Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders

Key Points

  • NMDARs (N-methyl-D-aspartate receptors) have long been known for their role in neuropathology, and inappropriate activity is implicated in neuronal loss in acute disorders such as stroke and traumatic brain injury. Certain chronic neurodegenerative diseases are also associated with abnormal NMDAR activity, including Huntington's and Alzheimer's diseases. However, the destructive effects of NMDAR activity are in striking contrast to the observation that the survival and resistance to trauma of several neuronal types is boosted by physiological synaptic NMDAR activity and function. Thus, there is a dichotomy of NMDAR signalling.

  • Recent studies have shown that cellular responses to NMDAR activation can depend on the receptor location. Activation of synaptic NMDARs, particularly when activated trans-synaptically, promotes neuronal health, whereas chronic activation of extrasynaptic NMDARs couples to cell death pathways. Differences are observed even when the overall Ca2+ loads triggered via the two routes are similar.

  • Synaptic NMDAR activity strongly promotes neuronal health by initiating a programme of transcriptional changes that promote resistance to various traumatic stimuli. Synaptic NMDARs control a nuclear Ca2+-regulated multi-gene program that protects against excitotoxic and apoptotic insults. Transcriptional suppression of key components of the intrinsic apoptosis pathway also restricts the apoptotic potential of neurons. Moreover, synaptic NMDAR activity promotes resistance to oxidative insults by boosting intrinsic antioxidant defences through transcriptional changes of proteins encoding antioxidant genes and regulatory factors.

  • Extrasynaptic NMDAR activity is coupled to several signalling pathways that promote neuronal death or vulnerability to trauma. These include the dephosphorylation and inactivation of the pro-survival transcription factor cyclic-AMP response element binding protein (CREB), nuclear import of the pro-death transcription factor forkhead box protein O (FOXO), inactivation of extracellular signal-regulated kinase 1/2 (ERK1/2) mitogen-activated protein (MAP) kinase, and calpain-dependent striatal enriched tyrosine phosphatase (STEP) cleavage and activation of p38 MAP kinase.

  • A shift in the balance from synaptic towards extrasynaptic NMDAR signalling may be an important factor in the aetiology of neurodegenerative diseases. In Huntington's disease, mutant huntingtin causes a specific increase in extrasynaptic NMDAR currents. Furthermore, extrasynaptic NMDAR activity in turn promotes the toxicity of mutant huntingtin and synaptic NMDAR activity reduces mutant huntingtin toxicity by promoting the formation of non-toxic inclusions. In acute ischaemic trauma, cell death may be caused in part by an upregulation and activation of extrasynaptic NMDARs

  • In treating disorders associated with abnormal NMDAR activity, therapies aimed at selectively blocking chronic extrasynaptic NMDAR activity without interfering with normal synaptic NMDAR activity may be better tolerated and more efficacious than conventional antagonists. The NMDAR antagonist memantine is well suited to this role, which may explain its tolerance in humans and its recently demonstrated efficacy in preclinical models of Huntington's disease.

Abstract

There is a long-standing paradox that NMDA (N-methyl-D-aspartate) receptors (NMDARs) can both promote neuronal health and kill neurons. Recent studies show that NMDAR-induced responses depend on the receptor location: stimulation of synaptic NMDARs, acting primarily through nuclear Ca2+ signalling, leads to the build-up of a neuroprotective 'shield', whereas stimulation of extrasynaptic NMDARs promotes cell death. These differences result from the activation of distinct genomic programmes and from opposing actions on intracellular signalling pathways. Perturbations in the balance between synaptic and extrasynaptic NMDAR activity contribute to neuronal dysfunction in acute ischaemia and Huntington's disease, and could be a common theme in the aetiology of neurodegenerative diseases. Neuroprotective therapies should aim to both enhance the effect of synaptic activity and disrupt extrasynaptic NMDAR-dependent death signalling.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: χ-shaped model of NMDAR-dependent excitotoxicity.
Figure 2: Neuroprotective pathways activated by synaptic NMDAR activity.
Figure 3: Opposing effects of synaptic and extrasynaptic NMDAR signalling on gene expression.
Figure 4: Synaptic NMDAR-dependent transcriptional changes have both anti-apoptotic and antioxidant effects.
Figure 5: Activators and effectors of extrasynaptic NMDAR activity.
Figure 6: Clinical relevance of the balance between synaptic and extrasynaptic NMDAR activity.

Similar content being viewed by others

References

  1. Aamodt, S. M. & Constantine-Paton, M. The role of neural activity in synaptic development and its implications for adult brain function. Adv. Neurol. 79, 133–144 (1999).

    CAS  PubMed  Google Scholar 

  2. Bliss, T. V. & Collingridge, G. L. A synaptic model of memory: long-term potentiation in the hippocampus. Nature 361, 31–39 (1993).

    Article  CAS  PubMed  Google Scholar 

  3. Tovar, K. R. & Westbrook, G. L. The incorporation of NMDA receptors with a distinct subunit composition at nascent hippocampal synapses in vitro. J. Neurosci. 19, 4180–4188 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rosenmund, C., Feltz, A. & Westbrook, G. L. Synaptic NMDA receptor channels have a low open probability. J. Neurosci. 15, 2788–2795 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cottrell, J. R., Dube, G. R., Egles, C. & Liu, G. Distribution, density, and clustering of functional glutamate receptors before and after synaptogenesis in hippocampal neurons. J. Neurophysiol. 84, 1573–1587 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. Petralia, R. S. et al. Organization of NMDA receptors at extrasynaptic locations. Neuroscience 167, 68–87 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Rusakov, D. A. & Kullmann, D. M. Extrasynaptic glutamate diffusion in the hippocampus: ultrastructural constraints, uptake, and receptor activation. J. Neurosci. 18, 3158–3170 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Massey, P. V. et al. Differential roles of NR2A and NR2B-containing NMDA receptors in cortical long-term potentiation and long-term depression. J. Neurosci. 24, 7821–7828 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhao, J. P. & Constantine-Paton, M. NR2A−/− mice lack long-term potentiation but retain NMDA receptor and L-type Ca2+ channel-dependent long-term depression in the juvenile superior colliculus. J. Neurosci. 27, 13649–13654 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lucas, D. R. & Newhouse, J. P. The toxic effect of sodium L-glutamate on the inner layers of the retina. AMA Arch. Ophthalmol. 58, 193–201 (1957).

    Article  CAS  PubMed  Google Scholar 

  11. Curtis, D. R., Phillis, J. W. & Watkins, J. C. Chemical excitation of spinal neurones. Nature 183, 611–612 (1959).

    Article  CAS  PubMed  Google Scholar 

  12. Olney, J. W. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science 164, 719–721 (1969).

    Article  CAS  PubMed  Google Scholar 

  13. Choi, D. W. Ionic dependence of glutamate neurotoxicity. J. Neurosci. 7, 369–379 (1987). In one of several seminal contributions to the field, Choi demonstrated that an excitotoxic insult triggers temporally distinct phases of injury: acute Na+-dependent swelling and more delayed Ca2+-dependent injury. He showed that the Ca2+-dependent mechanism dominates at modest insult intensity, and discussed the potential role of NMDARs in this process.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Choi, D. W., Maulucci-Gedde, M. & Kriegstein, A. R. Glutamate neurotoxicity in cortical cell culture. J. Neurosci. 7, 357–368 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Choi, D. W., Koh, J. Y. & Peters, S. Pharmacology of glutamate neurotoxicity in cortical cell culture: attenuation by NMDA antagonists. J. Neurosci. 8, 185–196 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tymianski, M., Charlton, M. P., Carlen, P. L. & Tator, C. H. Source specificity of earlly calcium neurotoxicity in cultures embryonic spinal neurons. J. Neurosci. 13, 2085–2104 (1993). This paper was the first to show that Ca2+-mediated excitotoxicity was 'source specific'. It was shown that Ca2+ influx through bath-activation of NMDARs was more effective at killing neurons than Ca2+ influx through voltage-gated Ca2+ channels, taking into account overall Ca2+ load.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rothman, S. M. & Olney, J. W. Glutamate and the pathophysiology of hypoxic-ischemic brain damage. Ann. Neurol. 19, 105–111 (1986).

    Article  CAS  PubMed  Google Scholar 

  18. Choi, D. W. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1, 623–634 (1988).

    Article  CAS  PubMed  Google Scholar 

  19. Lipton, S. A. & Rosenberg, P. A. Excitatory amino acids as a final common pathway for neurologic disorders. N. Engl. J. Med. 330, 613–621 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Arundine, M. & Tymianski, M. Molecular mechanisms of glutamate-dependent neurodegeneration in ischemia and traumatic brain injury. Cell. Mol. Life Sci. 61, 657–668 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Dawson, T. M., Zhang, J., Dawson, V. L. & Snyder, S. H. Nitric oxide: cellular regulation and neuronal injury. Prog. Brain Res. 103, 365–369 (1994).

    Article  CAS  PubMed  Google Scholar 

  22. Camacho, A. & Massieu, L. Role of glutamate transporters in the clearance and release of glutamate during ischemia and its relation to neuronal death. Arch. Med. Res. 37, 11–18 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Fan, M. M. & Raymond, L. A. N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease. Prog. Neurobiol. 81, 272–293 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Kalia, L. V., Kalia, S. K. & Salter, M. W. NMDA receptors in clinical neurology: excitatory times ahead. Lancet Neurol. 7, 742–755 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ikonomidou, C. & Turski, L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury? Lancet Neurol. 1, 383–386 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Hetman, M. & Kharebava, G. Survival signaling pathways activated by NMDA receptors. Curr. Top. Med. Chem. 6, 787–799 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Hardingham, G. E. Pro-survival signalling from the NMDA receptor. Biochem. Soc. Trans. 34, 936–938 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gould, E., Cameron, H. A. & McEwen, B. S. Blockade of NMDA receptors increases cell death and birth in the developing rat dentate gyrus. J. Comp. Neurol. 340, 551–565 (1994).

    Article  CAS  PubMed  Google Scholar 

  29. Ikonomidou, C. et al. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science 283, 70–74 (1999). This study provided an elegant demonstration of the widespread vulnerability of the developing nervous system to NMDAR blockade. Subsequent studies would characterize the effect of NMDAR blockade in the mature CNS and the extent to which NMDAR blockade exacerbates neuronal loss following injury of metabolic inhibition.

    Article  CAS  PubMed  Google Scholar 

  30. Pohl, D. et al. NMDA antagonists and apoptotic cell death triggered by head trauma in developing rat brain. Proc. Natl Acad. Sci. USA 96, 2508–2513 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Monti, B. & Contestabile, A. Blockade of the NMDA receptor increases developmental apoptotic elimination of granule neurons and activates caspases in the rat cerebellum. Eur. J. Neurosci. 12, 3117–3123 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Adams, S. M., de Rivero Vaccari, J. C. & Corriveau, R. A. Pronounced cell death in the absence of NMDA receptors in the developing somatosensory thalamus. J. Neurosci. 24, 9441–9450 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ikonomidou, C., Stefovska, V. & Turski, L. Neuronal death enhanced by N-methyl-D-aspartate antagonists. Proc. Natl Acad. Sci. USA 97, 12885–12890 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tashiro, A., Sandler, V. M., Toni, N., Zhao, C. & Gage, F. H. NMDA-receptor-mediated, cell-specific integration of new neurons in adult dentate gyrus. Nature 442, 929–933 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Lipton, S. A. & Kater, S. B. Neurotransmitter regulation of neuronal outgrowth, plasticity and survival. Trends Neurosci. 12, 265–270 (1989).

    Article  CAS  PubMed  Google Scholar 

  36. Lipton, S. A. & Nakanishi, N. Shakespeare in Love - with NMDA receptors? Nature Med. 5, 270–271 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Hardingham, G. E. & Bading, H. The Yin and Yang of NMDA receptor signalling. Trends Neurosci. 26, 81–89 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Hardingham, G. E., Fukunaga, Y. & Bading, H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nature Neurosci. 5, 405–414 (2002). The concept that signalling resulting from synaptic and extrasynaptic NMDAR activation is linked to survival and death, respectively, was first shown here.

    Article  CAS  PubMed  Google Scholar 

  39. Leveille, F. et al. Neuronal viability is controlled by a functional relation between synaptic and extrasynaptic NMDA receptors. FASEB J. 22, 4258–4271 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Stanika, R. I. et al. Coupling diverse routes of calcium entry to mitochondrial dysfunction and glutamate excitotoxicity. Proc. Natl Acad. Sci. USA 106, 9854–9859 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ivanov, A. et al. Opposing role of synaptic and extrasynaptic NMDA receptors in regulation of the ERK activity in cultured rat hippocampal neurons. J. Physiol. 572, 789–798 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang, S. J. et al. Decoding NMDA Receptor Signaling: identification of genomic programs specifying neuronal survival and death. Neuron 53, 549–562 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Papadia, S. et al. Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nature Neurosci. 11, 476–487 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Okamoto, S. et al. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nature Med. 15, 1407–1413 (2009). This paper demonstrated that the balance between synaptic and extrasynaptic NMDAR signalling controls the formation of mutant huntingtin inclusions, neurotoxicity, and disease severity and progression in models of Huntington's disease. It also showed the therapeutic benefit of targeting extrasynaptic NMDAR activation pharmacalogically (with low-dose memantine).

    Article  CAS  PubMed  Google Scholar 

  45. Soriano, F. X. et al. Preconditioning doses of NMDA promote neuroprotection by enhancing neuronal excitability. J. Neurosci. 26, 4509–4518 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Isacson, O. On neuronal health. Trends Neurosci. 16, 306–308 (1993).

    Article  CAS  PubMed  Google Scholar 

  47. Papadia, S., Stevenson, P., Hardingham, N. R., Bading, H. & Hardingham, G. E. Nuclear Ca2+ and the cAMP response element-binding protein family mediate a late phase of activity-dependent neuroprotection. J. Neurosci. 25, 4279–4287 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Morgan, J. I., Cohen, D. R., Hempstead, J. L. & Curran, T. Mapping patterns of c-fos expression in the central nervous system after seizure. Science 237, 192–197 (1987).

    Article  CAS  PubMed  Google Scholar 

  49. Cole, A. J., Saffen, D. W., Baraban, J. M. & Worley, P. F. Rapid increase of an immediate early gene messenger RNA in hippocampal neurons by synaptic NMDA receptor activation. Nature 340, 474–476 (1989). One of the first demonstrations that synaptic NMDARs are an important mediator of activity-dependent gene transcription. Physiologically relevant, LTP-inducing stimuli were found to trigger robust induction of immediate early gene expression that was dependent on NMDAR activity.

    Article  CAS  PubMed  Google Scholar 

  50. Hardingham, G. E., Arnold, F. J. & Bading, H. Nuclear calcium signaling controls CREB-mediated gene expression triggered by synaptic activity. Nature Neurosci. 4, 261–267 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Zhang, S. J. et al. Nuclear calcium signaling controls expression of a large gene pool: identification of a gene program for acquired neuroprotection induced by synaptic activity. PLoS Genet. 5, e1000604 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Chrivia, J. C. et al. Phosphorylated CREB binds specifically to the nuclear protein CBP. Nature 365, 855–859 (1993).

    Article  CAS  PubMed  Google Scholar 

  53. Enslen, H. et al. Characterization of Ca2+/calmodulin-dependent protein kinase IV. Role in transcriptional regulation. J. Biol. Chem. 269, 15520–15527 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Kwok, R. P. S. et al. Nuclear protein CBP is coactivator for the transcription factor CREB. Nature 370, 223–226 (1994).

    Article  CAS  PubMed  Google Scholar 

  55. Matthews, R. P. et al. Calcium/calmodulin-dependent protein kinase types II and IV differentially regulate CREB-dependent gene expression. Mol. Cell. Biol. 14, 6107–6116 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Sun, P., Enslen, H., Myung, P. S. & Maurer, R. A. Differential activation of CREB by Ca2+/calmodulin-dependent protein kinases type II and type IV involves phosphorylation of a site that negatively regulates activity. Genes Dev. 8, 2527–2539 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. Hardingham, G. E., Chawla, S., Johnson, C. M. & Bading, H. Distinct functions of nuclear and cytoplasmic calcium in the control of gene expression. Nature 385, 260–265 (1997).

    Article  CAS  PubMed  Google Scholar 

  58. Chawla, S., Hardingham, G. E., Quinn, D. R. & Bading, H. CBP: a signal-regulated transcriptional coactivator controlled by nuclear calcium and CaM kinase IV. Science 281, 1505–1509 (1998).

    Article  CAS  PubMed  Google Scholar 

  59. Bonni, A. et al. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science 286, 1358–1362 (1999).

    Article  CAS  PubMed  Google Scholar 

  60. Mayr, B. & Montminy, M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nature Rev. Mol. Cell Biol. 2, 599–609 (2001).

    Article  CAS  Google Scholar 

  61. Lonze, B. E., Riccio, A., Cohen, S. & Ginty, D. D. Apoptosis, axonal growth defects, and degeneration of peripheral neurons in mice lacking CREB. Neuron 34, 371–385 (2002).

    Article  CAS  PubMed  Google Scholar 

  62. Mantamadiotis, T. et al. Disruption of CREB function in brain leads to neurodegeneration. Nature Genetics 31, 47–54 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Carlezon, W. A., Jr,, Duman, R. S. & Nestler, E. J. The many faces of CREB. Trends Neurosci. 28, 436–445 (2005).

    Article  CAS  PubMed  Google Scholar 

  64. Lee, B., Butcher, G. Q., Hoyt, K. R., Impey, S. & Obrietan, K. Activity-dependent neuroprotection and cAMP response element-binding protein (CREB): kinase coupling, stimulus intensity, and temporal regulation of CREB phosphorylation at serine 133. J. Neurosci. 25, 1137–1148 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Limback-Stokin, K., Korzus, E., Nagaoka-Yasuda, R. & Mayford, M. Nuclear calcium/calmodulin regulates memory consolidation. J. Neurosci. 24, 10858–10867 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Lau, D. & Bading, H. Synaptic activity-mediated suppression of p53 and induction of nuclear calcium-regulated neuroprotective genes promote survival through inhibition of mitochondrial permeability transition. J. Neurosci. 29, 4420–4429 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Leveille, F. et al. Suppression of the intrinsic apoptosis pathway by synaptic activity. J. Neurosci. 30, 2623–2635 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Favaron, M. et al. NMDA-stimulated expression of BDNF mRNA in cultured cerebellar granule neurones. Neuroreport 4, 1171–1174 (1993).

    Article  CAS  PubMed  Google Scholar 

  69. Jiang, X. et al. The excitoprotective effect of N-methyl-D-aspartate receptors is mediated by a brain-derived neurotrophic factor autocrine loop in cultured hippocampal neurons. J. Neurochem. 94, 713–722 (2005).

    Article  CAS  PubMed  Google Scholar 

  70. Thoenen, H., Barde, Y. A., Davies, A. M. & Johnson, J. E. Neurotrophic factors and neuronal death. Ciba Found. Symp. 126, 82–95 (1987).

    CAS  PubMed  Google Scholar 

  71. Hansen, H. H. et al. Mechanisms leading to disseminated apoptosis following NMDA receptor blockade in the developing rat brain. Neurobiol. Dis. 16, 440–453 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Vashishta, A. et al. Nuclear factor of activated T-cells isoform c4 (NFATc4/NFAT3) as a mediator of antiapoptotic transcription in NMDA receptor-stimulated cortical neurons. J. Neurosci. 29, 15331–15340 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Zheng, S. et al. NMDA-induced neuronal survival is mediated through nuclear factor I-A in mice. J. Clin. Invest. 120, 2446–2456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Shinoda, S. et al. Bim regulation may determine hippocampal vulnerability after injurious seizures and in temporal lobe epilepsy. J. Clin. Invest. 113, 1059–1068 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Lehtinen, M. K. et al. A conserved MST–FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell 125, 987–1001 (2006).

    Article  CAS  PubMed  Google Scholar 

  76. Salih, D. A. & Brunet, A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr. Opin. Cell Biol. 20, 126–136 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Dick, O. & Bading, H. Synaptic activity and nuclear calcium signaling protects hippocampal neurons from death signal-associated nuclear translocation of FoxO3a induced by extrasynaptic NMDA receptors. 19 Apr 2010 J. Biol. Chem. (doi:10.1074/jbc.M110.127654).

    Article  CAS  Google Scholar 

  78. Al-Mubarak, B., Soriano, F. X. & Hardingham, G. E. Synaptic NMDAR activity suppresses FOXO1 expression via a cis-acting FOXO binding site: FOXO1 is a FOXO target gene. Channels (Austin) 3, 233–238 (2009).

    Article  CAS  Google Scholar 

  79. Biteau, B., Labarre, J. & Toledano, M. B. ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin. Nature 425, 980–984 (2003).

    Article  CAS  PubMed  Google Scholar 

  80. Chang, T. S. et al. Characterization of mammalian sulfiredoxin and its reactivation of hyperoxidized peroxiredoxin through reduction of cysteine sulfinic acid in the active site to cysteine. J. Biol. Chem. 279, 50994–51001 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Budanov, A. V., Sablina, A. A., Feinstein, E., Koonin, E. V. & Chumakov, P. M. Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of bacterial AhpD. Science 304, 596–600 (2004).

    Article  CAS  PubMed  Google Scholar 

  82. Soriano, F. X. et al. Induction of sulfiredoxin expression and reduction of peroxiredoxin hyperoxidation by the neuroprotective Nrf2 activator 3H-1, 2-dithiole-3-thione. J. Neurochem. 107, 533–543 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Rhee, S. G., Woo, H. A., Bae, S. H. & Park, S. Sestrin 2 is not a reductase for cysteine sulfinic acid of peroxiredoxins. Antioxid. Redox Signal. 11, 739–745 (2009).

    Article  PubMed  CAS  Google Scholar 

  84. Hardingham, G. E. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem. Soc. Trans. 37, 1147–1160 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Bading, H., Ginty, D. D. & Greenberg, M. E. Regulation of gene expression in hippocampal neurons by distinct calcium signaling pathways. Science 260, 181–186 (1993). This was the first paper to show that Ca2+ regulation of gene expression can depend on the site of Ca2+ entry in neurons. Membrane depolarization, inducing Ca2+ influx through voltage-gated Ca2+ channels, was shown to activate overlapping but different promoter elements compared to Ca2+ influx triggered by bath-glutamate activation of NMDARs. This turned out to be due to activation of extrasynaptic NMDARs but this was not discovered until 2002. See also reference 38.

    Article  CAS  PubMed  Google Scholar 

  86. Hardingham, G. E., Chawla, S., Cruzalegui, F. H. & Bading, H. Control of recruitment and transcription-activating function of CBP determines gene regulation by NMDA receptors and L-type calcium channels. Neuron 22, 789–798 (1999).

    Article  CAS  PubMed  Google Scholar 

  87. Sala, C., Rudolph-Correia, S. & Sheng, M. Developmentally regulated NMDA receptor-dependent dephosphorylation of cAMP response element-binding protein (CREB) in hippocampal neurons. J. Neurosci. 20, 3529–3536 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ghosh, A. & Greenberg, M. E. Calcium signalling in neurons: molecular mechanisms and cellular consequences. Science 268, 239–247 (1995).

    Article  CAS  PubMed  Google Scholar 

  89. Hardingham, G. E. & Bading, H. Coupling of extrasynaptic NMDA receptors to a CREB shut-off pathway is developmentally regulated. Biochim. Biophys. Acta 1600, 148–153 (2002).

    Article  CAS  PubMed  Google Scholar 

  90. Dieterich, D. C. et al. Caldendrin–Jacob: a protein liaison that couples NMDA receptor signalling to the nucleus. PLoS Biol. 6, e34 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Chandler, L. J., Sutton, G., Dorairaj, N. R. & Norwood, D. N-methyl D-aspartate receptor-mediated bidirectional control of extracellular signal-regulated kinase activity in cortical neuronal cultures. J. Biol. Chem. 276, 2627–2636 (2001).

    Article  CAS  PubMed  Google Scholar 

  92. Bading, H. & Greenberg, M. E. Stimulation of protein tyrosine phosphorylation by NMDA receptor activation. Science 253, 912–914 (1991).

    Article  CAS  PubMed  Google Scholar 

  93. Kim, M. J., Dunah, A. W., Wang, Y. T. & Sheng, M. Differential roles of NR2A- and NR2B-containing NMDA receptors in Ras-ERK signaling and AMPA receptor trafficking. Neuron 46, 745–760 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Mulholland, P. J., Luong, N. T., Woodward, J. J. & Chandler, L. J. Brain-derived neurotrophic factor activation of extracellular signal-regulated kinase is autonomous from the dominant extrasynaptic NMDA receptor extracellular signal-regulated kinase shutoff pathway. Neuroscience 151, 419–427 (2008).

    Article  CAS  PubMed  Google Scholar 

  95. Xu, J. et al. Extrasynaptic NMDA receptors couple preferentially to excitotoxicity via calpain-mediated cleavage of STEP. J. Neurosci. 29, 9330–9343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Bano, D. et al. Cleavage of the plasma membrane Na+/Ca2+ exchanger in excitotoxicity. Cell 120, 275–285 (2005).

    Article  CAS  PubMed  Google Scholar 

  97. Kawasaki, H. et al. Activation and involvement of p38 mitogen-activated protein kinase in glutamate-induced apoptosis in rat cerebellar granule cells. J. Biol. Chem. 272, 18518–18521 (1997).

    Article  CAS  PubMed  Google Scholar 

  98. Cao, J. et al. Distinct requirements for p38α and c-Jun N-terminal kinase stress-activated protein kinases in different forms of apoptotic neuronal death. J. Biol. Chem. 279, 35903–35913 (2004).

    Article  CAS  PubMed  Google Scholar 

  99. Soriano, F. X. et al. Specific targeting of pro-death NMDA receptor signals with differing reliance on the NR2B PDZ ligand. J. Neurosci. 28, 10696–10710 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Okamoto, S., Krainc, D., Sherman, K. & Lipton, S. A. Antiapoptotic role of the p38 mitogen-activated protein kinase-myocyte enhancer factor 2 transcription factor pathway during neuronal differentiation. Proc. Natl Acad. Sci. USA 97, 7561–7566 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Soriano, F. X., Leveille, F., Papadia, S., Bell, K. F. & Hardingham, G. E. Neuronal activity controls the antagonistic balance between PGC-1α and SMRT in regulating antioxidant defences. Antioxid. Redox Signal. (in the press).

  102. Paul, S., Nairn, A. C., Wang, P. & Lombroso, P. J. NMDA-mediated activation of the tyrosine phosphatase STEP regulates the duration of ERK signaling. Nature Neurosci. 6, 34–42 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Wahl, A. S. et al. Hypoxic/ischemic conditions induce expression of the putative pro-death gene Clca1 via activation of extrasynaptic N-methyl-D-aspartate receptors. Neuroscience 158, 344–352 (2009).

    Article  CAS  PubMed  Google Scholar 

  104. Wittmann, M. et al. Synaptic activity induces dramatic changes in the geometry of the cell nucleus: interplay between nuclear structure, histone H3 phosphorylation, and nuclear calcium signaling. J. Neurosci. 29, 14687–14700 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Taddei, A. et al. Nuclear pore association confers optimal expression levels for an inducible yeast gene. Nature 441, 774–778 (2006).

    Article  CAS  PubMed  Google Scholar 

  106. Akhtar, A. & Gasser, S. M. The nuclear envelope and transcriptional control. Nature Rev. Genet. 8, 507–517 (2007).

    Article  CAS  PubMed  Google Scholar 

  107. Collins, M. O. & Grant, S. G. Supramolecular signalling complexes in the nervous system. Subcell. Biochem. 43, 185–207 (2007).

    Article  CAS  PubMed  Google Scholar 

  108. Aarts, M. et al. Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions. Science 298, 846–850 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. Cao, J. et al. The PSD95-nNOS interface: a target for inhibition of excitotoxic p38 stress-activated protein kinase activation and cell death. J. Cell Biol. 168, 117–126 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Sheng, M., Cummings, J., Roldan, L. A., Jan, Y. N. & Jan, L. Y. Changing subunit composition of heteromeric NMDA receptors during development of rat cortex. Nature 368, 144–147 (1994).

    Article  CAS  PubMed  Google Scholar 

  111. Zhong, J., Russell, S. L., Pritchett, D. B., Molinoff, P. B. & Williams, K. Expression of mRNAs encoding subunits of the N-methyl-D-aspartate receptor in cultured cortical neurons. Mol. Pharmacol. 45, 846–853 (1994).

    CAS  PubMed  Google Scholar 

  112. Steigerwald, F. et al. C-Terminal truncation of NR2A subunits impairs synaptic but not extrasynaptic localization of NMDA receptors. J. Neurosci. 20, 4573–4581 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Groc, L. et al. NMDA receptor surface mobility depends on NR2A-2B subunits. Proc. Natl Acad. Sci. USA 103, 18769–18774 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Martel, M., Wyllie, D. J. & Hardingham, G. E. In developing hippocampal neurons, NR2B-containing NMDA receptors can mediate signalling to neuronal survival and synaptic potentiation, as well as neuronal death. Neuroscience 158, 334–343 (2009).

    Article  CAS  PubMed  Google Scholar 

  115. Thomas, C. G., Miller, A. J. & Westbrook, G. L. Synaptic and extrasynaptic NMDA receptor NR2 subunits in cultured hippocampal neurons. J. Neurophysiol. 95, 1727–1734 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Harris, A. Z. & Pettit, D. L. Extrasynaptic and synaptic NMDA receptors form stable and uniform pools in rat hippocampal slices. J. Physiol. 584, 509–519 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liu, Y. et al. NMDA receptor subunits have differential roles in mediating excitotoxic neuronal death both in vitro and in vivo. J. Neurosci. 27, 2846–2857 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Weitlauf, C. et al. Activation of NR2A-containing NMDA receptors is not obligatory for NMDA receptor-dependent long-term potentiation. J. Neurosci. 25, 8386–8390 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Berberich, S. et al. Lack of NMDA receptor subtype selectivity for hippocampal long-term potentiation. J. Neurosci. 25, 6907–6910 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Neyton, J. & Paoletti, P. Relating NMDA receptor function to receptor subunit composition: limitations of the pharmacological approach. J. Neurosci. 26, 1331–1333 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Frizelle, P. A., Chen, P. E. & Wyllie, D. J. A. Equilibrium constants for NVP-AAM077 acting at recombinant NR1/NR2A and NR1/NR2B NMDA receptors: implications for studies of synaptic transmission. Mol. Pharmacol. 70, 1022–1032 (2006).

    Article  CAS  PubMed  Google Scholar 

  122. de Marchena, J. et al. NMDA receptor antagonists reveal age-dependent differences in the properties of visual cortical plasticity. J. Neurophysiol. 100, 1936–1948 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. von Engelhardt, J. et al. Excitotoxicity in vitro by NR2A- and NR2B-containing NMDA receptors. Neuropharmacology 53, 10–17 (2007).

    Article  CAS  PubMed  Google Scholar 

  124. Hatton, C. J. & Paoletti, P. Modulation of triheteromeric NMDA receptors by N-terminal domain ligands. Neuron 46, 261–274 (2005).

    Article  CAS  PubMed  Google Scholar 

  125. Harris, A. Z. & Pettit, D. L. Recruiting extrasynaptic NMDA receptors augments synaptic signaling. J. Neurophysiol. 99, 524–533 (2008).

    Article  CAS  PubMed  Google Scholar 

  126. Groc, L., Bard, L. & Choquet, D. Surface trafficking of N-methyl-D-aspartate receptors: physiological and pathological perspectives. Neuroscience 158, 4–18 (2009).

    Article  CAS  PubMed  Google Scholar 

  127. Tovar, K. R. & Westbrook, G. L. Mobile NMDA receptors at hippocampal synapses. Neuron 34, 255–264 (2002).

    Article  CAS  PubMed  Google Scholar 

  128. Chen, H. S. & Lipton, S. A. The chemical biology of clinically tolerated NMDA receptor antagonists. J. Neurochem. 97, 1611–1626 (2006).

    Article  CAS  PubMed  Google Scholar 

  129. Rossi, D. J., Oshima, T. & Attwell, D. Glutamate release in severe brain ischaemia is mainly by reversed uptake. Nature 403, 316–321 (2000).

    Article  CAS  PubMed  Google Scholar 

  130. Choi, D. W. Calcium-mediated neurotoxicity: relationship to specific channel types and role in ischemic damage. Trends Neurosci. 11, 465–469 (1988).

    Article  CAS  PubMed  Google Scholar 

  131. Waagepetersen, H. S., Shimamoto, K. & Schousboe, A. Comparison of effects of DL-threo-β-benzyloxyaspartate (DL-TBOA) and L-trans-pyrrolidine-2, 4-dicarboxylate (t-2, 4-PDC) on uptake and release of [3h]D-aspartate in astrocytes and glutamatergic neurons. Neurochem. Res. 26, 661–666 (2001).

    Article  CAS  PubMed  Google Scholar 

  132. Gouix, E. et al. Reverse glial glutamate uptake triggers neuronal cell death through extrasynaptic NMDA receptor activation. Mol. Cell. Neurosci. 40, 463–473 (2009).

    Article  CAS  PubMed  Google Scholar 

  133. Tu, W. et al. DAPK1 interaction with NMDA receptor NR2B subunits mediates brain damage in stroke. Cell 140, 222–234 (2010). This study showed that activation of death-associated protein kinase 1 (DAPK1) by ischaemia contributes to neuronal injury by enhancing injurious Ca2+ influx though extrasynaptic NMDARs. Peptide-mediated disruption of DAPK1-GluN2B interactions was shown to be protective in models of stroke.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Milnerwood, A. et al. Early increase in extrasynaptic NMDA receptor signalling and expression contributes to phenotype onset in Huntington's disease mice. Neuron 65, 178–190 (2010). Enhanced NMDAR activity and NMDAR-dependent excitotoxicity had for some years been associated with Huntington's disease pathogenesis. This paper showed that specific increases in extrasynaptic (but not synaptic) NMDAR activity contributes to phenotype onset, and demonstrated the beneficial effects of memantine treatment.

    Article  CAS  PubMed  Google Scholar 

  135. Subramaniam, S., Sixt, K. M., Barrow, R. & Snyder, S. H. Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity. Science 324, 1327–1330 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Nucifora, F. C. et al. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 291, 2423–2428 (2001).

    Article  CAS  PubMed  Google Scholar 

  137. Jiang, H. et al. Depletion of CBP is directly linked with cellular toxicity caused by mutant huntingtin. Neurobiol. Dis. 23, 543–551 (2006).

    Article  CAS  PubMed  Google Scholar 

  138. McGill, J. K. & Beal, M. F. PGC-1α, a new therapeutic target in Huntington's disease? Cell 127, 465–468 (2006).

    Article  CAS  PubMed  Google Scholar 

  139. St-Pierre, J. et al. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127, 397–408 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Wareski, P. et al. PGC-1α and PGC-1β regulate mitochondrial density in neurons. J. Biol. Chem. 284, 21379–21385 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Weydt, P. et al. Thermoregulatory and metabolic defects in Huntington's disease transgenic mice implicate PGC-1α in Huntington's disease neurodegeneration. Cell Metab. 4, 349–362 (2006).

    Article  CAS  PubMed  Google Scholar 

  142. Cui, L. et al. Transcriptional repression of PGC-1α by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 127, 59–69 (2006).

    Article  CAS  PubMed  Google Scholar 

  143. Lin, J. et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1α null mice. Cell 119, 121–135 (2004).

    Article  CAS  PubMed  Google Scholar 

  144. Muir, K. W. Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr. Opin. Pharmacol. 6, 53–60 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Albers, G. W., Goldstein, L. B., Hall, D. & Lesko, L. M. Aptiganel hydrochloride in acute ischemic stroke: a randomized controlled trial. JAMA 286, 2673–2682 (2001).

    Article  CAS  PubMed  Google Scholar 

  146. Biegon, A. et al. Dynamic changes in N-methyl-D-aspartate receptors after closed head injury in mice: implications for treatment of neurological and cognitive deficits. Proc. Natl Acad. Sci. USA 101, 5117–5122 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Yaka, R. et al. D-cycloserine improves functional recovery and reinstates long-term potentiation (LTP) in a mouse model of closed head injury. FASEB J. 21, 2033–2041 (2007).

    Article  CAS  PubMed  Google Scholar 

  148. Lipton, S. A. Pathologically activated therapeutics for neuroprotection. Nature Rev. Neurosci. 8, 803–808 (2007).

    Article  CAS  Google Scholar 

  149. Chen, H. S. et al. Neuroprotective concentrations of the N-methyl-D-aspartate open-channel blocker memantine are effective without cytoplasmic vacuolation following post-ischemic administration and do not block maze learning or long-term potentiation. Neuroscience 86, 1121–1132 (1998).

    Article  CAS  PubMed  Google Scholar 

  150. Xia, P., Chen, H. S., Zhang, D. & Lipton, S. A. Memantine preferentially blocks extrasynaptic over synaptic NMDA receptor currents in hippocampal autapses. J. Neurosci. 30, 11246–11250 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Kotermanski, S. E. & Johnson, J. W. Mg2+ imparts NMDA receptor subtype selectivity to the Alzheimer's drug memantine. J. Neurosci. 29, 2774–2779 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Chen, S. & Diamond, J. S. Synaptically released glutamate activates extrasynaptic NMDA receptors on cells in the ganglion cell layer of rat retina. J. Neurosci. 22, 2165–2173 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Clark, B. A. & Cull-Candy, S. G. Activity-dependent recruitment of extrasynaptic NMDA receptor activation at an AMPA receptor-only synapse. J. Neurosci. 22, 4428–4436 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Scimemi, A., Fine, A., Kullmann, D. M. & Rusakov, D. A. NR2B-containing receptors mediate cross talk among hippocampal synapses. J. Neurosci. 24, 4767–4777 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Bengtson, C. P., Dick, O. & Bading, H. A quantitative method to assess extrasynaptic NMDA receptor function in the protective effect of synaptic activity against neurotoxicity. BMC Neurosci. 9, 11 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Wu, G. Y., Deisseroth, K. & Tsien, R. W. Activity-dependent CREB phosphorylation: convergence of a fast, sensitive calmodulin kinase pathway and a slow, less sensitive mitogen-activated protein kinase pathway. Proc. Natl Acad. Sci. USA 98, 2808–2813 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Hardingham, G. E., Arnold, F. J. & Bading, H. A calcium microdomain near NMDA receptors: on switch for ERK-dependent synapse-to-nucleus communication. Nature Neurosci. 4, 565–566 (2001).

    Article  CAS  PubMed  Google Scholar 

  158. Impey, S. et al. Phosphorylation of CBP mediates transcriptional activation by neural activity and CaM kinase IV. Neuron 34, 235–244 (2002).

    Article  CAS  PubMed  Google Scholar 

  159. Screaton, R. A. et al. The CREB coactivator TORC2 functions as a calcium- and cAMP-sensitive coincidence detector. Cell 119, 61–74 (2004).

    Article  CAS  PubMed  Google Scholar 

  160. Kovacs, K. A. et al. TORC1 is a calcium- and cAMP-sensitive coincidence detector involved in hippocampal long-term synaptic plasticity. Proc. Natl Acad. Sci. USA 104, 4700–4705 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Ravnskjaer, K. et al. Cooperative interactions between CBP and TORC2 confer selectivity to CREB target gene expression. EMBO J. 26, 2880–2889 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Luo, Y., Zhu, W., Jia, J., Zhang, C. & Xu, Y. NMDA receptor dependent PGC-1α up-regulation protects the cortical neuron against oxygen-glucose deprivation/reperfusion injury. J. Mol. Neurosci. 39, 262–268 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Bell and C. P. Bengston for comments on the manuscript, and R. Petralia for supplying the electronmicrographs in Box 1. Work in the authors' laboratories is supported by the Wellcome Trust, the Medical Research Council (MRC), the Biotechnology and Biological Sciences Research Council, the Royal Society (G.E.H.) and by the Alexander von Humboldt Foundation, the European Research Council Advanced Grant, the Deutsche Forschungsgemeinschaft, the EU Network of Excellence NeuroNE and the EU Project Glutamate Receptor Interacting Proteins as Novel Neuroprotective Targets (GRIPANNT) (H.B.). G.E.H. is an MRC senior non-clinical research fellow. H.B. is a member of the Excellence Cluster CellNetworks at Heidelberg University.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Giles E. Hardingham's homepage

Hilmar Bading's homepage

Glossary

Pro-apoptotic Bcl2 homology domain 3 (BH3)-only member gene

(Also known as Puma.) A gene encoding a pro-apoptotic member of the Bcl2 family that contains only a BH3 domain, as opposed to those that contain multiple BH domains (for example, Bax and Bak)

Calpain

A family of Ca2+-dependent cysteine proteases.

Reversed uptake

Describes the action of glutamate transporters in pumping glutamate out of the cell, as opposed to their usual function of taking glutamate up from the extracellular space. Ischaemic conditions cause reversed uptake owing to membrane depolarization.

Hetero-exchange

The process by which an inwardly transported molecule leads to the efflux of a different molecule by the same transporter.

Huntingtin

A gene that contains an elevated number of CAG trinucleotide repeats in Huntington's disease and is the disease-causing agent.

Sumoylation

The process of covalent attachment of small ubiquitin-like modifier (SUMO) protein onto another protein. This modification typically alters the activity, stability or localization of the modified protein.

3-nitropropanoic acid

A mitochondrial toxin that inhibits succinate dehydrogenase (part of complex II).

Transactivating capacity

The ability of a transcription factor or co-activator to enhance gene transcription when associated with that gene's promoter. This may be influenced by post-translational modifications that determine the association of accessory factors, including chromatin-modifying enzymes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hardingham, G., Bading, H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci 11, 682–696 (2010). https://doi.org/10.1038/nrn2911

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn2911

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing