ArticlePDF Available

MOG cell-based assay detects non-MS patients with inflammatory neurologic disease

Authors:

Abstract and Figures

To optimize sensitivity and disease specificity of a myelin oligodendrocyte glycoprotein (MOG) antibody assay. Consecutive sera (n = 1,109) sent for aquaporin-4 (AQP4) antibody testing were screened for MOG antibodies (Abs) by cell-based assays using either full-length human MOG (FL-MOG) or the short-length form (SL-MOG). The Abs were initially detected by Alexa Fluor goat anti-human IgG (H + L) and subsequently by Alexa Fluor mouse antibodies to human IgG1. When tested at 1:20 dilution, 40/1,109 sera were positive for AQP4-Abs, 21 for SL-MOG, and 180 for FL-MOG. Only one of the 40 AQP4-Ab-positive sera was positive for SL-MOG-Abs, but 10 (25%) were positive for FL-MOG-Abs (p = 0.0069). Of equal concern, 48% (42/88) of sera from controls (patients with epilepsy) were positive by FL-MOG assay. However, using an IgG1-specific secondary antibody, only 65/1,109 (5.8%) sera were positive on FL-MOG, and AQP4-Ab- positive and control sera were negative. IgM reactivity accounted for the remaining anti-human IgG (H + L) positivity toward FL-MOG. The clinical diagnoses were obtained in 33 FL-MOG-positive patients, blinded to the antibody data. IgG1-Abs to FL-MOG were associated with optic neuritis (n = 11), AQP4-seronegative neuromyelitis optica spectrum disorder (n = 4), and acute disseminated encephalomyelitis (n = 1). All 7 patients with probable multiple sclerosis (MS) were MOG-IgG1 negative. The limited disease specificity of FL-MOG-Abs identified using Alexa Fluor goat anti-human IgG (H + L) is due in part to detection of IgM-Abs. Use of the FL-MOG and restricting to IgG1-Abs substantially improves specificity for non-MS demyelinating diseases. This study provides Class II evidence that the presence of serum IgG1- MOG-Abs in AQP4-Ab-negative patients distinguishes non-MS CNS demyelinating disorders from MS (sensitivity 24%, 95% confidence interval [CI] 9%-45%; specificity 100%, 95% CI 88%-100%).
Content may be subject to copyright.
Patrick Waters, PhD*
Mark Woodhall, PhD*
Kevin C. OConnor, PhD
Markus Reindl, PhD
Bethan Lang, PhD
Douglas K. Sato, MD
Maciej Jurynczyk, MD
George Tackley, MBBCh
Joao Rocha, MD
Toshiyuki Takahashi, MD
Tatsuro Misu, MD
Ichiro Nakashima, MD
Jacqueline Palace, MD
Kazuo Fujihara, MD
M. Isabel Leite, DPhil
Angela Vincent, FRS
Correspondence to
Dr. Waters:
paddy.waters@ndcn.ox.ac.uk
Supplemental data
at Neurology.org/nn
MOG cell-based assay detects non-MS
patients with inflammatory neurologic
disease
ABSTRACT
Objective: To optimize sensitivity and disease specificity of a myelin oligodendrocyte glycoprotein
(MOG) antibody assay.
Methods: Consecutive sera (n 51,109) sent for aquaporin-4 (AQP4) antibody testing were
screened for MOG antibodies (Abs) by cell-based assays using either full-length human MOG
(FL-MOG) or the short-length form (SL-MOG). The Abs were initially detected by Alexa Fluor goat
anti-human IgG (H 1L) and subsequently by Alexa Fluor mouse antibodies to human IgG1.
Results: When tested at 1:20 dilution, 40/1,109 sera were positive for AQP4-Abs, 21 for SL-
MOG, and 180 for FL-MOG. Only one of the 40 AQP4-Abpositive sera was positive for SL-
MOG-Abs, but 10 (25%) were positive for FL-MOG-Abs (p50.0069). Of equal concern, 48%
(42/88) of sera from controls (patients with epilepsy) were positive by FL-MOG assay. However,
using an IgG1-specific secondary antibody, only 65/1,109 (5.8%) sera were positive on FL-MOG,
and AQP4-Abpositive and control sera were negative. IgM reactivity accounted for the remain-
ing anti-human IgG (H 1L) positivity toward FL-MOG. The clinical diagnoses were obtained in 33
FL-MOGpositive patients, blinded to the antibody data. IgG1-Abs to FL-MOG were associated
with optic neuritis (n 511), AQP4-seronegative neuromyelitis optica spectrum disorder (n 54),
and acute disseminated encephalomyelitis (n 51). All 7 patients with probable multiple sclerosis
(MS) were MOG-IgG1 negative.
Conclusions: The limited disease specificity of FL-MOG-Abs identified using Alexa Fluor goat anti-
human IgG (H 1L) is due in part to detection of IgM-Abs. Use of the FL-MOG and restricting to
IgG1-Abs substantially improves specificity for non-MS demyelinating diseases.
Classification of evidence: This study provides Class II evidence that the presence of serum
IgG1- MOG-Abs in AQP4-Abnegative patients distinguishes non-MS CNS demyelinating
disorders from MS (sensitivity 24%, 95% confidence interval [CI] 9%45%; specificity
100%, 95% CI 88%100%). Neurol Neuroimmunol Neuroinflamm 2015;2:e89; doi: 10.1212/
NXI.0000000000000089
GLOSSARY
Abs 5antibodies; ADEM 5acute disseminated encephalomyelitis; AQP4 5aquaporin-4; CBA 5cell-based assay; CI 5
confidence interval; EDTA 5ethylenediaminetetraacetic acid; FACS 5fluorescent-activated cell sorting; FL-MOG 5full-
length human MOG; LETM 5longitudinally extensive TM; MOG 5myelin oligodendrocyte glycoprotein; MS 5multiple scle-
rosis; NMO 5neuromyelitis optica; NMOSD 5NMO spectrum disorder; ON 5optic neuritis; PEI 5polyethylenimine; SL-
MOG 5short-length MOG; TM 5transverse myelitis.
Antibodies (Abs) that bind the CNS-restricted membrane protein myelin oligodendrocyte gly-
coprotein (MOG) were first described by ELISA or Western blot predominantly in patients with
multiple sclerosis (MS), but they have also been described in patients with bacterial or viral CNS
inflammation or neuromyelitis optica (NMO).
111
These findings were not reproducible using
*These authors contributed equally to the manuscript.
From the Nuffield Department of Clinical Neurosciences (P.W., M.W., B.L., M.J., G.T., J.R., J.P., M.I.L., A.V.), John Radcliffe Hospital, Oxford,
UK; Department of Neurology (K.C.O.), Yale School of Medicine, New Haven, CT; Clinical Department of Neurology (M.R.), Innsbruck
Medical University, Innsbruck, Austria; Department of Neurology (D.K.S., I.N.) and Department of Multiple Sclerosis Therapeutics (T.M., K.F.)
Tohoku University School of Medicine, Sendai, Japan; and Department of Neurology (T.T.), Yonezawa National Hospital, Yonezawa, Japan.
Funding information and disclosures are provided at the end of the article. Go to Neurology.org/nn for full disclosure forms. The Article Processing
Charge was paid by the authors.
This is an open access article distributed under the terms of the Creative Commons Attribution-Noncommercial No Derivative 3.0 License, which
permits downloading and sharing the work provided it is properly cited. The work cannot be changed in any way or used commercially.
Neurology.org/nn © 2015 American Academy of Neurology 1
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
similar methods,
1218
but serologic findings and
different experimental approaches suggested
that MOG-Abs may be pathogenic.
1923
More-specific assays using soluble, tetramerized
extracellular domain of native MOG identified
Abs in a subset of patients with acute dissemi-
nated encephalomyelitis (ADEM) but rarely in
adult-onset MS cases, now suggesting that the
test could be of relevance for discriminating MS
from other demyelinating syndromes.
24
This
was confirmed by cell-based assay (CBA) that
also used a truncated MOG, in which MOG-
Abs were found in patients with aquaporin-4
(AQP4)seronegative NMO but not those
with MS.
25,26
CBA using full-length human
MOG (FL-MOG) appears to be more sensi-
tive, and a clinical phenotype of ADEM and
AQP4-seronegative NMO spectrum disorder
(NMOSD), often optic neuritis (ON), is
emerging.
2736
However, positivity in healthy
individuals and patients with MS, even at rela-
tively high serum dilutions (up to 1:640), af-
fects its clinical use.
Here we confirm that C-terminal truncation
of the MOG antigen reduces assay sensitivity
and that many of the low positive Abs found
to bind to FL-MOG result from cross-
reactivity of the anti-human IgG secondary
antibody with IgM Abs. Using IgG1-specific
secondary antibody allows use of lower serum
dilutions with FL-MOG, with improved spec-
ificity for patients with ON, transverse myelitis
(TM), AQP4-Abnegative NMO, or ADEM.
METHODS Patients. Consecutive serum samples from 1,109
individuals sent for routine AQP4-Ab testing over 3 months were
studied. Samples are sent to Oxford via clinical immunology
laboratories with very limited or no clinical information. Sera from
118 of the 180 FL-MOGpositive samples were used to assess
different secondary Abs, and 15/180 FL-MOGseropositive
samples were used for flow cytometry (a flow diagram of which
samples were tested on the different assays is shown in figure 1).
To assess the clinical relevance, a brief anonymized questionnaire was
sent after the analyses to 48 identifiable referring clinicians requesting
patient diagnosis, treatment responses, and relapses, if any. Controls
were sera from previously archived cohorts. To validate the results, 2
other cohorts were screened. Patients seen at the National NMO
Specialised Services who had already been tested for AQP4-Abs were
tested for MOG-IgG1-Abs. After testing was completed, the
diagnoses and follow-up times from the seropositive patients were
obtained from a database. A further cohort of 101 Japanese patients
with a range of demyelinating diagnoses (see Results) followed by or
referred to Tohoku University Hospital and who had been previously
tested for AQP4-Abs were tested for MOG-IgG1-Abs. All assays
were carried out blinded to the clinical diagnoses.
Ethics. Ethics have been approved for the study of any patients
whose samples have been referred to the Neuroimmunology lab-
oratory in Oxford for diagnostic testing (Oxfordshire REC A;
07/Q1604/28 Immune factors in neurological disease). Since
January 2010, data on all patients seen within the Oxford clinical
NMO service have been entered prospectively into a clinical data-
base and patient serum samples have been routinely tested for
AQP4-Abs and MOG-Abs. The ethics committee of Tohoku
University Graduate School of Medicine approved this study,
and all participants provided written informed consent.
Constructs. The cloning of M23 isoform of human AQP4 has
been described previously.
7
FL-MOG was cloned into
pIRES2-DsRed2 using the forward primer (59-39):
gatcctcgagccaccatggcaagcttatcaagaccctctctg and the reverse
primer (59-39): gatccccgggtcagaagggatttcgtagctcttcaagg. A
C-terminaltruncated MOG construct was created from the
full-length construct by insertion of a stop codon after Gly155
and excision of the remainder of the C-terminus. The 2 forms
differ only in the intracytoplasmic domain (figure 2A).
Cell-based assays. HEK293T cells, polyethylenimine (PEI)
transfected with human M23-AQP4, FL-MOG, or C-
terminaltruncated human MOG (short-length MOG; SL-
MOG) were used as the substrate for live CBAs, which were
performed as described elsewhere.
79
Patient sera were tested at
1:20 dilution. The Alexa Fluor 488 goat anti-human IgG (H 1L)
from Invitrogen (A1013, Carlsbad, CA) was used at 1:750 dilution.
A semiquantitative scoring system was used: 0, no binding;
1, low-level binding; 24, increasing level of specific binding.
Figure 1 Flow diagram of the assays and the samples that were evaluated
A total of 1,109 samples were initially screened at a serum dilution of 1:20 for antibodies to
aquaporin-4 (AQP4), short-length MOG (SL-MOG), and full-length human MO G (FL-MOG). Dif-
ferent secondary antibodies were then evaluated on FL-MOGpositive serum samples by FL-
MOG cell-based assay (CBA) or flow cytometry. When the assay was established, 2 patient
cohorts with clinical diagnoses from Oxford, UK and Sendai, Japan were used to calculate
assay metrics.
2Neurology: Neuroimmunology & Neuroinflammation
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
Samples scoring .1 were considered positive. The average of 2
individualsscoresisplotted(M.W.,P.W.).
Subclasses. Alexa Fluor 488 mouse anti-human IgG1 (A10631,
Invitrogen) and mouse anti-human IgM (A21215, Invitrogen),
both at 1:500 dilution, or anti-human IgG3 (1:50 dilution,
I7260, Sigma-Aldrich, Gillingham, UK) followed by Alexa
Fluor 488 goat anti-mouse IgG (H 1L; A11001, Invitrogen)
were used as secondary or tertiary Abs. These assays were carried
out as described previously except cells were fixed after the final
antibody incubation.
Flow cytometry. A similar method to that used for detection of
AQP4-Abs (as described in Waters et al.
37
) was used here for FL-
MOG detection. Briefly, HEK293T cells were transfected using
PEI for 16 hours with the pIRES2-DsRed2 plasmid that contained
the complementary DNA for FL-MOG. After washing and
incubation for 24 hours at 37°C in 5% CO
2
, the cells were
trypsinized and resuspended in DulbeccosmodifiedEagles
medium, 1% fetal calf serum, 1 mM ethylenediaminetetraacetic
acid (EDTA) (fluorescent-activated cell sorting [FACS] buffer) at
1.0 310
6
cells/mL. The cells were rotated at 4°C for 1 hour. All
further steps were carried outat 4°C. Patient serum (diluted 1:10 in
FACS buffer) was mixed with 1.0 310
5
cells (100 mL). After
rocking for 1 hour, the cells were washed, and bound IgG was
detected with Alexa Fluor 488 goat anti-human IgG (diluted
1:500 in FACS buffer), Alexa Fluor 488 anti-human IgG1, or
Alexa Fluor anti-human IgM for 3045 minutes. The cells were
washed, resuspended in 400 mL phosphate-buffered saline/2 mM
EDTA, and analyzed by FACScalibur. The level of transfection was
determined by measuring DsRed intensity (PE-Texas red channel)
in live cells (figure 3D, y-axis). Two gates were created: the upper
gate captured cells expressing high levels of DsRed; the lower gate
captured untransfected or poorly transfected cells and served as a
negative control for each sample (figure 3Da). Bound IgG was
measured in the green channel (a shift to the right on the x-axis).
A score for each serum was determined by subtracting the median
green fluorescence in the lower gate from the median green
fluorescence in the upper gate.
Statistics. A 2-tailed Wilcoxon matched-pairs signed-rank test
was used to compare the FL-MOG and SL-MOG assays. The
Mann-Whitney unpaired 2-tailed ttest or Fisher exact test was
used to compare groups (p,0.05 was considered significant).
Primary research question. Does this MOG assay using an
anti-human IgG1-specific secondary antibody identify a
Figure 2 Antibodies to MOG detected with anti-human IgG (H 1L) as the secondary antibody
(A) Schematic of the human MOG proteins tested. The extracellular and transmembrane domains are identical, but the short-length MOG (SL-MOG) is 73
amino acids shorter at the C-terminus than full-length MOG (FL-MOG). (B) Screening 1,109 consecutive samples sent for aquaporin-4 (AQP4) antibody test-
ing. With anti-human IgG (H 1L) as the secondary antibody, 21 SL-MOGpositive samples and 180 FL-MOGpositive samples were identified; however, a
cohort of epilepsy sera demonstrates the striking lack of specificity in the FL-MOG assay. Comparing the AQP4 seropositivity in the 2 MOG assays, 1/38
AQP4-positive samples were also positive for SL-MOG antibodies (C), compared with 10/38 for FL-MOG antibodies (D). CBA 5cell-based assay.
Neurology: Neuroimmunology & Neuroinflammation 3
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
subgroup of AQP4-antibodyseronegative patients with a non-
MS CNS demyelinating disease?
This study provides Class II evidence that the presence of
serum IgG1-Abs specific for MOG can distinguish AQP4-Ab
negative patients with non-MS CNS demyelinating diseases from
those with MS. The Japanese patients were used to calculate the
assay metrics: 6 of 25 AQP4-Abnegative patients with non-MS
demyelinating diseases were MOG-IgG1 positive, for a sensitivity
of 24% (95% confidence interval [CI] 9%45%), and 0 of 27
patients with MS were MOG-IgG1 positive, for a specificity of
100% (95% CI 88%100%).
RESULTS Out of 1,109 samples sent for diagnostic
testing for AQP4-Abs, 40 sera were positive at 1:20
dilution. The SL-MOG assay detected Abs in 21
patients, including 1 (low positive) who was strongly
positive for AQP4 (figure 2, B and C). However, the
FL-MOG assays detected antibodies in 180 sera (16%
of the test cohort), and 10 of these sera were also
positive for AQP4-Abs (figure 2D). Positive results
for FL-MOG were also found in 42/88 sera from
patients with epilepsy (48%, figure 2B).
Control groups and 118/180 FL-MOGpositive
sera that were available were retested by CBA using
either anti-IgG1 or anti-IgM class-specific secondary
Abs (figure 3). With anti-IgM, 101/118 test sera,
7/10 healthy individuals, and 11/17 patients with MS
were positive. The secondary antibody alone did not
bind to FL-MOGtransfected HEK cells, and the con-
trol sera were negative on AQP4-transfected cells. With
anti-IgG1, by contrast, only 65 of 118 sera had scores
of greater than 1, and negative results were found in 49
patients with MS, 13 healthy sera, and 14 AQP4-Ab
positive controls (figure 3C).
Figure 3 Antibodies to MOG using different secondary antibodies: Anti-human IgG (H 1L), IgG1, or IgM
(A) Comparison of binding to full-length myelin oligodendrocyte glycoprotein (FL-MOG) using anti-human IgG (H 1L), anti-IgM, or anti-IgG1 secondary
antibodies with 3 different test sera (a-c) and a healthy control serum (con). (B) IgM and (C) IgG1 binding scores for patients and healthy controls (HC).
(D.a) PIRES2-DsRed2-FL-MOG transiently transfected HEK cells are separated into cells that express MOG and DsRed2 well (in the upper section of the
graph) or poorly or not at all (lowest section of the graph). (D.b) Healthy control sera (upper panels) causes a specific shift in the MOG-transfected cells
compared to the untransfected cells when anti-human IgG (H 1L) or anti-human IgM secondary antibodies are used (arrows), but not when anti-human IgG1
secondary antibodies are used. The lower panels show higher shifts in sera positive for FL-MOG antibodies compared to controls in the upper panel. (E)
Fifteen samples that were IgG (H 1L) positive and 5 healthy controls were tested on flow cytometry with anti-IgM or IgG1. A high cutoff is generated with
anti-human IgM secondary antibody (DMFI of 270) vs a DMFI of 2.5 for the anti-human IgG1 antibody. Of note, one IgM-positive patient is IgG1 negative (blue
circle). Ab 5antibody; AQP4 5aquaporin-4; CBA 5cell-based assay; MFI 5mean fluorescence intensity; MS 5multiple sclerosis.
4Neurology: Neuroimmunology & Neuroinflammation
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
Table 1 Antibody subclass specificity, IgG (H 1L) endpoint titration, SL-MOG cell-based assay score, sex, age at testing, and clinical description of 33 patients
Isotype
IgG (H 1L)
endpoint titer
SL-MOG
score Sex, age, y
Presentation or initial
diagnosis Diagnosis Treatment Recovery Relapse
IgG1 1,200 4.0 M, 32 NMO NMOSD IVMP and steroid taper, PEX Substantial No
800 0 M, 23 BON rON IVMP and steroid taper Partial No
1,600 0 F, 36 Sequential BON BON IVMP and steroid taper No Yes
800 0 F, 38 BON BON IVMP Good No
200 0 F, 54 CRION CRION Steroids, cyclosporin, MMF Yes Yes
300 0 M, 55 NMO historic case NMOSD pre AQP4 Steroids, azathioprine Yes Yes
3,200 3.0 F, 48 RON rON Steroids, azathioprine Incomplete No
3,200 1.0 F, 45 NMOSD-like NMOSD None Substantial No
200 0 M, 47 BON BON Steroids only Yes No
200 0 F, 57 Tumefactive lesions, ? CNS vasculitis Vasculitis? Steroids, cylcophosphamide Yes No
800 2.5 F, 39 ON ON Steroids only Very good No
1,200 0 F, 3.3 ADEM ADEM IVMP Full No
1,600 0 F, 29 ON ON None Partial Not clear
800 3.5 F, 67 NMO-like but AQP4 antibody negative rLETM Steroids only Good Not after
treatment
300 0 F, 33 BON BON Steroids only Very good No
800 0 F, 28 Sequential BON rBON Steroids only Yes (third episode) No
100 0 M, 30 rBON rON None Spontaneous Yes
IgG3 3,200 0 M, 51 ON, myelitis, patchy cord lesions Probable MS IVMP Full No
IgM 25 0 F, 50 6 years pain, aching, fatigue, visual
disturbance, ? TM
Other Steroids only No
75 0 F, 30 Single episode ON, some focal
WM lesions
ON None Spontaneous No
20 0 F, 34 Tumefactive MS, homonymous
hemianopia, ON, cord lesions;
OCB positive
Probable MS Steroids and DMT Yes Yes
20 0 F, 30 Probable MS Probable MS None Yes but not full Yes
20 0 M, 44 Probable MS, many previous minor events Probable MS None Partial Yes
20 0 F, 24 Pain and tingling, perineal numbness,
bladder disturbance, patchy cord
lesions
Probable MS,
probable CIS
Steroids only Yes No
Continued
Neurology: Neuroimmunology & Neuroinflammation 5
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
To further examine the lack of specificity in this
assay, a group of 15 FL-MOGseropositive samples
and 5 healthy controls were tested by flow cytometry
(figure 3D). IgG in healthy control sera bound to FL-
MOGtransfected cells when compared to the un-
transfected control cells in the same test sample when
using anti-human IgG (H 1L) or anti-human IgM
secondary Abs, but not with the anti-human IgG1
secondary antibody (figure 3D, horizontal arrows).
Using the median score 16 SDs of the 5 healthy
control sera, very different cutoffs were generated:
270 for the IgM antibody and 2.5 for the IgG1 anti-
body. One sera (large bluecircle) demonstrated strong
positivity using anti-human IgM secondary antibody
but was negative for IgG-Abs (IgG3, 4 were also neg-
ative on this sample; data not shown).
Clinical phenotypes. Questionnaires on 38/48 patients
(selected because the referring clinician could be iden-
tified) who were positive for IgG (H 1L) Abs were
returned, but complete IgG1 and IgM antibody re-
sults were only available in 33 (17 IgG1-specific,
1 IgG3, and 15 IgM only). The isotype, FL-MOG
endpoint titers, SL-MOG scores, and clinical
diagnoses are shown in table 1. Seven patients with
MS were positive with anti-human IgG (H 1L) or
anti-IgM but not with anti-IgG1. In contrast, all of
the anti-IgG1positive patients had a clinical
diagnosis of non-MS inflammatory demyelinating
CNS disease. ON was more common with IgG1-
MOG-Abs (11/17 vs 3/15; p50.02). In addition,
one 51-year-old male patient with ON and myelitis
with patchy cord lesions and high levels of IgG3
antibodies (endpoint titer of 3,200) was diagnosed
with probable MS.
The majority of patients in each group substan-
tially improved (13/17 IgG1 group vs 7/13 IgM on-
ly), and relapses were found in both groups (5/16
IgG1 and 6/12 IgM only).
Confirmatory cohorts. AQP4-seropositive NMOSD
patients (37 NMO, 11 TM, 33 ON) seen by the
Oxford NMO service were negative for MOG-IgG1-
Abs; however, 23 AQP4-seronegative patients
(8NMO,1TM,9ON,1ON1TM, 4 ADEM)
were MOG-IgG1 positive (figure 4A, table 2).
Thirteen patients with NMO were double
seronegative. Hence, of the 58 patients seen in
Oxford that fulfill the 2006 Wingerchuk criteria for
NMO, 37 (63.8%) are AQP4 seropositive, 8 (13.8%)
MOG-IgG1 positive, and 13 (22.4%) double
seronegative.
A second cohort of 101 Japanese patients with
inflammatory CNS diseases was screened with anti-
IgG1/FL-MOG without knowledge of the clinical
phenotype or AQP4 status. None of the AQP4-
seropositive patients (28 NMO, 5 recurrent ON, 6
Table 1 Continued
Isotype
IgG (H 1L)
endpoint titer
SL-MOG
score Sex, age, y
Presentation or initial
diagnosis Diagnosis Treatment Recovery Relapse
75 0 F, 33 MRI multiple lesions, progressive
disease
Progressive MS Steroids only Yes Relapses now
progressing
100 0 F, 19 Myelitis and ON thickening, ? NMOSD rLETM/? MS Steroids only Yes No
100 0 M, 77 LETM LETM Steroids only Partial No
100 0 M, 42 TM, not LETM TM (thoracic) IVMP Very good No
75 0 F, 28 BON, 2 cerebellar lesions, CSF:
18 WBC
BON IV steroids Yes No
400 0 F, 56 Recurrent TM Not clear Steroids Partial Yes
100 0 M, 32 Probable atypical MS,
myelitis but only small patch
Probable MS IVMP Partial Stepwise progression
150 0 F, 22 Probable relapsing MS Probable MS Steroids only No Yes
20 0 M, 55 Left visual loss, atypical ON ON None Partial Not clear
Abbreviations: ADEM 5acute disseminated encephalomyelitis; AQP4 5aquaporin-4; BON 5bilateral ON; CIS 5clinically isolated syndrome; CRION 5chronic relapsing inflammatory ON; DMT 5disease-modifying
therapy; IVMP 5IV methylprednisolone; LETM 5longitudinally extensive TM; MMF 5mycophenolate mofetil; MS 5multiple sclerosis; NMO 5neuromyelitis optica; NMOSD 5NMO spectrum disorder; OCB 5
oligoclonal band; ON 5optic neuritis; PEX 5plasma exchange; rBON 5recurrent BON; rLETM 5recurrent LETM; rON 5recurrent ON; SL-MOG 5short-length myelin oligodendrocyte glycoprotein; TM 5transverse
myelitis; WBC 5white blood cell; WM 5white matter.
All patients were seronegative for AQP4 antibodies.
6Neurology: Neuroimmunology & Neuroinflammation
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
monophasic longitudinally extensive TM [LETM], 10
recurrent LETM) were MOG-IgG1 positive, but 6
samples had IgG1-MOG-Abs, with a clinical diagnosis
of bilateral ON (3/3), monophasic LETM (1/10), or
ADEM (2/11). The remainder of the cohort was dou-
ble seronegative (3 NMO, 4 recurrent ON, 3 mono-
phasic LETM, 27 MS, and 9 ADEM (figure 4). In
contrast to the Oxford cohort, in which 8/23 AQP4-
seronegative NMO patients were MOG-IgG1 posi-
tive, none of the 3 AQP4-seronegative NMO patients
from Tohoku were MOG-IgG1 positive. The majority
of the MOG-IgG1seropositive patients had a single
attack and good recovery after steroid treatment, but 2
children with ADEM relapsed (table 2).
DISCUSSION MOG-Abs have been detected using
different methods, which affects the patient groups
that are identified as seropositive. Initially, using
peptide Western blots or ELISAs, patients with MS
or viral or bacterial encephalitis were identified as
MOG seropositive. More recently, the extracellular
domain of native MOG has been used in immuno-
precipitation assays in which the majority of patients
with MS were seronegative but one-third of patients
with ADEM were seropositive. The advent of the
CBA enabled native human MOG to be expressed
on the cell surface as a target for these Abs.
Unfortunately, sera from many healthy individuals
diluted 1:20 were seropositive using this assay;
therefore, a high-titerserum cutoff of 1:160 is
used to differentiate patient cohorts from healthy
individuals. A few patients with MS, AQP4-
seropositive patients, and healthy controls are still
positive using this high-titercutoff.
30
We confirm the lack of disease specificity of the
MOG CBA at 1:20: 16% of sera sent for AQP4
Figure 4 Confirmatory cohorts to assess MOG-IgG1 assay
(A) All 81 aquaporin-4 (AQP4)- seropositive patients (blue) from the Oxford National neuromyelitis optica (NMO) service
were negative for IgG1 antibodies to myelin oligodendrocyte glycoprotein (MOG); however, 23 AQP4-seronegative patients
were identified as MOG-IgG1 seropositive (red). Of the 58 patients with NMO, 37 (63.4%) were AQP4 seropositive, 8
(13.8%) were MOG-IgG1 seropositive, and 13 (22%) were double seronegative. (B) A second cohort from Japan was
screened blinded to clinical information. None of the 49 AQP4-seropositive patients (blue) or 27 patients with multiple scle-
rosis (MS) were positive for MOG antibodies, but 6/25 patients with acute disseminated encephalomyelitis (ADEM), trans-
verse myelitis (TM), optic neuritis (ON), or AQP4-seronegative NMO were MOG antibody positive (red). CBA 5cell-based
assay.
Neurology: Neuroimmunology & Neuroinflammation 7
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
testing and nearly 50% of patients with epilepsy were
MOG positive. Similar positivity is seen in healthy
control sera (data not shown). The secondary anti-
body IgG (H 1L) binds to more than the IgG anti-
body class, which appears to affect the MOG CBA
more than CBAs in which other targets are expressed
(e.g., AQP4, GlyR). When examined by flow cytom-
etry, the low- levelbinding of healthy control sera
visualized by CBA is replicated by a specific shift in
the MOG-transfected cells when compared to the
untransfected or poorly transfected cells. Two advan-
tages of this quantitative system are that the
background binding can be quantified and a cutoff
can be generated based on healthy sera. Using 6 SDs
above the mean of a group of healthy control sera, the
IgG (H 1L) antibody gave a cutoff of 470, with the
top of the assay 10 times this cutoff value (data not
shown). Using the same control and test samples, the
anti-human IgM secondary antibody gave a cutoff of
270, with the top of the assay only 3 times the cutoff
and with very few positive samples, whereas the anti-
human IgG1 antibody cutoff was just 2.5 and the top
of the assay was 220 times this cutoff. The specificity
of the MOG-IgG1 assay was confirmed by CBA in
Table 2 Demographics, diagnoses, treatment, and response to treatment in the confirmatory cohorts tested
from Oxford and Japan
Sex, age, y Diagnosis Treatment Recovery Relapse
Oxford F, 12 ADEM IVMP 1steroid tapering Complete No
F, 6 ADEM Acyclovir 1IVIg followed by oral steroids Substantial No
M, 3 ADEM ON Steroids, IVIg, and PEX, then azathioprine
and prednisolone
Substantial Yes
M, 27 ADEM, LETM IVMP then oral steroids Partial Yes
F, 37 BON IVMP and oral steroids Substantial Yes
M, 33 BON IVMP 1steroid tapering and PEX Partial No
M, 4 BON IVMP 1steroid tapering Substantial No
F, 59 LETM IVMP 1steroid tapering Complete No
F, 34 NMO IVMP, oral steroids Substantial Yes
F, 23 NMO None Complete Yes
M, 16 NMO IVMP 1steroid tapering Partial Yes
M, 36 NMO IVMP 1steroid tapering, azathioprine Partial Yes
M, 24 NMO IVMP, oral steroids, azathioprine Partial Yes
M, 31 NMO IVMP, PEX, oral steroids Complete No
F, 34 NMO IVMP, oral steroids Substantial Yes
M, 17 NMO IVMP, oral steroids Substantial Yes
F, 14 ON IVMP, oral steroids Substantial No
F, 54 ON brain IVMP, oral steroids, MMF Partial Yes
M, 27 ON TM brain IVMP, oral steroids, interferon bPartial Yes
F, 43 RION IVMP, oral steroids, methotrexate None Yes
F, 42 RION IVMP, methotrexate, oral steroids Partial Yes
M, 8 RION IVMP, oral steroids, PEX, azathioprine Partial Yes
M, 34 RION None Partial Yes
Japan M, 28 BON IVMP 1steroid tapering Yes No
M, 70 BON IVMP 1steroid tapering Yes No
M, 37 BON IVMP 1steroid tapering Yes No
M, 50 Myelitis IVMP Yes No
M, 1.3 ADEM IVMP 1steroids Yes Yes
F, 9 ADEM IVMP 1steroids Yes Yes
Abbreviations: ADEM 5acute disseminated encephalomyelitis; BON 5bilateral ON; Brain 5changes seen on brain MRI;
IVIg 5IV immunoglobulin; IVMP 5IV methylprednisolone; LETM 5longitudinally extensive TM; MMF 5mycophenolate
mofetil; NMO 5neuromyelitis optica; ON 5optic neuritis; PEX 5plasma exchange; RION 5relapsing inflammatory ON;
TM 5transverse myelitis.
None of the patients were aquaporin-4 antibody positive.
8Neurology: Neuroimmunology & Neuroinflammation
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
which 49 patients with MS, 13 healthy control sera,
and 14 AQP4-seropositive serum samples were all neg-
ative at a dilution of 1:20, whereas 65 of the 118
samples that were positive using IgG (H 1L) second-
ary antibody remained positive using the IgG1-specific
antibody. None of the MOG-IgG1positive patients
with an available clinical diagnosis had MS, suggesting
that this assay may be valuable to help distinguish
patients with MS from those with ADEM or AQP4-
Abnegative NMOSD. Furthermore, 60/65 (92%)
IgG1-positive samples had IgG (H 1L) endpoint
titers $1:200, indicating that the IgG1 assay identifies
not only the patients above cutoff with the anti-IgG
(H 1L) but also disease-relevant Abs that fall below
this cutoff. These findings are consistent with a previ-
ous report that high-titer MOG-Abs were exclusively
of the IgG1 isotype.
30,36,38
Detection of IgM by CBA at a serum dilution of
1:20 did not distinguish different patient groups from
healthy controls, limiting its diagnostic use. The flow
cytometry data show that something in healthy and
patient sera binds to the surface of MOG-
transfected cells at low levels and is detected by
anti-human IgG (H 1L) or IgM antibodies. This
is consistent with other studies reporting high levels
of MOG-IgM-Abs using immunoblot or ELISA
1,18,39
and might be explained by the observation that MOG
binds to components of the immune system such as
C1q or DC-SIGN.
40,41
The SL-MOG assay was previously shown to be
negative in patients with MS and healthy controls,
26,27
but here it only identified 32.3% of the IgG1 FL-
MOG-Abspositive samples (see table 1). As the extra-
cellular domains are identical in the 2 constructs, the
differences in assay sensitivity may be due to expression
level on the surface, glycosylation, or ability to multi-
merize. Two of the 21 SL-MOGpositive patients
were IgM positive only. The low sensitivity of the
SL-MOG assay limits its use in clinical practice.
Although this work is retrospective with limited
clinical descriptions of the patients, it does suggest
that the anti-IgG1/FL-MOG antibody assay can be
useful in identifying MOG-Abs in patients with
demyelinating diseases who are unlikely to have
MS. Prospective studies with longer-term follow-up
are needed to establish the clinical utility of this assay.
AUTHOR CONTRIBUTIONS
Drafting/revising the manuscript: all authors. Study concept or design:
P.W. Analysis or interpretation of data: P.W., A.V. Contribution of vital
reagents/tools/patients: K.C.O., M.R., D.K.S., M.J., G.T., J.R., T.T.,
T.M., I.N., K.F. Acquisition of data: P.W., M.W., D.K.S., A.V. Statis-
tical analysis: P.W., A.V. Study supervision or coordination: P.W.
Obtaining funding: J.P., A.V.
STUDY FUNDING
NHS National Specialised Services for Neuromyelitis Optica (P.W.,
M.W., J.P., M.I.L., A.V.), the Oxford Biomedical Research Centre
(M.I.L., P.W., A.V.), the ERA-net E-rare EDEN project (P.W., M.R.,
A.V.), KAKENHI (22229008) of The Ministry of Education, Culture,
Sports, Science and Technology (MEXT) of Japan, and the Health and
Labour Sciences Research Grant on Intractable Diseases (Neuroimmuno-
logical Diseases) from the Ministry of Health, Labour and Welfare of
Japan (D.K.S., T.T., T.M., I.N., K.F.). M.J. received research fellowship
from the Polish Ministry of Science and Higher Education program
Moblinosc Plus (1070/MOB/2013/0).
DISCLOSURE
P. Waters has received speaker honoraria from Biogen Idec Japan and
Euroimmun AG; has been a review editor for Frontiers in Molecular
Innate Immunity; holds a patent for assays for the detection and anti-
bodies to lGi1, Caspr2, and tag-1; and has submitted a patent for
GABARR. M. Woodhall reports no disclosures. K.C. OConnor has
received travel funding and speaker honorarium from ACTRIMS-
CMSC and has received research support from NIH and Nancy Davis
Foundation for Multiple Sclerosis. M. Riendl is an academic editor for
PLOS ONE; is on the editorial board for Current Medicinal Chemistry
and Autoimmune Diseases; and has received research support from
Austrian Science Fund, Austrian Federal Ministry of Science, and Jubi-
laeumsfonds of the Austrian National Bank. M. Reindl and Medical
University of Innsbruck receive payments for antibody assays (AQP4
and antineuronal antibodies) and for AQP4 antibody validation experi-
ments organized by Euroimmun. B. Lang is a member of the Medical
Committee of Mayaware; holds a patent for use of LGI1 as an antigen in
detection of autoantibodies and use of GABAa gamma subunit in detec-
tion of autoantibodies; receives research support from Epilepsy Research
UK; and received royalties for use of LGI1 as an antigen in detection of
autoantibodies. Her department receives payment for running diagnostic
assays for a range of autoantibodies. D.K. Sato has received research
support from Ministry of Education, Culture, Sports, Science & Tech-
nology (MEXT) in Japan, Japanese Government Scholarship Program,
and Ichiro Kanehara Foundation. M. Jurynczyk has received research
support from the Polish Ministry of Science and Higher Education.
G. Tackley and J. Rocha report no disclosures. T. Takahashi has received
speaker honoraria from Biogen Idec and Cosmic Corporation. T. Misu
has received speaker honoraria from Bayer Schering Pharma, Biogen Idec,
and Mitsubishi Pharma; has received research support from Bayer
Schering Pharma, Biogen Idec Japan, Asahi Kasei Kuraray Medical
Co., The Chemo-Sero-Therapeutic Research Institute, Teva Pharmaceu-
tical K.K., Mitsubishi Tanabe Pharma Corporation, and Teijin Pharma;
and has received Grants-in-Aid for Scientific Research from the Ministry
of Education, Science and Technology and the Ministry of Health, Labor
and Welfare of Japan. I. Nakashima has received travel funding/and or
speaker honoraria from Biogen Idec Japan, Tanabe Mitsubishi, and
Novartis Pharma; is an editorial board member for Multiple Sclerosis
International; and received research support from LSI Medience Corpo-
ration. J. Palace has been a UK advisory board participant for Merck
Serono, Bayer Schering Pharma, Biogen Idec, Teva Pharmaceutical
Industries Ltd, Novartis Pharmaceuticals UK Ltd, Sanofi-Aventis, and
Alexion; has received travel funding and/or speaker honoraria from
ECTRIMS, Merck Serono, Novartis, Biogen Idec, Bayer Schering, and
ISIS Innovation Limited, a wholly owned subsidiary of the University of
Oxford; has filed a patent application to protect for the use of metab-
olomics as a method to diagnose and stage disease in multiple sclerosis;
has consulted for Ono Pharmaceuticals Ltd, Chigai Pharma Ltd, CI
Consulting, Biogen Idec, and GlaxoSmithKline; and has received research
support from Bayer Schering, Merck Serono, Novartis, Department of
Health, MS Society, and UK Guthy Jackson Foundation. K. Fujihara
serves on scientific advisory boards for Bayer Schering Pharma, Biogen
Idec, Mitsubishi Tanabe Pharma Corporation, Novartis Pharma, Chugai
Pharmaceutical, Ono Pharmaceutical, Nihon Pharmaceutical, Merck
Serono, Alexion Pharmaceuticals, Medimmune, and Medical Review;
has received funding for travel and speaker honoraria from Bayer Schering
Pharma, Biogen Idec, Eisai Inc., Mitsubishi Tanabe Pharma Corporation,
Novartis Pharma, Astellas Pharma Inc., Takeda Pharmaceutical Company
Limited, Asahi Kasei Medical Co., Daiichi Sankyo, Nihon Pharmaceuti-
cal, and Cosmic Corporation; is on the editorial board for Clinical and
Experimental Neuroimmunology; is an advisory board member of Sri
Lanka Journal of Neurology; and has received research support from Bayer
Neurology: Neuroimmunology & Neuroinflammation 9
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
Schering Pharma, Biogen Idec Japan, Asahi Kasei Medical, The Chemo-
Sero-Therapeutic Research Institute, Teva Pharmaceutical, Mitsubishi
Tanabe Pharma, Teijin Pharma, Chugai Pharmaceutical, Ono Pharma-
ceutical, Nihon Pharmaceutical, Genzyme Japan, Ministry of Education,
Science and Technology of Japan, and Ministry of Health, Welfare and
Labor of Japan. M.I. Leite has received travel funding and/or speaker
honoraria from Biogen Idec and has received research support from NHS
specialised commissioning group for neuromyelitis optica, UK and
NIHR Oxford Biomedical Research Centre. A. Vincent has received
travel funding and speaker honoraria from Baxter International Inc and
Biogen Inc; is on the editorial board for Neurology; was an associate editor
for Brain; holds a patent with Oxford University for LGI1/CASPR2
antibodies, licensed to Euroimmun AG, and for GABAAR antibodies,
in negotiation with Euroimmun AG; received royalties from Athena
Diagnostics, Euroimmun AG, Blackwell Publishing, and Mac Keith
Press; has consulted with Athena Diagnostics; and has received research
support from NIHR. Go to Neurology.org/nn for full disclosure forms.
Received December 13, 2014. Accepted in final form January 20, 2015.
REFERENCES
1. Berger T, Rubner P, Schautzer F, et al. Antimyelin antibod-
iesasapredictorofclinicallydefinite multiple sclerosis after a
first demyelinating event. N Engl J Med 2003;349:139145.
2. Klawiter EC, Piccio L, Lyons JA, et al. Elevated intrathecal
myelin oligodendrocyte glycoprotein antibodies in multi-
ple sclerosis. Arch Neurol 2010;67:11021108.
3. Zhou D, Srivastava R, Nessler S, et al. Identification of a
pathogenic antibody response to native myelin oligoden-
drocyte glycoprotein in multiple sclerosis. Proc Natl Acad
Sci USA 2006;103:1905719062.
4. Zadro I, Brinar V, Horvat G, Brinar M. Clinical relevance
of antibodies against myelin oligodendrocyte glycoprotein
in different clinical types of multiple sclerosis. Clin Neurol
Neurosurg 2007;109:2326.
5. Lalive PH, Menge T, Delarasse C, et al. Antibodies to
native myelin oligodendrocyte glycoprotein are serologic
markers of early inflammation in multiple sclerosis. Proc
Natl Acad Sci USA 2006;103:22802285.
6. Gaertner S, de Graaf KL, Greve B, Weissert R. Antibodies
against glycosylated native MOG are elevated in patients
with multiple sclerosis. Neurology 2004;63:23812383.
7. Markovic M, Trajkovic V, Drulovic J, et al. Antibodies
against myelin oligodendrocyte glycoprotein in the cere-
brospinal fluid of multiple sclerosis patients. J Neurol Sci
2003;211:6773.
8. Reindl M, Linington C, Brehm U, et al. Antibodies
against the myelin oligodendrocyte glycoprotein and the
myelin basic protein in multiple sclerosis and other neu-
rological diseases: a comparative study. Brain 1999;122:
20472056.
9. Bernard CC, Johns TG, Slavin A, et al. Myelin oligoden-
drocyte glycoprotein: a novel candidate autoantigen in
multiple sclerosis. J Mol Med (Berl) 1997;75:7788.
10. Xiao BG, Linington C, Link H. Antibodies to myelin-
oligodendrocyte glycoprotein in cerebrospinal fluid
from patients with multiple sclerosis and controls.
J Neuroimmunol 1991;31:9196.
11. Haase CG, Schmidt S. Detection of brain-specific autoan-
tibodies to myelin oligodendrocyte glycoprotein, S100beta
and myelin basic protein in patients with Devics neuro-
myelitis optica. Neurosci Lett 2001;307:131133.
12. Pittock SJ, Reindl M, Achenbach S, et al. Myelin oligo-
dendrocyte glycoprotein antibodies in pathologically
proven multiple sclerosis: frequency, stability and clinico-
pathologic correlations. Mult Scler 2007;13:716.
13. Karni A, Bakimer-Kleiner R, Abramsky O, Ben-Nun A.
Elevated levels of antibody to myelin oligodendrocyte gly-
coprotein is not specific for patients with multiple sclero-
sis. Arch Neurol 1999;56:311315.
14. Lim ET, Berger T, Reindl M, et al. Anti-myelin antibodies
do not allow earlier diagnosis of multiple sclerosis. Mult
Scler 2005;11:492494.
15. Wang H, Munger KL, Reindl M, et al. Myelin oligoden-
drocyte glycoprotein antibodies and multiple sclerosis in
healthy young adults. Neurology 2008;71:11421146.
16. Pelayo R, Tintoré M, Montalban X, et al. Anti-myelin
antibodies with no progression to multiple sclerosis. N
Engl J Med 2007;356:426428.
17. Kuhle J, Pohl C, Mehling M, et al. Lack of association
between anti-myelin antibodies and progression to multi-
ple sclerosis. N Engl J Med 2007;356:371378.
18. Lampasona V, Franciotta D, Furlan R, et al. Similar low
frequency of anti-MOG IgG and IgM in MS patients and
healthy subjects. Neurology 2004;62:20922094.
19. DaleRC,TantsisEM,MerhebV,etal.AntibodiestoMOG
have a demyelination phenotype and affect oligodendrocyte
cytoskeleton. Neurol Neuroimmunol Neuroinflamm 2014;
1:e12; doi: 10.1212/NXI.0000000000000012.
20. Ikeda K, Kiyota N, Kuroda H, et al. Severe demyelination
but no astrocytopathy in clinically definite neuromyelitis
optica with anti-myelin-oligodendrocyte glycoprotein anti-
body. Mult Scler Epub 2014 Sep 25.
21. Marta CB, Bansal R, Pfeiffer SE. Microglial Fc receptors
mediate physiological changes resulting from antibody
cross-linking of myelin oligodendrocyte glycoprotein.
JNeuroimmunol 2008;196:3540.
22. vonBüdingenHC,HauserSL,FuhrmannA,etal.Molecular
characterization of antibody specificities against myelin/oligo-
dendrocyte glycoprotein in autoimmune demyelination. Proc
Natl Acad Sci USA 2002;99:82078212.
23. Marta CB, Oliver AR, Sweet RA, Pfeiffer SE, Ruddle NH.
Pathogenic myelin oligodendrocyte glycoprotein antibod-
ies recognize glycosylated epitopes and perturb oligoden-
drocyte physiology. Proc Natl Acad Sci USA 2005;102:
1399213997.
24. OConnor KC, McLaughlin KA, De Jager PL, et al. Self-
antigen tetramers discriminate between myelin autoanti-
bodies to native or denatured protein. Nat Med 2007;13:
211217.
25. Kitley J, Woodhall M, Waters P, et al. Myelin-oligoden-
drocyte glycoprotein antibodies in adults with a neuromy-
elitis optica phenotype. Neurology 2012;79:12731277.
26. Kitley J, Waters P, Woodhall M, et al. Neuromyelitis
optica spectrum disorders with aquaporin-4 and myelin-
oligodendrocyte glycoprotein antibodies: a comparative
study. JAMA Neurol 2014;71:276283.
27. Sato DK, Callegaro D, Lana-Peixoto MA, et al. Distinc-
tion between MOG antibody-positive and AQP4
antibody-positive NMO spectrum disorders. Neurology
2014;82:474481.
28. Reindl M, Di Pauli F, Rostasy K, Berger T. The spectrum
of MOG autoantibody-associated demyelinating diseases.
Nat Rev Neurol 2013;9:455461.
29. Rostásy K, Mader S, Hennes EM, et al. Persisting myelin
oligodendrocyte glycoprotein antibodies in aquaporin-4
antibody negative pediatric neuromyelitis optica. Mult
Scler 2013;19:10521059.
30. Mader S, Gredler V, Schanda K, et al. Complement
activating antibodies to myelin oligodendrocyte
10 Neurology: Neuroimmunology & Neuroinflammation
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
glycoprotein in neuromyelitis optica and related disor-
ders. J Neuroinflammation 2011;8:184.
31. Ramanathan S, Reddel SW, Henderson A, et al. Antibodies
to myelin oligodendrocyte glycoprotein in bilateral and recur-
rent optic neuritis. Neurol Neuroimmunol Neuroinflamm
2014;1:e40; doi: 10.1212/NXI.0000000000000040.
32. Mayer MC, Breithaupt C, Reindl M, et al. Distinction
and temporal stability of conformational epitopes on mye-
lin oligodendrocyte glycoprotein recognized by patients
with different inflammatory central nervous system dis-
eases. J Immunol 2013;191:35943604.
33. Rostasy K, Mader S, Schanda K, et al. Anti-myelin oligo-
dendrocyte glycoprotein antibodies in pediatric patients
with optic neuritis. Arch Neurol 2012;69:752756.
34. Kezuka T, Usui Y, Yamakawa N, et al. Relationship
between NMO-antibody and anti-MOG antibody in
optic neuritis. J Neuroophthalmol 2012;32:107110.
35. Brilot F, Dale RC, Selter RC, et al. Antibodies to native
myelin oligodendrocyte glycoprotein in children with
inflammatory demyelinating central nervous system dis-
ease. Ann Neurol 2009;66:833842.
36. Probstel AK, Dornmair K, Bittner R, et al. Antibodies to
MOG are transient in childhood acute disseminated
encephalomyelitis. Neurology 2011;77:580588.
37. Waters PJ, McKeon A, Leite MI, et al. Serologic diagnosis
of NMO: a multicenter comparison of aquaporin-4-IgG
assays. Neurology 2012;78:665671.
38. McLaughlin KA, Chitnis T, Newcombe J, et al. Age-
dependent B cell autoimmunity to a myelin surface anti-
gen in pediatric multiple sclerosis. J Immunol 2009;183:
40674076.
39. Egg R, Reindl M, Deisenhammer F, Linington C,
Berger T. Anti-MOG and anti-MBP antibody subclasses
in multiple sclerosis. Mult Scler 2001;7:285289.
40. Johns TG, Bernard CC. Binding of complement compo-
nent Clq to myelin oligodendrocyte glycoprotein: a novel
mechanism for regulating CNS inflammation. Mol Immu-
nol 1997;34:3338.
41. García-Vallejo JJ, Ilarregui JM, Kalay H, et al. CNS mye-
lin induces regulatory functions of DC-SIGN-expressing,
antigen-presenting cells via cognate interaction with
MOG. J Exp Med 2014;211:14651483.
Neurology: Neuroimmunology & Neuroinflammation 11
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
DOI 10.1212/NXI.0000000000000089
2015;2; Neurol Neuroimmunol Neuroinflamm
Patrick Waters, Mark Woodhall, Kevin C. O'Connor, et al.
MOG cell-based assay detects non-MS patients with inflammatory neurologic disease
This information is current as of March 19, 2015
Services
Updated Information & http://nn.neurology.org/content/2/3/e89.full.html
including high resolution figures, can be found at:
Supplementary Material http://nn.neurology.org/content/suppl/2015/03/19/2.3.e89.DC1.html
Supplementary material can be found at:
References http://nn.neurology.org/content/2/3/e89.full.html##ref-list-1
This article cites 40 articles, 12 of which you can access for free at:
Subspecialty Collections
http://nn.neurology.org//cgi/collection/multiple_sclerosis
Multiple sclerosis http://nn.neurology.org//cgi/collection/devics_syndrome
Devic's syndrome
myelitis
http://nn.neurology.org//cgi/collection/acute_disseminated_encephalo
Acute disseminated encephalomyelitis
following collection(s):
This article, along with others on similar topics, appears in the
Permissions & Licensing
http://nn.neurology.org/misc/about.xhtml#permissions
its entirety can be found online at:
Information about reproducing this article in parts (figures,tables) or in
Reprints http://nn.neurology.org/misc/addir.xhtml#reprintsus
Information about ordering reprints can be found online:
2015 American Academy of Neurology. All rights reserved. Online ISSN: 2332-7812.
Published since April 2014, it is an open-access, online-only, continuous publication journal. Copyright ©
is an official journal of the American Academy of Neurology.Neurol Neuroimmunol Neuroinflamm

Supplementary resource (1)

... MOG-IgG3 antibodies have previously been reported in three patients with negative results for total MOG-IgG, as determined by a non-subclass-specific assay, in one study [24]; in three out of 15 patients with discordant results from three different non-subclass-specific CBAs in a second study [6]; and, at high titre (1:3200), in a single patient with ON and myelitis with patchy cord lesions [32]. However, in contrast to the present study, no unequivocally MOG-IgGpositive earlier or follow-up samples from the same patients that would corroborate the specificity of those findings were available. ...
... However, in contrast to the present study, no unequivocally MOG-IgGpositive earlier or follow-up samples from the same patients that would corroborate the specificity of those findings were available. Moreover, MOG-IgG3 titres were below the cutoff commonly applied in that type of CBA in one of these studies [24] and no healthy controls were included in any of the three previous studies [6,24,32]. ...
Article
Full-text available
Background Myelin oligodendrocyte glycoprotein antibody-associated encephalomyelitis (MOG-EM; also termed MOG antibody-associated disease, MOGAD) is the most important differential diagnosis of both multiple sclerosis and neuromyelitis optica spectrum disorders. A recent proposal for new diagnostic criteria for MOG-EM/MOGAD explicitly recommends the use of immunoglobulin G subclass 1 (IgG1)- or IgG crystallizable fragment (Fc) region-specific assays and allows the use of heavy-and-light-chain-(H+L) specific assays for detecting MOG-IgG. By contrast, the utility of MOG-IgG3-specific testing has not been systematically evaluated. Objective To assess whether the use of MOG-IgG3-specific testing can improve the sensitivity of MOG-IgG testing. Methods Re-testing of 22 patients with a definite diagnosis of MOG-EM/MOGAD and clearly positive MOG-IgG status initially but negative or equivocal results in H+L- or Fc-specific routine assays later in the disease course (i.e. patients with spontaneous or treatment-driven seroreversion). Results In accordance with previous studies that had used MOG-IgG1-specific assays, IgG subclass-specific testing yielded a higher sensitivity than testing by non-subclass-specific assays. Using subclass-specific secondary antibodies, 26/27 supposedly seroreverted samples were still clearly positive for MOG-IgG, with MOG-IgG1 being the most frequently detected subclass (25/27 [93%] samples). However, also MOG-IgG3 was detected in 14/27 (52%) samples (from 12/22 [55%] patients). Most strikingly, MOG-IgG3 was the predominant subclass in 8/27 (30%) samples (from 7/22 [32%] patients), with no unequivocal MOG-IgG1 signal in 2 and only a very weak concomitant MOG-IgG1 signal in the other six samples. By contrast, no significant MOG-IgG3 reactivity was seen in 60 control samples (from 42 healthy individuals and 18 patients with MS). Of note, MOG-IgG3 was also detected in the only patient in our cohort previously diagnosed with MOG-IgA ⁺ /IgG – MOG-EM/MOGAD, a recently described new disease subvariant. MOG-IgA and MOG-IgM were negative in all other patients tested. Conclusions In some patients with MOG-EM/MOGAD, MOG-IgG is either exclusively or predominantly MOG-IgG3. Thus, the use of IgG1-specific assays might only partly overcome the current limitations of MOG-IgG testing and—just like H+L- and Fcγ-specific testing—might overlook some genuinely seropositive patients. This would have potentially significant consequences for the management of patients with MOG-EM/MOGAD. Given that IgG3 chiefly detects proteins and is a strong activator of complement and other effector mechanisms, MOG-IgG3 may be involved in the immunopathogenesis of MOG-EM/MOGAD. Studies on the frequency and dynamics as well as the clinical and therapeutic significance of MOG-IgG3 seropositivity are warranted.
... Earlier studies reported higher proportions of MOG positivity among patients with MS (up to 21%), other inflammatory neurological diseases, and also healthy controls [4,63]; whereas these rates were found to be about 1% in MS and none in healthy controls in more recent studies [64,65]. This appears to be due to the differences in antibody detection methodologies. ...
... Since only those anti-MOG antibodies that direct against native MOG in their conformational state, rather than linear or denatured MOG, are pathogenic [61], live CBAs have been considered the gold standard in the diagnosis of MOGAD [3]. There is an acceptable concordance between the fixed and the live CBAs, and therefore the fixed CBAs are considered to be a reasonable alternative when live CBA is not available [65]. However, the positive predictive value of live CBAs is higher when compared to that of fixed-CBAs [66,67]. ...
Article
Full-text available
Over the last two decades, immunoglobulin G (IgG) antibodies against myelin oligodendrocyte glycoprotein (MOG), previously thought to be a biomarker of multiple sclerosis (MS), have been shown to cause a distinct disease called MOG antibody-associated disease (MOGAD). MOGAD accounts for approximately one-third of all demyelinating syndromes in children and is the second most common central nervous system (CNS) demyelinating disease after MS. The diagnosis is made by detecting anti-MOG IgG antibodies against the natural MOG antigen, in the presence of compatible clinical and neuroradiological features. However, due to controversies in the methodologies for detecting anti-MOG antibodies and their diagnostic cutoff values, as well as the expanding clinical spectrum, accurate diagnosis may be challenging, at least in a subset of patients. Clinical presentations of MOGAD vary by age; the highest rates are seen in acute disseminated encephalomyelitis in younger children and optic neuritis, myelitis, or brainstem symptoms in older children. Although it was previously thought to be a milder demyelinating disorder and to have a monophasic course in the majority of patients, recent studies have shown that relapses occur in about half of the patients and sequelae develop in a significant proportion of them, especially in those with persistently high antibody titers, leukodystrophy-like magnetic resonance imaging (MRI) lesions, and spinal cord involvement. However, due to the monophasic course in about half of the patients, long-term treatment is not recommended after the first clinical episode but is recommended for patients who experience relapse. Accurate and early diagnosis of MOGAD is essential for proper management and better outcome. This review covers the challenges in the diagnosis of MOGAD in children.
Article
Full-text available
Introduction We aimed to assess the frequency, duration, and severity of area postrema syndrome (APS) during follow-up in neuromyelitis optica spectrum disorder (NMOSD) patients, as well as its association with inflammatory activity and prognostic factors of APS severity in a real-world setting. Methods We conducted a retrospective study on a cohort of Latin American (LATAM) NMOSD patients who had experienced APS during their follow-up. Patients from Mexico, Peru, Brazil, Colombia, Panama, Chile and Argentina patients who met 2015 NMOSD criteria were included. We evaluated data on symptom type (nausea, vomiting and/or hiccups), frequency, duration, severity (measured by APS severity scale), association with other NMOSD core relapses, and acute treatments (symptomatic and immunotherapy or plasmapheresis). Logistic regression was conducted to evaluate factors associated with APS severity (vs. mild-moderate). Results Out of 631 NMOSD patients, 116 (18.3%) developed APS during their follow-up. The most common APS phenotype was severe. Inflammatory activity (i.e., relapses) significantly decreased after the onset of APS. Half of the patients experienced isolated APS with a median duration of 10 days, and the most frequently used acute treatment was IV steroids. All three symptoms were present in 44.6% of the patients. APS symptoms resolved following immunotherapy. Logistic regression did not identify independent factors associated with the severity of APS. Conclusions Our findings indicate that 18.3% of NMOSD patients developed APS during the follow-up period, with most patients fulfilling criteria for severe APS. The inflammatory activity decreased after the onset of APS compared to the previous year.
Article
Full-text available
Recent reports indicated that myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) might be a rare complication after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. It is unclear whether this is an unspecific sequel of infection or vaccination or caused by possible immunological cross-reactivity of SARS-CoV-2 proteins and MOG. The aim of this study was therefore to elucidate whether there is an immunological cross-reactivity between SARS-CoV-2 spike (S) or nucleocapsid (N) proteins and MOG and to explore the relation of antibody responses against MOG and SARS-CoV-2 and other coronaviruses. We analysed serum samples from patients with SARS-CoV-2 infection and neurological symptoms with (MOGAD, n = 12) or without MOG-antibodies (n = 10); SARS-CoV-2 infection without neurological symptoms (n = 32); vaccinated patients with no history of SARS-CoV-2 infection and neurological symptoms with (MOGAD, n = 10) or without MOG-antibodies (n = 9); and SARS-CoV-2 negative/naïve unvaccinated patients with neurological symptoms with (MOGAD, n = 47) or without MOG-antibodies (n = 20). All samples were analysed for serum antibody responses to MOG, SARS-CoV-2, and other common coronaviruses (CoV-229E, CoV-HKU1, CoV-NL63 and CoV-OC43). Based on sample amount and antibody titres, 21 samples were selected for analysis of antibody cross-reactivity between MOG and SARS-CoV-2 S and N proteins using affinity purification and pre-absorption. Whereas we found no association of 2 immunoglobulin G (IgG) and A (IgA) MOG antibodies with coronavirus antibodies, infections with SARS-CoV-2 correlated with an increased immunoglobulin M (IgM) MOG antibody response. Purified antibodies showed no cross-reactivity between SARS-CoV-2 S protein and MOG. However, one sample of a patient with MOGAD following SARS-CoV-2 infection showed a clear IgG antibody cross-reactivity to SARS-CoV-2 N protein and MOG. This patient was also seropositive for other coronaviruses and showed immunological cross-reactivity of SARS-CoV-2 and CoV-229E N proteins. Overall, our results indicate that an IgG antibody cross-reactivity between MOG and SARS-CoV-2 proteins is rare. The presence of increased MOG-IgM antibodies after SARS-CoV-2 infection may either be a consequence of a previous infection with other coronaviruses or arise as an unspecific sequel after viral infection. Furthermore, our data indicate that MOG-IgA and particularly MOG-IgM antibodies are a rather unspecific sequel of viral infections. Finally, our findings do not support a causative role of coronavirus infections for the presence of MOG-IgG antibodies.
Article
This study was designed to analyze clinical and radiographic features of adult patients coexisting with NMDAR-IgG and MOG-IgG. Eleven adult patients coexisting with NMDAR-IgG and MOG-IgG were collected from Xiangya Hospital, Central South University, between June 2017 and December 2021. Fifty-five patients with anti-NMDAR encephalitis and 49 with MOG-AD were served as controls. Onset age was 27 (IQR 20–34) years old. Seizures and psychotic symptoms were prominent symptoms. Ten of eleven patients presented abnormal T2/FLAIR hyperintensity, mainly involving the cortex, brainstem, and optic nerve. Compared with the NMDAR IgG ( +)/MOG IgG ( −) group, the NMDAR IgG ( +)/MOG IgG ( +) group showed more ataxia symptoms (27.3% vs. 3.6%, P = 0.037), while more T2/FLAIR hyperintensity lesions were found in the brainstem (54.5% vs. 7.3%, P < 0.001) and optic nerve (27.3% vs. 1.8%, P = 0.011) with more abnormal MRI patterns (90.9% vs. 41.8%, P = 0.003). In comparison with the NMDAR IgG ( −)/MOG IgG ( +) group, the NMDAR IgG ( +)/MOG IgG ( +) group had more seizures (72.7% vs. 24.5%, P = 0.007) and mental symptoms (45.5% vs. 0, P < 0.001). The NMDAR IgG ( +)/MOG IgG ( +) group tended to be treated with corticosteroids alone (63.6% vs. 20.0%, P = 0.009), more prone to recur (36.5% vs. 7.3%, P = 0.028) and lower mRS score (P = 0.036) at the last follow-up than pure anti-NMDAR encephalitis. The symptoms of the NMDAR IgG ( +)/MOG IgG ( +) group were more similar to anti-NMDAR encephalitis, while MRI patterns overlapped more with MOG-AD. Detecting both NMDAR-IgG and MOG-IgG maybe warranted in patients with atypical encephalitis symptoms and demyelinating lesions in infratentorial regions.
Article
Full-text available
Background Myelin oligodendrocyte glycoprotein antibody (MOG) immunoglobulin G (IgG)-associated disease (MOGAD) has clinical and pathophysiological features that are similar to but distinct from those of aquaporin-4 antibody (AQP4-IgG)-positive neuromyelitis optica spectrum disorders (AQP4-NMOSD). MOG-IgG and AQP4-IgG, mostly of the IgG1 subtype, can both activate the complement system. Therefore, we investigated whether the levels of serum complement components, regulators, and activation products differ between MOGAD and AQP4-NMOSD, and if complement analytes can be utilized to differentiate between these diseases. Methods The sera of patients with MOGAD (from during an attack and remission; N =19 and N =9, respectively) and AQP4-NMOSD ( N =35 and N =17), and healthy controls ( N =38) were analyzed for C1q-binding circulating immune complex (CIC-C1q), C1 inhibitor (C1-INH), factor H (FH), C3, iC3b, and soluble terminal complement complex (sC5b-9). Results In attack samples, the levels of C1-INH, FH, and iC3b were higher in the MOGAD group than in the NMOSD group (all, p <0.001), while the level of sC5b-9 was increased only in the NMOSD group. In MOGAD, there were no differences in the concentrations of complement analytes based on disease status. However, within AQP4-NMOSD, remission samples indicated a higher C1-INH level than attack samples (p=0.003). Notably, AQP4-NMOSD patients on medications during attack showed lower levels of iC3b ( p <0.001) and higher levels of C3 ( p =0.008), C1-INH ( p =0.004), and sC5b-9 ( p <0.001) compared to those not on medication. Among patients not on medication at the time of attack sampling, serum MOG-IgG cell-based assay (CBA) score had a positive correlation with iC3b and C1-INH levels (rho=0.764 and p =0.010, and rho=0.629 and p =0.049, respectively), and AQP4-IgG CBA score had a positive correlation with C1-INH level (rho=0.836, p =0.003). Conclusions This study indicates a higher prominence of complement pathway activation and subsequent C3 degradation in MOGAD compared to AQP4-NMOSD. On the other hand, the production of terminal complement complexes (TCC) was found to be more substantial in AQP4-NMOSD than in MOGAD. These findings suggest a strong regulation of the complement system, implying its potential involvement in the pathogenesis of MOGAD through mechanisms that extend beyond TCC formation.
Article
Full-text available
Purpose Optic neuritis (ON) is a relatively common ophthalmic disease that has recently received renewed attention owing to immunological breakthroughs. We studied the profile of patients with ON with special reference to antibody-mediated ON and the challenges faced in its management. Methods Case records of patients with ON presenting to a tertiary eye-care center in South India were analyzed. Data on demographics, presenting visual acuity (VA), clinical features, seropositivity for aquaporin-4 immunoglobulin G (AQP4-IgG) and myelin oligodendrocyte glycoprotein immunoglobulin G (MOG-IgG), details of magnetic resonance imaging (MRI) of orbits and brain, and treatment were collected. Results Among 138 cases with acute ON, male: female ratio was 1:2. Isolated ON was present in 41.3% of cases. Antibody testing of sera was performed in 68 patients only due to financial limitations. Among these, 48.5% were MOG-IgG-seropositive, 11.76% were AQP4-IgG-seropositive, and 30.88% samples were double seronegative. Other causes included multiple sclerosis (n = 4), lactational ON (n = 4), tuberculosis (n = 2), invasive perineuritis (n = 2), COVID-19 vaccination (n = 2), and COVID-19 (n = 1). The mean presenting best corrected visual acuity (BCVA) was 1.31 ± 1.16 logMAR (logarithm of the minimum angle of resolution). The mean BCVA at 3 months was 0.167 ± 0.46 logMAR. Only initial VA ≤ ‘Counting fingers’ (CF) had a significant association with the visual outcome for final VA worse than CF. The steep cost of investigations and treatment posed challenges for many patients in the management of ON. Conclusion MOG-IgG-associated ON is common in India. Unfortunately, financial constraints delay the diagnosis and timely management of ON, adversely affecting the outcome.
Article
Background and objectives: We aimed to evaluate the mortality of patients with AQP4 antibody-seropositive (AQP4-Ab+) neuromyelitis optica spectrum disorder (NMOSD) in Denmark compared with that in the general population. Methods: We identified patients with AQP4-Ab+ NMOSD fulfilling the 2015 International Panel for Neuromyelitis Optica Diagnosis (IPND) criteria from multiple sources (laboratories and the Danish Multiple Sclerosis Registry). We obtained detailed information about patients from hospital records and about the general population matched on age, sex, and calendar year from Statistics Denmark. We calculated standardized mortality ratio (SMR), excess number of deaths per 1,000 person-years (EDR), and life expectancies compared with those of the matched general population. We examined predictive factors of mortality and the cause of death. Results: Of 66 patients with AQP4-Ab+ NMOSD between 2008 and 2020, 15 died. Overall, the SMR was 2.54 (95% CI 1.47-4.09), and the EDR was 16.8 (95% CI 4.6-34.3). The median life expectancy for patients with AQP4-Ab+ NMOSD was 64.08 years (95% CI 53.02-83.9), compared with 83.07 years for the general population. Risk of death over time was increased in the patient population with a hazard ratio (HR) of 2.22 (1.34-3.68; p = 0.002). The cause of death was directly related to NMOSD in 93% of the cases. The age at disease onset was an independent predictor of death (HR 1.042; 95% CI 1.006-1.079; p = 0.02). Discussion: AQP4-Ab+ NMOSD is associated with increased mortality and shorter life expectancy compared with that in the general population, underlining the need for highly effective treatment approaches.
Article
Full-text available
Myelin oligodendrocyte glycoprotein (MOG) is a myelin-specific protein restricted to the central nervous system (CNS). While MOG is considered a putative autoantigen in MS, its function(s) in myelin is unknown. As CNS myelin is able to activate the classical complement pathway, it must contain a Clq-binding/activating protein but the identity of this protein has not been reported. The data in this paper clearly demonstrate that MOG specifically binds Clq in a dose-dependent and saturating manner. This calcium-dependent interaction is mediated by the extracellular immunoglobulin-like domain of MOG. This MOG domain contains an amino acid motif similar to the core Clq-binding sequence previously identified in IgG antibodies. Purified MOG also inhibited the antibody-dependent lysis of RBC by complement. Taken together, these results demonstrate that MOG binds Clq near the IgG binding site and may be the protein responsible for complement activation in myelin. This direct interaction between a myelin-specific protein and Clq has significant implications for CNS inflammation and could be particularly important in demyelinating diseases such as multiple sclerosis.
Article
Full-text available
Objective: We examined a cohort of adults with aquaporin-4 (AQP4) antibody–negative neuromyelitis optica/neuromyelitis optica spectrum disorder (NMO/NMOSD) for antibodies to myelin oligodendrocyte glycoprotein (MOG). Methods: We performed a flow cytometry cell-based assay using live human lentivirus–transduced cells expressing full-length surface MOG. Serum was tested in 23 AQP4 antibody–negative NMO/NMOSD patients with bilateral and/or recurrent optic neuritis (BON, n = 11), longitudinally extensive transverse myelitis (LETM, n = 10), and sequential BON and LETM (n = 2), as well as in patients with multiple sclerosis (MS, n = 76) and controls (n = 52). Results: MOG antibodies were detected in 9/23 AQP4 antibody–negative patients with NMO/NMOSD, compared to 1/76 patients with MS and 0/52 controls (p < 0.001). MOG antibodies were detected in 8/11 patients with BON, 0/10 patients with LETM, and 1/2 patients with sequential BON and LETM. Six of 9 MOG antibody–positive patients had a relapsing course. MOG antibody–positive patients had prominent optic disc swelling and were more likely to have a rapid response to steroid therapy and relapse on steroid cessation than MOG antibody–negative patients (p = 0.034 and p = 0.029, respectively). While 8/9 MOG antibody–positive patients had good follow-up visual acuity, one experienced sustained visual impairment, 3 had retinal nerve fiber layer thinning, and one had residual spinal disability.
Article
Full-text available
Objective: To examine the clinical features of pediatric CNS demyelination associated with positive myelin oligodendrocyte glycoprotein (MOG) antibodies and to examine the functional effects of MOG antibody on oligodendrocyte cytoskeleton. Methods: We measured MOG antibody using a fluorescence-activated cell sorting live cell-based assay in acute sera of 73 children with CNS demyelination (DEM) (median age 8 years, range 1.3-15.3) followed for a median of 4 years. We used MO3.13 cells to examine immunoglobulin (Ig) G effects on oligodendrocyte cytoskeleton using 3D deconvolution imaging. Results: MOG antibodies were found in 31/73 patients with DEM (42%) but in 0/24 controls. At first presentation, MOG antibody-positive patients were more likely to have bilateral than unilateral optic neuritis (ON) (9/10 vs 1/5, respectively, p = 0.03), less likely to have brainstem findings (2/31 vs 16/42, p = 0.005), more likely to have a raised erythrocyte sedimentation rate >20 mm/h (9/19 vs 3/21, p = 0.05), less likely to have intrathecal oligoclonal bands (0/16 vs 5/27, p = 0.18), and less likely to be homozygous or heterozygous for human leukocyte antigen DRB1*1501 (3/18 vs 7/22, p = 0.46). MOG antibody positivity varied according to clinical phenotype, with ON and relapsing ON most likely to be seropositive. Two relapsing MOG antibody-positive patients treated with mycophenolate mofetil remain in remission and have become MOG antibody seronegative. Oligodendrocytes incubated with purified IgG from MOG antibody-positive patients showed a striking loss of organization of the thin filaments and the microtubule cytoskeleton, as evidenced by F-actin and β-tubulin immunolabelings. Conclusions: MOG antibody may define a separate demyelination syndrome, which has therapeutic implications. MOG antibody has functional effects on oligodendrocyte cytoskeleton.
Article
Full-text available
We report a patient with neuromyelitis optica (NMO) presenting anti-myelin-oligodendrocyte glycoprotein (MOG)-seropositive, in whom biomarkers of demyelination and astrocyte damage were measured during an acute attack. A 31-year-old man developed right optic neuritis followed by longitudinally extensive transverse myelitis, fulfilling the criteria for definite NMO. He was anti-MOG-seropositive and anti-aquaporin-4 seronegative. The myelin basic protein level was markedly elevated whereas glial fibrillary acidic protein was not detectable in cerebrospinal fluid during an acute attack. His symptoms quickly improved after high-dose methylprednisolone therapy. This case suggests that NMO patients with anti-MOG may have severe demyelination in the absence of astrocyte injury.
Article
Full-text available
Myelin oligodendrocyte glycoprotein (MOG), a constituent of central nervous system myelin, is an important autoantigen in the neuroinflammatory disease multiple sclerosis (MS). However, its function remains unknown. Here, we show that, in healthy human myelin, MOG is decorated with fucosylated N-glycans that support recognition by the C-type lectin receptor (CLR) DC-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) on microglia and DCs. The interaction of MOG with DC-SIGN in the context of simultaneous TLR4 activation resulted in enhanced IL-10 secretion and decreased T cell proliferation in a DC-SIGN-, glycosylation-, and Raf1-dependent manner. Exposure of oligodendrocytes to proinflammatory factors resulted in the down-regulation of fucosyltransferase expression, reflected by altered glycosylation at the MS lesion site. Indeed, removal of fucose on myelin reduced DC-SIGN-dependent homeostatic control, and resulted in inflammasome activation, increased T cell proliferation, and differentiation toward a Th17-prone phenotype. These data demonstrate a new role for myelin glycosylation in the control of immune homeostasis in the healthy human brain through the MOG-DC-SIGN homeostatic regulatory axis, which is comprised by inflammatory insults that affect glycosylation. This phenomenon should be considered as a basis to restore immune tolerance in MS.
Article
Full-text available
Importance Most patients with neuromyelitis optica (NMO) and many with NMO spectrum disorder have autoantibodies against aquaporin-4 (AQP4-Abs), but recently, myelin-oligodendrocyte glycoprotein antibodies (MOG-Abs) have been found in some patients. Here, we showed that patients with NMO/NMOSD with MOG-Abs demonstrate differences when compared with patients with AQP4-Abs.Objective To characterize the features of patients with NMO/NMOSD with MOG-Abs and compare them with patients with AQP4-Ab–positive NMO/NMOSD.Design, Setting, and Participants This observational study was conducted at a single UK specialist center for NMO. Patients with a first demyelinating event between January 1, 2010, and April 1, 2013, seen within the Oxford NMO service and who tested positive for MOG-Abs or AQP4-Abs were included in the study.Exposure Cell-based assays using C-terminal–truncated human MOG and full-length M23-AQP4 were used to test patient serum samples for AQP4-Abs and MOG-Abs.Main Outcomes and Measures Demographic, clinical, and disability data, and magnetic resonance imaging findings.Results Twenty AQP4-Ab–positive patients and 9 MOG-Ab–positive patients were identified. Most patients in both groups were white. Ninety percent of AQP4-Ab–positive patients but only 44% MOG-Ab–positive patients were females (P = .02) with a trend toward older age at disease onset in AQP4-Ab–positive patients (44.9 vs 32.3 years; P = .05). MOG-Ab–positive patients more frequently presented with simultaneous/sequential optic neuritis and myelitis (44% vs 0%; P = .005). Onset episode severity did not differ between the 2 groups, but patients with MOG-Abs had better outcomes from the onset episode, with better recovery Expanded Disability Status Scale scores and a lower risk for visual and motor disability. Myelin-oligodendrocyte glycoprotein antibody–positive patients were more likely to have conus involvement on spinal magnetic resonance imaging (75% vs 17%; P = .02) and involvement of deep gray nuclei on brain magnetic resonance imaging (P = .03). Cerebrospinal fluid characteristics were similar in the 2 groups. A higher proportion of AQP4-Ab–positive patients relapsed (40% vs 0%; P = .03) despite similar follow-up durations.Conclusions and Relevance Despite the fact that patients with MOG-Abs can fulfill the diagnostic criteria for NMO, there are differences when compared with those with AQP4-Abs. These include a higher proportion of males, younger age, and greater likelihood of involvement of the conus and deep gray matter structures on imaging. Additionally, patients with MOG-Abs had more favorable outcomes. Patients with AQP4-Ab–negative NMO/NMOSD should be tested for MOG-Abs.
Article
Full-text available
To evaluate clinical features among patients with neuromyelitis optica spectrum disorders (NMOSD) who have myelin oligodendrocyte glycoprotein (MOG) antibodies, aquaporin-4 (AQP4) antibodies, or seronegativity for both antibodies. Sera from patients diagnosed with NMOSD in 1 of 3 centers (2 sites in Brazil and 1 site in Japan) were tested for MOG and AQP4 antibodies using cell-based assays with live transfected cells. Among the 215 patients with NMOSD, 7.4% (16/215) were positive for MOG antibodies and 64.7% (139/215) were positive for AQP4 antibodies. No patients were positive for both antibodies. Patients with MOG antibodies represented 21.1% (16/76) of the patients negative for AQP4 antibodies. Compared with patients with AQP4 antibodies or patients who were seronegative, patients with MOG antibodies were more frequently male, had a more restricted phenotype (optic nerve more than spinal cord), more frequently had bilateral simultaneous optic neuritis, more often had a single attack, had spinal cord lesions distributed in the lower portion of the spinal cord, and usually demonstrated better functional recovery after an attack. Patients with NMOSD with MOG antibodies have distinct clinical features, fewer attacks, and better recovery than patients with AQP4 antibodies or patients seronegative for both antibodies.
Article
Neuromyelitis optica (NMO) is a rare syndrome characterized by the combination of acute optic neuritis and transverse myelitis, usually not seen in Multiple Sclerosis (MS) and other demyelinating syndromes of the central nervous system (CNS). A high prevalence of various autoantibodies has been described in patients with NMO suggesting a polyclonal activation of the humoral immune system. We examined autoantibody responses to myelin (MBP, MOG with isotypes and epitopes) and astroglial (S100beta) antigens in four patients with NMO by ELISA and Immunoblot. All patients showed a positive anti-MOG response, with one showing reaction to the MOG epitope corresponding to amino acid sequence 63-87. MBP-autoantibodies were only detected in two and S100beta-autoantibodies in one patient. Despite the limited number of samples, these findings suggest a predominant anti-MOG rather than anti-MBP or anti-S100beta autoantibody response in NMO, though no NMO-specific antibody pattern was found, which is in keeping with a widespread acute immune activation, including a strong B-cell response.