Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-9pm4c Total loading time: 0 Render date: 2024-04-25T13:36:01.026Z Has data issue: false hasContentIssue false

Chapter 19 - Autoimmune Psychosis

from Section 4 - Autoimmunity in Neurological and Psychiatric Diseases

Published online by Cambridge University Press:  27 January 2022

Josep Dalmau
Affiliation:
Universitat de Barcelona
Francesc Graus
Affiliation:
Universitat de Barcelona
Get access

Summary

In this chapter we critically trace the concept of autoimmune psychosis, and review several well-defined autoimmune diseases that can manifest with psychosis. After the discovery of anti-NMDAR encephalitis, several studies suggested that NMDAR antibodies could occur in patients with psychosis caused by primary psychiatric diseases. This led to a generalized NMDAR antibody testing without much consideration for validation of results, the use of appropriate controls, or whether the antibodies were also present in CSF (most studies only partially examined serum). Two consequences of this uncontrolled testing were: (1) the wide range in prevalence of serum NMDAR antibodies (from 0% to 20% in patients with many different diseases, including healthy controls) among different laboratories often using the same commercial diagnostic test; and (2) the lack of clinical significance of the findings. These inconclusive studies refocused the attention of investigators to search for a specific psychiatric phenotype of anti-NMDAR encephalitis, but no specific phenotype could be identified. However, there are several important clinical clues that suggest when a first episode of psychosis is autoimmune, and we provide a diagnostic algorithm to identify these cases. Aside from anti-NMDAR encephalitis, psychiatric manifestations are prominent in three other disorders: paediatric autoimmune neuropsychiatric disorders associated with streptococcal infection (PANDAS), Hashimoto encephalopathy, and neuropsychiatric manifestations of systemic lupus erythematosus (SLE). In the first two disorders the autoimmune basis is unclear and no pathogenic antibodies have been identified. In SLE, none of the antibodies reported to be associated with neuropsychiatric manifestations has shown neuropsychiatric symptom specificity. Moreover, none of the SLE antibodies has shown properties similar to those of neuronal surface antibodies related to autoimmune encephalitis, which associate with specific syndromes, alter neuronal function by direct interaction with the cell surface target, and cause symptoms in animal models, including psychotic-like behaviour.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2022

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Javitt, DC, Zukin, SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 1991;148:13011308.Google Scholar
Patkar, AA, Mago, R, Masand, PS. Psychotic symptoms in patients with medical disorders. Curr Psychiatry Rep 2004;6:216224.Google Scholar
Khandaker, GM, Cousins, L, Deakin, J, et al. Inflammation and immunity in schizophrenia: implications for pathophysiology and treatment. Lancet Psychiatry 2015;2:258270.Google Scholar
Dalmau, J, Tuzun, E, Wu, HY, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 2007;61:2536.CrossRefGoogle ScholarPubMed
Vitaliani, R, Mason, W, Ances, B, et al. Paraneoplastic encephalitis, psychiatric symptoms, and hypoventilation in ovarian teratoma. Ann Neurol 2005;58:594604.Google Scholar
Florance, NR, Davis, RL, Lam, C, et al. Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis in children and adolescents. Ann Neurol 2009;66:1118.CrossRefGoogle ScholarPubMed
Dalmau, J, Gleichman, AJ, Hughes, EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008;7:10911098.CrossRefGoogle ScholarPubMed
Dhossche, D, Fink, M, Shorter, E, Wachtel, LE. Anti-NMDA receptor encephalitis versus pediatric catatonia. Am J Psychiatry 2011;168:749750.CrossRefGoogle ScholarPubMed
Steiner, J, Walter, M, Glanz, W, 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;70:271278.CrossRefGoogle ScholarPubMed
Castillo-Gomez, E, Oliveira, B, Tapken, D, et al. All naturally occurring autoantibodies against the NMDA receptor subunit NR1 have pathogenic potential irrespective of epitope and immunoglobulin class. Mol Psychiatry 2017;22:17761784.CrossRefGoogle ScholarPubMed
Castillo-Gomez, E, Kastner, A, Steiner, J, et al. The brain as immunoprecipitator of serum autoantibodies against N-Methyl-D-aspartate receptor subunit NR1. Ann Neurol 2016;79:144151.Google Scholar
Zerche, M, Weissenborn, K, Ott, C, et al. Preexisting serum autoantibodies against the NMDAR subunit NR1 modulate evolution of lesion size in acute ischemic stroke. Stroke 2015;46:11801186.CrossRefGoogle ScholarPubMed
Dahm, L, Ott, C, Steiner, J, et al. Seroprevalence of autoantibodies against brain antigens in health and disease. Ann Neurol 2014;76:8294.CrossRefGoogle ScholarPubMed
Ehrenreich, H, Steiner, J. Serum autoantibodies against N-methyl-D-aspartate receptor subunit NR1 are no disease indicator. Ann Neurol 2014;31:306312.Google Scholar
Hammer, C, Stepniak, B, Schneider, A, et al. Neuropsychiatric disease relevance of circulating anti-NMDA receptor autoantibodies depends on blood–brain barrier integrity. Mol Psychiatry 2014;19:11431149.Google Scholar
Sperber, PS, Siegerink, B, Huo, S, et al. Serum Anti-NMDA (N-methyl-D-aspartate)-receptor antibodies and long-term clinical outcome after stroke (PROSCIS-B). Stroke 2019;50:32133219.Google Scholar
Zandi, MS, Irani, SR, Lang, B, et al. Disease-relevant autoantibodies in first episode schizophrenia. J Neurol 2011;258:686688.Google Scholar
Tsutsui, K, Kanbayashi, T, Tanaka, K, et al. Anti-NMDA-receptor antibody detected in encephalitis, schizophrenia, and narcolepsy with psychotic features. BMC Psychiatry 2012;12:37.CrossRefGoogle ScholarPubMed
Zandi, MS, Paterson, RW, Ellul, MA, et al. Clinical relevance of serum antibodies to extracellular N-methyl-D-aspartate receptor epitopes. J Neurol Neurosurg Psychiatry 2015;86:708713.CrossRefGoogle ScholarPubMed
Hara, M, Martinez-Hernandez, E, Arino, H, et al. Clinical and pathogenic significance of IgG, IgA, and IgM antibodies against the NMDA receptor. Neurology 2018;90:e1386e1394.Google Scholar
Steiner, J, Teegen, B, Schiltz, K, et al. Prevalence of N-methyl-D-aspartate receptor autoantibodies in the peripheral blood: healthy control samples revisited. JAMA Psychiatry 2014;71:838839.Google Scholar
Doss, S, Wandinger, KP, Hyman, BT, et al. High prevalence of NMDA receptor IgA/IgM antibodies in different dementia types. Ann Clin Transl Neurol 2014;1:822832.Google Scholar
Lancaster, E, Leypoldt, F, Titulaer, MJ, et al. Immunoglobulin G antibodies to the N-methyl-D-aspartate receptor are distinct from immunoglobulin A and immunoglobulin M responses. Ann Neurol 2015;77:183.Google Scholar
Titulaer, MJ, Dalmau, J. Antibodies to NMDA receptor, blood–brain barrier disruption and schizophrenia: a theory with unproven links. Mol Psychiatry 2014;19:1054.CrossRefGoogle ScholarPubMed
Mane-Damas, M, Hoffmann, C, Zong, S, et al. Autoimmunity in psychotic disorders. Where we stand, challenges and opportunities. Autoimmun Rev 2019;18:102348.Google Scholar
Deakin, J, Lennox, BR, Zandi, MS. Antibodies to the N-methyl-D-aspartate receptor and other synaptic proteins in psychosis. Biol Psychiatry 2014;75:284291.CrossRefGoogle Scholar
Pollak, TA, McCormack, R, Peakman, M, Nicholson, TR, David, AS. Prevalence of anti-N-methyl-d-aspartate (NMDA) antibodies in patients with schizophrenia and related psychoses: a systematic review and meta-analysis. Psychol Med 2013;44:24752487.CrossRefGoogle ScholarPubMed
Al-Diwani, A, Handel, A, Townsend, L, et al. The psychopathology of NMDAR-antibody encephalitis in adults: a systematic review and phenotypic analysis of individual patient data. Lancet Psychiatry 2019;6:235246.CrossRefGoogle ScholarPubMed
Sarkis, RA, Coffey, MJ, Cooper, JJ, Hassan, I, Lennox, B. Anti-N-methyl-D-aspartate receptor encephalitis: a review of psychiatric phenotypes and management considerations – a report of the American Neuropsychiatric Association Committee on Research. J Neuropsychiatry Clin Neurosci 2019;31:137142.Google Scholar
Warren, N, Siskind, D, O’Gorman, C. Refining the psychiatric syndrome of anti-N-methyl-d-aspartate receptor encephalitis. Acta Psychiatr Scand 2018;138:401408.CrossRefGoogle ScholarPubMed
Olney, JW, Farber, NB. Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry 1995;52:9981007.Google Scholar
Kehrer, C, Maziashvili, N, Dugladze, T, Gloveli, T. Altered excitatory-inhibitory balance in the NMDA-hypofunction model of schizophrenia. Front Mol Neurosci 2008;1:6.CrossRefGoogle ScholarPubMed
Masdeu, JC, Dalmau, J, Berman, KF. NMDA receptor internalization by autoantibodies: a reversible mechanism underlying psychosis? Trends Neurosci 2016;39:300–310.Google Scholar
Gunduz-Bruce, H. The acute effects of NMDA antagonism: from the rodent to the human brain. Brain Res Rev 2009;60:279286.Google Scholar
Endres, D, Perlov, E, Baumgartner, A, et al. Immunological findings in psychotic syndromes: a tertiary care hospital’s CSF sample of 180 patients. Front Hum Neurosci 2015;9:476.Google Scholar
Oviedo-Salcedo, T, de Witte, L, Kumpfel, T, et al. Absence of cerebrospinal fluid antineuronal antibodies in schizophrenia spectrum disorders. Br J Psychiatry 2018;212:318320.Google Scholar
Lennox, BR, Palmer-Cooper, EC, Pollak, T, et al. Prevalence and clinical characteristics of serum neuronal cell surface antibodies in first-episode psychosis: a case–control study. Lancet Psychiatry 2017;4:4248.Google Scholar
Kelleher, E, McNamara, P, Dunne, J, et al. Prevalence of N-methyl-D-aspartate receptor antibody (NMDAR-Ab) encephalitis in patients with first episode psychosis and treatment resistant schizophrenia on clozapine, a population based study. Schizophr Res 2020;222:455461.CrossRefGoogle ScholarPubMed
Schou, MB, Saether, SG, Drange, OK, et al. The significance of anti-neuronal antibodies for acute psychiatric disorders: a retrospective case-controlled study. BMC Neurosci 2018;19:68.CrossRefGoogle ScholarPubMed
Jézéquel, J, Rogemond, V, Pollak, T, et al. Cell- and single molecule-based methods to detect anti-N-methyl-D-aspartate receptor autoantibodies in patients with first-episode psychosis from the OPTiMiSE project. Biol Psychiatry 2017;82:766772.Google Scholar
Hoffmann, C, Zong, S, Mane-Damas, M, et al. Absence of autoantibodies against neuronal surface antigens in sera of patients with psychotic disorders. JAMA Psychiatry 2019;7:322325.Google Scholar
de Witte, LD, Hoffmann, C, van Mierlo, HC, et al. Absence of N-methyl-D-aspartate receptor IgG autoantibodies in schizophrenia: the importance of cross-validation studies. JAMA Psychiatry 2015;72:731733.Google Scholar
Masopust, J, Andrys, C, Bazant, J, et al. Anti-NMDA receptor antibodies in patients with a first episode of schizophrenia. Neuropsychiatr Dis Treat 2015;11:619623.Google ScholarPubMed
Gaughran, F, Lally, J, Beck, K, et al. Brain-relevant antibodies in first-episode psychosis: a matched case-control study. Psychol Med 2018;48:12571263.CrossRefGoogle ScholarPubMed
Pollak, TA, Vincent, A, Iyegbe, C, et al. Relationship between serum NMDA receptor antibodies and response to antipsychotic treatment in first-episode psychosis. Biol Psychiatry 2020;90:915.Google Scholar
Rhoads, J, Guirgis, H, McKnight, C, Duchemin, AM. Lack of anti-NMDA receptor autoantibodies in the serum of subjects with schizophrenia. Schizophr Res 2011;129:213214.Google Scholar
Vincent, A, Bien, CG, Irani, SR, Waters, P. Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol 2011;10:759772.Google Scholar
Gresa-Arribas, N, Titulaer, MJ, Torrents, A, et al. Antibody titres at diagnosis and during follow-up of anti-NMDA receptor encephalitis: a retrospective study. Lancet Neurol 2014;13:167177.Google Scholar
Bien, CG, Bien, CI, Dogan Onugoren, M, et al. Routine diagnostics for neural antibodies, clinical correlates, treatment and functional outcome. J Neurol 2020;267:21012114.Google Scholar
Zandi, MS, Deakin, JB, Morris, K, et al. Immunotherapy for patients with acute psychosis and serum N-methyl D-aspartate receptor (NMDAR) antibodies: a description of a treated case series. Schizophr Res 2014;160:193195.Google Scholar
Pollak, TA, Lennox, BR, Muller, S, et al. Autoimmune psychosis: an international consensus on an approach to the diagnosis and management of psychosis of suspected autoimmune origin. Lancet Psychiatry 2020;7:93108.Google Scholar
Scott, JG, Gillis, D, Swayne, A, Blum, S. Testing for antibodies to N-methyl-d-aspartate receptor and other neuronal cell surface antigens in patients with early psychosis. Aust N Z J Psychiatry 2018;52:727729.Google Scholar
Graus, F, Titulaer, MJ, Balu, R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016;15:391404.Google Scholar
Warren, N, Swayne, A, Siskind, D, et al. Serum and CSF Anti-NMDAR antibody testing in psychiatry. J Neuropsychiatry Clin Neurosci 2020;32:154160.Google Scholar
Herken, J, Pruss, H. Red flags: clinical signs for identifying autoimmune encephalitis in psychiatric patients. Front Psychiatry 2017;8:25.CrossRefGoogle ScholarPubMed
Irani, SR, Bien, CG, Lang, B. Autoimmune epilepsies. Curr Opin Neurol 2011;24:146153.Google Scholar
Damato, V, Balint, B, Kienzler, AK, Irani, SR. The clinical features, underlying immunology, and treatment of autoantibody-mediated movement disorders. Mov Disord 2018;33:13761389.CrossRefGoogle ScholarPubMed
Dubey, D, Kothapalli, N, McKeon, A, et al. Predictors of neural-specific autoantibodies and immunotherapy response in patients with cognitive dysfunction. J Neuroimmunol 2018;323:6272.CrossRefGoogle ScholarPubMed
Dubey, D, Singh, J, Britton, JW, et al. Predictive models in the diagnosis and treatment of autoimmune epilepsy. Epilepsia 2017;58:11811189.CrossRefGoogle ScholarPubMed
Dubey, D, Farzal, Z, Hays, R, Brown, LS, Vernino, S. Evaluation of positive and negative predictors of seizure outcomes among patients with immune-mediated epilepsy: a meta-analysis. Ther Adv Neurol Disord 2016;9:369377.CrossRefGoogle ScholarPubMed
Moussa, T, Afzal, K, Cooper, J, et al. Pediatric anti-NMDA receptor encephalitis with catatonia: treatment with electroconvulsive therapy. Pediatr Rheumatol Online J 2019;17:8.Google Scholar
Coffey, MJ, Cooper, JJ. Electroconvulsive therapy in anti-N-methyl-D-aspartate receptor encephalitis: a case report and review of the literature. J ECT 2016;32:225229.CrossRefGoogle ScholarPubMed
Endres, D, Dersch, R, Hochstuhl, B, et al. Intrathecal thyroid autoantibody synthesis in a subgroup of patients with schizophreniform syndromes. J Neuropsychiatry Clin Neurosci 2017;29:365374.CrossRefGoogle Scholar
Endres, D, Perlov, E, Riering, AN, et al. Steroid-responsive chronic schizophreniform syndrome in the context of mildly increased antithyroid peroxidase antibodies. Front Psychiatry 2017;8:64.Google Scholar
Scott, JG, Gillis, D, Ryan, AE, et al. The prevalence and treatment outcomes of antineuronal antibody-positive patients admitted with first episode of psychosis. BJPsych Open 2018;4:6974.Google Scholar
Leboyer, M, Berk, M, Yolken, RH, et al. Immuno-psychiatry: an agenda for clinical practice and innovative research. BMC Med 2016;14:173.Google Scholar
Ezeoke, A, Mellor, A, Buckley, P, Miller, B. A systematic, quantitative review of blood autoantibodies in schizophrenia. Schizophr Res 2013;150:245251.CrossRefGoogle ScholarPubMed
Lejuste, F, Thomas, L, Picard, G, et al. Neuroleptic intolerance in patients with anti-NMDAR encephalitis. Neurol Neuroimmunol Neuroinflamm 2016;3:e280.Google Scholar
Sansing, LH, Tuzun, E, Ko, MW, et al. A patient with encephalitis associated with NMDA receptor antibodies. Nat Clin Pract Neurol 2007;3:291296.Google Scholar
Dalmau, J, Armangue, T, Planaguma, J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol 2019;18:10451057.Google Scholar
Matsumoto, T, Matsumoto, K, Kobayashi, T, Kato, S. Electroconvulsive therapy can improve psychotic symptoms in anti-NMDA-receptor encephalitis. Psychiatry Clin Neurosci 2012;66:242243.CrossRefGoogle ScholarPubMed
Lai, M, Hughes, EG, Peng, X, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 2009;65:424434.Google Scholar
Lancaster, E, Lai, M, Peng, X, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 2010;9:6776.Google Scholar
Masdeu, JC, Gonzalez-Pinto, A, Matute, C, et al. Serum IgG antibodies against the NR1 subunit of the NMDA receptor not detected in schizophrenia. Am J Psychiatry 2012;169:11201121.Google Scholar
Jézéquel, J, Johansson, EM, Dupuis, JP, et al. Dynamic disorganization of synaptic NMDA receptors triggered by autoantibodies from psychotic patients. Nat Commun 2017;8:1791.CrossRefGoogle ScholarPubMed
Aviv, R. The death debate; what does it mean to die? The New Yorker, 5 February 2018.Google Scholar
Lehmann-Facius, H. Über die Liquordiagnose der Schizophrenien. Klinische Wochenschrift 1937;16:16461648.CrossRefGoogle Scholar
Smith, RS. A comprehensive macrophage-T-lymphocyte theory of schizophrenia. Med Hypotheses 1992;39:248257.Google Scholar
Stein, M, Miller, AH, Trestman, RL. Depression, the immune system, and health and illness: findings in search of meaning. Arch Gen Psychiatry 1991;48:171177.Google Scholar
Pariante, CM. Psychoneuroimmunology or immunopsychiatry? Lancet Psychiatry 2015;2:197199.Google Scholar
Yirmiya, R. Endotoxin produces a depressive-like episode in rats. Brain Res 1996;711:163174.Google Scholar
Udina, M, Navines, R, Egmond, E, et al. Glucocorticoid receptors, brain-derived neurotrophic factor, serotonin and dopamine neurotransmission are associated with interferon-induced depression. Int J Neuropsychopharmacol 2016;19:pyv135.Google Scholar
Breitbart, W, Rosenfeld, B, Tobias, K, et al. Depression, cytokines, and pancreatic cancer. Psychooncology 2014;23:339345.Google Scholar
Pawar, DS, Molinaro, JR, Knight, JM, Heinrich, TW. Toxicities of CAR T-cell therapy and the role of the consultation-liaison psychiatrist. Psychosomatics 2019;60:519523.Google Scholar
Bodro, M, Compta, Y, Llanso, L, Esteller, D, et al. Increased CSF levels of IL-1b, IL-6, and ACE in SARS-CoV-2 associated encephalitis. Neurol Neuroimmunol Neuroimflamm 2020;7:e821.CrossRefGoogle Scholar
Brown, AS. The Kraepelinian dichotomy from the perspective of prenatal infectious and immunologic insults. Schizophr Bull 2015;41:786791.Google Scholar
Danese, A, Pariante, CM, Caspi, A, Taylor, A, Poulton, R. Childhood maltreatment predicts adult inflammation in a life-course study. Proc Natl Acad Sci USA 2007;104:13191324.Google Scholar
Copeland, WE, Wolke, D, Lereya, ST, et al. Childhood bullying involvement predicts low-grade systemic inflammation into adulthood. Proc Natl Acad Sci USA 2014;111:75707575.Google Scholar
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry 2011;70:663671.Google Scholar
Girgis, RR, Kumar, SS, Brown, AS. The cytokine model of schizophrenia: emerging therapeutic strategies. Biol Psychiatry 2014;75:292299.Google Scholar
Black, C, Miller, BJ. Meta-analysis of cytokines and chemokines in suicidality: distinguishing suicidal versus nonsuicidal patients. Biol Psychiatry 2015;78:2837.Google Scholar
Berk, M, Williams, LJ, Jacka, FN, et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med 2013;11:200.Google Scholar
Rosenblat, JD, McIntyre, RS. Are medical comorbid conditions of bipolar disorder due to immune dysfunction? Acta Psychiatr Scand 2015;132:180191.Google Scholar
Jeppesen, R, Benros, ME. Autoimmune diseases and psychotic disorders. Front Psychiatry 2019;10:131.Google Scholar
Benros, ME, Nielsen, PR, Nordentoft, M, et al. Autoimmune diseases and severe infections as risk factors for schizophrenia: a 30-year population-based register study. Am J Psychiatry 2011;168:13031310.Google Scholar
Benros, ME, Pedersen, MG, Rasmussen, H, et al. A nationwide study on the risk of autoimmune diseases in individuals with a personal or a family history of schizophrenia and related psychosis. Am J Psychiatry 2014;171:218226.Google Scholar
Chen, SW, Zhong, XS, Jiang, LN, et al. Maternal autoimmune diseases and the risk of autism spectrum disorders in offspring: a systematic review and meta-analysis. Behav Brain Res 2016;296:6169.Google Scholar
Brimberg, L, Mader, S, Jeganathan, V, et al. Caspr2-reactive antibody cloned from a mother of an ASD child mediates an ASD-like phenotype in mice. Mol Psychiatry 2016;21:16631671.Google Scholar
Coutinho, E, Jacobson, L, Pedersen, MG, et al. CASPR2 autoantibodies are raised during pregnancy in mothers of children with mental retardation and disorders of psychological development but not autism. J Neurol Neurosurg Psychiatry 2017;88:718721.Google Scholar
Atladottir, HO, Thorsen, P, Ostergaard, L, et al. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J Autism Dev Disord 2010;40:14231430.Google Scholar
Marder, SR, Cannon, TD. Schizophrenia. N Engl J Med 2019;381:17531761.Google Scholar
Schizophrenia Working Group of the Psychiatric Genomics C. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014;511:421427.CrossRefGoogle Scholar
Purcell, SM, Moran, JL, Fromer, M, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 2014;506:185190.Google Scholar
Sundararajan, T, Manzardo, AM, Butler, MG. Functional analysis of schizophrenia genes using GeneAnalytics program and integrated databases. Gene 2018;641:2534.Google Scholar
Fromer, M, Pocklington, AJ, Kavanagh, DH, et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 2014;506:179184.Google Scholar
Butler, MG, McGuire, AB, Masoud, H, Manzardo, AM. Currently recognized genes for schizophrenia: high-resolution chromosome ideogram representation. Am J Med Genet B Neuropsychiatr Genet 2016;171B:181202.Google Scholar
Tost, H, Alam, T, Meyer-Lindenberg, A. Dopamine and psychosis: theory, pathomechanisms and intermediate phenotypes. Neurosci Biobehav Rev 2010;34:689700.Google Scholar
Carlsson, A. Biochemical and pharmacological aspects of Parkinsonism. Acta Neurol Scand Suppl 1972;51:1142.Google Scholar
Yaryura-Tobias, JA, Diamond, B, Merlis, S. The action of L-dopa on schizophrenic patients (a preliminary report). Curr Ther Res Clin Exp 1970;12:528531.Google Scholar
Kim, JS, Kornhuber, HH, Schmid-Burgk, W, Holzmuller, B. Low cerebrospinal fluid glutamate in schizophrenic patients and a new hypothesis on schizophrenia. Neurosci Lett 1980;20:379382.CrossRefGoogle Scholar
Krystal, JH, Karper, LP, Seibyl, JP, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 1994;51:199214.Google Scholar
Kayser, MS, Titulaer, MJ, Gresa-Arribas, N, Dalmau, J. Frequency and characteristics of isolated psychiatric episodes in anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2013;70:11331139.Google Scholar
Martucci, L, Wong, AH, De Luca, V, et al. N-methyl-D-aspartate receptor NR2B subunit gene GRIN2B in schizophrenia and bipolar disorder: polymorphisms and mRNA levels. Schizophr Res 2006;84:214221.Google Scholar
Javitt, DC. Neurophysiological models for new treatment development in schizophrenia: early sensory approaches. Ann N Y Acad Sci 2015;1344:92104.Google Scholar
Lewis, DA, Hashimoto, T, Volk, DW. Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci 2005;6:312324.Google Scholar
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;12:125135.Google Scholar
Lisman, JE, Pi, HJ, Zhang, Y, Otmakhova, NA. A thalamo-hippocampal-ventral tegmental area loop may produce the positive feedback that underlies the psychotic break in schizophrenia. Biol Psychiatry 2010;68:1724.Google Scholar
Anticevic, A, Gancsos, M, Murray, JD, et al. NMDA receptor function in large-scale anticorrelated neural systems with implications for cognition and schizophrenia. Proc Natl Acad Sci USA 2012;109:1672016725.Google Scholar
Pilowsky, LS, Bressan, RA, Stone, JM, et al. First in vivo evidence of an NMDA receptor deficit in medication-free schizophrenic patients. Mol Psychiatry 2006;11:118119.Google Scholar
Poels, EM, Kegeles, LS, Kantrowitz, JT, et al. Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry 2014;19:2029.Google Scholar
Weickert, CS, Fung, SJ, Catts, VS, et al. Molecular evidence of N-methyl-D-aspartate receptor hypofunction in schizophrenia. Mol Psychiatry 2013;18:11851192.Google Scholar
Law, AJ, Deakin, JF. Asymmetrical reductions of hippocampal NMDAR1 glutamate receptor mRNA in the psychoses. Neuroreport 2001;12:29712974.Google Scholar
Mohn, AR, Gainetdinov, RR, Caron, MG, Koller, BH. Mice with reduced NMDA receptor expression display behaviors related to schizophrenia. Cell 1999;98:427436.Google Scholar
Papaleo, F, Lipska, BK, Weinberger, DR. Mouse models of genetic effects on cognition: relevance to schizophrenia. Neuropharmacology 2012;62:12041220.Google Scholar
Moghaddam, B, Javitt, D. From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 2012;37:415.Google Scholar
Manto, M, Dalmau, J, Didelot, A, Rogemond, V, Honnorat, J. In vivo effects of antibodies from patients with anti-NMDA receptor encephalitis: further evidence of synaptic glutamatergic dysfunction. Orphanet J Rare Dis 2010;5:31.Google Scholar
Dalmau, J. NMDA receptor encephalitis and other antibody-mediated disorders of the synapse: The 2016 Cotzias Lecture. Neurology 2016;87:24712482.Google Scholar
Planaguma, J, Leypoldt, F, Mannara, F, et al. Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain 2015;138:94109.Google Scholar
Haussleiter, IS, Emons, B, Schaub, M, et al. Investigation of antibodies against synaptic proteins in a cross-sectional cohort of psychotic patients. Schizophr Res 2012;140:258259.Google Scholar
Chen, CH, Cheng, MC, Liu, CM, et al. Seroprevalence survey of selective anti-neuronal autoantibodies in patients with first-episode schizophrenia and chronic schizophrenia. Schizophr Res 2017;190:2831.CrossRefGoogle ScholarPubMed
Endres, D, Meixensberger, S, Dersch, R, et al. Cerebrospinal fluid, antineuronal autoantibody, EEG, and MRI findings from 992 patients with schizophreniform and affective psychosis. Transl Psychiatry 2020;10:279.Google Scholar
Saether, SG, Schou, M, Kondziella, D. What is the significance of onconeural antibodies for psychiatric symptomatology? A systematic review. BMC Psychiatry 2017;17:161.Google Scholar
Saether, SG, Schou, M, Stoecker, W, et al. Onconeural antibodies in acute psychiatric inpatient care. J Neuropsychiatry Clin Neurosci 2017;29:7476.Google Scholar
Guasp, M, Gine-Serven, E, Rosa-Justicia, M, et al. Clinical, neuro-immunological, and CSF investigations in first episode psychosis. Neurology 2021;97:e61e75.Google Scholar
Pathmanandavel, K, Starling, J, Merheb, V, 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;77:537547.Google Scholar
Mantere, O, Saarela, M, Kieseppa, T, et al. Anti-neuronal anti-bodies in patients with early psychosis. Schizophr Res 2018;192:404407.Google Scholar
Engen, K, Wortinger, LA, Jorgensen, KN, et al. Autoantibodies to the N-methyl-D-aspartate receptor in adolescents with early onset psychosis and healthy controls. Front Psychiatry 2020;11:666.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013;12:157165.CrossRefGoogle ScholarPubMed
van Sonderen, A, Petit-Pedrol, M, Dalmau, J, Titulaer, MJ. The value of LGI1, Caspr2 and voltage-gated potassium channel antibodies in encephalitis. Nat Rev Neurol 2017;13:290301.Google Scholar
Tobin, WO, Lennon, VA, Komorowski, L, et al. DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients. Neurology 2014;83:17971803.Google Scholar
Arino, H, Armangue, T, Petit-Pedrol, M, et al. Anti-LGI1-associated cognitive impairment: presentation and long-term outcome. Neurology 2016;87:759765.Google Scholar
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;90:e1964e1972.Google Scholar
Graus, F, Boronat, A, Xifro, X, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010;74:857859.Google Scholar
Boronat, A, Gelfand, JM, Gresa-Arribas, N, et al. Encephalitis and antibodies to dipeptidyl-peptidase-like protein-6, a subunit of Kv4.2 potassium channels. Ann Neurol 2013;73:120128.Google Scholar
Petit-Pedrol, M, Armangue, T, Peng, X, et al. Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol 2014;13:276286.Google Scholar
Sonderen, AV, Arends, S, Tavy, DLJ, et al. Predictive value of electroencephalography in anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2018;89:11011106.Google Scholar
Armangue, T, Spatola, M, Vlagea, A, et al. Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: a prospective observational study and retrospective analysis. Lancet Neurol 2018;17:760772.Google Scholar
Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med 2018;378:840851.Google Scholar
Hansen, HC, Klingbeil, C, Dalmau, J, et al. Persistent intrathecal antibody synthesis 15 years after recovering from anti-N-methyl-D-aspartate receptor encephalitis. JAMA Neurol 2013;70:117119.Google Scholar
Swedo, SE, Leonard, HL, Garvey, M, et al. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: clinical description of the first 50 cases. Am J Psychiatry 1998;155:264271.Google Scholar
Swedo, SE, Rapoport, JL, Leonard, H, Lenane, M, Cheslow, D. Obsessive-compulsive disorder in children and adolescents: clinical phenomenology of 70 consecutive cases. Arch Gen Psychiatry 1989;46:335341.Google Scholar
Swedo, SE, Rapoport, JL, Cheslow, DL, et al. High prevalence of obsessive-compulsive symptoms in patients with Sydenham’s chorea. Am J Psychiatry 1989;146:246249.Google Scholar
Swedo, SE, Leonard, HL, Schapiro, MB, et al. Sydenham’s chorea: physical and psychological symptoms of St Vitus dance. Pediatrics 1993;91:706713.Google ScholarPubMed
Swedo, SE. Sydenham’s chorea: a model for childhood autoimmune neuropsychiatric disorders. JAMA 1994;272:17881791.Google Scholar
Rettew, DC, Swedo, SE, Leonard, HL, Lenane, MC, Rapoport, JL. Obsessions and compulsions across time in 79 children and adolescents with obsessive-compulsive disorder. J Am Acad Child Adolesc Psychiatry 1992;31:10501056.Google Scholar
Leonard, HL, Lenane, MC, Swedo, SE, et al. Tics and Tourette’s disorder: a 2- to 7-year follow-up of 54 obsessive-compulsive children. Am J Psychiatry 1992;149:12441251.Google Scholar
Swedo, SE, Leonard, HL, Kiessling, LS. Speculations on antineuronal antibody-mediated neuropsychiatric disorders of childhood. Pediatrics 1994;93:323326.Google Scholar
Chang, K, Frankovich, J, Cooperstock, M, et al. Clinical evaluation of youth with pediatric acute-onset neuropsychiatric syndrome (PANS): recommendations from the 2013 PANS Consensus Conference. J Child Adolesc Psychopharmacol 2015;25:313.CrossRefGoogle ScholarPubMed
Swedo, SE, Leckman, JF, Rose, NR. From research subgroup to clinical syndrome: modifying the PANDAS criteria to describe PANS (pediatric acute-onset neuropsychiatric syndrome). Pediatr Ther 2012;2:2.Google Scholar
Wilbur, C, Bitnun, A, Kronenberg, S, et al. PANDAS/PANS in childhood: controversies and evidence. Paediatr Child Health 2019;24:8591.Google Scholar
Singer, HS, Gilbert, DL, Wolf, DS, Mink, JW, Kurlan, R. Moving from PANDAS to CANS. J Pediatr 2012;160:725731.Google Scholar
Allen, AJ, Leonard, HL, Swedo, SE. Case study: a new infection-triggered, autoimmune subtype of pediatric OCD and Tourette’s syndrome. J Am Acad Child Adolesc Psychiatry 1995;34:307311.Google Scholar
Hesselmark, E, Bejerot, S. Clinical features of paediatric acute-onset neuropsychiatric syndrome: findings from a case–control study. BJPsych Open 2019;5:e25.Google Scholar
Hesselmark, E, Bejerot, S. Biomarkers for diagnosis of Pediatric Acute Neuropsychiatric Syndrome (PANS): sensitivity and specificity of the Cunningham Panel. J Neuroimmunol 2017;312:3137.Google Scholar
Silverman, M, Frankovich, J, Nguyen, E, et al. Psychotic symptoms in youth with Pediatric Acute-onset Neuropsychiatric Syndrome (PANS) may reflect syndrome severity and heterogeneity. J Psychiatr Res 2019;110:93102.Google Scholar
Bernstein, GA, Victor, AM, Pipal, AJ, Williams, KA. Comparison of clinical characteristics of pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections and childhood obsessive-compulsive disorder. J Child Adolesc Psychopharmacol 2010;20:333340.Google Scholar
Giedd, JN, Rapoport, JL, Garvey, MA, Perlmutter, S, Swedo, SE. MRI assessment of children with obsessive-compulsive disorder or tics associated with streptococcal infection. Am J Psychiatry 2000;157:281283.Google Scholar
Cunningham, MW. Rheumatic fever, autoimmunity, and molecular mimicry: the streptococcal connection. Int Rev Immunol 2014;33:314329.Google Scholar
Shimasaki, C, Frye, RE, Trifiletti, R, et al. Evaluation of the Cunningham Panel in pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection (PANDAS) and pediatric acute-onset neuropsychiatric syndrome (PANS): changes in antineuronal antibody titers parallel changes in patient symptoms. J Neuroimmunol 2020;339:577138.Google Scholar
Kirvan, CA, Swedo, SE, Heuser, JS, Cunningham, MW. Mimicry and autoantibody-mediated neuronal cell signaling in Sydenham chorea. Nat Med 2003;9:914920.Google Scholar
Sigra, S, Hesselmark, E, Bejerot, S. Treatment of PANDAS and PANS: a systematic review. Neurosci Biobehav Rev 2018;86:5165.Google Scholar
Brown, KD, Farmer, C, Freeman, GM Jr, et al. Effect of early and prophylactic nonsteroidal anti-inflammatory drugs on flare duration in pediatric acute-onset neuropsychiatric syndrome: an observational study of patients followed by an academic community-based pediatric acute-onset neuropsychiatric syndrome clinic. J Child Adolesc Psychopharmacol 2017;27:619628.Google Scholar
Murphy, TK, Storch, EA, Lewin, AB, Edge, PJ, Goodman, WK. Clinical factors associated with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections. J Pediatr 2012;160:314319.Google Scholar
Armangue, T, Titulaer, MJ, Malaga, I, et al. Pediatric anti-N-methyl-D-aspartate receptor encephalitis: clinical analysis and novel findings in a series of 20 patients. J Pediatr 2013;162:850856.Google Scholar
Mohammad, SS, Jones, H, Hong, M, et al. Symptomatic treatment of children with anti-NMDAR encephalitis. Dev Med Child Neurol 2016;58:376384.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Armangue, T, Olive-Cirera, G, Martinez-Hernandez, E, et al. Associations of paediatric demyelinating and encephalitic syndromes with myelin oligodendrocyte glycoprotein antibodies: a multicentre observational study. Lancet Neurol 2020;19:234246.CrossRefGoogle ScholarPubMed
Schiess, N, Pardo, CA. Hashimoto’s encephalopathy. Ann N Y Acad Sci 2008;1142:254265.Google Scholar
Hollowell, JG, Staehling, NW, Flanders, WD, et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab 2002;87:489499.Google Scholar
Mattozzi, S, Sabater, L, Escudero, D, et al. Hashimoto encephalopathy in the 21st century. Neurology 2020;94:e217e224.Google Scholar
Tiosano, S, Nir, Z, Gendelman, O, et al. The association between systemic lupus erythematosus and bipolar disorder: a big data analysis. Eur Psychiatry 2017;43:116119.Google Scholar
Tiosano, S, Farhi, A, Watad, A, et al. Schizophrenia among patients with systemic lupus erythematosus: population-based cross-sectional study. Epidemiol Psychiatr Sci 2017;26:424429.Google Scholar
American College of Rheumatology. The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Arthritis Rheum 1999;42:599608.Google Scholar
Ainiala, H, Hietaharju, A, Loukkola, J, et al. Validity of the new American College of Rheumatology criteria for neuropsychiatric lupus syndromes: a population-based evaluation. Arthritis Rheum 2001;45:419423.Google Scholar
Petri, M, Orbai, AM, Alarcon, GS, et al. Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus. Arthritis Rheum 2012;64:26772686.Google Scholar
Appenzeller, S, Cendes, F, Costallat, LT. Acute psychosis in systemic lupus erythematosus. Rheumatol Int 2008;28:237243.Google Scholar
Pego-Reigosa, JM, Isenberg, DA. Psychosis due to systemic lupus erythematosus: characteristics and long-term outcome of this rare manifestation of the disease. Rheumatology (Oxford) 2008;47:14981502.Google Scholar
Hanly, JG, Li, Q, Su, L, et al. Psychosis in systemic lupus erythematosus: results from an international inception cohort study. Arthritis Rheumatol 2019;71:281289.Google Scholar
Khajezadeh, MA, Zamani, G, Moazzami, B, et al. Neuropsychiatric involvement in juvenile-onset systemic lupus erythematosus. Neurol Res Int 2018;2018:2548142.Google Scholar
Sibbitt, WL Jr, Brandt, JR, Johnson, CR, et al. The incidence and prevalence of neuropsychiatric syndromes in pediatric onset systemic lupus erythematosus. J Rheumatol 2002;29:15361542.Google Scholar
Tay, SH, Mak, A. Comment on: Diagnosing and attributing neuropsychiatric events to systemic lupus erythematosus: time to untie the Gordian Knot?: Reply. Rheumatology (Oxford) 2017;56:857859.Google Scholar
Hanly, JG, Su, L, Urowitz, MB, et al. Mood disorders in systemic lupus erythematosus: results from an international inception cohort study. Arthritis Rheumatol 2015;67:18371847.Google Scholar
Tay, SH, Cheung, PP, Mak, A. Active disease is independently associated with more severe anxiety rather than depressive symptoms in patients with systemic lupus erythematosus. Lupus 2015;24:13921399.Google Scholar
Sherer, Y, Gorstein, A, Fritzler, MJ, Shoenfeld, Y. Autoantibody explosion in systemic lupus erythematosus: more than 100 different antibodies found in SLE patients. Semin Arthritis Rheum 2004;34:501537.Google Scholar
Zandman-Goddard, G, Chapman, J, Shoenfeld, Y. Autoantibodies involved in neuropsychiatric SLE and antiphospholipid syndrome. Semin Arthritis Rheum 2007;36:297315.Google Scholar
Hanly, JG, Urowitz, MB, Su, L, et al. Autoantibodies as biomarkers for the prediction of neuropsychiatric events in systemic lupus erythematosus. Ann Rheum Dis 2011;70:17261732.Google Scholar
Press, J, Palayew, K, Laxer, RM, et al. Antiribosomal P antibodies in pediatric patients with systemic lupus erythematosus and psychosis. Arthritis Rheum 1996;39:671676.Google Scholar
Sciascia, S, Bertolaccini, ML, Roccatello, D, Khamashta, MA, Sanna, G. Autoantibodies involved in neuropsychiatric manifestations associated with systemic lupus erythematosus: a systematic review. J Neurol 2014;261:17061714.Google Scholar
Gaburo, N Jr, de Carvalho, JF, Timo-Iaria, CIM, et al. Electrophysiological dysfunction induced by anti-ribosomal P protein antibodies injection into the lateral ventricle of the rat brain. Lupus 2017;26:463469.Google Scholar
Huerta, PT, Kowal, C, DeGiorgio, LA, Volpe, BT, Diamond, B. Immunity and behavior: antibodies alter emotion. Proc Natl Acad Sci USA 2006;103:678683.Google Scholar
Tin, SK, Xu, Q, Thumboo, J, et al. Novel brain reactive autoantibodies: prevalence in systemic lupus erythematosus and association with psychoses and seizures. J Neuroimmunol 2005;169:153160.Google Scholar
Varley, JA, Andersson, M, Grant, E, et al. Absence of neuronal autoantibodies in neuropsychiatric systemic lupus erythematosus. Ann Neurol 2020;88:12441250.Google Scholar
Haselmann, H, Mannara, F, Werner, C, et al. Human autoantibodies against the AMPA receptor subunit GluA2 induce receptor reorganization and memory dysfunction. Neuron 2018;100:91105.Google Scholar
Petit-Pedrol, M, Sell, J, Planaguma, J, et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018;141:31443159.Google Scholar
Carceles-Cordon, M, Mannara, F, Aguilar, E, et al. NMDAR antibodies alter dopamine receptors and cause psychotic behavior in mice. Ann Neurol 2020;88:603613.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Autoimmune Psychosis
  • Josep Dalmau, Universitat de Barcelona, Francesc Graus, Universitat de Barcelona
  • Book: Autoimmune Encephalitis and Related Disorders of the Nervous System
  • Online publication: 27 January 2022
  • Chapter DOI: https://doi.org/10.1017/9781108696722.020
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Autoimmune Psychosis
  • Josep Dalmau, Universitat de Barcelona, Francesc Graus, Universitat de Barcelona
  • Book: Autoimmune Encephalitis and Related Disorders of the Nervous System
  • Online publication: 27 January 2022
  • Chapter DOI: https://doi.org/10.1017/9781108696722.020
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Autoimmune Psychosis
  • Josep Dalmau, Universitat de Barcelona, Francesc Graus, Universitat de Barcelona
  • Book: Autoimmune Encephalitis and Related Disorders of the Nervous System
  • Online publication: 27 January 2022
  • Chapter DOI: https://doi.org/10.1017/9781108696722.020
Available formats
×