NMDA Receptor Internalization by Autoantibodies: A Reversible Mechanism Underlying Psychosis?

Joseph C Masdeu, Josep Dalmau, Karen F Berman, Joseph C Masdeu, Josep Dalmau, Karen F Berman

Abstract

Since the early 1990s it has been postulated that hypofunction of N-methyl-d-aspartate (NMDA) receptors in brain networks supporting perception and cognition underlies schizophrenic psychosis. Recently, NMDA receptor hypofunction was described in patients with psychotic manifestations who exhibited autoantibodies binding the GluN1 subunit of the receptor, and who improved when the level of these antibodies was lowered by immunomodulation. In this disorder, NMDA receptor antibodies decrease the availability of NMDA receptors by internalizing them. In this opinion article, we review this mechanism as well as data supporting or refuting the possibility that this disorder or similar autoimmune disorders affecting synaptic proteins, which are therefore treatable with immunomodulation, could account for some cases of idiopathic psychosis. We also suggest methodological approaches to clarify this issue.

Keywords: NMDA receptor; antibodies; immunotherapy; psychosis; schizophrenia; synapsis.

Conflict of interest statement

Conflict of Interest: JCM and KFB report no conflicts of interest.

Copyright © 2016 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Effect of NMDA receptor (NMDAR) hypofunction. This is a simplified diagram of some of the cognitive networks that are affected by NMDAR hypofunction. Three critical nodes are shown: (1) The subiculum, effector region of the hippocampal formation; (2) the prefrontal cortex (PFC), supporting working memory and executive function; and (3) the ventral tegmental area (VTA), which houses dopaminergic neurons involved in facilitating episodic and working memory, as well as motivation. (A) At each of these nodes, glutamatergic excitatory inputs to pyramidal or dopaminergic neurons (these from multiple brain regions, prominently lateral hypothalamus [101]) provide collaterals to NMDARs on gabaergic neurons, which in their turn inhibit excessive pyramidal firing. Gabaergic interneurons in the subiculum and PFC are parvalbumin positive (PV) and, in the PFC are identified with the fast-spiking cells, critical for the generation of synchronous gamma oscillations [53, 102]. In the VTA, gabaergic interneurons also harbor NMDARs [103] and stain with glutamic acid decarboxylase (GAD); by contrast VTA PV neurons are mostly projection neurons [104]. Dopaminergic VTA neurons project to the nucleus accumbens (NAc), PFC and hippocampus (not shown). The NAc inhibits the globus pallidus medialis (GPm), which in turn inhibits tonically VTA dopaminergic neurons [10, 54]. (B) NMDAR hypofunction is associated with increased pyramidal firing, which in turn increases the inhibitory activity of the NAc over the GPm and lessens its inhibitory tone over the VTA dopaminergic neurons. As a result, there is an increased production of dopamine, as found in psychoses, which are also attended by impaired working memory associated with abnormal functioning of PV interneurons [39, 54]. Increased pyramidal firing has also been documented with NMDAR ablation restricted to the NMDARs on frontal pyramidal neurons [105].
Figure 2
Figure 2
Simplified diagram of some of the synaptic receptors known to modulate NMDARs. The excitatory neurotransmitter glutamate (Glu) is secreted at the presynaptic terminal and activates the NMDA receptor on the postsynaptic membrane. The NMDAR is a tetramer composed of two GluN1 and two NR2 subunits. It is modulated by a number of synaptic proteins. On the presynaptic membrane, metabotropic glutamate type 2/3 receptors (Glu 2/3), which bind glutamate, downregulate Glu production, while in the postsynaptic membrane metabotropic glutamate type 5 receptors (Glu 5) upregulate the NMDAR via the Gq/11 protein and phospholipase C enzyme [106]. Src kinase (Src kinase) contributes to stabilize NR2 though its action on the postsynaptic density (PSD) complex [107, 108]. Enhanced ErbB4 signaling through PSD-95 and neuregulin 1 may cause NMDAR hypofunction [109]. NMDAR activation requires the presence of glycine (Gly) or D-serine (D-ser) occupying a binding site in the GluN1 subunit [110].
Figure 3
Figure 3
Age at onset of NMDA receptor antibody synaptopathy (NMDARAS) and of schizophrenic psychosis. Graphic built with data from [20] for NMDARAS and from [111, 112] for schizophrenic psychoses.
Figure 4
Figure 4
Diagram illustrating how more severe clinical findings in patients with anti-NMDAR disease are associated with lesser NMDA receptor availability on the postsynaptic region and higher levels of brain antibodies. The clinical picture resembles that caused by phencyclidine (PCP, green bar), in which the profile of symptoms correlates with the circulating concentration of the drug. Post-synaptic receptors depicted in blue represent α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR), those in green N-methyl-D-aspartate receptors (NMDAR). The small round vesicles that are released in the synapse represent glutamate. Modified from Dalmau et al [74].

References

    1. Hansen KB, et al. Distinct functional and pharmacological properties of Triheteromeric GluN1/GluN2A/GluN2B NMDA receptors. Neuron. 2014;81:1084–1096.
    1. Karakas E, Furukawa H. Crystal structure of a heterotetrameric NMDA receptor ion channel. Science. 2014;344:992–997.
    1. Lee CH, et al. NMDA receptor structures reveal subunit arrangement and pore architecture. Nature. 2014;511:191–197.
    1. Paoletti P, et al. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383–400.
    1. Wyllie DJ, et al. Influence of GluN2 subunit identity on NMDA receptor function. Neuropharmacology. 2013;74:4–17.
    1. Pachernegg S, et al. GluN3 subunit-containing NMDA receptors: not just one-trick ponies. Trends Neurosci. 2012;35:240–249.
    1. Lau CG, Zukin RS. NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nat Rev Neurosci. 2007;8:413–426.
    1. Traynelis SF, et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62:405–496.
    1. Wang M, Arnsten AF. Contribution of NMDA receptors to dorsolateral prefrontal cortical networks in primates. Neurosci Bull. 2015;31:191–197.
    1. Modinos G, et al. Translating the MAM model of psychosis to humans. Trends Neurosci. 2015;38:129–138.
    1. de Souza Silva MA, et al. Evidence for a specific integrative mechanism for episodic memory mediated by AMPA/kainate receptors in a circuit involving medial prefrontal cortex and hippocampal CA3 region. Cereb Cortex 2015
    1. Wang M, et al. NMDA receptors subserve persistent neuronal firing during working memory in dorsolateral prefrontal cortex. Neuron. 2013;77:736–749.
    1. Javitt DC, Freedman R. Sensory processing dysfunction in the personal experience and neuronal machinery of schizophrenia. Am J Psychiatry. 2015;172:17–31.
    1. Krystal JH, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214.
    1. Javitt DC, Zukin SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry. 1991;148:1301–1308.
    1. Fromer M, et al. De novo mutations in schizophrenia implicate synaptic networks. Nature. 2014;506:179–184.
    1. Wen Z, et al. Synaptic dysregulation in a human iPS cell model of mental disorders. Nature. 2014;515:414–418.
    1. Dalmau J, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol. 2008;7:1091–1098.
    1. Dalmau J, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol. 2007;61:25–36.
    1. Titulaer MJ, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol. 2013;12:157–165.
    1. Irani SR, et al. N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain. 2010;133:1655–1667.
    1. Viaccoz A, et al. Clinical specificities of adult male patients with NMDA receptor antibodies encephalitis. Neurology. 2014;82:556–563.
    1. Pathmanandavel K, et al. Antibodies to surface dopamine-2 receptor and N-methyl-d-aspartate receptor in the first episode of acute psychosis in children. Biol Psychiatry. 2015;77:537–547.
    1. Kayser MS, et al. Frequency and characteristics of isolated psychiatric episodes in anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol. 2013;70:1133–1139.
    1. Zandi MS, et al. Disease-relevant autoantibodies in first episode schizophrenia. J Neurol. 2011;258:686–688.
    1. Kendler KS, et al. Lifetime prevalence, demographic risk factors, and diagnostic validity of nonaffective psychosis as assessed in a US community sample. The National Comorbidity Survey. Arch Gen Psychiatry. 1996;53:1022–1031.
    1. Jablensky A, et al. Psychotic disorders in urban areas: an overview of the Study on Low Prevalence Disorders. Aust N Z J Psychiatry. 2000;34:221–236.
    1. de Witte LD, et al. Absence of N-methyl-D-aspartate receptor IgG autoantibodies in schizophrenia: the importance of cross-validation studies. JAMA Psychiatry. 2015;72:731–733.
    1. Masdeu JC, et al. Serum IgG antibodies against the NR1 subunit of the NMDA receptor not detected in schizophrenia. Am J Psychiatry. 2012;169:1120–1121.
    1. Rhoads J, et al. Lack of anti-NMDA receptor autoantibodies in the serum of subjects with schizophrenia. Schizophr Res. 2011;129:213–214.
    1. Hammer C, et al. Neuropsychiatric disease relevance of circulating anti-NMDA receptor autoantibodies depends on blood-brain barrier integrity. Mol Psychiatry 2013
    1. Steiner J, et al. Increased prevalence of diverse N-methyl-D-aspartate glutamate receptor antibodies in patients with an initial diagnosis of schizophrenia: specific relevance of IgG NR1a antibodies for distinction from N-methyl-D-aspartate glutamate receptor encephalitis. JAMA Psychiatry. 2013;70:271–278.
    1. Dahm L, et al. Seroprevalence of autoantibodies against brain antigens in health and disease. Ann Neurol. 2014;76:82–94.
    1. Kapur S, et al. Relationship between dopamine D(2) occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry. 2000;157:514–520.
    1. Kegeles LS, et al. Increased synaptic dopamine function in associative regions of the striatum in schizophrenia. Arch Gen Psychiatry. 2010;67:231–239.
    1. Meyer-Lindenberg A, et al. Reduced prefrontal activity predicts exaggerated striatal dopaminergic function in schizophrenia. Nat Neurosci. 2002;5:267–271.
    1. Fusar-Poli P, et al. Abnormal prefrontal activation directly related to pre-synaptic striatal dopamine dysfunction in people at clinical high risk for psychosis. Mol Psychiatry. 2011;16:67–75.
    1. Eisenberg DP, et al. Catechol-o-methyltransferase valine(158)methionine genotype and resting regional cerebral blood flow in medication-free patients with schizophrenia. Biol Psychiatry. 2010;67:287–290.
    1. Moghaddam B, Javitt D. From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology. 2012;37:4–15.
    1. Rodriguez-Santiago B, et al. Association of common copy number variants at the glutathione S-transferase genes and rare novel genomic changes with schizophrenia. Mol Psychiatry. 2010;15:1023–1033.
    1. Martucci L, et al. N-methyl-D-aspartate receptor NR2B subunit gene GRIN2B in schizophrenia and bipolar disorder: Polymorphisms and mRNA levels. Schizophr Res. 2006;84:214–221.
    1. Javitt DC. Neurophysiological models for new treatment development in schizophrenia: early sensory approaches. Ann N Y Acad Sci. 2015;1344:92–104.
    1. Lewis DA, et al. Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci. 2005;6:312–324.
    1. Naatanen R, Kahkonen S. Central auditory dysfunction in schizophrenia as revealed by the mismatch negativity (MMN) and its magnetic equivalent MMNm: a review. Int J Neuropsychopharmacol. 2009;12:125–135.
    1. Lisman JE, et al. A thalamo-hippocampal-ventral tegmental area loop may produce the positive feedback that underlies the psychotic break in schizophrenia. Biol Psychiatry. 2010;68:17–24.
    1. Anticevic A, et al. NMDA receptor function in large-scale anticorrelated neural systems with implications for cognition and schizophrenia. Proc Natl Acad Sci U S A. 2012;109:16720–16725.
    1. Pilowsky LS, et al. First in vivo evidence of an NMDA receptor deficit in medication-free schizophrenic patients. Mol Psychiatry. 2006;11:118–119.
    1. Poels EM, et al. Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry. 2014;19:20–29.
    1. Weickert CS, et al. Molecular evidence of N-methyl-D-aspartate receptor hypofunction in schizophrenia. Mol Psychiatry. 2013;18:1185–1192.
    1. Law AJ, Deakin JF. Asymmetrical reductions of hippocampal NMDAR1 glutamate receptor mRNA in the psychoses. Neuroreport. 2001;12:2971–2974.
    1. Mohn AR, et al. Mice with reduced NMDA receptor expression display behaviors related to schizophrenia. Cell. 1999;98:427–436.
    1. Papaleo F, et al. Mouse models of genetic effects on cognition: relevance to schizophrenia. Neuropharmacology. 2012;62:1204–1220.
    1. Rotaru DC, et al. The role of glutamatergic inputs onto parvalbumin-positive interneurons: relevance for schizophrenia. Rev Neurosci. 2012;23:97–109.
    1. Lisman JE, et al. Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends Neurosci. 2008;31:234–242.
    1. Tandon R, et al. Schizophrenia, "just the facts" 4. Clinical features and conceptualization. Schizophr Res. 2009;110:1–23.
    1. Buckley C, et al. Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann Neurol. 2001;50:73–78.
    1. Lancaster E, Dalmau J. Neuronal autoantigens--pathogenesis, associated disorders and antibody testing. Nat Rev Neurol. 2012;8:380–390.
    1. Lai M, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol. 2009;65:424–434.
    1. Petit-Pedrol M, et al. Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterization of the antigen, and analysis of the effects of antibodies. Lancet Neurol. 2014;13 in press.
    1. Bien CG, et al. Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain. 2012;135:1622–1638.
    1. Camdessanche JP, et al. Brain immunohistopathological study in a patient with anti-NMDAR encephalitis. Eur J Neurol. 2011;18:929–931.
    1. Martinez-Hernandez E, et al. Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis. Neurology. 2011;77:589–593.
    1. Lim M, et al. Autoimmune encephalopathies. Pediatr Clin North Am. 2015;62:667–685.
    1. Gleichman AJ, et al. Anti-NMDA receptor encephalitis antibody binding is dependent on amino acid identity of a small region within the GluN1 amino terminal domain. J Neurosci. 2012;32:11082–11094.
    1. Gresa-Arribas N, et al. Antibody titres at diagnosis and during follow-up of anti-NMDA receptor encephalitis: a retrospective study. Lancet Neurol. 2014;13:167–177.
    1. Hughes EG, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci. 2010;30:5866–5875.
    1. Moscato EH, et al. Acute mechanisms underlying antibody effects in anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol. 2014;76:108–119.
    1. Mikasova L, et al. Disrupted surface cross-talk between NMDA and Ephrin-B2 receptors in anti-NMDA encephalitis. Brain. 2012;135:1606–1621.
    1. Planaguma J, et al. Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain. 2015;138:94–109.
    1. Rose NR, Bona C. Defining criteria for autoimmune diseases (Witebsky's postulates revisited) Immunol Today. 1993;14:426–430.
    1. Pruss H, et al. Retrospective analysis of NMDA receptor antibodies in encephalitis of unknown origin. Neurology. 2010;75:1735–1739.
    1. Granerod J, et al. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis. 2010;10:835–844.
    1. Gable MS, et al. The frequency of autoimmune N-methyl-D-aspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project. Clin Infect Dis. 2012;54:899–904.
    1. Dalmau J, et al. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol. 2011;10:63–74.
    1. Hafner H, et al. ABC Schizophrenia study: an overview of results since 1996. Soc Psychiatry Psychiatr Epidemiol. 2013;48:1021–1031.
    1. Novotna M, et al. Poor early relapse recovery affects onset of progressive disease course in multiple sclerosis. Neurology. 2015;85:722–729.
    1. Lebon S, et al. Anti-N-methyl-D-aspartate (NMDA) receptor encephalitis mimicking a primary psychiatric disorder in an adolescent. J Child Neurol. 2012;27:1607–1610.
    1. Tidswell J, et al. Early recognition of anti-N-methyl D-aspartate (NMDA) receptor encephalitis presenting as acute psychosis. Australas Psychiatry. 2013;21:596–599.
    1. Barry H, et al. Anti-NMDA receptor encephalitis: an important differential diagnosis in psychosis. Br J Psychiatry. 2011;199:508–509.
    1. Khandaker GM, et al. Inflammation and immunity in schizophrenia: implications for pathophysiology and treatment. Lancet Psychiatry. 2015;2:258–270.
    1. Knight JG, et al. Rationale for a trial of immunosuppressive therapy in acute schizophrenia. Mol Psychiatry. 2007;12:424–431.
    1. Häfner H, an der Heiden W. Course and outcome of schizophrenia. In: Hirsch SRI, Weinberger DR, editors. Schizophrenia. Blackwell; 2003. pp. 101–141.
    1. Fillman SG, et al. Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol Psychiatry. 2013;18:206–214.
    1. Saetre P, et al. Inflammation-related genes up-regulated in schizophrenia brains. BMC Psychiatry. 2007;7:46.
    1. Bloomfield PS, et al. Microglial activity in people at ultra high risk of psychosis and in schizophrenia: An [C]PBR28 PET brain imaging study. Am J Psychiatry. 2015 appiajp201514101358.
    1. Schizophrenia Working Group of the Psychiatric Genomics C. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511:421–427.
    1. Pollak TA, et al. Prevalence of anti-N-methyl-D-aspartate (NMDA) receptor [corrected] antibodies in patients with schizophrenia and related psychoses: a systematic review and meta-analysis. Psychol Med. 2014;44:2475–2487.
    1. Tsutsui K, et al. Anti-NMDA-receptor antibody detected in encephalitis, schizophrenia, and narcolepsy with psychotic features. BMC Psychiatry. 2012;12:37.
    1. Hammer C, et al. Neuropsychiatric disease relevance of circulating anti-NMDA receptor autoantibodies depends on blood-brain barrier integrity. Mol Psychiatry. 2014;19:1143–1149.
    1. Ransohoff RM, Engelhardt B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol. 2012;12:623–635.
    1. Leypoldt F, et al. Autoimmune encephalopathies. Ann N Y Acad Sci. 2015;1338:94–114.
    1. Louveau A, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–341.
    1. Zandi MS, et al. N-methyl-D-aspartate receptor autoantibodies in psychiatric illness. Biol Psychiatry 2015
    1. Joubert B, et al. Clinical spectrum of encephalitis associated with antibodies against the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor: Case series and review of the literature. JAMA Neurol. 2015;72:1163–1169.
    1. Bataller L, et al. Reversible paraneoplastic limbic encephalitis associated with antibodies to the AMPA receptor. Neurology. 2010;74:265–267.
    1. Lancaster E, et al. Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology. 2011;77:1698–1701.
    1. Jarius S, et al. Psychotic syndrome associated with anti-Ca/ARHGAP26 and voltage-gated potassium channel antibodies. J Neuroimmunol. 2015;286:79–82.
    1. Hedegaard M, et al. Molecular pharmacology of human NMDA receptors. Neurochem Int. 2012;61:601–609.
    1. Zandi MS, et al. Clinical relevance of serum antibodies to extracellular N-methyl-D-aspartate receptor epitopes. J Neurol Neurosurg Psychiatry. 2015;86:708–713.
    1. Najjar S, et al. Neuropsychiatric autoimmune encephalitis without VGKC-complex, NMDAR, and GAD autoantibodies: case report and literature review. Cogn Behav Neurol. 2013;26:36–49.
    1. Watabe-Uchida M, et al. Whole-brain mapping of direct inputs to midbrain dopamine neurons. Neuron. 2012;74:858–873.
    1. Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007;27:11496–11500.
    1. Kodangattil JN, et al. Spike timing-dependent plasticity at GABAergic synapses in the ventral tegmental area. J Physiol. 2013;591:4699–4710.
    1. Olson VG, Nestler EJ. Topographical organization of GABAergic neurons within the ventral tegmental area of the rat. Synapse. 2007;61:87–95.
    1. Tatard-Leitman VM, et al. Pyramidal cell selective ablation of N-methyl-D-aspartate receptor 1 causes increase in cellular and network excitability. Biol Psychiatry. 2015;77:556–568.
    1. Ellaithy A, et al. Positive allosteric modulators of metabotropic glutamate 2 receptors in schizophrenia treatment. Trends Neurosci 2015
    1. Banerjee A, et al. Src kinase as a mediator of convergent molecular abnormalities leading to NMDAR hypoactivity in schizophrenia. Mol Psychiatry. 2015;20:1091–1100.
    1. Pitcher GM, et al. Schizophrenia susceptibility pathway neuregulin 1-ErbB4 suppresses Src upregulation of NMDA receptors. Nat Med. 2011;17:470–478.
    1. Hahn CG, et al. Altered neuregulin 1-erbB4 signaling contributes to NMDA receptor hypofunction in schizophrenia. Nat Med. 2006;12:824–828.
    1. Labrie V, Roder JC. The involvement of the NMDA receptor D-serine/glycine site in the pathophysiology and treatment of schizophrenia. Neurosci Biobehav Rev. 2010;34:351–372.
    1. Hafner H, Nowotny B. Epidemiology of early-onset schizophrenia. Eur Arch Psychiatry Clin Neurosci. 1995;245:80–92.
    1. Hafner H, et al. Sex differences in schizophrenia. Psychiatria Fennica. 1991;22:123–156.

Source: PubMed

3
S'abonner