Ghezzi and Mennini, 2001, Hermann et al., 2001, Pickering et al., 2005). One injury promoting effect is the rapid insertion of AMPA type glutamate receptors into the plasma membrane (Beattie et al., 2002, Hermann et al., 2001, Stellwagen and Malenka, 2006, Yu et al., 2002). Interestingly, the majority of these AMPA receptors contain GluA1 (Beattie et al., 2002, Leonoudakis et al., 2008, Ogoshi et al., 2005, Stellwagen et al., 2005). Furthermore, although some studies showed membrane increases in GluA2 as well, TNF-α preferentially caused the rapid insertion of GluA2 lacking, Ca2 +-permeable AMPA receptors (Ca-perm AMPAr) in hippocampal pyramidal neurons (which normally have relatively low levels of these receptors) (Ogoshi et al., 2005, Stellwagen et al., 2005) and spinal dorsal horn neurons (Choi et al., 2010), resulting in increased susceptibility to excitotoxic injury (Leonoudakis et al., 2008).

Embryonic spinal MNs possess significant numbers of Ca-perm AMPAr under basal conditions (Carriedo et al., 1995, Carriedo et al., 1996, Van Den Bosch et al., 2000, Vandenberghe et al., 2000), there is indirect evidence that adult MNs possess them as well (Corona and Tapia, 2007, Darman et al., 2004, Tateno et al., 2004, Williams et al., 1997, Yin et al., 2007), and it is likely that their presence plays a role in the high susceptibility of MNs to excitotoxic injury. Past studies concerning effects of TNF-α on MNs, and consequences for disease have mixed results. One recent study found spinal cord trauma to cause a rapid appearance of Ca-perm AMPAr on neurons via a TNF-α dependent mechanism, which contributed to the resultant injury (Ferguson et al., 2008). However, another study reported TNF-α to reduce numbers of Ca-perm AMPAr on cultured MNs (Rainey-Smith et al., 2010). Regarding possible roles in models of ALS, TNF-α appeared to contribute to MN loss in the wobbler mouse model (Bigini et al., 2008), but knock out of the TNF-α gene had no effect on disease progression in superoxide dismutase type 1 (SOD1) mutant mouse ALS models (Gowing et al., 2006).

The present study employs a histochemical stain based upon kainate-stimulated uptake of Co2 + ions (“Co2 + labeling”) to confirm the presence of functional Ca-perm AMPAr on adult ventral horn MNs in lumbar spinal cord slices, and examine effects of TNF-α on their numbers. We further make use of organotypic spinal cord slice cultures to determine effects of low levels of TNF-α on slow excitotoxic MN degeneration caused by subtoxic exposures to the glutamate uptake blocker, trans-pyrrolidine-2,4-dicarboxylic acid (PDC). Present results may be relevant to ALS or other conditions in which slow excitotoxicity and inflammation contribute to MN damage.

Section snippets

Animal tissue collection and handling

All animal procedures were conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animal and approved by the University of California Irvine Institutional Care and Use committee.

Ca-perm AMPAr are present in spinal MNs from developing and adult rats

As discussed above, numerous studies have used techniques including kainate stimulated Co2 + uptake labeling (“Co2 + labeling”) or electrophysiological recording to demonstrate the presence of Ca-perm AMPAr on embryonic and neonatal MNs. Although it has been widely assumed that adult MNs are likely to possess these receptors as well, the evidence to date for their presence in adults is largely indirect, based upon measurements of relative AMPA subunit mRNA or protein levels (Shaw and Ince, 1997,

Summary of principal findings

In the present study, we use a well-established histochemical technique to directly demonstrate, for the first time, the presence of functional Ca-perm AMPAr on adult MNs, and find that acute TNF-α exposure induces a rapid increase in the numbers of these receptors in the neuronal membrane, via a signal transduction pathway that includes both PI3K and PKA. We further find that MNs in organotypic spinal cord slice cultures possess these Ca-perm AMPAr under basal conditions (after 10 days in

Conclusions

It is clear that the survival of MNs depends critically upon interrelationships with neighboring cells, and neuroinflammation, with astrocyte dysfunction, likely contributes to MN damage in a number of conditions including ALS. TNF-α is an important cytokine that accumulates in diseased spinal cord, and has complex effects that could well modulate MN survival. However, as discussed above, it has differing effects in different in vitro and animal models, and its net effects on MN survival in ALS

Acknowledgments

This work was supported by NIH grants NS36548 (JHW) and NS065219 (LSS), and a grant from the Muscular Dystrophy Association (JHW).

References (65)

  • I. Nagano et al.

    Ventral root avulsion leads to downregulation of GluR2 subunit in spinal motoneurons in adult rats

    Neuroscience

    (2003)
  • F. Ogoshi et al.

    Tumor necrosis-factor-alpha (TNF-alpha) induces rapid insertion of Ca2 +-permeable alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA)/kainate (Ca-A/K) channels in a subset of hippocampal pyramidal neurons

    Exp. Neurol.

    (2005)
  • T. Philips et al.

    Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease

    Lancet Neurol.

    (2011)
  • R.M. Pruss et al.

    Agonist-activated cobalt uptake identifies divalent cation-permeable kainate receptors on neurons and glial cells

    Neuron

    (1991)
  • C. Richichi et al.

    Mechanisms of seizure-induced ‘transcriptional channelopathy’ of hyperpolarization-activated cyclic nucleotide gated (HCN) channels

    Neurobiol. Dis.

    (2008)
  • L. Tolosa et al.

    TNF-alpha potentiates glutamate-induced spinal cord motoneuron death via NF-kappaB

    Mol. Cell. Neurosci.

    (2011)
  • L. Van Den Bosch et al.

    Ca(2 +)-permeable AMPA receptors and selective vulnerability of motor neurons

    J. Neurol. Sci.

    (2000)
  • J.H. Weiss et al.

    Ca2 +–Zn2 + permeable AMPA or kainate receptors: possible key factors in selective neurodegeneration

    Trends Neurosci.

    (2000)
  • H.Z. Yin et al.

    Marked synergism between mutant SOD1 and glutamate transport inhibition in the induction of motor neuronal degeneration in spinal cord slice cultures

    Brain Res.

    (2012)
  • H. Yin et al.

    Cortical neurones with Ca2 + permeable AMPA/kainate channels display distinct receptor immunoreactivity and are GABAergic

    Neurobiol. Dis.

    (1994)
  • H.Z. Yin et al.

    Intrathecal infusion of a Ca2 + permeable AMPA channel blocker slows loss of both motor neurons and of the astrocyte glutamate transporter, GLT-1 in a mutant SOD1 rat model of ALS

    Exp. Neurol.

    (2007)
  • Z. Yu et al.

    Tumor necrosis factor alpha increases neuronal vulnerability to excitotoxic necrosis by inducing expression of the AMPA-glutamate receptor subunit GluR1 via an acid sphingomyelinase- and NF-kappaB-dependent mechanism

    Neurobiol. Dis.

    (2002)
  • J.Y. Zou et al.

    TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition

    Brain Res.

    (2005)
  • M.E. Alexianu et al.

    The role of calcium-binding proteins in selective motoneuron vulnerability in amyotrophic lateral sclerosis

    Ann. Neurol.

    (1994)
  • E.C. Beattie et al.

    Control of synaptic strength by glial TNFalpha

    Science

    (2002)
  • J.D. Bell et al.

    PICK1-mediated GluR2 endocytosis contributes to cellular injury after neuronal trauma

    Cell Death Differ.

    (2009)
  • S.G. Carriedo et al.

    In vitro kainate injury to large, SMI-32(+) spinal neurons is Ca2 + dependent

    Neuroreport

    (1995)
  • S.G. Carriedo et al.

    Motor neurons are selectively vulnerable to AMPA/kainate receptor-mediated injury in vitro

    J. Neurosci.

    (1996)
  • S.G. Carriedo et al.

    AMPA exposures induce mitochondrial Ca(2 +) overload and ROS generation in spinal motor neurons in vitro

    J. Neurosci.

    (2000)
  • J. Darman et al.

    Viral-induced spinal motor neuron death is non-cell-autonomous and involves glutamate excitotoxicity

    J. Neurosci.

    (2004)
  • J.L. Elliott et al.

    Parvalbumin is a marker of ALS-resistant motor neurons

    Neuroreport

    (1995)
  • A.R. Ferguson et al.

    Cell death after spinal cord injury is exacerbated by rapid TNF alpha-induced trafficking of GluR2-lacking AMPARs to the plasma membrane

    J. Neurosci.

    (2008)
  • Cited by (43)

    • Glaucoma and neuroinflammation: An overview

      2021, Survey of Ophthalmology
      Citation Excerpt :

      In another study, the inhibition of TNF-α in an animal model of glaucoma induced a reduction in the number of activated microglial cells, which ultimately was associated with a decreased loss of RGCs.168 Moreover, TNF-α upregulates the expression of the receptor for a-amino-3-hydroxy-5-methyl4-isoxazolepropionic acid (AMPAR), which represents a major excitatory subgroup of glutamate receptors in neuronal cells and synergizes with TNF-α in triggering excitotoxic cell death.232 One more key topic in deciphering how RGCs survival is affected by neuro-inflammation and the cascade of molecular events following its excessive activation is extracellular matrix (ECM) remodeling.

    • A potential role for microglia in stress- and drug-induced plasticity in the nucleus accumbens: A mechanism for stress-induced vulnerability to substance use disorder

      2019, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Silent synapses become functional upon the insertion of AMPARs into the synapse (Dong and Nestler, 2014). Interestingly, it seems microglia are capable of regulating AMPA receptor expression on neurons via the secretion of the pro-inflammatory cytokine tumor necrosis factor- alpha (TNF-α) (Beattie et al., 2002; He et al., 2012; Leonoudakis et al., 2004, 2008; Lewitus et al., 2016; Yin et al., 2012). The literature is somewhat mixed, with some researchers finding that TNF-α enhances AMPAR signaling through the insertion of calcium permeable AMPARs (He et al., 2012; Leonoudakis et al., 2008); and other studies finding decreased AMPAR signaling in response to TNF-α (Lewitus et al., 2016).

    • Excitotoxicity

      2018, Comprehensive Toxicology: Third Edition
    View all citing articles on Scopus
    View full text