The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ReviewsFull Access

Autoimmune Encephalitis

Abstract

Antibodies to brain cell-surface proteins have recently been identified in patients whose symptoms cross the boundary between neurology and psychiatry and are associated with syndromes that are responsive to treatment with immunosuppression-based treatments. This has led to an expansion in the field of autoimmune encephalitis. The common syndromes, with particular relevance to psychiatry, are discussed, in particular the N-methyl-d-aspartate receptor antibody encephalitis syndrome, which is most often associated with a psychosis and cognitive prodrome resembling acute or first-episode psychosis. The recognition, clinical diagnosis and associations, and evidence for disease mechanisms are discussed in this brief review.

Definition

Autoimmune encephalitis is a clinical label applied to a group of disorders of the brain associated with a self-reactive immune response causing brain dysfunction. The group of disorders includes cellular-immunity mediated syndromes—such as acute disseminated encephalomyelitis (1), Rasmussen’s encephalitis (2), and intracellular-paraneoplastic antibody–associated encephalomyelitis (3)—but also disorders associated with autoantibodies to synaptic proteins (“autoimmune synaptopathies” [4]), the poststreptococcal neurological syndromes (5), encephalitis lethargica (6), and a combination of mechanisms (antibody, cytokine, vascular) in syndromes including neuropsychiatric systemic lupus erythematosus (7), neuro-Behçet’s disease (8), and neurosarcoidosis (9). Antibody-associated encephalitis often presents with a syndrome of psychiatric and cognitive features, which are often but not exclusively accompanied by seizures and movement disorders. This article focuses on antibody-associated synaptic disorders of the brain, which are of the most relevance to psychiatrists.

Assessment and Differential Diagnosis

Mental status change, fever, and seizures are hallmarks of classic encephalitis, and the identification and empirical treatment of viral infection of the brain, commonly herpes-simplex virus (HSV), is the first action required of physicians to avoid the devastating brain destruction that occurs from viral encephalitis and to identify bacterial causes of illness, including Listeria meningitis. Patients with autoimmune encephalitis may have a similar clinical presentation, often associated with fever, which in itself is not reliable to distinguish between infective and autoimmune encephalitis (10). Computed tomography and magnetic resonance imaging of the brain are often abnormal in infectious encephalitis, although often after a delay of 72 hours, and cerebrospinal fluid (CSF) examination for pleocytosis and HSV polymerase chain reaction may be falsely negative if done fewer than 72 hours into the illness; therefore, a strong index of suspicion is required to ensure that a diagnosis of infective encephalitis is not missed. Extensive investigation and consideration of alternative diagnoses is necessary, which can range across most differential diagnoses in neurology and psychiatry (drug-induced states, hereditary disease; e.g., C9ORF72-related frontotemporal dementia, intravascular and primary central nervous system [CNS] lymphoma, low-grade tumors of the brain, HIV infection).

Autoimmune disorders of brain synapses often produce symptoms that result in review by a psychiatrist first, particularly the N-methyl-d-aspartate receptor (NMDAR) antibody encephalitis: psychosis, mania, amnesia, and subcortical cognitive symptoms, catatonia-like movement disorders, and affective symptoms (4). Alternative symptoms, including seizures, headache, and cognitive dysfunction with prominent anterograde amnesia and frontal dysexecutive dysfunction, may be the prominent features at presentation. The nature of symptoms may be subacute or acute, often worsening over a period of weeks or months. The identification of different autoantibodies has been useful in allowing classification into “cell-surface antibody syndromes,” although there is considerable overlap (Table 1). Older clinical diagnoses—including Hashimoto’s encephalopathy (an encephalopathy first described by Lord Brain in 1966, associated with thyroid autoimmunity and rapid corticosteroid treatment responsiveness) (11) and Bickerstaff’s brainstem encephalitis (an encephalopathy associated with GQ1b ganglioside antibodies and the peripheral nerve Fisher syndrome [areflexia, ophthalmoplegia, and ataxia]) (12)—are likely to represent disorders on a clinical spectrum, and much work is under way to try to reorganize the classification on the basis of disease mechanisms and hence allow tailored therapy and the discovery of novel therapies.

Table 1. Autoimmune Neuropsychiatric Syndromesa

SyndromeNeuropsychiatric ManifestationsKnown Autoimmune Mechanism or Target
NMDAR encephalitisPsychosis, cognitive dysfunction, movement disorders, seizures (see Box 1)IgG antibodies to the NMDAR, which probably cause internalization of the NMDAR and disrupt surface trafficking of the receptors
VGKC-complex antibody encephalopathy (LGI1, caspr2 antibody related)Psychosis, amnesia, seizures, choreaIgG antibodies to LGI1 or caspr2. Low levels (<400 pM) of VGKC-complex antibodies determined by radioimmunoprecipitation assay alone may be nonpathogenic.
Hashimoto’s encephalopathyA syndrome of behavioral and cognitive change, rapidly responsive to steroidsMany cases include NMDAR antibodies. Thyroid peroxidase antibodies can be a nonspecific marker of this nonspecific syndromic diagnosis.
Bickerstaff’s brainstem encephalitisComa and the Fisher syndrome (ataxia, ophthalmoplegia, areflexia)GQ1b ganglioside antibodies
Encephalitis lethargicaPostinfective encephalopathy with movement, a broad range of psychiatric symptoms, and hyper- and hypokinetic movement disorders, e.g., highly prevalent in the 1920sMany cases with hyperkinetic movements have NMDAR antibodies.
CNS lupusPsychosis, cognitive dysfunction, seizures, and chorea, often at the presentation stage of systemic lupus erythematosusA heterogeneous group of conditions—vascular disease (associated with phospholipid antibodies), symptoms due to previous structural and often vascular damage, and probable antibody and cytokine-mediated dysfunction (no specific marker known)
Poststreptococcal neuropsychiatric disorders, including Sydenham’s choreaAffective symptoms, choreiform movement disorderA range of possible targets, including the dopamine–2 receptor
Autoimmune epilepsy syndromes (e.g., with antibodies to GABA and AMPA receptors), Rasmussen’s encephalitisExplosive-onset epilepsy, resistant to antiepileptic drugs, often with other features of an autoimmune encephalopathyAntibodies to GABA and AMPA receptors and cellular infiltration— less well characterized; can be tumor associated (paraneoplastic)
Progressive encephalomyelitis with rigidity and myoclonus Anxiety and affective symptoms, rigidity, encephalopathy, and seizuresAssociated with glycine receptor and GAD antibodies (mechanisms unclear)

aThere is considerable overlap in these disorders and classifications. NMDAR, N-methyl-d-aspartate receptor; VGKC, voltage-gated potassium channel; LGI1, leucine-rich glioma-inactivated 1; CNS, central nervous system; GABA, gamma amino butyric acid; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; GAD, glutamic acid decarboxylase.

Table 1. Autoimmune Neuropsychiatric Syndromesa

Enlarge table

NMDAR Antibody Encephalitis

Dalmau in 2005 described a group of young women with ovarian teratoma with an encephalitis characterized by a psychosis prodrome, rapid descent into profound coma, and rhythmic movement disorder, with antibodies to the NMDAR described in this group in 2007 (13, 14). The original patients had ovarian teratoma, likely leading to a paraneoplastic phenomenon, because the teratoma express neuronal tissue; but subsequently, it has been shown that most patients do not have an underlying tumor. People of all ages, from infants to the elderly, may be affected, although young adults are most commonly affected, and the disease is as common, if not more common, than infective encephalitis among those under age 30, for example, as seen in the California Encephalitis Project (15). In NMDAR encephalitis, a hyperkinetic movement disorder may be present more often than a parkinsonian syndrome; dysautonomia with sweating changes, tachycardia, and syncope may be present, although more often among patients with severe disease resulting in intensive care admission (1618). Brain imaging may be normal, reflecting the disease mechanism of antibody-mediated neuronal dysfunction rather than gross cellular infiltration. Untreated, the NMDAR encephalitis syndrome is associated with spontaneous remission and frequent relapse (18), as well as with long-term cognitive dysfunction and atrophy, likely as a result of damage induced by prolonged disease activity, although there is little research to date on long-term outcomes, particularly related to the other antibody associations. Electroencephalogram is often abnormal, although it may be normal for 20% of patients. CSF examination is useful for excluding mimics of autoimmune encephalitis and may reveal a cellular pleocytosis, often lymphocytic, or oligoclonal bands; but a normal spinal fluid examination does not exclude the disease. Low levels of NMDAR antibodies are seen perhaps as a secondary phenomenon in degenerative diseases such as motor neuron disease, prion disease, and Alzheimer’s disease; the clinical relevance of these antibodies is not clear (19). The NMDAR antibodies are likely pathogenic, reducing NMDAR currents in vitro and inducing appropriate effects in animal models with passive transfer. The likely mechanism is that of internalization of NMDAR antibodies and disruption of trafficking of the NMDAR at synapses (4). Patients with the full syndrome may have a protracted course, with some requiring ventilation in the intensive care unit for many months, but then making excellent recovery. A range of therapies have been tried—including intravenous immunoglobulins, plasmapheresis, oral immunosuppressant agents, cyclophosphamide, and B cell depletion with rituximab (18)—though clinical trials are required to identify the most effective and safe treatment regimens.

We and others have looked for NMDAR and related antibodies in patients with psychosis for three main reasons: two-thirds of patients with NMDAR encephalitis first see a psychiatrist (11, 17), NMDAR hypofunction is a good model of psychosis and schizophrenia (20), and the neuropathology of schizophrenia (21) has similarities to the neuropathology of NMDAR encephalitis (22). One caveat with patients experiencing psychosis is that they often have clinical or subclinical movement disorders and autonomic and cognitive dysfunction, although not at a level that would normally prompt referral to a neurologist. We and others have found a small percentage (2%−7%) of patients with relatively pure acute psychosis to have NMDAR or related antibodies (2325). These patients often have a psychosis refractory to conventional therapies, although they are highly sensitive to immunotherapies (26). Different prevalence rates in different epidemiological studies looking for these antibodies in psychosis are likely related to the methodology of antibody measurement as well as to the demographic characteristics of those tested (27). In particular, caution is called for in the interpretation of prefixed cell-based assay slides and in the high prevalence of IgA and IgM (unlikely to be pathogenic) found, for instance, in a large number of disease controls and in dementias (28). The possibility that antibody-associated autoimmunity underlies the symptoms of some patients with psychotic illness, who may improve with available therapies, is very exciting, although further study and treatment require care, particularly with the risks of immunotherapies and immunosuppressives, and well-designed randomized clinical trials are now required.

A further discrete group is younger patients, often infants, with HSV-1 encephalitis who recover with treatment but relapse with a hyperkinetic movement disorder and who have an absence of HSV-1 in CSF by polymerase chain reaction and a presence of NMDAR antibodies (29). The association of HSV-1 infection with cognitive symptoms in schizophrenia is a potential link here and deserves further study (30). Further detailed reviews are provided by Graus et al. (31), Irani et al. (32), and Crisp et al. (4).

LGI1, Caspr2, and Antibodies to the Voltage-Gated Potassium Channel (VGKC) Complex

In 2001, before the NMDAR antibody discovery, Angela Vincent and colleagues at Oxford University described anterograde amnesia and seizures with high-intensity signal change in mesial temporal lobes (overall, a “limbic” picture) in two patients with high levels of antibodies to the VGKC determined by radioimmunoprecipitation assay (33). One of the original patients had coexistent myasthenia gravis. The radioimmunoprecipitation assay identifies antibodies to proteins complexed to the VGKC, commonly leucine-rich glioma-inactivated 1 (LGI1) (34) and caspr2 (35). Low levels of antibodies to the VGKC measured by radioimmunoprecipitation assay may therefore detect intracellular binding and reflect an epiphenomenal finding or be found at low levels in healthy aging (36). A serum level of 400 pM or above is more likely to be disease relevant in the appropriate clinical setting, although in the absence of LGI1 or caspr2 antibodies it should be treated with caution. Caspr2 antibodies are more likely to be associated with an occult underlying malignancy, and a thorough search, including with positron emission tomography imaging, is recommended. Caspr2 is expressed in the juxtaparanode, and patients with this antibody are more likely to experience muscle cramps and sweating, reflecting peripheral nerve hyperexcitability (the Isaac syndrome in isolation [37], or Morvan’s syndrome when in combination with a “limbic” encephalitis [35]). Brief stereotyped focal dystonic seizures (faciobrachial dystonic seizures [FBDS]) involving the face and arm, and sometimes leg, can occur from tens to hundreds of times a day, refractory to anticonvulsant therapy (38, 39) in patients with LGI1 antibodies. FBDS can precede the amnesia in this syndrome, and corticosteroid treatment is associated with a better response and probably prevention of atrophy and damage in retrospective series (40), although no randomized trials have been undertaken in this rare disorder. The amnesia may be subtle, and the study of these patients has furthered insight into basic memory mechanisms in the temporal lobe, for example, demonstrating impaired binding together of disparate features of an object in memory (41). Isolated psychosis may occur, often in relapse of patients with LGI1 antibodies (39), and psychiatric symptoms, including affective and psychotic and cognitive symptoms, are common (unpublished data). Cardiac arrhythmias occur in NMDAR and LGI1 antibody-associated encephalitis, and ECG monitoring is recommended, particularly in the severe stages of the illness (42).

Less Common Antibody Associations

Novel antibodies are being discovered at a rapid pace, some with clues to clinical syndromes (4). Glycine receptor antibodies, the target highly expressed in the spinal cord and brainstem, are associated with progressive encephalomyelitis, rigidity, and myoclonus and with opsoclonus and encephalitis with seizures among some patients (43). Some of these patients have antibodies to the intracellular protein glutamic acid decarboxylase, an antibody usually associated with stiff-person syndrome and type 1 diabetes (44). Dopamine D2-receptor antibodies are associated with a brainstem encephalitis with prominent psychosis and hypo- and hyperkinetic movement disorders (45); antidipeptidyl-peptidase–like protein-6 antibodies are associated with profuse diarrhea prior to an encephalitic illness (46); the gamma-aminobutyric acid (GABA)-(A) and GABA(B) receptor antibodies are associated with refractory status epilepticus (47). Descriptions of these antibody associations are few and require replication and description in larger populations. Table 1 summarizes these and related disorders, including poststreptococcal neuropsychiatric disorders, CNS lupus, and encephalitis lethargica (Box 1).

BOX 1. COMMON NEUROPSYCHIATRIC MANIFESTATIONS OF N-METHYL-D-ASPARTATE RECEPTOR ENCEPHALITIS

Good epidemiologic data are lacking on the prevalence of each manifestation, but those listed below are all commonly seen in patients with NMDAR encephalitis.

Cognitive dysfunction (including frontal dysexecutive syndromes, temporal lobe amnestic syndromes, and subcortical processing dysfunction)

Positive psychotic symptoms (delusions, hallucinations)—often resistant to conventional therapies

Negative psychotic symptoms—often resistant to conventional therapies

Affective symptoms (e.g., depressed mood, mania)

Movement disorder, usually hyperkinetic

Seizures—often resistant to conventional therapies

Sleep disturbance, including REM sleep disturbance

Future Directions

The disease-causing mechanisms of antibodies to brain proteins are under study in this novel field, including passive-transfer experiments (injection of antibodies into the blood or spinal fluid in mice or rats to determine the effects of the antibodies, followed by a washout period to see whether the effects can be reversed) (48), in-depth analysis of the origins of the immunoglobulins and immunizing mechanisms (49), and biomarker discovery (e.g., protein markers measurable in blood or spinal fluid that can help track disease activity with treatments, or predict disease activity states). A hunt is under way for the discovery of novel antigen targets that may help categorize subtypes of illness and aid rapid clinical diagnosis, through extensive testing of possible cases but also with neurological, immunological, and healthy controls to independently judge the specificity of an antibody test. Gold-standard tests, such as live cell-based assays, will have advantages over rapid fixed immunoassays and enzyme-linked immunosorbent assay tests, although there will be a role for the latter in terms of providing rapid results to clinicians—within a day of blood sampling, which is a situation seen in rheumatology in the antinuclear antibody testing for suspected systemic lupus erythematosus. The field is in need of in-depth clinical and phenotypic analysis (50) to provide independent biomarkers to aid in clinical diagnosis and research studies, given the nonspecific nature of many of the phenotypes—for instance, advanced brain imaging or CSF biomarkers of inflammation or damage. Greater integrative working between psychiatrists and neurologists is required to take forward work in synaptic neuropsychiatric disorders, which heavily relies on collaboration, sample collection, and engagement of patients and charities in research, including natural history studies and clinical trials. Immune mechanisms are increasingly implicated in the pathogenesis of degenerative diseases, including Alzheimer’s disease and Parkinson’s disease. The ability to modify and target immune mechanisms through the repurposing of existing therapies to allow modification of the disease course in neurodegenerative disease is a topic of great interest.

Dr. Zandi is with the Institute of Neurology, University College London, London, United Kingdom, and with the National Institute for Health Research, University College London Hospitals Biomedical Research Centre. Address correspondence to Dr. Zandi at the UCL Institute of Neurology, Queen Square House, Queen Square, London, United Kingdom.

Dr. Zandi is supported by the University College London Biomedical Resource Centre.

Dr. Zandi reports receiving honoraria for lecturing from Eisei.

References

1 Tenembaum S, Chitnis T, Ness J, et al.: Acute disseminated encephalomyelitis. Neurology 2007; 68(Suppl 2):S23–S36CrossrefGoogle Scholar

2 Bauer J, Elger CE, Hans VH, et al.: Astrocytes are a specific immunological target in Rasmussen’s encephalitis. Ann Neurol 2007; 62:67–80CrossrefGoogle Scholar

3 Dalmau J, Rosenfeld MR: Paraneoplastic syndromes of the CNS. Lancet Neurol 2008; 7:327–340CrossrefGoogle Scholar

4 Crisp SJ, Kullmann DM, Vincent A: Autoimmune synaptopathies. Nat Rev Neurosci 2016; 17:103–117CrossrefGoogle Scholar

5 Swedo SE, Seidlitz J, Kovacevic M, et al.: Clinical presentation of pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections in research and community settings. J Child Adolesc Psychopharmacol 2015; 25:26–30CrossrefGoogle Scholar

6 Dale RC, Irani SR, Brilot F, et al.: N-methyl-D-aspartate receptor antibodies in pediatric dyskinetic encephalitis lethargica. Ann Neurol 2009; 66:704–709CrossrefGoogle Scholar

7 Hanly JG, Urowitz MB, Su L, et al.: Prospective analysis of neuropsychiatric events in an international disease inception cohort of patients with systemic lupus erythematosus. Ann Rheum Dis 2010; 69:529–535CrossrefGoogle Scholar

8 Al-Araji A, Kidd DP: Neuro-Behçet’s disease: epidemiology, clinical characteristics, and management. Lancet Neurol 2009; 8:192–204CrossrefGoogle Scholar

9 Hoitsma E, Faber CG, Drent M, et al.: Neurosarcoidosis: a clinical dilemma. Lancet Neurol 2004; 3:397–407CrossrefGoogle Scholar

10 Granerod J, Ambrose HE, Davies NW, et al.: Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis 2010; 10:835–844CrossrefGoogle Scholar

11 Brain L, Jellinek EH, Ball K: Hashimoto’s disease and encephalopathy. Lancet 1966; 2:512–514CrossrefGoogle Scholar

12 Odaka M, Yuki N, Yamada M, et al.: Bickerstaff’s brainstem encephalitis: clinical features of 62 cases and a subgroup associated with Guillain-Barré syndrome. Brain 2003; 126:2279–2290CrossrefGoogle Scholar

13 Ances BM, Vitaliani R, Taylor RA, et al.: Treatment-responsive limbic encephalitis identified by neuropil antibodies: MRI and PET correlates. Brain 2005; 128:1764–1777CrossrefGoogle Scholar

14 Dalmau J, Tüzün E, Wu HY, et al.: Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 2007; 61:25–36CrossrefGoogle Scholar

15 Gable MS, Sheriff H, Dalmau J, 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; 54:899–904CrossrefGoogle Scholar

16 Dalmau J, Gleichman AJ, Hughes EG, et al.: Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008; 7:1091–1098CrossrefGoogle Scholar

17 Irani SR, Bera K, Waters P, 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; 133:1655–1667CrossrefGoogle Scholar

18 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; 12:157–165CrossrefGoogle Scholar

19 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; 86:708–713CrossrefGoogle Scholar

20 Javitt DC, Zukin SR, Heresco-Levy U, et al.: Has an angel shown the way? Etiological and therapeutic implications of the PCP/NMDA model of schizophrenia. Schizophr Bull 2012; 38:958–966CrossrefGoogle Scholar

21 Harrison PJ: The neuropathology of schizophrenia: a critical review of the data and their interpretation. Brain 1999; 122:593–624CrossrefGoogle Scholar

22 Bien CG, Vincent A, Barnett MH, et al.: Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 2012; 135:1622–1638CrossrefGoogle Scholar

23 Zandi MS, Irani SR, Lang B, et al.: Disease-relevant autoantibodies in first episode schizophrenia. J Neurol 2011; 258:686–688CrossrefGoogle Scholar

24 Kayser MS, Titulaer MJ, Gresa-Arribas N, et al.: Frequency and characteristics of isolated psychiatric episodes in anti–N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2013; 70:1133–1139CrossrefGoogle Scholar

25 Pollak TA, McCormack R, Peakman M, et al.: Prevalence of anti-N-methyl-d-aspartate (NMDA) antibodies in patients with schizophrenia and related psychoses: a systematic review and meta-analysis. Psychol Med 2014; 44:2475–2487CrossrefGoogle Scholar

26 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; 160:193–195CrossrefGoogle Scholar

27 Deakin J, Lennox BR, Zandi MS: Antibodies to the N-methyl-D-aspartate receptor and other synaptic proteins in psychosis. Biol Psychiatry 2014; 75:284–291CrossrefGoogle Scholar

28 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; 1:822–832CrossrefGoogle Scholar

29 Hacohen Y, Deiva K, Pettingill P, et al.: N-methyl-D-aspartate receptor antibodies in post-herpes simplex virus encephalitis neurological relapse. Mov Disord 2014; 29:90–96CrossrefGoogle Scholar

30 Dickerson FB, Boronow JJ, Stallings C, et al.: Association of serum antibodies to herpes simplex virus 1 with cognitive deficits in individuals with schizophrenia. Arch Gen Psychiatry 2003; 60:466–472CrossrefGoogle Scholar

31 Graus F, Titulaer MJ, Balu R, et al.: A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016; 15:391–404CrossrefGoogle Scholar

32 Irani SR, Gelfand JM, Al-Diwani A, et al.: Cell-surface central nervous system autoantibodies: clinical relevance and emerging paradigms. Ann Neurol 2014; 76:168–184CrossrefGoogle Scholar

33 Buckley C, Oger J, Clover L, et al.: Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann Neurol 2001; 50:73–78CrossrefGoogle Scholar

34 Irani SR, Alexander S, Waters P, et al.: Antibodies to Kv1 potassium channel–complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010; 133:2734–2748CrossrefGoogle Scholar

35 Irani SR, Pettingill P, Kleopa KA, et al.: Morvan syndrome: clinical and serological observations in 29 cases. Ann Neurol 2012; 72:241–255CrossrefGoogle Scholar

36 Paterson RW, Zandi MS, Armstrong R, et al.: Clinical relevance of positive voltage-gated potassium channel (VGKC)-complex antibodies: experience from a tertiary referral centre. J Neurol Neurosurg Psychiatry 2014; 85:625–630CrossrefGoogle Scholar

37 Newsom-Davis J, Buckley C, Clover L, et al.: Autoimmune disorders of neuronal potassium channels. Ann NY Acad Sci 2003; 998:202–210CrossrefGoogle Scholar

38 Irani SR, Michell AW, Lang B, et al.: Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011; 69:892–900CrossrefGoogle Scholar

39 Brown JW, Martin PJ, Thorpe JW, et al.: Long-term remission with rituximab in refractory leucine-rich glioma inactivated 1 antibody encephalitis. J Neuroimmunol 2014; 271:66–68CrossrefGoogle Scholar

40 Irani SR, Stagg CJ, Schott JM, et al.: Faciobrachial dystonic seizures: the influence of immunotherapy on seizure control and prevention of cognitive impairment in a broadening phenotype. Brain 2013; 136:3151–3162CrossrefGoogle Scholar

41 Pertzov Y, Miller TD, Gorgoraptis N, et al.: Binding deficits in memory following medial temporal lobe damage in patients with voltage-gated potassium channel complex antibody-associated limbic encephalitis. Brain 2013; 136:2474–2485CrossrefGoogle Scholar

42 Naasan G, Irani SR, Bettcher BM, et al.: Episodic bradycardia as neurocardiac prodrome to voltage-gated potassium channel complex/leucine-rich, glioma inactivated 1 antibody encephalitis. JAMA Neurol 2014; 71:1300–1304CrossrefGoogle Scholar

43 Carvajal-González A, Leite MI, Waters P, et al.: Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain 2014; 137:2178–2192CrossrefGoogle Scholar

44 Saiz A, Blanco Y, Sabater L, et al.: Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 2008; 131:2553–2563CrossrefGoogle Scholar

45 Dale RC, Merheb V, Pillai S, et al.: Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain 2012; 135:3453–3468CrossrefGoogle Scholar

46 Piepgras J, Höltje M, Michel K, et al.: Anti-DPPX encephalitis: pathogenic effects of antibodies on gut and brain neurons. Neurology 2015; 85:890–897CrossrefGoogle Scholar

47 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; 13:276–286CrossrefGoogle Scholar

48 Planagumà J, Leypoldt F, Mannara F, et al.: Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain 2015; 138:94–109CrossrefGoogle Scholar

49 Irani SR, Lehmann-Horn K, Geschwind M, et al.: The active intrathecal B-cell response in LGI1-antibody encephalitis. Lancet 2015; 385(Suppl 1):S46CrossrefGoogle Scholar

50 Pollak TA, Beck K, Irani SR, et al.: Autoantibodies to central nervous system neuronal surface antigens: psychiatric symptoms and psychopharmacological implications. Psychopharmacology (Berl) 2016; 233:1605–1621CrossrefGoogle Scholar