Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2014 Apr 9;176(2):149–164. doi: 10.1111/cei.12271

Neuromyelitis optica: clinical features, immunopathogenesis and treatment

S Jarius *, B Wildemann *, F Paul
PMCID: PMC3992027  PMID: 24666204

Abstract

The term ‘neuromyelitis optica’ (‘Devic's syndrome’, NMO) refers to a syndrome characterized by optic neuritis and myelitis. In recent years, the condition has raised enormous interest among scientists and clinical neurologists, fuelled by the detection of a specific serum immunoglobulin (Ig)G reactivity (NMO-IgG) in up to 80% of patients with NMO. These autoantibodies were later shown to target aquaporin-4 (AQP4), the most abundant water channel in the central nervous system (CNS). Here we give an up-to-date overview of the clinical and paraclinical features, immunopathogenesis and treatment of NMO. We discuss the widening clinical spectrum of AQP4-related autoimmunity, the role of magnetic resonance imaging (MRI) and new diagnostic means such as optical coherence tomography in the diagnosis of NMO, the role of NMO-IgG, T cells and granulocytes in the pathophysiology of NMO, and outline prospects for new and emerging therapies for this rare, but often devastating condition.

Other Articles published in this series

  • Paraneoplastic neurological syndromes. Clinical and Experimental Immunology 2014, 175: 336–48.

  • Disease-modifying therapy in multiple sclerosis and chronic inflammatory demyelinating polyradiculoneuropathy: common and divergent current and future strategies. Clinical and Experimental Immunology 2014, 175: 359–72.

  • Monoclonal antibodies in treatment of multiple sclerosis. Clinical and Experimental Immunology 2014, 175: 373–84.

  • CLIPPERS: chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids. Review of an increasingly recognized entity within

  • the spectrum of inflammatory central nervous system disorders. Clinical and Experimental Immunology 2014, 175: 385–96.

  • Requirement for safety monitoring for approved multiple sclerosis therapies: an overview. Clinical and Experimental Immunology 2014, 175: 397–407.

  • Myasthenia gravis: an update for the clinician. Clinical and Experimental Immunology 2014, 175: 408–18.

  • Cerebral vasculitis in adults: what are the steps in order to establish the diagnosis? Red flags and pitfalls. Clinical and Experimental Immunology 2014, 175: 419–24.

  • Multiple sclerosis treatment and infectious issues: update 2013. Clinical and Experimental Immunology 2014, 175: 425–38.

  • Diagnosis, pathogenesis and treatment of myositis: recent advances 2014, 175: 349–58.

  • Management of disease-modifying treatments in neurological autoimmune diseases of the central nervous system 2014, 176: 135–48.

Keywords: aquaporin-4 antibodies (AQP4), Devic syndrome, diagnosis, neuromyelitis optica, NMO-IgG, pathogenesis, pathophysiology, treatment

Introduction

Neuromyelitis optica (NMO, Devic's syndrome) is an inflammatory disorder of the central nervous system (CNS) that presents typically with relapses of optic neuritis (ON) or myelitis 14. In recent years, the condition has raised enormous interest among scientists and clinical neurologists, fuelled by the detection of a highly specific serum immunoglobulin (Ig)G autoantibody (NMO-IgG) targeting the most abundant astrocytic water channel aquaporin-4 (AQP4) 58. NMO-IgG/AQP4-antibodies are present in up to 80% of patients with NMO 811. This seminal discovery has – together with previous neuropathological work that had already suggested humoral mechanisms to be relevant in the disease pathogenesis 12 – made clear that in most cases NMO is not a subform of multiple sclerosis (MS), as had been assumed for decades, but rather an autoimmune condition with an immunopathogenesis distinct from that of MS despite considerable overlap in clinical presentation and paraclinical findings. AQP4-antibody-positive NMO is part of an expanding spectrum of humorally mediated autoimmune diseases of the CNS that have been identified over the last few years 13,14. Several studies suggest that optimum treatment options may differ between NMO and MS, which underscores the necessity for a timely and accurate diagnosis. Another important advance was the discovery that AQP4 autoimmunity is associated with a much broader range of CNS symptoms than just NMO; this prompted the proposal to refer to the condition by terms such as ‘NMO spectrum disorder’ (NMOSD) 15, ‘autoimmune AQP4 channelopathy’ 16, ‘AQP4 autoimmune syndrome’ 17 or ‘AQP4 encephalomyelitis’ 18.

The aim of this review paper is to summarize current knowledge on the pathogenesis of AQP4-antibody-related NMO and to provide an update on the widening clinical spectrum, relevant paraclinical findings and current treatments.

History

First reports on patients with myelitis and amaurosis date back to the early 19th century 1824. However, neurologists and ophthalmologists only developed sustained interest in this rare syndrome after Eugène Devic and his student Fernand Gault published a review in 1894 [25,26]. Devic and Gault also coined the term neuro-myélite optique aiguë 25,26. In 1907 the Turkish physician Acchioté suggested naming the syndrome after Devic 18.

Epidemiology

Epidemiological and population-based studies suggest that the prevalence of NMO ranges from <1/100 000 to 4·4/100 000 in Europe and North America 2731. However, the true number of cases may be higher, as some studies reported a rate of patients misdiagnosed with MS as high as 30–40%, especially before tests for AQP4 antibodies became broadly available 1,32. Typical age at onset peaks at approximately 35–45 years, but NMO may also become manifest in children and the elderly 1,3339. Female preponderance is substantially higher in seropositive (∼9–10:1) than in seronegative patients (∼2:1) 1,40. The majority of NMO cases are sporadic, although rare familial cases indistinguishable from the former with respect to clinical presentation, age and sex distribution have been reported 41.

Clinical presentation, disease course and prognosis

In more than 90% of patients, NMO is a relapsing disease with attacks of ON, myelitis or both, occurring unpredictably 1. A monophasic course accounts for the remaining 10% and is more often associated with simultaneous ON and myelitis 1,36, while a progressive course seems to be extremely uncommon 42. Attacks of ON and myelitis are often more disabling and, if untreated, remission is poorer than in MS, which leads to a faster accrual of irreversible neurological disability. Following older studies, approximately 60% of patients exhibited severely impaired ambulation [expanded disability status scale (EDSS) 43 ≥6] or blindness in at least one eye after a disease course of 7–8 years 36. Five-year survival rate was reported to be as low as 68% in a North American study on patients seen between 1977 and 1997, which is in strong contrast to more recent studies that report 5-year survival rates of more than 90% 1,44. In a small subset of patients the disease may follow a benign course, with only minor disability after up to 10 years 1,45. The majority of NMO-related deaths result from severe ascending cervical myelitis or brainstem involvement leading to respiratory failure 1,36. The discrepancy between older and newer studies with respect to prognosis and survival rate may be explained by increased awareness of the disease subsequent to the detection and availability of AQP testing in clinical routine. Thus, it can be assumed that the rate of misdiagnoses may have dropped and that more patients are diagnosed and treated earlier. Moreover, treatment options have improved. Nevertheless, NMO remains a potentially life-threatening and severely disabling condition that usually requires prompt and consequent immunosuppressive treatment. Clinical decision-making with respect to diagnosis and treatment initiation remains challenging when a patient presents with ON or myelitis only, or with other clinical symptoms, such as brainstem encephalitis with intractable hiccups and vomiting or a syndrome of inappropriate anti-diuretic hormone secretion 1,4650. In such cases, testing for AQP4-antibody by means of a both highly sensitive and highly specific assay can be essential 51. Other symptoms and syndromes that have occasionally been reported in association with AQP4 autoimmunity include seizures 52, posterior reversible encephalopathy syndrome 53, myeloradiculitis 54, meningoencephalitis 55, findings related to brainstem involvement, such as hearing loss, diplopia, olfactory dysfunction and other cranial nerve palsies, or endocrinological abnormalities due to diencephalic lesions 1,5658. Moreover, pain syndromes 1,59,60 and cognitive dysfunction 6163 seem to develop more frequently than appreciated previously.

In contrast to MS, a higher proportion of NMO patients (30–50%) exhibit laboratory findings or clinical signs of other systemic or organ-specific autoimmunity, such as systemic lupus erythematosus, Sjögren's syndrome, autoimmune thyroid disease, myasthenia gravis or, possibly, autoimmune-mediated vitamin B12 deficiency 6474. The invariable association with myelitis and/or ON suggests that AQP4 antibodies in patients with rheumatic diseases do not represent an unspecific epiphenomenon, but rather points to the existence of two concomitant autoimmune conditions.

Two studies found an increase in relapse rate in the first or the first and second trimenon, respectively, after delivery 75,76. Preliminary data suggest that AQP4-antibodies might also be capable of causing damage in AQP4-expressing organs and tissues outside the CNS (e.g. placentitis with the risk of miscarriage 7779, myositis 8083, internal otitis 56 or gastritis 74).

Diagnostic criteria

In 2006, the diagnostic criteria for NMO were revised after NMO-IgG were detected. In addition to including this novel and highly specific marker, the absolute restriction of CNS involvement beyond the optic nerves and spinal cord was removed and the specificity of longitudinally extensive spinal cord lesions emphasized 84,85. In addition to the two clinical index events of ON and acute myelitis, a diagnosis of NMO now requires that two of the three following supportive criteria be fulfilled:

  • Contiguous spinal cord magnetic resonance imaging (MRI) lesion extending over three or more vertebral segments;

  • Brain MRI not meeting diagnostic criteria for MS according to Paty 86; and

  • NMO-IgG seropositive status.

Given the rapid expansion of our knowledge on NMO, it is to be expected that these diagnostic criteria may be modified or replaced in the nearer future.

Immunopathogenesis

Several lines of evidence from clinical, pathological and immunological studies indicate that AQP4-antibodies have a decisive role in the pathogenesis of NMO 8790:

  • (a) NMO-IgG/AQP4-IgG is highly specific for NMO and its limited forms 9,51,88. The largest study performed thus far found the antibody in only 0·6% of 1672 controls using a tissue-based assay (TBA) 29. Similarly, specificity rates as high as 99·83% (n = 604; TBA) 91, 99·57% [n = 234; cell-based assay (CBA)] 92, 99·27% (n = 137; TBA) 7, 99·71% (n = 695, TBA) 93, 98·69% [n = 153, enzyme-linked immunosorbent assay (ELISA)] 10, 100% (n = 100, CBA 9, n = 85, CBA 11, n = 114, fluorescence activated cell sorter (FACS) 94, n = 178, ELISA 94, n = 85, immunoprecipitation 11) were reported in a number of recent studies (see references 88 and 51 for a comprehensive summary).

  • (b) AQP4-IgG serum levels were found to correlate with NMO disease activity in several independent studies 10,9597, serum levels increasing shortly before relapse and declining during recovery 9597.

  • (c) Similarly, AQP4-antibody-positive plasmablasts are selectively increased in the blood of NMO patients and peak at relapse 98.

  • (d) In patients with isolated ON and in patients with isolated longitudinally extensive transverse myelitis (LETM), AQP4-antibodies have been shown to predict conversion to NMO, i.e. the development of additional LETM or additional ON, respectively 10,99101.

  • (e) In patients with NMO or its limited forms, the presence of AQP4-antibodies predicts future relapse 1,99101.

  • (f) Clinically, the presence of AQP4-antibodies in patients with NMO is associated with distinct phenotypic features. AQP4-antibody-positive patients are more often female (∼10:1), show a relapsing disease course more often than seronegative patients and frequently exhibit signs of co-existing autoimmunity 10,102,103. By contrast, seronegative NMO is more frequently monophasic and shows only a slight female preponderance (∼1:2) 10,102.

  • (g) Some studies found a correlation between disease severity and AQP4-antibody status and/or titres 9597,102: the presence of AQP4-antibodies may be associated with more extensive spinal involvement as assessed by MRI 1,97, more severe attack-related optic and/or motor disability 1,97 and, possibly, more severe disability in the long-term course 102. Seronegative NMO was also reported to take a milder disease course in paediatric patients 35. In addition to AQP4-antibodies, genetic background and age at onset also seem to determine prognosis 103.

  • (h) B cell or antibody-targeted treatments have been found to be effective in NMO, including plasma exchange (PE) 104109, B cell depletion by rituximab 87,110114 and the interleukin (IL)-6 receptor inhibitor tocilizumab 115117.

  • (i) Incomplete B cell depletion or recurrence of B cells is associated with breakthrough attacks 95,114.

  • (j) Successful treatment is usually associated with a decline in AQP4-antibody serum concentrations and stable suppression during remission 10,9597.

  • (k) AQP4 is expressed at highest levels in opticospinal tissues, which also appear to contain higher amounts of supramolecular AQP4 aggregates 118. An immune response targeting AQP4 could thus well explain why the optic nerve and the spinal cord are predilection sites in NMO.

  • (l) NMO lesions are characterized by a marked loss of astrocytic AQP4 and by prominent deposits of IgG and IgM concentrated around blood vessels, i.e. at the main sites of AQP4 expression 12,119121.

  • (m) In some lesions astroglial loss is not associated with loss of myelin and neuronal axons 119,120,122, suggesting that the initial immune response in NMO is directed against astrocytes, a cell population that expresses AQP4 at high levels.

  • (n) While AQP4 is lost in parallel with glial fibrillary acidic protein (GFAP), indicating astrocyte loss, GFAP is preserved in other lesions, indicating that AQP4 is the primary target of the anti-astrocytic immune response in NMO 119,120,122. This initial loss of AQP4 might reflect internalization (and possibly endolysosomal degradation 123) of AQP4 (either of M1-AQP4 124 or of both isoforms 125), as demonstrated both in transfected cell lines and in cultured astrocytes 123,124,126128, and might thus still be reversible. However, as a limitation, a more recent study could not find evidence of AQP4 endocytosis in vivo after injecting fluorescent AQP4-antibodies 129.

  • (o) Most importantly, passive transfer animal experiments using IgG from AQP4-antibody-positive patients were able to reproduce the neuropathological features of NMO. Intracerebral injection of IgG from AQP4-antibody-positive patients, together with human complement, caused a marked loss of astrocytes 130. However, the fact that pretreatment with complete Freund's adjuvant or pre-existing experimental autoimmune encephalomyelitis (EAE) was required for inducing tissue damage in studies administering IgG intravenously or intraperitoneally suggests that a disrupted blood–brain barrier (BBB) and, possibly, an inflammatory environment is necessary for AQP4-IgG to exert its pathogenic effects in vivo 131134. As in human lesions, AQP4 preceded astrocyte loss, demyelination and neuronal necrosis in those models 130,133.

  • (p) Strong direct evidence for a pathogenic role of AQP4-IgG comes from the finding that AQP4-antibody belongs mainly to the complement-activating IG1 subclass 11,94,135,136. In line with this finding, the presence of the terminal membrane attack complex, indicating complement activation, at sites of AQP4 loss has been described as a key feature of NMO lesions in humans 12,119,120; anaphylatoxin C5a levels in the cerebrospinal fluid (CSF) of AQP4-antibody-seropositive patients have been found to be elevated 137; and eculizumab, a C5 inhibitor, was recently shown to substantially reduce the relapse rates in patients with NMO 138,139. In several independent in-vitro studies it was shown that sera from NMO-IgG-positive patients, but not from controls, can induce (according to some studies, titre-dependent) death of AQP4-transfected cell lines in the presence of human complement 11,123,136,140,141 (possibly more effectively after transfection with M23-AQP4 than M1-AQP4 142). One of these studies even reported a correlation between the percentage of damaged cells by AQP4-IgG-positive sera and the severity of clinical relapses 140. Similarly, co-administration of (human) complement was necessary to induce lesion pathology in AQP4-IgG-driven animal models of NMO, whereas a C1 complement inhibitor prevented tissue damage 130. As in human lesions, complement deposits have been found within spinal cord lesions in these animal models 130,132,133. This observation is corroborated by ex-vivo and animal models of NMO. Exposure to AQP4-antibody-positive NMO sera or recombinant NMO antibody in the presence of human complement reproduced the loss of AQP4, GFAP and myelin that characterizes human NMO lesions in cultured mouse spinal cord slices or optic nerves 143. Lesions were not seen in spinal cord slices from AQP4 null mice 143. Verkman and colleagues performed a number of sophisticated experiments that provide further strong evidence for an essential role of AQP4-antibody-and complement-dependent cytotoxicity (CDC): a high-affinity monoclonal antibody (termed aquaporumab) from recombinant monoclonal antibodies derived from AQP4-IgG-positive CSF plasmablasts of a patient with NMO and rendered non-pathogenic by introducing IgG1Fc mutations at locations required for the induction of CDC 144, cleavage of IgG from NMO patients by means of an IgG-degrading enzyme of Streptococcus pyogenesto (IdeS) to yield Fc and F(ab')2 fragments 145, selectively deglycosylating the heavy chain of natural AQP4-IgG with bacteria-derived endoglycosidase S to render it non-pathogenic 146, and preincubation with small molecules (identified by automated high-throughput screening) that sterically block interaction between AQP4-antibody and its target antigen 147,148 have all been shown to prevent lesion formation in both slice cultures and mice exposed to AQP4-antibody-positive sera and human complement.

While AQP4-antibody-mediated CDC may play a major role in the pathogenesis of NMO, there is abundant evidence suggesting that additional immunological players are involved:

  • (a) NMO lesions have been shown to contain large numbers of macrophages, eosinophils and neutrophils, which often display signs of degranulation, as well as a few T cells 12,149.

  • (b) Numerous proinflammatory cytokines have been found to be elevated in the serum and CSF of patients with NMO: serum levels of IL-6 are increased significantly and have been implicated in the maintenance of AQP4-antibody-positive plasmablasts in the blood 98,150. IL-6 is also elevated in the CSF 150,151, as are the B cell recruiting and activating factor (BAFF), a proliferation-inducing ligand (APRIL) and C-X-C motif chemokine 13 (CXCL13) 152154, indicating the presence of a B cell-friendly environment in the CNS as well. However, IL-6 also promotes development and maintenance of IL-17-producing T helper type 17 (Th17) cells by inhibiting the conversion of conventional T cells to forkhead box protein 3 (FoxP3)+ T regulatory cells 155,156. Whether Th17 cells contribute to NMO pathogenesis is currently being studied 157161. IL-17 levels are indeed increased in patients with NMO during acute attacks 157,162, and an IL-17 gene polymorphism has been reported recently in Chinese patients with NMO 162. Intrathecal activation of the IL-17/IL-8 axis is thought to promote recruitment of neutrophils 163. IL-4, a major secreted cytokine of eosinophils, is known to cause a shift towards a type 2 helper T cell (Th2) response 164. Both neutrophils and eosinophils have been found in the CSF and are present in NMO lesions 12,165. In spinal cord slice cultures a number of proinflammatory cytokines, including IL-6, were shown to enhance AQP4-antibody-mediated cytotoxicity 143.

  • (c) AQP4-IgG could act on macrophages, neutrophils and eosinophils by binding to Fc receptors. Direct evidence for a role of neutrophils in the pathogenesis of NMO comes from the finding that neutrophil elastase inhibitors can ameliorate lesion formation in mouse models of NMO 166, and from the observation that disease was exacerbated both in a mouse model of NMO and in an NMO patient treated with granulocyte colony-stimulating factor 166,167. In spinal cord slice cultures treated with NMO-IgG and human complement, the severity of lesions is increased markedly by including macrophages, neutrophils or eosinophils (or their granule toxins) and could be ameliorated by antihistamines such as cetirizine and ketotifen, which have eosinophil-stabilizing effects 143,168. Antihistamines were also effective in an animal model of NMO 168, and lesion severity has been shown to be increased in transgenic hypereosinophilic mice. By contrast, reduced lesion severity was observed in mice rendered hypoeosinophilic by anti-IL-5 antibody or by gene deletion 168. Neutrophil and eosinophil infiltration has also been noted in animal models of NMO following peripheral or intracerebral injection of AQP4-IgG and complement 133,168. Complement-dependent attraction has been suggested to be involved in granulocyte trafficking through the BBB 128. Granulocyte trafficking is not inhibited by natalizumab, a drug employed successfully in MS; accordingly, natalizumab does not seem to be effective in NMO 169171.

  • (d) The contribution of natural killer (NK) cells to NMO pathology is controversial. In one study, NMO-IgG binding to human fetal astrocytes was found to result in NK cell degranulation and astrocyte killing by antibody-dependent cellular cytotoxicity (ADCC) 128. NK cell-mediated ADCC following exposure to AQP4-antibodies was also observed in mouse astrocyte and spinal cord slice cultures and in a mouse passive transfer model 143,172. An AQP4-antibody mutant lacking ADCC effector function and Fc fragments generated by IdeS-mediated cleavage of recombinant AQP4-specific IgG significantly reduced tissue damage 145,172. However, the relevance of these findings in human disease is somewhat doubtful, given the rarity of granzyme B+ and perforin+ NK cells and cytotoxic T cells in active human NMO lesions reported in a recent study 149.

  • (e) B cells are surely involved in the pathophysiology of NMO as progenitors of AQP4-antibody-producing plasma cells. In addition, B cells could contribute to the pathogenesis of NMO by producing IL-6 and as antigen-presenting cells for AQP4-specific T cells. BAFF, APRIL and CXCL13 levels are elevated in the CSF and the percentage of BAFF-R-and CXCR5-expressing peripheral B cells is higher in NMO 152154. A recent study reported possibly impaired immunoregulatory B cell properties, as indicated by lowered CD19+CD24highCD38high regulatory B cell levels and reduced B cell expression of regulatory IL-10 154.

  • (f) T cells are also certainly relevant, because T helper cells (including Th17 cells 173) are involved in B cell isotype switching and affinity maturation. Furthermore, CD3+ and CD8+ T cells were detected directly within NMO lesions, albeit at low numbers 12. Moreover, NMO has been reported to be associated with human leucocyte antigen D-related (HLA-DR)B1*03 in Brazilians 174; this allele group has also been associated with other autoimmune disorders, including systemic lupus erythematosus, a disease which frequently co-exists with NMO 6467. Similarly, a French group found NMO-IgG-positive NMO to be associated with a high frequency of HLA-DRB1*01*03 alleles, mainly of the DR3 pattern 175. HLA-DR3 heterodimers enhance T cell stimulation and stabilize T cell/CD4/class II interaction 175. Like DR1, DR3 has been reported to increase the T cell response by enhancing B and T cell co-operation 175. The exact role of T cells in NMO is currently being investigated intensively, and several studies have attempted to identify immunodominant (T cell-activating) determinants of human AQP4 157,159,160,176180. T cells have also been proposed to be involved in molecular mimicry (e.g. involving bacterial aquaporins), which is currently discussed as a potential trigger of NMO attacks 160,181184.

  • (g) In addition, a role for glutamate-mediated excitotoxicity has been discussed. While one study reported that membrane AQP4 in human embryonic kidney (HEK) cells transfected with human AQP4 is endocytosed together with the excitatory amino acid transporter 2 (EAAT2) following incubation with NMO-IgG in the absence of complement, resulting in disruption of glutamate homeostasis, another study could not confirm these findings in cultures of mouse astrocytes 126,129. An increase in extracellular glutamate could result in overstimulation of neurones and oligodendrocytes, and could render the latter more susceptible to Ig-independent complement attack 126,185.

  • (h) Similarly, it is still controversial whether AQP4-antibodies exert some of their pathogenic effects by compromising the water homeostatic function of AQP4 either by blocking the water pore or by endocytosing the protein. According to a time-to-lysis assay using AQP4-transfected Xenopus oocytes, one study reported that NMO-IgG impairs water influx 124; however, other studies which utilized cultured astrocytes 127 or plasma membrane vesicles isolated from AQP4-expressing Chinese hamster ovary (CHO) cells did not find an effect on the water transport capability of AQP4 90,186,187.

  • (i) As mentioned above, IgG deposits in NMO lesions are accompanied by prominent IgM deposits 12. IgM is an even more efficient activator of complement than IgG. Serum AQP4-IgM antibodies can be detected in approximately 10% of NMO patients 188.

Depending on the detection method used, 10–50% of patients with NMO are negative for AQP4-IgG 51. Insufficient assay sensitivity is certainly a common cause of AQP4-IgG seronegativity, as shown in a number of recent comparative studies 9,10,51,189191. Moreover, AQP4-antibody titres have been shown to vary strongly over the course of disease depending, among other factors, on disease activity and treatment status. Retesting in a second, more sensitive assay and at follow-up visits, in particular during acute relapses, is thus advisable in seronegative cases (see reference 51 for a comprehensive overview and comparison of the currently available assays and a discussion of diagnostic pitfalls). However, the fact that approximately 10–20% of patients are seronegative even in the most up-to-date assays, as well as the recent demonstration of significant epidemiological and clinical differences between seropositive and seronegative patients 1,102,189, suggests that NMO might indeed be an aetiologically heterogeneous syndrome, i.e. a common phenotype shared by various autoimmune, (para)infectious 183,192,193 and metabolic diseases affecting the optic nerve and spinal cord. Indirect evidence for a role of so far unknown autoantibodies in seronegative NMO comes from reports that PE also has a therapeutic effect in some seronegative NMO patients 104 and from studies finding that complement-dependent astrocyte cell death induced by serum from AQP4-IgG-seronegative patients with NMO is more pronounced than that induced by serum from patients with MS or healthy donors 141. Recently, antibodies to myelin oligodendrocyte glycoprotein (MOG) have been identified in a subset of patients with seronegative NMOSD 194197; the pathogenic, prognostic and therapeutic relevance of these antibodies is currently being investigated. Moreover, anti-CV2/CRMP5 and, possibly, NMDA receptor autoimmunity have been shown to mimic NMO in single patients 198,199. In addition, connective tissue disorders (CTD), in particular systemic lupus erythematosus and Sjögren's syndrome, have been implicated in the pathogenesis of NMOSD in some patients 64,65,67. A broad summary of the differential diagnosis of NMO is provided in the reference list 200202. It should be kept in mind that a lack of NMO-IgG/AQP4-antibody seropositivity does not rule out a diagnosis of NMO, according to the currently most widely adopted diagnostic criteria 84. As will be discussed in the following sections, CSF analysis and spinal cord and brain imaging can facilitate the differential diagnosis of seronegative NMO and MS.

Paraclinical findings

Cerebrospinal fluid

CSF findings in NMO and MS differ markedly. CSF-restricted oligoclonal bands (OCB), a diagnostic mainstay in MS, are present in only approximately 18% of AQP4-antibody-positive cases and frequently disappear during remission 1,165. Similarly, quantitative evidence for intrathecal IgG synthesis, i.e. an elevated IgG CSF/serum ratio, is only present in approximately 8% of CSF samples and exclusively during relapse 165. By contrast, OCB are present in far more than 90% of cases in classical MS 203,204 and can be detected over the entire course of the disease 205. A positive, polyspecific, intrathecal immune reaction to measles, rubella and varicella zoster virus (also termed MRZ reaction 206208) – as defined by at least two out of three positive antibody indices – is present in 60–80% of MS patients, but absent in approximately 97% of NMO patients 1,209. CSF white cell counts (WCC) are often normal or only mildly elevated in NMO (median 19/μl during acute disease, 3/μl during remission 165). However, cell counts >100/μl are possible 1,165, especially during relapse 165. In addition to lymphocytes and monocytes, cytology often reveals neutrophilic and eosinophilic granulocytes 1,36,165, cell types which are usually absent in MS. An elevated albumin CSF/serum ratio, indicating blood–CSF barrier (BCB) disruption, and an increase in total protein is present in approximately 50% of cases, more often during acute attacks. CSF lactate levels are elevated during acute myelitis in approximately 40%, but normal during remission 165,210. In rare AQP4-antibody-positive NMOSD patients, elevated lactate with marked neutrophilic pleocytosis may be taken falsely as bacterial CNS infection, all the more if the meninges are also involved 55,165,211. A positive correlation was reported between QAlb values, CSF total protein levels and CSF L-lactate levels, on one hand, and the spinal cord lesion load as determined by MRI, on the other hand 165. Importantly, CSF findings in AQP4-antibody-positive NMOSD vary significantly both between relapse and remission and – probably reflecting both differences in lesion volume and the rostrocaudal CSF gradient – between acute myelitis and acute ON 165; in fact, normal CSF findings are not unusual in patients presenting with acute AQP4-antibody-positive ON 165. No significant differences were found between seropositive and seronegative patients with regard to OCB, MRZ reaction and WCC in a recent multicentre study 1.

AQP4-antibodies are produced mainly by plasma cells in the peripheral blood. The trigger underlying AQP4-antibody production is unknown, although molecular mimicry has been suggested 160,181184,212215. By contrast, intrathecal synthesis to an extent detectable by antibody index calculation is very rare 131,136,216,217. AQP4-antibodies may enter the CNS by passive diffusion and, in addition, at sites lacking a proper BBB, such as the area postrema 47, or through a disrupted BBB, caused possibly by acute infections, which were shown to precede NMO attacks in 15–35% of patients 1,36,44,103,218. Notably, AQP4, the target antigen of NMO-IgG, is itself an integral constituent of the BBB.

Magnetic resonance imaging

Spinal MRI is crucial for diagnosis and differential diagnosis. Long cord lesions extending over three or more vertebral segments, often with patchy and inhomogeneous contrast enhancement over weeks or even months or, less frequently, central necrosis and cavitation, are characteristic features and highly suggestive of an NMOSD 1,37,84,219. However, it is important to keep in mind that, depending on the timing of spinal MRI to onset of clinical symptoms, NMOSD patients may well exhibit shorter spinal lesions 1,32 and that other, mostly rare differential diagnoses of long cord lesions need to be considered, including spinal ischaemia, neurosarcoidosis and others 201,202. Despite their often dramatic appearance, cord lesions in NMO may improve substantially upon treatment and even recover fully. Conversely, severe inflammation may cause irreversible cord atrophy, which may be a negative predictive factor for response to PE in case of subsequent attacks 220. Recently, so-called spinal ‘bright spotty lesions’ have been suggested as an additional criterion to distinguish NMOSD from MS 221. Moreover, advanced imaging techniques such as magnetic resonance spectroscopy and diffusion tensor imaging that are not applied regularly in clinical routine have confirmed severe spinal tissue injury and also suggest astrocytic damage that may help to distinguish NMO from MS 222224.

Substantial new insights on brain involvement in NMO result from a multitude of MRI studies performed in the past few years. These studies were encouraged by the seminal work by Pittock and colleagues who showed that, contrary to previous thinking, the majority of NMO patients (up to 60%) exhibit (mostly unspecific) lesions on serial cranial MRI during the course of the disease. Some of these lesions are typical of MS and may even fulfill the so-called ‘Barkhof criteria’ 1,225. Similar findings have been reported by other groups, with approximately 15% of patients fulfilling the Barkhof criteria 1,226. Thus, it is widely accepted nowadays that, although many patients have normal cranial MRI findings at disease onset, brain lesions – including even those resembling typical MS lesions – do not rule out an NMO diagnosis 227. However, ultrahigh-field imaging studies reported that, in contrast to MS, NMO lesions do not typically show central veins and a hypointense rim and lack visible cortical lesions 228,229. This is in line with other imaging and neuropathological reports that indicate the absence of cortical demyelination in NMO 63,230,231. Brain lesions tend to be located at sites of high aquaporin-4 expression, such as the diencephalon, the hypothalamus and the aqueduct 232234, and may also appear large and oedematous in the corpus callosum 235,236. Contrast enhancement on brain MRI with a cloudlike shape and pencil-thin ependymal enhancement were reported to be typical of NMO 237,238. Recent diffusion, perfusion and brain volume studies, including voxel-based morphometry, revealed diffuse and widespread white matter and grey matter alterations in NMO 239243. Thus, brain damage is probably more severe than can be estimated from conventional MR images.

While there is now compelling evidence that AQP4-Ab-positive ‘Asian opticospinal MS’ (OSMS) is identical to Western NMO, a small proportion of Asian patients still cannot be easily classified as NMO or MS, e.g. seronegative patients presenting with LETM and a secondary progressive course or OSMS patients with LETM and peripheral spinal cord lesions 244,245. However, re-evaluation using more up-to-date assays, together with strict MRI criteria distinguishing between confluent (as sometimes seen in MS) and contiguous (as typically seen in NMO) longitudinal lesions, may help to clarify the nosological status of those patients.

Optical coherence tomography and visual-evoked potentials

Optical coherence tomography (OCT) is a non-invasive technique by which unmyelinated retinal CNS axons (the so-called retinal nerve fibre layer RNFL) and their neurons, the retinal ganglion cells, can be visualized. Neuroaxonal retinal damage has been shown widely in MS and ON and is currently under investigation in many other neurological conditions 246254). In NMO, OCT studies have been consistent with the clinical experience of a more severe visual dysfunction and poorer visual outcome than for MS and more profound damage to the RNFL 246,255257. Whereas progressive reduction of the RNFL, independent of clinical attacks of ON, has been reported in MS, retinal axonal loss in NMO is probably associated predominantly with clinical relapses 258261. The utility of OCT for distinguishing NMO from MS and other inflammatory conditions with ocular involvement is currently being investigated. Visual evoked potentials show either reduced amplitudes or prolonged latencies, or both; in more severe cases there may be no response at all 262. Delayed P100 latencies may indicate that the optic nerve is subclinically affected in patients presenting with LETM, but with no history of clinically apparent ON.

Treatment

NMO is still an incurable disease. The goal of treating acute NMO events is to improve relapse symptoms and restore neurological functions; long-term immunosuppression aims to prevent further attacks 4,263,264. Any treatment recommendations are limited by the small size of most studies, which were mostly retrospective case-series. No prospective controlled trials in NMO have been conducted, and most study designs with long placebo treatment would probably be considered unethical. Relapses are treated with high-dose intravenous methylprednisolone; if response is insufficient, patients may benefit from PE 265. If a patient has previously responded well to PE, PE may be considered as initial treatment in case of another relapse. In patients in whom both steroids and PE do not improve symptoms, treatment with intravenous immunoglobulins 266 or an escalation to cytoablative therapy such as cyclophosphamide may be considered 264.

For long-term immunosuppression, patients usually receive either B cell-targeted therapies such as intravenous rituximab or oral azathioprine and/or prednisone 87,110,113,267272. Other possible options include mycophenolate mofetil 273, methotrexate 274 or mitoxantrone which, however, is limited by major side effects such as cardiotoxicity or leukaemia and thus generally not considered as initial treatment 264,275280. It is beyond the scope of this paper to provide details on dosing schemes and monitoring of the various NMO drugs, and therefore we refer the reader to two recent, excellent overviews on treatment recommendations 264,281. However, one aspect deserves mention: less severe lesions have been found in type I interferon (IFN) receptor-deficient mice, suggesting that type I IFNs might be involved in the pathogenesis of NMO. Accordingly, IFN-β, a therapeutic mainstay in MS, has been repeatedly reported to exacerbate disease or to be ineffective in patients with NMO. The use of IFN-β in the treatment of NMO is therefore strongly discouraged. Similarly, lack of efficacy or disease exacerbation has also been reported following treatment with other typical MS drugs such as natalizumab and, in single cases, also fingolimod and alemtuzumab 169171,282290.

Further prospects

A recent, small, open-label study with the monoclonal antibody eculizumab, an inhibitor of the complement component C5 approved for the treatment of paroxysmal nocturnal haemoglobinuria, reported an impressive reduction of relapse rates in 14 NMO patients with disease activity 138,139. This finding has stimulated a larger trial that is expected to begin in late 2013 or early 2014. Given the role of IL-6 in NMO, IL-6-targeted therapy with the monoclonal anti-IL-6-receptor antibody tocilizumab might represent another future treatment strategy, following encouraging case reports 115117. Further preliminary but intriguing experimental approaches are competitive, non-pathogenic, AQP4-specific antibodies, neutrophil elastase inhibitors or antihistamines with eosinophil-stabilizing properties 144,166,168,291.

Acknowledgments

The work of B. W. was supported by a research grant from Merck Serono. The work of S. J. was supported by a research fellowship from the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS).

Dicslosure

B. W. has served on a scientific advisory board for Novartis and Biogen Idec, has received funding for travel and speaker honoraria from Biogen Idec, Bayer Schering Pharma, Merck Serono, Teva Pharmaceutical Industries Ltd and Genzyme-A Sanofi Company and has received research support from Bayer Schering Pharma, Merck Serono, Biotest Pharmaceuticals Corporation, Teva Pharmaceutical Industries Ltd and the Bundesministerium für Bildung und Forschung (BMBF). S. J. has no conflicts of interest. F. P. has received speaker honoraria, travel grants and research grants from Teva, Sanofi/Genzyme, Bayer, Merck-Serono, Biogen Idec and Novartis. He serves on the Novartis advisory board of the OCTIMS study. He is supported by the German ministry of education and research (BMBF/KKNMS, Competence Network Multiple Sclerosis). F. P. is also supported by the German Research Foundation (Exc 257) and has received travel reimbursement from the Guthy Jackson Charitable Foundation.

References

  • 1.Jarius S, Ruprecht K, Wildemann B, et al. Contrasting disease patterns in seropositive and seronegative neuromyelitis optica: a multicentre study of 175 patients. J Neuroinflammation. 2012;9:14. doi: 10.1186/1742-2094-9-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wildemann B, Jarius S, Paul F. [Neuromyelitis optica] Nervenarzt. 2013;84:436–441. doi: 10.1007/s00115-012-3602-x. [DOI] [PubMed] [Google Scholar]
  • 3.Jarius S, Wildemann B. [Neuromyelitis optica] Nervenarzt. 2007;78:1365–1377. doi: 10.1007/s00115-007-2262-8. [DOI] [PubMed] [Google Scholar]
  • 4.Trebst C, Berthele A, Jarius S, et al. [Diagnosis and treatment of neuromyelitis optica. Consensus recommendations of the Neuromyelitis Optica Study Group] Nervenarzt. 2011;82:768–777. doi: 10.1007/s00115-010-3192-4. [DOI] [PubMed] [Google Scholar]
  • 5.Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS. IgG marker of optic–spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med. 2005;202:473–477. doi: 10.1084/jem.20050304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lennon VA, Wingerchuk DM, Kryzer TJ, et al. A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet. 2004;364:2106–2112. doi: 10.1016/S0140-6736(04)17551-X. [DOI] [PubMed] [Google Scholar]
  • 7.Jarius S, Franciotta D, Bergamaschi R, et al. NMO-IgG in the diagnosis of neuromyelitis optica. Neurology. 2007;68:1076–1077. doi: 10.1212/01.wnl.0000256822.01222.bd. [DOI] [PubMed] [Google Scholar]
  • 8.Paul F, Jarius S, Aktas O, et al. Antibody to aquaporin 4 in the diagnosis of neuromyelitis optica. PLOS Med. 2007;4:e133. doi: 10.1371/journal.pmed.0040133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jarius S, Probst C, Borowski K, et al. Standardized method for the detection of antibodies to aquaporin-4 based on a highly sensitive immunofluorescence assay employing recombinant target antigen. J Neurol Sci. 2010;291:52–56. doi: 10.1016/j.jns.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 10.Jarius S, Franciotta D, Paul F, et al. Testing for antibodies to human aquaporin-4 by ELISA: sensitivity, specificity, and direct comparison with immunohistochemistry. J Neurol Sci. 2012;320:32–37. doi: 10.1016/j.jns.2012.06.002. [DOI] [PubMed] [Google Scholar]
  • 11.Waters P, Jarius S, Littleton E, et al. Aquaporin-4 antibodies in neuromyelitis optica and longitudinally extensive transverse myelitis. Arch Neurol. 2008;65:913–919. doi: 10.1001/archneur.65.7.913. [DOI] [PubMed] [Google Scholar]
  • 12.Lucchinetti CF, Mandler RN, McGavern D, et al. A role for humoral mechanisms in the pathogenesis of Devic's neuromyelitis optica. Brain. 2002;125:1450–1461. doi: 10.1093/brain/awf151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wildemann B, Jarius S. The expanding range of autoimmune disorders of the nervous system. Lancet Neurol. 2013;12:22–24. doi: 10.1016/S1474-4422(12)70301-0. [DOI] [PubMed] [Google Scholar]
  • 14.Wildemann B, Bien CG. [Immune-mediated encephalomyelitis] Nervenarzt. 2013;84:435. doi: 10.1007/s00115-012-3601-y. [DOI] [PubMed] [Google Scholar]
  • 15.Wingerchuk DM, Lennon VA, Lucchinetti CF, Pittock SJ, Weinshenker BG. The spectrum of neuromyelitis optica. Lancet Neurol. 2007;6:805–815. doi: 10.1016/S1474-4422(07)70216-8. [DOI] [PubMed] [Google Scholar]
  • 16.Lennon VA. 2006. Changing concepts in CNS demyelinating disorders: autoimmune water channelopathies. In: Program and abstracts of the American Academy of Neurology 58th Annual Meeting; 1–8 April 2006; San Diego, CA, USA. Plenary session.
  • 17.Matsushita T, Isobe N, Matsuoka T, et al. Aquaporin-4 autoimmune syndrome and anti-aquaporin-4 antibody-negative opticospinal multiple sclerosis in Japanese. Mult Scler. 2009;15:834–847. doi: 10.1177/1352458509104595. [DOI] [PubMed] [Google Scholar]
  • 18.Jarius S, Wildemann B. The history of neuromyelitis optica. J Neuroinflammation. 2013;10:8. doi: 10.1186/1742-2094-10-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jarius S, Wildemann B. ‘Noteomielite’ accompanied by acute amaurosis (1844). An early case of neuromyelitis optica. J Neurol Sci. 2012;313:182–184. doi: 10.1016/j.jns.2011.09.025. [DOI] [PubMed] [Google Scholar]
  • 20.Jarius S, Wildemann B. On the contribution of Thomas Clifford Allbutt, F.R.S., to the early history of neuromyelitis optica. J Neurol. 2013;260:100–104. doi: 10.1007/s00415-012-6594-3. [DOI] [PubMed] [Google Scholar]
  • 21.Jarius S, Wildemann B. An early British case of neuromyelitis optica (1850) BMJ. 2012;345:e6430. doi: 10.1136/bmj.e6430. [DOI] [PubMed] [Google Scholar]
  • 22.Jarius S, Wildemann B. An early case of neuromyelitis optica: on a forgotten report by Jacob Lockhart Clarke, FRS. Mult Scler. 2011;17:1384–1386. doi: 10.1177/1352458511411758. [DOI] [PubMed] [Google Scholar]
  • 23.Jarius S, Wildemann B. The case of the Marquis de Causan (1804): an early account of visual loss associated with spinal cord inflammation. J Neurol. 2012;259:1354–1357. doi: 10.1007/s00415-011-6355-8. [DOI] [PubMed] [Google Scholar]
  • 24.Jarius S, Wildemann B. ‘Spinal amaurosis’ (1841). On the early contribution of Edward Hocken to the concept of neuromyelitis optica. J Neurol. 2014;261:400–404. doi: 10.1007/s00415-013-7210-x. [DOI] [PubMed] [Google Scholar]
  • 25.Devic E. 1895. pp. 434–439. [Myélite aiguë dorso-lombaire avec névrite optique – Autopsie.Congrès français de médecine (Premiere Session; Lyon, 1894; Procès-verbaux, mémoires et discussions; Publiés par M le Dr L Bard)]. Lyon: Asselin et Houzeau, Louis Savy,
  • 26.Gault F. 1894. p. 102. [De la neuromyélite optique aiguë. Thése.Faculté de Médecine et de Pharmacie]. Lyon,
  • 27.Asgari N, Khorooshi R, Lillevang ST, Owens T. Complement-dependent pathogenicity of brain-specific antibodies in cerebrospinal fluid. J Neuroimmunol. 2013;254:76–82. doi: 10.1016/j.jneuroim.2012.09.010. [DOI] [PubMed] [Google Scholar]
  • 28.Jacob A, Panicker J, Lythgoe D, et al. The epidemiology of neuromyelitis optica amongst adults in the Merseyside county of United Kingdom. J Neurol. 2013;260:2134–2137. doi: 10.1007/s00415-013-6926-y. [DOI] [PubMed] [Google Scholar]
  • 29.Bizzoco E, Lolli F, Repice AM, et al. Prevalence of neuromyelitis optica spectrum disorder and phenotype distribution. J Neurol. 2009;256:1891–1898. doi: 10.1007/s00415-009-5171-x. [DOI] [PubMed] [Google Scholar]
  • 30.Ketelslegers IA, Catsman-Berrevoets CE, Neuteboom RF, et al. Incidence of acquired demyelinating syndromes of the CNS in Dutch children: a nationwide study. J Neurol. 2012;259:1929–1935. doi: 10.1007/s00415-012-6441-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cossburn M, Tackley G, Baker K, et al. The prevalence of neuromyelitis optica in South East Wales. Eur J Neurol. 2012;19:655–659. doi: 10.1111/j.1468-1331.2011.03529.x. [DOI] [PubMed] [Google Scholar]
  • 32.Mealy MA, Wingerchuk DM, Greenberg BM, Levy M. Epidemiology of neuromyelitis optica in the United States: a multicenter analysis. Arch Neurol. 2012;69:1176–1180. doi: 10.1001/archneurol.2012.314. [DOI] [PubMed] [Google Scholar]
  • 33.Yamakawa K, Kuroda H, Fujihara K, et al. Familial neuromyelitis optica (Devic's syndrome) with late onset in Japan. Neurology. 2000;55:318–320. doi: 10.1212/wnl.55.2.318. [DOI] [PubMed] [Google Scholar]
  • 34.Nouh A, Ali AM, Wichter MD, Messer E. One of the oldest patients with neuromyelitis optica in the literature. Mult Scler. 2013;19:256. doi: 10.1177/1352458512457848. [DOI] [PubMed] [Google Scholar]
  • 35.Huppke P, Bluthner M, Bauer O, et al. Neuromyelitis optica and NMO-IgG in European pediatric patients. Neurology. 2010;75:1740–1744. doi: 10.1212/WNL.0b013e3181fc2823. [DOI] [PubMed] [Google Scholar]
  • 36.Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of neuromyelitis optica (Devic's syndrome) Neurology. 1999;53:1107–1114. doi: 10.1212/wnl.53.5.1107. [DOI] [PubMed] [Google Scholar]
  • 37.Lim BC, Hwang H, Kim KJ, et al. Relapsing demyelinating CNS disease in a Korean pediatric population: multiple sclerosis versus neuromyelitis optica. Mult Scler. 2011;17:67–73. doi: 10.1177/1352458510382685. [DOI] [PubMed] [Google Scholar]
  • 38.Krupp LB, Tardieu M, Amato MP, et al. International Pediatric Multiple Sclerosis Study Group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions. Mult Scler. 2013;19:1261–1267. doi: 10.1177/1352458513484547. [DOI] [PubMed] [Google Scholar]
  • 39.Banwell B, Tenembaum S, Lennon VA, et al. Neuromyelitis optica-IgG in childhood inflammatory demyelinating CNS disorders. Neurology. 2008;70:344–352. doi: 10.1212/01.wnl.0000284600.80782.d5. [DOI] [PubMed] [Google Scholar]
  • 40.Wingerchuk DM. Neuromyelitis optica: effect of gender. J Neurol Sci. 2009;286:18–23. doi: 10.1016/j.jns.2009.08.045. [DOI] [PubMed] [Google Scholar]
  • 41.Matiello M, Kim HJ, Kim W, et al. Familial neuromyelitis optica. Neurology. 2010;75:310–315. doi: 10.1212/WNL.0b013e3181ea9f15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wingerchuk DM, Pittock SJ, Lucchinetti CF, Lennon VA, Weinshenker BG. A secondary progressive clinical course is uncommon in neuromyelitis optica. Neurology. 2007;68:603–605. doi: 10.1212/01.wnl.0000254502.87233.9a. [DOI] [PubMed] [Google Scholar]
  • 43.Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS) Neurology. 1983;33:1444–1452. doi: 10.1212/wnl.33.11.1444. [DOI] [PubMed] [Google Scholar]
  • 44.Ghezzi A, Bergamaschi R, Martinelli V, et al. Clinical characteristics, course and prognosis of relapsing Devic's neuromyelitis optica. J Neurol. 2004;251:47–52. doi: 10.1007/s00415-004-0271-0. [DOI] [PubMed] [Google Scholar]
  • 45.Collongues N, Cabre P, Marignier R, et al. A benign form of neuromyelitis optica: does it exist? Arch Neurol. 2011;68:918–924. doi: 10.1001/archneurol.2011.127. [DOI] [PubMed] [Google Scholar]
  • 46.Iorio R, Lucchinetti CF, Lennon VA, et al. Intractable nausea and vomiting from autoantibodies against a brain water channel. Clin Gastroenterol Hepatol. 2013;11:240–245. doi: 10.1016/j.cgh.2012.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Popescu BF, Lennon VA, Parisi JE, et al. Neuromyelitis optica unique area postrema lesions: nausea, vomiting, and pathogenic implications. Neurology. 2011;76:1229–1237. doi: 10.1212/WNL.0b013e318214332c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kim SM, Kim JS, Heo YE, Yang HR, Park KS. Cortical oscillopsia without nystagmus, an isolated symptom of neuromyelitis optica spectrum disorder with anti-aquaporin 4 antibody. Mult Scler. 2012;18:244–247. doi: 10.1177/1352458511414149. [DOI] [PubMed] [Google Scholar]
  • 49.Kim W, Kim SH, Lee SH, Li XF, Kim HJ. Brain abnormalities as an initial manifestation of neuromyelitis optica spectrum disorder. Mult Scler. 2011;17:1107–1112. doi: 10.1177/1352458511404917. [DOI] [PubMed] [Google Scholar]
  • 50.Iorio R, Lucchinetti CF, Lennon VA, et al. Syndrome of inappropriate antidiuresis may herald or accompany neuromyelitis optica. Neurology. 2011;77:1644–1646. doi: 10.1212/WNL.0b013e3182343377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jarius S, Wildemann B. Aquaporin-4 antibodies (NMO-IgG) as a serological marker of neuromyelitis optica: a critical review of the literature. Brain Pathol. 2013;23:661–683. doi: 10.1111/bpa.12084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nakano H, Tanaka M, Kinoshita M, et al. Epileptic seizures in Japanese patients with multiple sclerosis and neuromyelitis optica. Epilepsy Res. 2012;104:175–180. doi: 10.1016/j.eplepsyres.2012.09.005. [DOI] [PubMed] [Google Scholar]
  • 53.Magana SM, Matiello M, Pittock SJ, et al. Posterior reversible encephalopathy syndrome in neuromyelitis optica spectrum disorders. Neurology. 2009;72:712–717. doi: 10.1212/01.wnl.0000343001.36493.ae. [DOI] [PubMed] [Google Scholar]
  • 54.Takai Y, Misu T, Nakashima I, et al. Two cases of lumbosacral myeloradiculitis with anti-aquaporin-4 antibody. Neurology. 2012;79:1826–1828. doi: 10.1212/WNL.0b013e3182703ff7. [DOI] [PubMed] [Google Scholar]
  • 55.Wang JY, Wang K, Chen XW, et al. Meningoencephalitis as an initial manifestation of neuromyelitis optica spectrum disorder. Mult Scler. 2012;19:639–643. doi: 10.1177/1352458512459785. [DOI] [PubMed] [Google Scholar]
  • 56.Jarius S, Lauda F, Wildemann B, Tumani H. Steroid-responsive hearing impairment in NMO-IgG/aquaporin-4-antibody-positive neuromyelitis optica. J Neurol. 2013;260:663–664. doi: 10.1007/s00415-012-6755-4. [DOI] [PubMed] [Google Scholar]
  • 57.Vernant JC, Cabre P, Smadja D, et al. Recurrent optic neuromyelitis with endocrinopathies: a new syndrome. Neurology. 1997;48:58–64. doi: 10.1212/wnl.48.1.58. [DOI] [PubMed] [Google Scholar]
  • 58.Schmidt F, Göktas Ö, Jarius S, et al. Olfactory dysfunction in patients with neuromyelitis optica. Mult Scler Int. 2013:654501. doi: 10.1155/2013/654501. doi: 10.1155/2013/654501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kanamori Y, Nakashima I, Takai Y, et al. Pain in neuromyelitis optica and its effect on quality of life: a cross-sectional study. Neurology. 2011;77:652–658. doi: 10.1212/WNL.0b013e318229e694. [DOI] [PubMed] [Google Scholar]
  • 60.Qian P, Lancia S, Alvarez E, Klawiter EC, Cross AH, Naismith RT. Association of neuromyelitis optica with severe and intractable pain. Arch Neurol. 2012;69:1482–1487. doi: 10.1001/archneurol.2012.768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Blanc F, Zephir H, Lebrun C, et al. Cognitive functions in neuromyelitis optica. Arch Neurol. 2008;65:84–88. doi: 10.1001/archneurol.2007.16. [DOI] [PubMed] [Google Scholar]
  • 62.Blanc F, Noblet V, Jung B, et al. White matter atrophy and cognitive dysfunctions in neuromyelitis optica. PLOS ONE. 2012;7:e33878. doi: 10.1371/journal.pone.0033878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Saji E, Arakawa M, Yanagawa K, et al. Cognitive impairment and cortical degeneration in neuromyelitis optica. Ann Neurol. 2013;73:65–76. doi: 10.1002/ana.23721. [DOI] [PubMed] [Google Scholar]
  • 64.Jarius S, Jacobi C, Seze de J, et al. Frequency and syndrome specificity of antibodies to aquaporin-4 in neurological patients with rheumatic disorders. Mult Scler. 2011;17:1067–1073. doi: 10.1177/1352458511403958. [DOI] [PubMed] [Google Scholar]
  • 65.Wandinger KP, Stangel M, Witte T, et al. Autoantibodies against aquaporin-4 in patients with neuropsychiatric systemic lupus erythematosus and primary Sjogren's syndrome. Arthritis Rheum. 2010;62:1198–1200. doi: 10.1002/art.27337. [DOI] [PubMed] [Google Scholar]
  • 66.Zavada J, Nytrova P, Wandinger KP, et al. Seroprevalence and specificity of NMO-IgG (anti-aquaporin 4 antibodies) in patients with neuropsychiatric systemic lupus erythematosus. Rheumatol Int. 2013;33:259–263. doi: 10.1007/s00296-011-2176-4. [DOI] [PubMed] [Google Scholar]
  • 67.Pittock SJ, Lennon VA, Seze de J, et al. Neuromyelitis optica and non organ-specific autoimmunity. Arch Neurol. 2008;65:78–83. doi: 10.1001/archneurol.2007.17. [DOI] [PubMed] [Google Scholar]
  • 68.Jarius S, Paul F, Franciotta D, et al. Neuromyelitis optica spectrum disorders in patients with myasthenia gravis: ten new aquaporin-4 antibody positive cases and a review of the literature. Mult Scler. 2012;18:1135–1143. doi: 10.1177/1352458511431728. [DOI] [PubMed] [Google Scholar]
  • 69.Jarius S, Paul F, Martins da Silva A, et al. Neuromyelitis optica and longitudinally extensive transverse myelitis following thymectomy for myasthenia gravis. Mult Scler. 2007;13:P534. [Google Scholar]
  • 70.Kay CS, Scola RH, Lorenzoni PJ, Jarius S, Arruda WO, Werneck LC. NMO-IgG positive neuromyelitis optica in a patient with myasthenia gravis but no thymectomy. J Neurol Sci. 2008;275:148–150. doi: 10.1016/j.jns.2008.06.038. [DOI] [PubMed] [Google Scholar]
  • 71.Leite MI, Coutinho E, Lana-Peixoto M, et al. Myasthenia gravis and neuromyelitis optica spectrum disorder: a multicenter study of 16 patients. Neurology. 2012;78:1601–1607. doi: 10.1212/WNL.0b013e31825644ff. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bergamaschi R, Jarius S, Robotti M, Pichiecchio A, Wildemann B, Meola G. Two cases of benign neuromyelitis optica in patients with celiac disease. J Neurol. 2009;256:2097–2099. doi: 10.1007/s00415-009-5288-y. [DOI] [PubMed] [Google Scholar]
  • 73.Jarius S, Jacob S, Waters P, Jacob A, Littleton E, Vincent A. Neuromyelitis optica in patients with gluten sensitivity associated with antibodies to aquaporin-4. J Neurol Neurosurg Psychiatry. 2008;79:1084. doi: 10.1136/jnnp.2007.143263. [DOI] [PubMed] [Google Scholar]
  • 74.Jarius S, Paul F, Ruprecht K, Wildemann B. Low vitamin B12 levels and gastric parietal cell antibodies in patients with aquaporin-4 antibody-positive neuromyelitis optica spectrum disorders. J Neurol. 2012;259:2743–2745. doi: 10.1007/s00415-012-6677-1. [DOI] [PubMed] [Google Scholar]
  • 75.Fragoso YD, Adoni T, Bichuetti DB, et al. Neuromyelitis optica and pregnancy. J Neurol. 2013;260:2614–2619. doi: 10.1007/s00415-013-7031-y. [DOI] [PubMed] [Google Scholar]
  • 76.Kim W, Kim SH, Nakashima I, et al. Influence of pregnancy on neuromyelitis optica spectrum disorder. Neurology. 2012;78:1264–1267. doi: 10.1212/WNL.0b013e318250d812. [DOI] [PubMed] [Google Scholar]
  • 77.Saadoun S, Waters P, Leite MI, Bennett JL, Vincent A, Papadopoulos MC. Neuromyelitis optica IgG causes placental inflammation and fetal death. J Immunol. 2013;191:2999–3005. doi: 10.4049/jimmunol.1301483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Reuss R, Rommer PS, Bruck W, et al. A woman with acute myelopathy in pregnancy: case outcome. BMJ. 2009;339:b4026. doi: 10.1136/bmj.b4026. [DOI] [PubMed] [Google Scholar]
  • 79.Reuss R, Rommer PS, Brueck W, Jarius S, Bolz M, Zettl UK. Anti-AQP4 Ab might be relevant in pregnancy. BMJ. Feb 8; online reply. [Google Scholar]
  • 80.Deguchi S, Deguchi K, Sato K, et al. HyperCKemia related to the initial and recurrent attacks of neuromyelitis optica. Intern Med. 2012;51:2617–2620. doi: 10.2169/internalmedicine.51.7898. [DOI] [PubMed] [Google Scholar]
  • 81.Di Filippo M, Franciotta D, Massa R, et al. Recurrent hyperCKemia with normal muscle biopsy in a pediatric patient with neuromyelitis optica. Neurology. 2012;79:1182–1184. doi: 10.1212/WNL.0b013e3182698d39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Suzuki N, Takahashi T, Aoki M, et al. Neuromyelitis optica preceded by hyperCKemia episode. Neurology. 2010;74:1543–1545. doi: 10.1212/WNL.0b013e3181dd445b. [DOI] [PubMed] [Google Scholar]
  • 83.Jeret JS, Suzuki N, Takahashi T, Fujihara K. Neuromyelitis optica preceded by hyperCKemia episode. Neurology. 2010;75:2253. doi: 10.1212/WNL.0b013e3181fac76b. author reply 4. [DOI] [PubMed] [Google Scholar]
  • 84.Wingerchuk DM, Lennon VA, Pittock SJ, Lucchinetti CF, Weinshenker BG. Revised diagnostic criteria for neuromyelitis optica. Neurology. 2006;66:1485–1489. doi: 10.1212/01.wnl.0000216139.44259.74. [DOI] [PubMed] [Google Scholar]
  • 85.Jarius S, Paul F, Franciotta D, et al. Revised diagnostic criteria for neuromyelitis optica – incorporation of NMO-IgG status. Nat Clin Pract Neurol. 2007;3:E1. doi: 10.1038/ncpneuro0501. [DOI] [PubMed] [Google Scholar]
  • 86.Paty DW, Oger JJ, Kastrukoff LF, et al. MRI in the diagnosis of MS: a prospective study with comparison of clinical evaluation, evoked potentials, oligoclonal banding, and CT. Neurology. 1988;38:180–185. doi: 10.1212/wnl.38.2.180. [DOI] [PubMed] [Google Scholar]
  • 87.Jarius S, Aboul-Enein F, Waters P, et al. Antibody to aquaporin-4 in the long-term course of neuromyelitis optica. Brain. 2008;131:3072–3080. doi: 10.1093/brain/awn240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jarius S, Wildemann B. AQP4 antibodies in neuromyelitis optica: diagnostic and pathogenetic relevance. Nat Rev Neurol. 2010;6:383–392. doi: 10.1038/nrneurol.2010.72. [DOI] [PubMed] [Google Scholar]
  • 89.Papadopoulos MC, Verkman AS. Aquaporin 4 and neuromyelitis optica. Lancet Neurol. 2012;11:535–544. doi: 10.1016/S1474-4422(12)70133-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ratelade J, Verkman AS. Neuromyelitis optica: aquaporin-4 based pathogenesis mechanisms and new therapies. Int J Biochem Cell Biol. 2012;44:1519–1530. doi: 10.1016/j.biocel.2012.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dellavance A, Alvarenga RR, Rodrigues SH, Kok F, Souza de AW, Andrade LE. Anti-aquaporin-4 antibodies in the context of assorted immune-mediated diseases. Eur J Neurol. 2012;19:248–252. doi: 10.1111/j.1468-1331.2011.03479.x. [DOI] [PubMed] [Google Scholar]
  • 92.Mader S, Lutterotti A, Di Pauli F, et al. Patterns of antibody binding to aquaporin-4 isoforms in neuromyelitis optica. PLOS ONE. 2010;5:e10455. doi: 10.1371/journal.pone.0010455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.McKeon A, Fryer JP, Apiwattanakul M, et al. Diagnosis of neuromyelitis spectrum disorders: comparative sensitivities and specificities of immunohistochemical and immunoprecipitation assays. Arch Neurol. 2009;66:1134–1138. doi: 10.1001/archneurol.2009.178. [DOI] [PubMed] [Google Scholar]
  • 94.Isobe N, Yonekawa T, Matsushita T, et al. Quantitative assays for anti-aquaporin-4 antibody with subclass analysis in neuromyelitis optica. Mult Scler. 2012;18:1541–1551. doi: 10.1177/1352458512443917. [DOI] [PubMed] [Google Scholar]
  • 95.Jarius S, Paul F, Franciotta D, et al. Mechanisms of disease: aquaporin-4 antibodies in neuromyelitis optica. Nat Clin Pract Neurol. 2008;4:202–214. doi: 10.1038/ncpneuro0764. [DOI] [PubMed] [Google Scholar]
  • 96.Kim W, Lee JE, Li XF, et al. Quantitative measurement of anti-aquaporin-4 antibodies by enzyme-linked immunosorbent assay using purified recombinant human aquaporin-4. Mult Scler. 2012;18:578–586. doi: 10.1177/1352458511424590. [DOI] [PubMed] [Google Scholar]
  • 97.Takahashi T, Fujihara K, Nakashima I, et al. Anti-aquaporin-4 antibody is involved in the pathogenesis of NMO: a study on antibody titre. Brain. 2007;130:1235–1243. doi: 10.1093/brain/awm062. [DOI] [PubMed] [Google Scholar]
  • 98.Chihara N, Aranami T, Sato W, et al. Interleukin 6 signaling promotes anti-aquaporin 4 autoantibody production from plasmablasts in neuromyelitis optica. Proc Natl Acad Sci USA. 2011;108:3701–3706. doi: 10.1073/pnas.1017385108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jarius S, Frederikson J, Waters P, et al. Frequency and prognostic impact of antibodies to aquaporin-4 in patients with optic neuritis. J Neurol Sci. 2010;298:158–162. doi: 10.1016/j.jns.2010.07.011. [DOI] [PubMed] [Google Scholar]
  • 100.Weinshenker BG, Wingerchuk DM, Vukusic S, et al. Neuromyelitis optica IgG predicts relapse after longitudinally extensive transverse myelitis. Ann Neurol. 2006;59:566–569. doi: 10.1002/ana.20770. [DOI] [PubMed] [Google Scholar]
  • 101.Matiello M, Lennon VA, Jacob A, et al. NMO-IgG predicts the outcome of recurrent optic neuritis. Neurology. 2008;70:2197–2200. doi: 10.1212/01.wnl.0000303817.82134.da. [DOI] [PubMed] [Google Scholar]
  • 102.Akman-Demir G, Tuzun E, Waters P, et al. Prognostic implications of aquaporin-4 antibody status in neuromyelitis optica patients. J Neurol. 2011;258:464–470. doi: 10.1007/s00415-010-5780-4. [DOI] [PubMed] [Google Scholar]
  • 103.Kitley J, Leite MI, Nakashima I, et al. Prognostic factors and disease course in aquaporin-4 antibody-positive patients with neuromyelitis optica spectrum disorder from the United Kingdom and Japan. Brain. 2012;135:1834–1849. doi: 10.1093/brain/aws109. [DOI] [PubMed] [Google Scholar]
  • 104.Bonnan M, Valentino R, Olindo S, Mehdaoui H, Smadja D, Cabre P. Plasma exchange in severe spinal attacks associated with neuromyelitis optica spectrum disorder. Mult Scler. 2009;15:487–492. doi: 10.1177/1352458508100837. [DOI] [PubMed] [Google Scholar]
  • 105.Khatri BO, Kramer J, Dukic M, Palencia M, Verre W. Maintenance plasma exchange therapy for steroid-refractory neuromyelitis optica. J Clin Apher. 2012;27:183–192. doi: 10.1002/jca.21215. [DOI] [PubMed] [Google Scholar]
  • 106.Kim SH, Kim W, Huh SY, Lee KY, Jung IJ, Kim HJ. Clinical efficacy of plasmapheresis in patients with neuromyelitis optica spectrum disorder and effects on circulating anti-aquaporin-4 antibody levels. J Clin Neurol. 2013;9:36–42. doi: 10.3988/jcn.2013.9.1.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Merle H, Olindo S, Jeannin S, et al. Treatment of optic neuritis by plasma exchange (add-on) in neuromyelitis optica. Arch Ophthalmol. 2012;130:858–862. doi: 10.1001/archophthalmol.2012.1126. [DOI] [PubMed] [Google Scholar]
  • 108.Watanabe S, Nakashima I, Misu T, et al. Therapeutic efficacy of plasma exchange in NMO-IgG-positive patients with neuromyelitis optica. Mult Scler. 2007;13:128–132. doi: 10.1177/1352458506071174. [DOI] [PubMed] [Google Scholar]
  • 109.Watanabe S, Nakashima I, Miyazawa I, et al. Successful treatment of a hypothalamic lesion in neuromyelitis optica by plasma exchange. J Neurol. 2007;254:670–671. doi: 10.1007/s00415-006-0329-2. [DOI] [PubMed] [Google Scholar]
  • 110.Bedi GS, Brown AD, Delgado SR, Usmani N, Lam BL, Sheremata WA. Impact of rituximab on relapse rate and disability in neuromyelitis optica. Mult Scler. 2011;17:1225–1230. doi: 10.1177/1352458511404586. [DOI] [PubMed] [Google Scholar]
  • 111.Cree BA, Lamb S, Morgan K, Chen A, Waubant E, Genain C. An open label study of the effects of rituximab in neuromyelitis optica. Neurology. 2005;64:1270–1272. doi: 10.1212/01.WNL.0000159399.81861.D5. [DOI] [PubMed] [Google Scholar]
  • 112.Jacob A, Weinshenker BG, Violich I, et al. Treatment of neuromyelitis optica with rituximab: retrospective analysis of 25 patients. Arch Neurol. 2008;65:1443–1448. doi: 10.1001/archneur.65.11.noc80069. [DOI] [PubMed] [Google Scholar]
  • 113.Kim SH, Kim W, Li XF, Jung IJ, Kim HJ. Repeated treatment with rituximab based on the assessment of peripheral circulating memory B cells in patients with relapsing neuromyelitis optica over 2 years. Arch Neurol. 2011;68:1412–1420. doi: 10.1001/archneurol.2011.154. [DOI] [PubMed] [Google Scholar]
  • 114.Pellkofer HL, Krumbholz M, Berthele A, et al. Long-term follow-up of patients with neuromyelitis optica after repeated therapy with rituximab. Neurology. 2011;76:1310–1315. doi: 10.1212/WNL.0b013e3182152881. [DOI] [PubMed] [Google Scholar]
  • 115.Araki M, Aranami T, Matsuoka T, Nakamura M, Miyake S, Yamamura T. Clinical improvement in a patient with neuromyelitis optica following therapy with the anti-IL-6 receptor monoclonal antibody tocilizumab. Mod Rheumatol. 2012;23:827–851. doi: 10.1007/s10165-012-0715-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ayzenberg I, Kleiter I, Schroder A, et al. Interleukin 6 receptor blockade in patients with neuromyelitis optica nonresponsive to anti-CD20 therapy. JAMA Neurol. 2013;70:394–397. doi: 10.1001/jamaneurol.2013.1246. [DOI] [PubMed] [Google Scholar]
  • 117.Kieseier BC, Stuve O, Dehmel T, et al. Disease amelioration with tocilizumab in a treatment-resistant patient with neuromyelitis optica: implication for cellular immune responses. Arch Neurol. 2013;70:390–393. doi: 10.1001/jamaneurol.2013.668. [DOI] [PubMed] [Google Scholar]
  • 118.Matiello M, Schaefer-Klein J, Sun D, Weinshenker BG. Aquaporin 4 expression and tissue susceptibility to neuromyelitis optica. JAMA Neurol. 2013;70:1118–1125. doi: 10.1001/jamaneurol.2013.3124. [DOI] [PubMed] [Google Scholar]
  • 119.Misu T, Fujihara K, Kakita A, et al. Loss of aquaporin 4 in lesions of neuromyelitis optica: distinction from multiple sclerosis. Brain. 2007;130:1224–1234. doi: 10.1093/brain/awm047. [DOI] [PubMed] [Google Scholar]
  • 120.Roemer SF, Parisi JE, Lennon VA, et al. Pattern-specific loss of aquaporin-4 immunoreactivity distinguishes neuromyelitis optica from multiple sclerosis. Brain. 2007;130:1194–1205. doi: 10.1093/brain/awl371. [DOI] [PubMed] [Google Scholar]
  • 121.Sinclair C, Kirk J, Herron B, Fitzgerald U, McQuaid S. Absence of aquaporin-4 expression in lesions of neuromyelitis optica but increased expression in multiple sclerosis lesions and normal-appearing white matter. Acta Neuropathol (Berl) 2007;113:187–194. doi: 10.1007/s00401-006-0169-2. [DOI] [PubMed] [Google Scholar]
  • 122.Misu T, Hoftberger R, Fujihara K, et al. Presence of six different lesion types suggests diverse mechanisms of tissue injury in neuromyelitis optica. Acta Neuropathol (Berl) 2013;125:815–827. doi: 10.1007/s00401-013-1116-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hinson SR, Pittock SJ, Lucchinetti CF, et al. Pathogenic potential of IgG binding to water channel extracellular domain in neuromyelitis optica. Neurology. 2007;69:2221–2231. doi: 10.1212/01.WNL.0000289761.64862.ce. [DOI] [PubMed] [Google Scholar]
  • 124.Hinson SR, Romero MF, Popescu BF, et al. Molecular outcomes of neuromyelitis optica (NMO)-IgG binding to aquaporin-4 in astrocytes. Proc Natl Acad Sci USA. 2012;109:1245–1250. doi: 10.1073/pnas.1109980108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Rossi A, Ratelade J, Papadopoulos MC, Bennett JL, Verkman AS. Neuromyelitis optica IgG does not alter aquaporin-4 water permeability, plasma membrane M1/M23 isoform content, or supramolecular assembly. Glia. 2012;60:2027–2039. doi: 10.1002/glia.22417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hinson SR, Roemer SF, Lucchinetti CF, et al. Aquaporin-4-binding autoantibodies in patients with neuromyelitis optica impair glutamate transport by down-regulating EAAT2. J Exp Med. 2008;205:2473–2481. doi: 10.1084/jem.20081241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Melamud L, Fernandez JM, Rivarola V, et al. Neuromyelitis optica immunoglobulin G present in sera from neuromyelitis optica patients affects aquaporin-4 expression and water permeability of the astrocyte plasma membrane. J Neurosci Res. 2012;90:1240–1248. doi: 10.1002/jnr.22822. [DOI] [PubMed] [Google Scholar]
  • 128.Vincent T, Saikali P, Cayrol R, et al. Functional consequences of neuromyelitis optica-IgG astrocyte interactions on blood–brain barrier permeability and granulocyte recruitment. J Immunol. 2008;181:5730–5737. doi: 10.4049/jimmunol.181.8.5730. [DOI] [PubMed] [Google Scholar]
  • 129.Ratelade J, Bennett JL, Verkman AS. Evidence against cellular internalization in vivo of NMO-IgG, aquaporin-4, and excitatory amino acid transporter 2 in neuromyelitis optica. J Biol Chem. 2011;286:45156–45164. doi: 10.1074/jbc.M111.297275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Saadoun S, Waters P, Bell BA, Vincent A, Verkman AS, Papadopoulos MC. Intra-cerebral injection of neuromyelitis optica immunoglobulin G and human complement produces neuromyelitis optica lesions in mice. Brain. 2010;133:349–361. doi: 10.1093/brain/awp309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bennett JL, Lam C, Kalluri SR, et al. Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica. Ann Neurol. 2009;66:617–629. doi: 10.1002/ana.21802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Bradl M, Misu T, Takahashi T, et al. Neuromyelitis optica: pathogenicity of patient immunoglobulin in vivo. Ann Neurol. 2009;66:630–643. doi: 10.1002/ana.21837. [DOI] [PubMed] [Google Scholar]
  • 133.Kinoshita M, Nakatsuji Y, Kimura T, et al. Neuromyelitis optica: passive transfer to rats by human immunoglobulin. Biochem Biophys Res Commun. 2009;386:623–627. doi: 10.1016/j.bbrc.2009.06.085. [DOI] [PubMed] [Google Scholar]
  • 134.Kinoshita M, Nakatsuji Y, Kimura T, et al. Anti-aquaporin-4 antibody induces astrocytic cytotoxicity in the absence of CNS antigen-specific T cells. Biochem Biophys Res Commun. 2010;394:205–210. doi: 10.1016/j.bbrc.2010.02.157. [DOI] [PubMed] [Google Scholar]
  • 135.Jarius S, Jacob S, Leite MI, Waters P, Vincent A. NMO-IgG/Aqp4-Ab belongs to the IgG1 subclass and activates complement in vitro. Mult Scler. 2007;13:P533. [Google Scholar]
  • 136.Kalluri SR, Illes Z, Srivastava R, et al. Quantification and functional characterization of antibodies to native aquaporin 4 in neuromyelitis optica. Arch Neurol. 2010;67:1201–1208. doi: 10.1001/archneurol.2010.269. [DOI] [PubMed] [Google Scholar]
  • 137.Kuroda H, Fujihara K, Takano R, et al. Increase of complement fragment C5a in cerebrospinal fluid during exacerbation of neuromyelitis optica. J Neuroimmunol. 2013;254:178–182. doi: 10.1016/j.jneuroim.2012.09.002. [DOI] [PubMed] [Google Scholar]
  • 138.Pittock SJ, Lennon VA, McKeon A, et al. Eculizumab in AQP4-IgG-positive relapsing neuromyelitis optica spectrum disorders: an open-label pilot study. Lancet Neurol. 2013;12:554–562. doi: 10.1016/S1474-4422(13)70076-0. [DOI] [PubMed] [Google Scholar]
  • 139.Paul F. Hope for a rare disease: eculizumab in neuromyelitis optica. Lancet Neurol. 2013;12:529–531. doi: 10.1016/S1474-4422(13)70089-9. [DOI] [PubMed] [Google Scholar]
  • 140.Hinson SR, McKeon A, Fryer JP, Apiwattanakul M, Lennon VA, Pittock SJ. Prediction of neuromyelitis optica attack severity by quantitation of complement-mediated injury to aquaporin-4-expressing cells. Arch Neurol. 2009;66:1164–1167. doi: 10.1001/archneurol.2009.188. [DOI] [PubMed] [Google Scholar]
  • 141.Sabater L, Giralt A, Boronat A, et al. Cytotoxic effect of neuromyelitis optica antibody (NMO-IgG) to astrocytes: an in vitro study. J Neuroimmunol. 2009;215:31–35. doi: 10.1016/j.jneuroim.2009.07.014. [DOI] [PubMed] [Google Scholar]
  • 142.Phuan PW, Ratelade J, Rossi A, Tradtrantip L, Verkman AS. Complement-dependent cytotoxicity in neuromyelitis optica requires aquaporin-4 protein assembly in orthogonal arrays. J Biol Chem. 2012;287:13829–13839. doi: 10.1074/jbc.M112.344325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhang H, Bennett JL, Verkman AS. Ex vivo spinal cord slice model of neuromyelitis optica reveals novel immunopathogenic mechanisms. Ann Neurol. 2011;70:943–954. doi: 10.1002/ana.22551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tradtrantip L, Zhang H, Saadoun S, et al. Anti-aquaporin-4 monoclonal antibody blocker therapy for neuromyelitis optica. Ann Neurol. 2012;71:314–322. doi: 10.1002/ana.22657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tradtrantip L, Asavapanumas N, Verkman AS. Therapeutic cleavage of anti-aquaporin-4 autoantibody in neuromyelitis optica by an IgG-selective proteinase. Mol Pharmacol. 2013;83:1268–1275. doi: 10.1124/mol.113.086470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Tradtrantip L, Ratelade J, Zhang H, Verkman AS. Enzymatic deglycosylation converts pathogenic neuromyelitis optica anti-aquaporin-4 immunoglobulin G into therapeutic antibody. Ann Neurol. 2013;73:77–85. doi: 10.1002/ana.23741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Phuan PW, Anderson MO, Tradtrantip L, et al. A small-molecule screen yields idiotype-specific blockers of neuromyelitis optica-immunoglobulin G binding to aquaporin-4. J Biol Chem. 2012;287:36837–36844. doi: 10.1074/jbc.M112.408716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Tradtrantip L, Zhang H, Anderson MO, et al. Small-molecule inhibitors of NMO-IgG binding to aquaporin-4 reduce astrocyte cytotoxicity in neuromyelitis optica. FASEB J. 2012;26:2197–2208. doi: 10.1096/fj.11-201608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Saadoun S, Bridges LR, Verkman AS, Papadopoulos MC. Paucity of natural killer and cytotoxic T cells in human neuromyelitis optica lesions. Neuroreport. 2012;23:1044–1047. doi: 10.1097/WNR.0b013e32835ab480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Icoz S, Tuzun E, Kurtuncu M, et al. Enhanced IL-6 production in aquaporin-4 antibody positive neuromyelitis optica patients. Int J Neurosci. 2010;120:71–75. doi: 10.3109/00207450903428970. [DOI] [PubMed] [Google Scholar]
  • 151.Uzawa A, Mori M, Ito M, et al. Markedly increased CSF interleukin-6 levels in neuromyelitis optica, but not in multiple sclerosis. J Neurol. 2009;256:2082–2084. doi: 10.1007/s00415-009-5274-4. [DOI] [PubMed] [Google Scholar]
  • 152.Wang H, Wang K, Zhong X, et al. Cerebrospinal fluid BAFF and APRIL levels in neuromyelitis optica and multiple sclerosis patients during relapse. J Clin Immunol. 2012;32:1007–1011. doi: 10.1007/s10875-012-9709-9. [DOI] [PubMed] [Google Scholar]
  • 153.Vaknin-Dembinsky A, Brill L, Orpaz N, Abramsky O, Karussis D. Preferential increase of B-cell activating factor in the cerebrospinal fluid of neuromyelitis optica in a white population. Mult Scler. 2010;16:1453–1457. doi: 10.1177/1352458510380416. [DOI] [PubMed] [Google Scholar]
  • 154.Quan C, Yu H, Qiao J, et al. Impaired regulatory function and enhanced intrathecal activation of B cells in neuromyelitis optica: distinct from multiple sclerosis. Mult Scler. 2013;19:289–298. doi: 10.1177/1352458512454771. [DOI] [PubMed] [Google Scholar]
  • 155.Korn T, Mitsdoerffer M, Croxford AL, et al. IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells. Proc Natl Acad Sci USA. 2008;105:18460–18465. doi: 10.1073/pnas.0809850105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Goodman WA, Levine AD, Massari JV, Sugiyama H, McCormick TS, Cooper KD. IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J Immunol. 2009;183:3170–3176. doi: 10.4049/jimmunol.0803721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wang HH, Dai YQ, Qiu W, et al. Interleukin-17-secreting T cells in neuromyelitis optica and multiple sclerosis during relapse. J Clin Neurosci. 2011;18:1313–1317. doi: 10.1016/j.jocn.2011.01.031. [DOI] [PubMed] [Google Scholar]
  • 158.Linhares UC, Schiavoni PB, Barros PO, et al. The ex vivo production of IL-6 and IL-21 by CD4+ T cells is directly associated with neurological disability in neuromyelitis optica patients. J Clin Immunol. 2013;33:179–189. doi: 10.1007/s10875-012-9780-2. [DOI] [PubMed] [Google Scholar]
  • 159.Arellano B, Hussain R, Zacharias T, et al. Human aquaporin 4281-300 is the immunodominant linear determinant in the context of HLA-DRB1*03:01: relevance for diagnosing and monitoring patients with Neuromyelitis optica. Arch Neurol. 2012;69:1125–1131. doi: 10.1001/archneurol.2012.1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Varrin-Doyer M, Spencer CM, Schulze-Topphoff U, et al. Aquaporin 4-specific T cells in neuromyelitis optica exhibit a Th17 bias and recognize Clostridium ABC transporter. Ann Neurol. 2012;72:53–64. doi: 10.1002/ana.23651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Quan C, Yu H, Qiao J, et al. Impaired regulatory function and enhanced intrathecal activation of B cells in neuromyelitis optica: distinct from multiple sclerosis. Mult Scler. 2012;19:289–298. doi: 10.1177/1352458512454771. [DOI] [PubMed] [Google Scholar]
  • 162.Wang H, Zhong X, Wang K, et al. Interleukin 17 gene polymorphism is associated with anti-aquaporin 4 antibody-positive neuromyelitis optica in the Southern Han Chinese – a case control study. J Neurol Sci. 2012;314:26–28. doi: 10.1016/j.jns.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • 163.Ishizu T, Osoegawa M, Mei FJ, et al. Intrathecal activation of the IL-17/IL-8 axis in opticospinal multiple sclerosis. Brain. 2005;128:988–1002. doi: 10.1093/brain/awh453. [DOI] [PubMed] [Google Scholar]
  • 164.Bjerke T, Gaustadnes M, Nielsen S, et al. Human blood eosinophils produce and secrete interleukin 4. Respir Med. 1996;90:271–277. doi: 10.1016/s0954-6111(96)90098-0. [DOI] [PubMed] [Google Scholar]
  • 165.Jarius S, Paul F, Franciotta D, et al. Cerebrospinal fluid findings in aquaporin-4 antibody positive neuromyelitis optica: Results from 211 lumbar punctures. J Neurol Sci. 2011;306:82–90. doi: 10.1016/j.jns.2011.03.038. [DOI] [PubMed] [Google Scholar]
  • 166.Saadoun S, Waters P, MacDonald C, et al. Neutrophil protease inhibition reduces neuromyelitis optica-immunoglobulin G-induced damage in mouse brain. Ann Neurol. 2012;71:323–333. doi: 10.1002/ana.22686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Jacob A, Saadoun S, Kitley J, et al. Detrimental role of granulocyte-colony stimulating factor in neuromyelitis optica: clinical case and histological evidence. Mult Scler. 2012;18:1801–1803. doi: 10.1177/1352458512443994. [DOI] [PubMed] [Google Scholar]
  • 168.Zhang H, Verkman AS. Eosinophil pathogenicity mechanisms and therapeutics in neuromyelitis optica. J Clin Invest. 2013;123:2306–2316. doi: 10.1172/JCI67554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Barnett MH, Prineas JW, Buckland ME, Parratt JD, Pollard JD. Massive astrocyte destruction in neuromyelitis optica despite natalizumab therapy. Mult Scler. 2012;18:108–112. doi: 10.1177/1352458511421185. [DOI] [PubMed] [Google Scholar]
  • 170.Kleiter I, Hellwig K, Berthele A, et al. Failure of natalizumab to prevent relapses in neuromyelitis optica. Arch Neurol. 2012;69:239–245. doi: 10.1001/archneurol.2011.216. [DOI] [PubMed] [Google Scholar]
  • 171.Jacob A, Hutchinson M, Elsone L, et al. Does natalizumab therapy worsen neuromyelitis optica? Neurology. 2012;79:1065–1066. doi: 10.1212/WNL.0b013e31826845fe. [DOI] [PubMed] [Google Scholar]
  • 172.Ratelade J, Zhang H, Saadoun S, Bennett JL, Papadopoulos MC, Verkman AS. Neuromyelitis optica IgG and natural killer cells produce NMO lesions in mice without myelin loss. Acta Neuropathol (Berl) 2012;123:861–872. doi: 10.1007/s00401-012-0986-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Mitsdoerffer M, Lee Y, Jager A, et al. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci USA. 2010;107:14292–14297. doi: 10.1073/pnas.1009234107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Brum DG, Barreira AA, Santos dos AC, et al. HLA-DRB association in neuromyelitis optica is different from that observed in multiple sclerosis. Mult Scler. 2010;16:21–29. doi: 10.1177/1352458509350741. [DOI] [PubMed] [Google Scholar]
  • 175.Zephir H, Fajardy I, Outteryck O, et al. Is neuromyelitis optica associated with human leukocyte antigen? Mult Scler. 2009;15:571–579. doi: 10.1177/1352458508102085. [DOI] [PubMed] [Google Scholar]
  • 176.Nelson PA, Khodadoust M, Prodhomme T, et al. Immunodominant T cell determinants of aquaporin-4, the autoantigen associated with neuromyelitis optica. PLOS ONE. 2010;5:e15050. doi: 10.1371/journal.pone.0015050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kalluri SR, Rothhammer V, Staszewski O, et al. Functional characterization of aquaporin-4 specific T cells: towards a model for neuromyelitis optica. PLOS ONE. 2011;6:e16083. doi: 10.1371/journal.pone.0016083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Vaknin-Dembinsky A, Brill L, Kassis I, et al. T-cell reactivity against AQP4 in neuromyelitis optica. Neurology. 2012;79:945–946. doi: 10.1212/WNL.0b013e318266fc2b. [DOI] [PubMed] [Google Scholar]
  • 179.Warabi Y, Yagi K, Hayashi H, Matsumoto Y. Characterization of the T cell receptor repertoire in the Japanese neuromyelitis optica: T cell activity is up-regulated compared to multiple sclerosis. J Neurol Sci. 2006;249:145–152. doi: 10.1016/j.jns.2006.06.011. [DOI] [PubMed] [Google Scholar]
  • 180.Matsuya N, Komori M, Nomura K, et al. Increased T-cell immunity against aquaporin-4 and proteolipid protein in neuromyelitis optica. Int Immunol. 2011;23:565–573. doi: 10.1093/intimm/dxr056. [DOI] [PubMed] [Google Scholar]
  • 181.Ren Z, Wang Y, Duan T, et al. Cross-immunoreactivity between bacterial aquaporin-Z and human aquaporin-4: potential relevance to neuromyelitis optica. J Immunol. 2012;189:4602–4611. doi: 10.4049/jimmunol.1200486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Jarius S, Wandinger KP, Platzer S, Wildemann B. Homology between Klebsiella pneumoniae and human aquaporin-4: no evidence for cross-reactivity in neuromyelitis optica. A study on 114 patients. J Neurol. 2011;258:929–931. doi: 10.1007/s00415-010-5839-2. [DOI] [PubMed] [Google Scholar]
  • 183.Sellner J, Hemmer B, Muhlau M. The clinical spectrum and immunobiology of parainfectious neuromyelitis optica (Devic) syndromes. J Autoimmun. 2010;34:371–379. doi: 10.1016/j.jaut.2009.09.013. [DOI] [PubMed] [Google Scholar]
  • 184.Menge T, Cree B, Saleh A, et al. Neuromyelitis optica following human papillomavirus vaccination. Neurology. 2012;79:285–287. doi: 10.1212/WNL.0b013e31825fdead. [DOI] [PubMed] [Google Scholar]
  • 185.Alberdi E, Sanchez-Gomez MV, Torre I, et al. Activation of kainate receptors sensitizes oligodendrocytes to complement attack. J Neurosci. 2006;26:3220–3228. doi: 10.1523/JNEUROSCI.3780-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Nicchia GP, Mastrototaro M, Rossi A, et al. Aquaporin-4 orthogonal arrays of particles are the target for neuromyelitis optica autoantibodies. Glia. 2009;57:1363–1373. doi: 10.1002/glia.20855. [DOI] [PubMed] [Google Scholar]
  • 187.Rossi A, Ratelade J, Papadopoulos MC, Bennett JL, Verkman AS. Consequences of NMO-IgG binding to aquaporin-4 in neuromyelitis optica. Proc Natl Acad Sci USA. 2012;109:E1511. doi: 10.1073/pnas.1203463109. author reply E2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Jarius S, Franciotta D, Bergamaschi R, Wildemann B, Wandinger KP. Immunoglobulin M antibodies to aquaporin-4 in neuromyelitis optica and related disorders. Clin Chem Lab Med. 2010;48:659–663. doi: 10.1515/CCLM.2010.127. [DOI] [PubMed] [Google Scholar]
  • 189.Jiao Y, Fryer JP, Lennon VA, et al. Updated estimate of AQP4-IgG serostatus and disability outcome in neuromyelitis optica. Neurology. 2013;81:1197–1204. doi: 10.1212/WNL.0b013e3182a6cb5c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Marignier R, Bernard-Valnet R, Giraudon P, et al. Aquaporin-4 antibody-negative neuromyelitis optica: distinct assay sensitivity-dependent entity. Neurology. 2013;80:2194–2200. doi: 10.1212/WNL.0b013e318296e917. [DOI] [PubMed] [Google Scholar]
  • 191.Waters PJ, McKeon A, Leite MI, et al. Serologic diagnosis of NMO: a multicenter comparison of aquaporin-4-IgG assays. Neurology. 2012;78:665–671. doi: 10.1212/WNL.0b013e318248dec1. discussion 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Mori M, Hosoya M, Hiwasa T, Hayakawa S, Uzawa A, Kuwabara S. Detection of mumps virus RNA in cerebrospinal fluid of patients with neuromyelitis optica. Neurol Sci. 2011;32:795–799. doi: 10.1007/s10072-011-0564-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Koga M, Takahashi T, Kawai M, Fujihara K, Kanda T. A serological analysis of viral and bacterial infections associated with neuromyelitis optica. J Neurol Sci. 2011;300:19–22. doi: 10.1016/j.jns.2010.10.013. [DOI] [PubMed] [Google Scholar]
  • 194.Mader S, Gredler V, Schanda K, et al. Complement activating antibodies to myelin oligodendrocyte glycoprotein in neuromyelitis optica and related disorders. J Neuroinflammation. 2011;8:184. doi: 10.1186/1742-2094-8-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Rostasy K, Mader S, Hennes E, et al. Persisting myelin oligodendrocyte glycoprotein antibodies in aquaporin-4 antibody negative pediatric neuromyelitis optica. Mult Scler. 2013;19:1052–1059. doi: 10.1177/1352458512470310. [DOI] [PubMed] [Google Scholar]
  • 196.Kitley J, Woodhall M, Waters P, et al. Myelin-oligodendrocyte glycoprotein antibodies in adults with a neuromyelitis optica phenotype. Neurology. 2012;79:1273–1277. doi: 10.1212/WNL.0b013e31826aac4e. [DOI] [PubMed] [Google Scholar]
  • 197.Mayer MC, Breithaupt C, Reindl M, et al. Distinction and temporal stability of conformational epitopes on myelin oligodendrocyte glycoprotein recognized by patients with different inflammatory central nervous system diseases. J Immunol. 2013;191:3594–3604. doi: 10.4049/jimmunol.1301296. [DOI] [PubMed] [Google Scholar]
  • 198.Jarius S, Wandinger KP, Borowski K, Stoecker W, Wildemann B. Antibodies to CV2/CRMP5 in neuromyelitis optica-like disease: case report and review of the literature. Clin Neurol Neurosurg. 2012;114:331–335. doi: 10.1016/j.clineuro.2011.10.048. [DOI] [PubMed] [Google Scholar]
  • 199.Kruer MC, Koch TK, Bourdette DN, et al. NMDA receptor encephalitis mimicking seronegative neuromyelitis optica. Neurology. 2010;74:1473–1475. doi: 10.1212/WNL.0b013e3181dc1a7f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Cree BA, Goodin DS, Hauser SL. Neuromyelitis optica. Semin Neurol. 2002;22:105–122. doi: 10.1055/s-2002-36534. [DOI] [PubMed] [Google Scholar]
  • 201.Trebst C, Raab P, Voss EV, et al. Longitudinal extensive transverse myelitis – it's not all neuromyelitis optica. Nat Rev Neurol. 2011;7:688–698. doi: 10.1038/nrneurol.2011.176. [DOI] [PubMed] [Google Scholar]
  • 202.Kitley JL, Leite MI, George JS, Palace JA. The differential diagnosis of longitudinally extensive transverse myelitis. Mult Scler. 2012;18:271–285. doi: 10.1177/1352458511406165. [DOI] [PubMed] [Google Scholar]
  • 203.Andersson M, Alvarez-Cermeno J, Bernardi G, et al. Cerebrospinal fluid in the diagnosis of multiple sclerosis: a consensus report. J Neurol Neurosurg Psychiatry. 1994;57:897–902. doi: 10.1136/jnnp.57.8.897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Reiber H, Teut M, Pohl D, Rostasy KM, Hanefeld F. Paediatric and adult multiple sclerosis: age-related differences and time course of the neuroimmunological response in cerebrospinal fluid. Mult Scler. 2009;15:1466–1480. doi: 10.1177/1352458509348418. [DOI] [PubMed] [Google Scholar]
  • 205.Walsh MJ, Tourtellotte WW. Temporal invariance and clonal uniformity of brain and cerebrospinal IgG, IgA, and IgM in multiple sclerosis. J Exp Med. 1986;163:41–53. doi: 10.1084/jem.163.1.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Reiber H, Ungefehr S, Jacobi C. The intrathecal, polyspecific and oligoclonal immune response in multiple sclerosis. Mult Scler. 1998;4:111–117. doi: 10.1177/135245859800400304. [DOI] [PubMed] [Google Scholar]
  • 207.Jarius S, Eichhorn P, Jacobi C, Wildemann B, Wick M, Voltz R. The intrathecal, polyspecific antiviral immune response: specific for MS or a general marker of CNS autoimmunity? J Neurol Sci. 2009;280:98–100. doi: 10.1016/j.jns.2008.08.002. [DOI] [PubMed] [Google Scholar]
  • 208.Jarius S, Eichhorn P, Wildemann B, Wick M. Usefulness of antibody index assessment in cerebrospinal fluid from patients negative for total-IgG oligoclonal bands. Fluids Barriers CNS. 2012;9:14. doi: 10.1186/2045-8118-9-14. Jul 31; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Jarius S, Franciotta D, Bergamaschi R, et al. Polyspecific, antiviral immune response distinguishes multiple sclerosis and neuromyelitis optica. J Neurol Neurosurg Psychiatry. 2008;79:1134–1136. doi: 10.1136/jnnp.2007.133330. [DOI] [PubMed] [Google Scholar]
  • 210.Jarius S, Wildemann B. Aquaporin-4 antibodies, CNS acidosis and neuromyelitis optica: a potential link. Med Hypotheses. 2013;81:1090–1095. doi: 10.1016/j.mehy.2013.10.011. [DOI] [PubMed] [Google Scholar]
  • 211.Lepur D, Peterkovic V, Kalabric-Lepur N. Neuromyelitis optica with CSF examination mimicking bacterial meningomyelitis. Neurol Sci. 2009;30:51–54. doi: 10.1007/s10072-008-0002-x. [DOI] [PubMed] [Google Scholar]
  • 212.Jarius S, Warth A, Wandinger KP, et al. Antibodies to aquaporin-4 in non-small cell lung cancer: a study on 50 patients. Neurol Sci. 2010;31:871–872. doi: 10.1007/s10072-010-0290-9. [DOI] [PubMed] [Google Scholar]
  • 213.Symmonds M, Nirmalananthan N, Taylor GP, Bridges LR, Schon F. Devastating aquaporin-4 and HTLV-1-associated necrotizing encephalopathy. J Neurol. 2012;259:2732–2735. doi: 10.1007/s00415-012-6651-y. [DOI] [PubMed] [Google Scholar]
  • 214.Glehn von F, Jarius S, Penalva de Oliveira AC, et al. Aquaporin-4 antibodies are not related to HTLV-1 associated myelopathy. PLOS ONE. 2012;7:e39372. doi: 10.1371/journal.pone.0039372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Wildemann B, Jarius S, Hartmann M, Regula JU, Hametner C. Acute disseminated encephalomyelitis following vaccination against human papilloma virus. Neurology. 2009;72:2132–2133. doi: 10.1212/WNL.0b013e3181aa53bb. [DOI] [PubMed] [Google Scholar]
  • 216.Jarius S, Franciotta D, Paul F, et al. Cerebrospinal fluid antibodies to aquaporin-4 in neuromyelitis optica and related disorders: frequency, origin, and diagnostic relevance. J Neuroinflammation. 2010;7:52. doi: 10.1186/1742-2094-7-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Dujmovic I, Mader S, Schanda K, et al. Temporal dynamics of cerebrospinal fluid anti-aquaporin-4 antibodies in patients with neuromyelitis optica spectrum disorders. J Neuroimmunol. 2011;234:124–130. doi: 10.1016/j.jneuroim.2011.01.007. [DOI] [PubMed] [Google Scholar]
  • 218.Heerlein K, Jarius S, Jacobi C, Rohde S, Storch-Hagenlocher B, Wildemann B. Aquaporin-4 antibody positive longitudinally extensive transverse myelitis following varicella zoster infection. J Neurol Sci. 2009;276:184–186. doi: 10.1016/j.jns.2008.08.015. [DOI] [PubMed] [Google Scholar]
  • 219.Kiyat-Atamer A, Ekizoglu E, Tuzun E, et al. Long-term MRI findings in neuromyelitis optica: seropositive versus seronegative patients. Eur J Neurol. 2013;20:781–787. doi: 10.1111/ene.12058. [DOI] [PubMed] [Google Scholar]
  • 220.Lim YM, Pyun SY, Kang BH, Kim J, Kim KK. Factors associated with the effectiveness of plasma exchange for the treatment of NMO-IgG-positive neuromyelitis optica spectrum disorders. Mult Scler. 2013;19:1216–1218. doi: 10.1177/1352458512471875. [DOI] [PubMed] [Google Scholar]
  • 221.Yonezu T, Ito S, Mori M, et al. ‘Bright spotty lesions’ on spinal magnetic resonance imaging differentiate neuromyelitis optica from multiple sclerosis. Mult Scler. 2013 doi: 10.1177/1352458513495581. Jul 4; epub ahead of print. doi: 10.1177/1352458513495581. [DOI] [PubMed] [Google Scholar]
  • 222.Klawiter EC, Xu J, Naismith RT, et al. Increased radial diffusivity in spinal cord lesions in neuromyelitis optica compared with multiple sclerosis. Mult Scler. 2012;18:1259–1268. doi: 10.1177/1352458512436593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Naismith RT, Xu J, Klawiter EC, et al. Spinal cord tract diffusion tensor imaging reveals disability substrate in demyelinating disease. Neurology. 2013;80:2201–2209. doi: 10.1212/WNL.0b013e318296e8f1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ciccarelli O, Thomas D, De Vita E, et al. Low myo-inositol indicating astrocytic damage in a case series of NMO. Ann Neurol. 2013 doi: 10.1002/ana.23909. Apr 3; epub ahead of print. doi: 10.1002/ana.23909. [DOI] [PubMed] [Google Scholar]
  • 225.Pittock SJ, Lennon VA, Krecke K, Wingerchuk DM, Lucchinetti CF, Weinshenker BG. Brain abnormalities in neuromyelitis optica. Arch Neurol. 2006;63:390–396. doi: 10.1001/archneur.63.3.390. [DOI] [PubMed] [Google Scholar]
  • 226.Matthews L, Marasco R, Jenkinson M, et al. Distinction of seropositive NMO spectrum disorder and MS brain lesion distribution. Neurology. 2013;80:1330–1337. doi: 10.1212/WNL.0b013e3182887957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kim W, Kim SH, Huh SY, Kim HJ. Brain abnormalities in neuromyelitis optica spectrum disorder. Mult Scler Int. 2012;2012:735486. doi: 10.1155/2012/735486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Sinnecker T, Dorr J, Pfueller CF, et al. Distinct lesion morphology at 7-T MRI differentiates neuromyelitis optica from multiple sclerosis. Neurology. 2012;79:708–714. doi: 10.1212/WNL.0b013e3182648bc8. [DOI] [PubMed] [Google Scholar]
  • 229.Kister I, Herbert J, Zhou Y, Ge Y. Ultrahigh-field MR (7 T) imaging of brain lesions in neuromyelitis optica. Mult Scler Int. 2013;2013:398259. doi: 10.1155/2013/398259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Popescu BF, Parisi JE, Cabrera-Gomez JA, et al. Absence of cortical demyelination in neuromyelitis optica. Neurology. 2010;75:2103–2109. doi: 10.1212/WNL.0b013e318200d80c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Calabrese M, Oh MS, Favaretto A, et al. No MRI evidence of cortical lesions in neuromyelitis optica. Neurology. 2012;79:1671–1676. doi: 10.1212/WNL.0b013e31826e9a96. [DOI] [PubMed] [Google Scholar]
  • 232.Pittock SJ, Weinshenker BG, Lucchinetti CF, Wingerchuk DM, Corboy JR, Lennon VA. Neuromyelitis optica brain lesions localized at sites of high aquaporin 4 expression. Arch Neurol. 2006;63:964–968. doi: 10.1001/archneur.63.7.964. [DOI] [PubMed] [Google Scholar]
  • 233.Zhang L, Wu A, Zhang B, et al. Comparison of deep gray matter lesions on magnetic resonance imaging among adults with acute disseminated encephalomyelitis, multiple sclerosis, and neuromyelitis optica. Mult Scler. 2014 doi: 10.1177/1352458513499420. Jan 22; epub ahead of print. doi: 10.1177/1352458513499420. [DOI] [PubMed] [Google Scholar]
  • 234.Chan KH, Tse CT, Chung CP, et al. Brain involvement in neuromyelitis optica spectrum disorders. Arch Neurol. 2011;68:1432–1439. doi: 10.1001/archneurol.2011.249. [DOI] [PubMed] [Google Scholar]
  • 235.Makino T, Ito S, Mori M, Yonezu T, Ogawa Y, Kuwabara S. Diffuse and heterogeneous T2-hyperintense lesions in the splenium are characteristic of neuromyelitis optica. Mult Scler. 2012;19:308–315. doi: 10.1177/1352458512454772. [DOI] [PubMed] [Google Scholar]
  • 236.Kim W, Park MS, Lee SH, et al. Characteristic brain magnetic resonance imaging abnormalities in central nervous system aquaporin-4 autoimmunity. Mult Scler. 2010;16:1229–1236. doi: 10.1177/1352458510376640. [DOI] [PubMed] [Google Scholar]
  • 237.Banker P, Sonni S, Kister I, Loh JP, Lui YW. Pencil-thin ependymal enhancement in neuromyelitis optica spectrum disorders. Mult Scler. 2012;18:1050–1053. doi: 10.1177/1352458511431730. [DOI] [PubMed] [Google Scholar]
  • 238.Ito S, Mori M, Makino T, Hayakawa S, Kuwabara S. ‘Cloud-like enhancement’ is a magnetic resonance imaging abnormality specific to neuromyelitis optica. Ann Neurol. 2009;66:425–428. doi: 10.1002/ana.21753. [DOI] [PubMed] [Google Scholar]
  • 239.Chanson JB, Lamy J, Rousseau F, et al. White matter volume is decreased in the brain of patients with neuromyelitis optica. Eur J Neurol. 2013;20:361–367. doi: 10.1111/j.1468-1331.2012.03867.x. [DOI] [PubMed] [Google Scholar]
  • 240.Sanchez-Catasus CA, Cabrera-Gomez J, Almaguer Melian W, et al. Brain tissue volumes and perfusion change with the number of optic neuritis attacks in relapsing neuromyelitis optica: a voxel-based correlation study. PLOS ONE. 2013;8:e66271. doi: 10.1371/journal.pone.0066271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Rueda Lopes FC, Doring T, Martins C, et al. The role of demyelination in neuromyelitis optica damage: diffusion-tensor MR imaging study. Radiology. 2012;263:235–242. doi: 10.1148/radiol.12111241. [DOI] [PubMed] [Google Scholar]
  • 242.Liu Y, Duan Y, He Y, et al. A tract-based diffusion study of cerebral white matter in neuromyelitis optica reveals widespread pathological alterations. Mult Scler. 2012;18:1013–1021. doi: 10.1177/1352458511431731. [DOI] [PubMed] [Google Scholar]
  • 243.Zhao DD, Zhou HY, Wu QZ, et al. Diffusion tensor imaging characterization of occult brain damage in relapsing neuromyelitis optica using 3.0T magnetic resonance imaging techniques. Neuroimage. 2012;59:3173–3177. doi: 10.1016/j.neuroimage.2011.11.022. [DOI] [PubMed] [Google Scholar]
  • 244.Matsuoka T, Matsushita T, Osoegawa M, et al. Heterogeneity and continuum of multiple sclerosis in Japanese according to magnetic resonance imaging findings. J Neurol Sci. 2008;266:115–125. doi: 10.1016/j.jns.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 245.Ishizu T, Kira J, Osoegawa M, et al. Heterogeneity and continuum of multiple sclerosis phenotypes in Japanese according to the results of the fourth nationwide survey. J Neurol Sci. 2009;280:22–28. doi: 10.1016/j.jns.2009.01.008. [DOI] [PubMed] [Google Scholar]
  • 246.Pfueller CF, Paul F. Imaging the visual pathway in neuromyelitis optica. Mult Scler Int. 2011;2011:869814. doi: 10.1155/2011/869814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Brandt AU, Zimmermann H, Kaufhold F, et al. Patterns of retinal damage facilitate differential diagnosis between Susac syndrome and MS. PLOS ONE. 2012;7:e38741. doi: 10.1371/journal.pone.0038741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Stricker S, Oberwahrenbrock T, Zimmermann H, et al. Temporal retinal nerve fiber loss in patients with spinocerebellar ataxia type 1. PLOS ONE. 2011;6:e23024. doi: 10.1371/journal.pone.0023024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Zimmermann H, Freing A, Kaufhold F, et al. Optic neuritis interferes with optical coherence tomography and magnetic resonance imaging correlations. Mult Scler. 2013;19:443–450. doi: 10.1177/1352458512457844. [DOI] [PubMed] [Google Scholar]
  • 250.Bock M, Brandt AU, Dorr J, et al. Time domain and spectral domain optical coherence tomography in multiple sclerosis: a comparative cross-sectional study. Mult Scler. 2010;16:893–896. doi: 10.1177/1352458510365156. [DOI] [PubMed] [Google Scholar]
  • 251.Brandt AU, Oberwahrenbrock T, Ringelstein M, et al. Primary retinal pathology in multiple sclerosis as detected by optical coherence tomography. Brain. 2011;134:e193. doi: 10.1093/brain/awr095. author reply e4. [DOI] [PubMed] [Google Scholar]
  • 252.Roth NM, Saidha S, Zimmermann H, et al. Optical coherence tomography does not support optic nerve involvement in amyotrophic lateral sclerosis. Eur J Neurol. 2013;20:1170–1176. doi: 10.1111/ene.12146. [DOI] [PubMed] [Google Scholar]
  • 253.Kaufhold F, Zimmermann H, Schneider E, et al. Optic neuritis is associated with inner nuclear layer thickening and microcystic macular edema independently of multiple sclerosis. PLOS ONE. 2013;8:e71145. doi: 10.1371/journal.pone.0071145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Kaufhold F, Kadas EM, Schmidt C, et al. Optic nerve head quantification in idiopathic intracranial hypertension by spectral domain OCT. PLOS ONE. 2012;7:e36965. doi: 10.1371/journal.pone.0036965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Naismith RT, Tutlam NT, Xu J, et al. Optical coherence tomography differs in neuromyelitis optica compared with multiple sclerosis. Neurology. 2009;72:1077–1082. doi: 10.1212/01.wnl.0000345042.53843.d5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Ratchford JN, Quigg ME, Conger A, et al. Optical coherence tomography helps differentiate neuromyelitis optica and MS optic neuropathies. Neurology. 2009;73:302–308. doi: 10.1212/WNL.0b013e3181af78b8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.von Glehn F, Jarius S, Cavalcanti Lira RP, Alves Ferreira MC, von Glehn FH, Costa E, Castro SM, Beltramini GC, Bergo FP, Farias AS, Brandão CO, Wildemann B, Damasceno BP, Cendes F, Santos LM, Yasuda CL. Structural brain abnormalities are related to retinal nerve fiber layer thinning and disease duration in neuromyelitis optica spectrum disorders. Mult Scler. 2014 doi: 10.1177/1352458513519838. Jan 29; epub ahead of print. doi: 10.1016/j.jns.2013.01.040. [DOI] [PubMed] [Google Scholar]
  • 258.Gelfand JM, Cree BA, Nolan R, Arnow S, Green AJ. Microcystic inner nuclear layer abnormalities and neuromyelitis optica. JAMA Neurol. 2013;70:629–633. doi: 10.1001/jamaneurol.2013.1832. [DOI] [PubMed] [Google Scholar]
  • 259.Sotirchos ES, Saidha S, Byraiah G, et al. In vivo identification of morphologic retinal abnormalities in neuromyelitis optica. Neurology. 2013;80:1406–1414. doi: 10.1212/WNL.0b013e31828c2f7a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Schneider E, Zimmermann H, Oberwahrenbrock T, et al. Optical coherence tomography reveals distinct patterns of retinal damage in neuromyelitis optica and multiple sclerosis. PLOS ONE. 2013;8:e66151. doi: 10.1371/journal.pone.0066151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Bouyon M, Collongues N, Zephir H, et al. Longitudinal follow-up of vision in a neuromyelitis optica cohort. Mult Scler. 2013;19:1320–1322. doi: 10.1177/1352458513476562. [DOI] [PubMed] [Google Scholar]
  • 262.Ringelstein M, Kleiter I, Ayzenberg I, et al. Visual evoked potentials in neuromyelitis optica and its spectrum disorders. Mult Scler. 2013 doi: 10.1177/1352458513503053. Sep 5; epub ahead of print. doi: 10.1177/1352458513503053. [DOI] [PubMed] [Google Scholar]
  • 263.Wildemann B, Jarius S, Paul F. [Neuromyelitis optica] Akt Neurol. 2012;39:33–41. [Google Scholar]
  • 264.Kimbrough DJ, Fujihara K, Jacob A, et al. Treatment of neuromyelitis optica: review and recommendations. Mult Scler Relat Disord. 2012;1:180–187. doi: 10.1016/j.msard.2012.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Magana SM, Keegan BM, Weinshenker BG, et al. Beneficial plasma exchange response in central nervous system inflammatory demyelination. Arch Neurol. 2011;68:870–878. doi: 10.1001/archneurol.2011.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Elsone L, Panicker J, Mutch K, Boggild M, Appleton R, Jacob A. Role of intravenous immunoglobulin in the treatment of acute relapses of neuromyelitis optica: experience in 10 patients. Mult Scler. 2013 doi: 10.1177/1352458513495938. Nov 29; epub ahead of print. doi: 10.1177/1352458513495938. [DOI] [PubMed] [Google Scholar]
  • 267.Greenberg BM, Graves D, Remington G, et al. Rituximab dosing and monitoring strategies in neuromyelitis optica patients: creating strategies for therapeutic success. Mult Scler. 2012;18:1022–1026. doi: 10.1177/1352458511432896. [DOI] [PubMed] [Google Scholar]
  • 268.Pellkofer H, Suessmair C, Schulze A, Hohlfeld R, Kuempfel T. Course of neuromyelitis optica during inadvertent pregnancy in a patient treated with rituximab. Mult Scler. 2009;15:1006–1008. doi: 10.1177/1352458509106512. [DOI] [PubMed] [Google Scholar]
  • 269.Costanzi C, Matiello M, Lucchinetti CF, et al. Azathioprine: tolerability, efficacy, and predictors of benefit in neuromyelitis optica. Neurology. 2011;77:659–666. doi: 10.1212/WNL.0b013e31822a2780. [DOI] [PubMed] [Google Scholar]
  • 270.Watanabe S, Misu T, Miyazawa I, et al. Low-dose corticosteroids reduce relapses in neuromyelitis optica: a retrospective analysis. Mult Scler. 2007;13:968–974. doi: 10.1177/1352458507077189. [DOI] [PubMed] [Google Scholar]
  • 271.Bichuetti DB, Lobato de Oliveira EM, Oliveira DM, Amorin de Souza N, Gabbai AA. Neuromyelitis optica treatment: analysis of 36 patients. Arch Neurol. 2010;67:1131–1136. doi: 10.1001/archneurol.2010.203. [DOI] [PubMed] [Google Scholar]
  • 272.Mandler RN, Ahmed W, Dencoff JE. Devic's neuromyelitis optica: a prospective study of seven patients treated with prednisone and azathioprine. Neurology. 1998;51:1219–1220. doi: 10.1212/wnl.51.4.1219. [DOI] [PubMed] [Google Scholar]
  • 273.Jacob A, Matiello M, Weinshenker BG, et al. Treatment of neuromyelitis optica with mycophenolate mofetil: retrospective analysis of 24 patients. Arch Neurol. 2009;66:1128–1133. doi: 10.1001/archneurol.2009.175. [DOI] [PubMed] [Google Scholar]
  • 274.Kitley J, Elsone L, George J, et al. Methotrexate is an alternative to azathioprine in neuromyelitis optica spectrum disorders with aquaporin-4 antibodies. J Neurol Neurosurg Psychiatry. 2013;84:918–921. doi: 10.1136/jnnp-2012-304774. [DOI] [PubMed] [Google Scholar]
  • 275.Kim SH, Kim W, Park MS, Sohn EH, Li XF, Kim HJ. Efficacy and safety of mitoxantrone in patients with highly relapsing neuromyelitis optica. Arch Neurol. 2011;68:473–479. doi: 10.1001/archneurol.2010.322. [DOI] [PubMed] [Google Scholar]
  • 276.Weinstock-Guttman B, Ramanathan M, Lincoff N, et al. Study of mitoxantrone for the treatment of recurrent neuromyelitis optica (Devic disease) Arch Neurol. 2006;63:957–963. doi: 10.1001/archneur.63.7.957. [DOI] [PubMed] [Google Scholar]
  • 277.Cabre P, Olindo S, Marignier R, Jeannin S, Merle H, Smadja D. Efficacy of mitoxantrone in neuromyelitis optica spectrum: clinical and neuroradiological study. J Neurol Neurosurg Psychiatry. 2013;84:511–516. doi: 10.1136/jnnp-2012-303121. [DOI] [PubMed] [Google Scholar]
  • 278.Dorr J, Bitsch A, Schmailzl KJ, et al. Severe cardiac failure in a patient with multiple sclerosis following low-dose mitoxantrone treatment. Neurology. 2009;73:991–993. doi: 10.1212/WNL.0b013e3181b878f6. [DOI] [PubMed] [Google Scholar]
  • 279.Stroet A, Hemmelmann C, Starck M, et al. Incidence of therapy-related acute leukaemia in mitoxantrone-treated multiple sclerosis patients in Germany. Ther Adv Neurol Disord. 2012;5:75–79. doi: 10.1177/1756285611433318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Stroet A, Gold R, Chan A. Acute myeloid leukemia in Italian patients with multiple sclerosis treated with mitoxantrone. Neurology. 2012;78:933. doi: 10.1212/01.wnl.0000413366.14351.54. author reply 4. [DOI] [PubMed] [Google Scholar]
  • 281.Trebst C, Jarius S, Berthele A, et al. Update on the diagnosis and treatment of neuromyelitis optica: recommendations of the Neuromyelitis Optica Study Group (NEMOS) J Neurol. 2013;261:1–16. doi: 10.1007/s00415-013-7169-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Kim SH, Kim W, Li XF, Jung IJ, Kim HJ. Does interferon beta treatment exacerbate neuromyelitis optica spectrum disorder? Mult Scler. 2012;78:1179–1185. doi: 10.1177/1352458512439439. [DOI] [PubMed] [Google Scholar]
  • 283.Palace J, Leite MI, Nairne A, Vincent A. Interferon beta treatment in neuromyelitis optica: increase in relapses and aquaporin 4 antibody titers. Arch Neurol. 2010;67:1016–1017. doi: 10.1001/archneurol.2010.188. [DOI] [PubMed] [Google Scholar]
  • 284.Papeix C, Vidal JS, Seze de J, et al. Immunosuppressive therapy is more effective than interferon in neuromyelitis optica. Mult Scler. 2007;13:256–259. doi: 10.1177/1352458506070732. [DOI] [PubMed] [Google Scholar]
  • 285.Shimizu Y, Yokoyama K, Misu T, et al. Development of extensive brain lesions following interferon beta therapy in relapsing neuromyelitis optica and longitudinally extensive myelitis. J Neurol. 2008;255:305–307. doi: 10.1007/s00415-007-0730-5. [DOI] [PubMed] [Google Scholar]
  • 286.Uzawa A, Mori M, Hayakawa S, Masuda S, Kuwabara S. Different responses to interferon beta-1b treatment in patients with neuromyelitis optica and multiple sclerosis. Eur J Neurol. 2010;17:672–676. doi: 10.1111/j.1468-1331.2009.02897.x. [DOI] [PubMed] [Google Scholar]
  • 287.Warabi Y, Matsumoto Y, Hayashi H. Interferon beta-1b exacerbates multiple sclerosis with severe optic nerve and spinal cord demyelination. J Neurol Sci. 2007;252:57–61. doi: 10.1016/j.jns.2006.10.008. [DOI] [PubMed] [Google Scholar]
  • 288.Min JH, Kim BJ, Lee KH. Development of extensive brain lesions following fingolimod (FTY720) treatment in a patient with neuromyelitis optica spectrum disorder. Mult Scler. 2012;18:113–115. doi: 10.1177/1352458511431973. [DOI] [PubMed] [Google Scholar]
  • 289.Izaki S, Narukawa S, Kubota A, Mitsui T, Fukaura H, Nomura K. A case of neuromyelitis optica spectrum disorder developing a fulminant course with multiple white-matter lesions following fingolimod treatment. Rinsho Shinkeigaku. 2013;53:513–517. doi: 10.5692/clinicalneurol.53.513. [DOI] [PubMed] [Google Scholar]
  • 290.Qian P, Cross AH, Naismith RT. Lack of response to monoclonal antibody therapy in neuromyelitis optica. Arch Neurol. 2011;68:1207–1209. doi: 10.1001/archneurol.2011.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Miyazaki K, Abe Y, Iwanari H, et al. Establishment of monoclonal antibodies against the extracellular domain that block binding of NMO-IgG to AQP4. J Neuroimmunol. 2013;260:107–116. doi: 10.1016/j.jneuroim.2013.03.003. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES