You are on page 1of 6

Autoimmunity Reviews 12 (2013) 918–923

Contents lists available at SciVerse ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Review

Pathophysiology of myasthenia gravis with antibodies to the


acetylcholine receptor, muscle-specific kinase and low-density
lipoprotein receptor-related protein 4
Jan J.G.M. Verschuuren a,⁎, Maartje G. Huijbers a, Jaap J. Plomp a, b, Erik H. Niks a, Peter C. Molenaar c,
Pilar Martinez-Martinez c, Alejandro M. Gomez c, Marc H. De Baets c, Mario Losen c,⁎
a
Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
b
Department of MCB-Neurophysiology, Leiden University Medical Centre, Leiden, The Netherlands
c
School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: Myasthenia gravis is caused by antibodies to the acetylcholine receptor, muscle-specific kinase, low-density
Accepted 12 March 2013 lipoprotein receptor-related protein 4, or possibly yet unidentified antibodies. The mechanisms by which
Available online 25 March 2013 these antibodies interfere with the function of postsynaptic proteins include complement activation, antigen-
ic modulation by crosslinking of the target proteins, competition with ligand binding sites, or steric hindrance
Keywords:
which inhibits conformational changes or binding to associated proteins. Screening for auto-antibodies to dif-
Myasthenia gravis
Neuromuscular junction
ferent postsynaptic targets, and also for low-affinity antibodies, is contributing to a more accurate diagnosis
MuSK of MG patients. Further studies into the specific pathophysiological pathways of the several MG subforms
AChR might help to develop new, more antigen specific, therapies.
Lrp4 © 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 918
2. Myasthenia gravis with antibodies against the acetylcholine receptor (AChR-MG) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 919
3. “Seronegative” myasthenia gravis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 919
4. Myasthenia gravis with MuSK antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 920
5. Myasthenia gravis with Lrp4 antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 920
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921
Take-home messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921
.

1. Introduction forms of myasthenia gravis (MG). The function of several proteins at


the neuromuscular junction can also be disturbed by genetic mutations,
Autoimmune reactions to proteins at the postsynaptic neuromuscu- leading to congenital myasthenic syndromes [4]. These syndromes,
lar synapse include antibodies to the acetylcholine receptor (AChR), which can be hereditary or caused by spontaneous mutations, are not
muscle-specific kinase (MuSK), and low-density lipoprotein receptor- associated with auto-antibody production, and not reviewed here. Auto-
related protein 4 (Lrp4) [1–3]. These antibodies define three distinct immune MG is virtually always sporadic with complex genetic and envi-
ronmental risk factors [5], although a few cases of familial autoimmune
MG with AChR or MuSK antibodies have been reported [6–9].
There are several mechanisms by which serum antibodies may
⁎ Corresponding authors.
E-mail addresses: J.J.G.M.Verschuuren@lumc.nl (J.J.G.M. Verschuuren), interfere with the function of postsynaptic proteins, including com-
M.Losen@maastrichtuniversity.nl (M. Losen). plement activation [10], antigenic modulation by crosslinking of the

1568-9972/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.autrev.2013.03.001
J.J.G.M. Verschuuren et al. / Autoimmunity Reviews 12 (2013) 918–923 919

target proteins, competition with ligand binding sites, or steric by stabilizing AChRs with rapsyn) or of complement activation (using
hindrance which inhibits conformational changes or binding to asso- complement inhibitors) can prevent neuromuscular junction damage
ciated proteins [11]. [48,61].
In different forms of myasthenia these mechanisms are not equally Besides from the immune attack by antibodies, it is conceivable that
important, and depend on factors like epitope specificity, antibody the very ability of the muscle to compensate for the loss of AChR and its
subclass, or antigen density. associated proteins during the attack by anti-AChR antibodies ultimate-
ly influences the severity of the disease [63]. Compensatory mecha-
2. Myasthenia gravis with antibodies against the acetylcholine nisms that change the expression levels of the AChR after antibody-
receptor (AChR-MG) mediated damage may vary between individuals [64,65]. There are
also presynaptic compensating changes in MG, e.g. a rise in the ACh
AChR-MG can be categorized according to the clinical symptoms synthesizing enzyme, choline acetyltransferase, and the ACh content
and pathogenesis in early-onset or late-onset MG, and MG with [66,67] as well as an increased release of ACh per nerve impulse
thymoma. AChR-MG with thymoma is mainly seen in elderly MG [68–70]. Most likely these compensatory increases are primarily caused
patients. Thymic abnormalities such as hyperplasia and thymoma by the reduction in the number of AChRs [71], triggering synaptic
are frequently associated with AChR-MG [12,13]. homeostatic mechanisms involving retrograde synaptic signaling [70].
Early-onset MG begins before the age of 40. It mostly starts with Most generalized AChR-MG patients have antibodies against the
ocular muscle weakness and patients subsequently often develop main immunogenic region (MIR) of the AChR alpha subunits. MIR
generalized muscle weakness. Early-onset patients usually have a antibody titers have been shown to correlate well with disease severity
hyperplastic thymus, containing germinal centers that may be sites of [72]. MIR antibodies are particularly pathogenic because 1) two alpha
intense B-cell responses against the AChR [14]. Females predominate subunits and thus two MIRs are present on each AChR, 2) the MIR is
and an association is found with HLA-B8DR3. exposed at an angle that allows antibody binding to two adjacent
In the late-onset AChR-MG subgroup of over 60 years of age, AChRs, and 3) the MIR is involved in AChR sensitivity to activation by
patients mostly have a normal (i.e. atrophic) thymus [15]. In this ACh [73].
group male patients are more frequent and an association with HLA Apart from AChR antibodies, auto-antibodies against other muscle
DRB1*15:01 has been described [16,17]. antigens are commonly found in AChR-MG patients [74]. For example,
Approximately 10% of generalized MG patients have a thymoma auto-antibodies against titin and the ryanodine receptor are frequently
[18]. Their age at onset varies considerably, although thymoma is present in MG patients with malignancies of the thymus [75]. The role
more frequent in the elderly patient [19–22]. The association between of these antibodies in the pathogenesis and their contribution to the
thymic pathology and MG, in addition to the fact that thymic lympho- clinical symptoms of MG are not completely understood yet, and
cytes from patients with follicular hyperplasia produce AChR antibodies some are not specific for MG since they also occur in patients with
in vitro [23], and that thymectomy improves the clinical symptoms in other autoimmune diseases [74].
some MG patients [24], suggests that the thymus has a special role in
triggering the anti-AChR immune response in MG [25].
AChR antibodies can highly differ in their subclass, type of light 3. “Seronegative” myasthenia gravis
chain, affinity and specificity to different AChR epitopes [26–32]. In
AChR-MG patients, the complement-fixing IgG1 and IgG3 AChR- Classically, AChR antibodies have been found in 85% of MG patients,
specific antibodies predominate; IgG2 and IgG4 are only present in while the remaining patients were indicated as ‘seronegative’. In 1986,
lower concentrations [33–35]. Generally, the total of the combined Mossman and colleagues showed that the crude immunoglobulin frac-
IgGs anti-AChR autoantibody titers correlate poorly with the severity tion, obtained by ammonium sulfate precipitation, of the serum of pa-
of disease [32,36–39], although after immunotherapy, such correlations tients with AChR-antibody-negative myasthenia gravis could transfer
have been found in individual AChR-MG patients [40,41] and also in MG the disease to mice [76]. Electrophysiological testing showed a signifi-
models [42]. This disparity could be accounted for by differences in the cant compound muscle action potential decrement upon repetitive
functional activities of the antibodies, i.e. the ability to accelerate the nerve stimulation. Surprisingly, the loss of AChR in the diaphragm was
rate of degradation of AChRs or to block acetylcholine binding sites only around 10%, and no significant amount of antibody bound to the
[43]. In addition, since IgG autoantibody subclasses have distinct immu- mouse AChRs was detected. Passive transfer of plasma or IgG from 7 se-
nological properties, another possible explanation for this weak correla- ronegative patients to mice caused reductions in miniature endplate
tion is variation in the isotype distribution. AChR IgG1 (but not IgG2, potential (MEPP) amplitudes and, in addition, the release of ACh
IgG3 and IgG4) concentration was found to be significantly correlated was significantly reduced by plasma or IgG from 4 of the 7 patients.
to severity of disease [33]. In rhesus monkeys, passive transfer of Unexpectedly, there were no changes in (exogenously applied)
human IgG1, but not IgG4 anti-AChR antibodies caused MG [44]. ACh-induced depolarization or single channel properties of the AChR,
IgG1 and IgG3 anti-AChR antibodies trigger three pathogenic mech- and 125I-alpha-bungarotoxin binding studies showed no effect on
anisms [11]: 1) they bind complement factors at the postsynaptic AChR number [77]. Together, these results pointed to the presence of
membrane, ultimately leading to formation of the membrane attack an antibody that interfered with neuromuscular transmission by bind-
complex [44–46], 2) they bind and crosslink postsynaptic AChRs, ing to non-AChR determinants at the neuromuscular junction. Interest-
resulting in an increased endocytosis and degradation (antigenic ingly, most patients had predominantly bulbar weakness and four of
modulation) [44,47–50], and 3) they may block the binding of ACh to them had assisted ventilation at some timepoint. This suggests that
the AChR [51–55] or inhibit the opening of this ion channel [56,57]. these patients might have suffered from MuSK-MG. Later publications
However, such AChR function impairing antibodies are not generally of the same group indeed indicated, that when retested, these sera
found in AChR-MG. The formation of the membrane attack complex in were predominantly MuSK antibody positive [78].
combination with antigenic modulation leads to: 1) damage of postsyn- The last decade ‘seronegative’ MG has become increasingly rare
aptic membrane folds, 2) removal of the AChR and 3) removal of with the discovery of anti-MuSK antibodies [79], antibodies against
AChR-associated proteins, including utrophin, rapsyn and voltage- clustered AChRs, detected in a sensitive cell-based assay [80] and,
gated sodium channels. Since all of these proteins are involved in very recently, antibodies to Lrp4 [81–83]. With the identification of
either the synaptic function or the formation and maintenance of the these antibodies as sensitive markers, the previously ‘seronegative’
neuromuscular junction, their loss underlies the myasthenic muscle MG patients have split up into subgroups with clear different clinical,
weakness [58–62]. Prevention of antigenic modulation (for example genetic and pharmacological characteristics.
920 J.J.G.M. Verschuuren et al. / Autoimmunity Reviews 12 (2013) 918–923

4. Myasthenia gravis with MuSK antibodies recombining half-antibodies with other IgG4 molecules (‘Fab-arm
exchange’). This produces hybrid antibodies which are bispecific and
IgG antibodies against MuSK were discovered in 2001 in serum of thus do not cross-link identical antigens [44,95,98].
70% of patients with ‘seronegative’ MG [79]. It is now known that These properties notwithstanding, IgG4 appears to be responsible
MuSK-MG differs from AChR-MG in several important aspects: for several autoimmune conditions, including pemphigus [99], idio-
pathic membranous glomerulonephritis with antibodies against
1) The clinical presentation. The distribution of muscle weakness is
M-type phospholipase-A2 receptors [100] and in thrombotic throm-
different, typically involving neck and respiratory muscles, often
bocytopenic purpura, with antibodies against a metalloproteinase
with respiratory crises; often an atrophied tongue is seen [84–86].
ADAMTS13 [101]. Except from pemphigus, the pathogenicity of the
The peak incidence lies in the fourth decade (vs. third decade in
antigen-specific IgG4 in these disorders has not been experimentally
AChR-MG). In contrast to AChR-MG, MuSK-MG has so far been rare-
shown [102].
ly described above the age of sixty years [86]. MuSK-MG patients
Shortly after their discovery it was debated if anti-MuSK antibodies
less often have pure ocular weakness at onset, with 5% never
were the pathogenic factor in MuSK-MG, or that they may only be a
experiencing any ocular weakness, but involvement of bulbar mus-
bystanding disease marker [103,104]. Initial studies could not detect
cles at the start of the disease is more frequent than in AChR-MG
abnormal AChR or MuSK intensities or clear IgG deposits at the neuro-
[2,3,86].
muscular junction [104]. Another study could not demonstrate AChR
2) Acetylcholinesterase inhibiting drugs often do not act at all or can
loss in motor point biopsies from biceps brachii muscles from
even be counterproductive [84–86]. Only 57% of MuSK-MG
MuSK-MG patients nor considerable deposition of immune complexes
patients treated with acetylcholinesterase inhibitor showed a favor-
at the neuromuscular junction [105]. The absence of complement
able effect. On the other hand, side effects of acetylcholinesterase
deposits in muscle tissue of most MuSK-MG patients is compatible
inhibitors are commonly reported, including marked fasciculations,
with IgG4 antibodies not being able to activate complement, suggesting
cramps, and worsening of symptoms [86]. Cholinesterase inhibitor
that other mechanisms play a role in MuSK pathogenesis.
nonresponsiveness was observed in 10 of 14 (71%) MuSK-MG
More recently, a number of studies demonstrated that MuSK-MG
patients, while it appeared in only 4 of 22 (18%) MuSK antibody-
can be transferred from man to mouse using serum or IgG: mice
negative patients and in 13 of 73 (18%) generalized AChR-MG
injected intraperitoneally with total IgG from MuSK-MG patients devel-
patients [87]. The majority of MuSK-MG patients does not experi-
oped weakness and considerable decrement of compound muscle ac-
ence long-term benefit from these drugs and stop to use them. Inter-
tion potentials upon repetitive nerve stimulation, suggesting severe
estingly, rituximab, a monoclonal antibody which depletes B-cells,
AChR loss from neuromuscular junctions [106]. Results from our labora-
seems to give long-term improvement in a more effective way in
tories indicated that mouse muscles regenerating de novo from satellite
MuSK-MG than in AChR-MG [88]. Moreover, rituximab has also
cells are especially prone to effects of MuSK plasma. Thus, MuSK plasma
proven to be very effective in pemphigus foliaceus and pemphigus
caused impairment of nerve stimulus-induced contraction of the flexor
vulgaris, even at a lower dose than is used to treat lymphoma [89].
digitorum brevis muscle and reduced neuromuscular junction size after
Like MuSK-MG, these disorders have been associated with IgG4
local administration of notexin, a potent myotoxin [107]. In detailed
[90]. All 15 pemphigus patients responded to therapy, and eight pa-
morphological analyses, Cole and colleagues demonstrated that passive
tients achieved complete remission in a median period of 51 weeks.
transfer of IgG from MuSK-MG patients to mice reduced the AChR
3) The HLA haplotype association of MuSK-MG is different from
density and that the alignment between the motor nerve terminal and
AChR-MG. MuSK-MG and early onset AChR-MG, which have distinct
the postsynaptic membrane was disturbed [106,108]. Another study
clinical characteristics and pathology of the thymus [91], significant-
indicated that MuSK IgG may interfere with the binding between
ly differ in HLA class II allele frequency. Early onset AChR-MG
collagen-Q and MuSK [109].
is linked with DR3 and DQ2 alleles, while MuSK-MG is associated
Recent work in our laboratories demonstrated that the IgG4 subclass
with DQ5 alleles [92,93]. Within the Northern Hemisphere, in West-
of human MuSK antibodies is myasthenogenic [110]. Mice injected with
ern populations the prevalence seems to correlate with distance
purified MuSK-MG IgG4 became weak and in vivo neuromuscular func-
from the equator (the prevalence being lower at northern latitudes),
tional analyses indicated disturbed neuromuscular transmission. In
suggesting a possible environmental factor or differences in the spe-
detailed electrophysiological studies of the neuromuscular junction
cific HLA frequencies among different populations [78].
we found clear post- and presynaptic deficits, i.e. loss of postsynaptic
4) The antibodies against MuSK in MuSK-MG are predominantly of the
ACh sensitivity and increased rundown of ACh release upon high-rate
IgG4 subclass as opposed to IgG1 and IgG3 antibodies in AChR-MG.
repetitive nerve stimulation. Similar synaptic electrophysiological
Using serially collected sera of six MuSK-MG patients it was abnormalities have recently been found in a total IgG MuSK-MG passive
shown that in individual MuSK-MG patients there is a correlation transfer mouse model [111,112], as well as after active MuSK immuni-
between IgG4 anti-MuSK antibody titer and clinical severity [94]. Of in- zation [113]. AChR loss seems to set in directly after commencing the
terest, one female patient had high titer IgG4 MuSK antibodies when daily IgG injections in the passive transfer mice, and progresses at a
she had severe clinical disease, but after going into clinical remission pace of ~3–4% per day. This matches the physiological rate by which
mostly IgG1 MuSK antibodies were found. Similar findings have been newly synthesized AChRs are inserted in maintenance turn-over at
described in pemphigus vulgaris, in which serum antibodies to peptides the neuromuscular junction, suggesting that MuSK IgG blocks this pro-
that spanned the extracellular domain of desmoglein-3, the antigen in cess [112].
pemphigus vulgaris, were studied [90]. Patients with active pemphigus
had high IgG4 and IgG1 titers against two desmoglein-3 peptide parts, 5. Myasthenia gravis with Lrp4 antibodies
termed Bos1 and Bos 6. Patients in remission had only IgG1 antibodies
to Bos 1, and IgG4 serum antibodies were no longer detectable. Lrp4 is a member of the low-density lipoprotein related receptor
Of the four human IgG subclasses, IgG4 displays remarkable distinct family which was recently identified as the long-sought postsynaptic
characteristics [95,96]: receptor for nerve-derived agrin and, consequently, as an activator of
1) it comprises less than 5% of the total IgG, 2) it is unable to bind MuSK [114]. Its crucial role for the formation of neuromuscular junc-
complement factor C1q and thus does not activate the complement tions was first demonstrated in Lrp4 −/− mice, which lacked AChR
cascade [97], 3) it has only weak Fcγ-receptor binding potency and clusters and died at birth due to respiratory paralysis; a phenotype
thus is not a strong activator of immune cells or antibody-dependent similar to those of MuSK and agrin knock-out mice. Lrp4 can dimerize
cell-mediated cytotoxicity, and 4) it has the unique property of and directly activate MuSK at the surface of the postsynaptic muscle
J.J.G.M. Verschuuren et al. / Autoimmunity Reviews 12 (2013) 918–923 921

Table 1
Frequency of Lrp4 antibodies in neurological diseases.

AChR-MG MuSK-MG “Double” Lambert-Eaton myasthenic syndrome Neuromyelitis Reference


‘seronegative’ MG (anti-calcium channel antibodies) optica

0/100 3/28 6/272 1/101 Higuchi [81]


1/11 19/38 Pevzner [82]
0/61 1/36 11/120 2/16 Zhang [83]

membrane and, along with Dok-7, it is required for the full activation facilitate diagnosis and probably also improve the treatment of these
of this essential kinase. Recently, three papers described the existence subgroups of patients. Moreover, screening for auto-antibodies to dif-
of Lrp4 antibodies in MG patients [81–83], but also in Lambert–Eaton ferent postsynaptic targets, and also for low-affinity antibodies, is
myasthenic syndrome (LEMS) and in neuromyelitis optica (NMO) already positively contributing to a more accurate diagnosis of MG
patients (Table 1). The majority of the patients are female with an patients. Further studies into the specific pathophysiological pathways
age that ranges from 17 to 79 (Table 2), and the involvement of of the several MG subforms will pave the way towards development
IgG1 subclass remind of AChR-MG [81]. However, Lrp4 forms a of new therapies that will be more antigen (and patient) specific.
multiprotein complex with MuSK, thus the clinical symptoms might
be similar to MuSK-MG. Indeed, a relatively high number of bulbar
and respiratory problems have been described in this small group. Take-home messages
Moreover, Lrp4 fulfills an important role in retrograde synaptic
signaling during development and possibly also in adulthood of the • AChR-MG and MuSK-MG present with clinically similar symptoms,
neuromuscular junction, which could also be disturbed by the Lrp4 but have a distinctly different immunopathogenesis.
immune response [81,82,115]. • In contrast to AChR and MuSK antibodies, Lrp4 autoantibodies have
Both Zhang et al. and Higuchi et al. demonstrated in enzyme- been found in more than one form of myasthenia or autoimmune
linked immunosorbent and cellular assays that some sera with Lrp4 disease, including MuSK-MG, neuromyelitis optica and Lambert
antibodies, but without AChR and MuSK antibodies, inhibit the Eaton myasthenic syndrome, and in ‘seronegative’ MG without
agrin-Lrp4 interaction and the agrin-induced AChR clustering [81,83]. AChR and MuSK autoantibodies.
Blocking of agrin signaling is certainly one of the most plausible patho- • IgG1 and IgG3 predominate in AChR-MG patients, IgG4 in MuSK-MG
logical mechanisms for Lrp4 antibodies in vivo, given that agrin is and IgG1 in MG with LRP4 autoantibodies.
fundamental for embryonic development of the neuromuscular • Thymic hyperplasia and thymoma are characteristic for AChR-MG,
junction and that it plays also a crucial role in postnatal synapse main- and not for MuSK MG.
tenance [116]. Indeed, impaired agrin signaling leads to less stable • The different types of MG require different treatment strategies;
neuromuscular junctions, with more dispersed and un-clustered MuSK MG patients may benefit from rituximab whereas AChE inhib-
AChRs, as found in cases of congenital myasthenic syndrome due to itors (regularly used in AChR-MG) often have a counterproductive
agrin gene mutations [117], and also in agrin knock-out mice models effect.
[118]. However, other pathophysiological mechanisms of Lrp4 antibod-
ies such as blocking the Lrp4-MuSK interaction or a role for comple- Acknowledgments
ment, which is implied by the involvement of IgG1 autoantibodies,
cannot be excluded. Our work is supported by the Prinses Beatrix Spierfonds (WAR09-19),
L'Association Française contre les Myopathies (#15363 and #15853) and
a Veni fellowship of the Netherlands Organization for Scientific Re-
6. Conclusion search (#91610148) and a fellowship of the Brain Foundation of the
Netherlands (#F2008(1)28).
It is increasingly evident that AChR-MG and MuSK-MG are different
autoimmune conditions that, despite sharing a number of clinical
symptoms, have a distinct pathophysiology. The presence of Lrp-4 auto- References
antibodies in ‘seronegative’ MG, but also MuSK-MG, LEMS and NMO is
[1] Zisimopoulou P, Brenner T, Trakas N, Tzartos SJ. Serological diagnostics in myasthenia
intriguing. Further research on the phenotypic characteristics of sero- gravis based on novel assays and recently identified antigens. Autoimmun Rev
negative and MuSK-MG patients with LRP-4 antibodies will likely 2013;12(9):924–30 (this issue).
[2] Mori S, Shigemoto K. Mechanisms associated with the pathogenicity of antibodies
against muscle-specific kinase in myasthenia gravis. Autoimmun Rev 2013;12(9):
912–7 (this issue).
[3] Evoli A, Padua L. Diagnosis and therapy of myasthenia gravis with antibodies to
muscle-specific kinase. Autoimmun Rev 2013;12(9):931–5 (this issue).
Table 2
[4] Engel AG. Congenital myasthenic syndromes in 2012. Curr Neurol Neurosci Rep
Characteristics of patients with Lrp4 antibodies.
2012;12:92–101.
[5] Ramanujam R, Pirskanen R, Ramanujam S, Hammarstrom L. Utilizing twins concor-
Clinical features
dance rates to infer the predisposition to myasthenia gravis. Twin Res Hum Genet
Age Sex Race Symptoms 2011;14:129–36.
F:M [6] Lavrnic D, Nikolic A, De Baets M, Verschuuren J, Verduyn W, Losen M, et al. Familial
occurrence of autoimmune myasthenia gravis with different antibody specificity.
n=9 26–72 4:5 n/a MGFA scale: Neurology 2008;70:2011–3.
(Higuchi [81]) 2a,2b [7] Evoli A, Batocchi AP, Zelano G, Uncini A, Palmisani MT, Tonali P. Familial autoim-
3a,3a,3b,3b mune myasthenia gravis: report of four families. J Neurol Neurosurg Psychiatry
5,5,5 1995;58:729–31.
n = 13 17–79 10:3 12/13 Ocular weakness 50% [8] Provenzano C, Arancio O, Evoli A, Rocca B, Bartoccioni E, de Grandis D, et al. Familial
(Pevzner [82]) Caucasian Facial/bulbar weakness 86% autoimmune myasthenia gravis with different pathogenetic antibodies. J Neurol
Neck weakness 86% Neurosurg Psychiatry 1988;51:1228–30.
Limb weakness 71% [9] Bergoffen J, Zmijewski CM, Fischbeck KH. Familial autoimmune myasthenia
Breathing difficulty 29% gravis. Neurology 1994;44:551–4.
[10] Tüzün E, Christadoss P. Complement Associated Pathogenic Mechanisms in My-
Myasthenic crisis in one patient
asthenia Gravis. Autoimmun Rev 2013;12(9):904–11 (this issue).
922 J.J.G.M. Verschuuren et al. / Autoimmunity Reviews 12 (2013) 918–923

[11] Gomez AM, Van Den Broeck J, Vrolix K, Janssen SP, Lemmens MA, Van Der Esch E, [43] Drachman DB, Adams RN, Josifek LF, Self SG. Functional activities of autoanti-
et al. Antibody effector mechanisms in myasthenia gravis — pathogenesis at the bodies to acetylcholine receptors and the clinical severity of myasthenia gravis.
neuromuscular junction. Autoimmunity 2010;43:353–70. N Engl J Med 1982;307:769–75.
[12] Shiono H, Wong YL, Matthews I, Liu JL, Zhang W, Sims G, et al. Spontaneous pro- [44] van der Neut Kolfschoten M, Schuurman J, Losen M, Bleeker WK,
duction of anti-IFN-alpha and anti-IL-12 autoantibodies by thymoma cells from Martinez-Martinez P, Vermeulen E, et al. Anti-inflammatory activity of human
myasthenia gravis patients suggests autoimmunization in the tumor. Int IgG4 antibodies by dynamic Fab arm exchange. Science 2007;317:1554–7.
Immunol 2003;15:903–13. [45] Engel AG, Lambert EH, Howard FM. Immune complexes (IgG and C3) at the
[13] Le Panse R, Bismuth J, Cizeron-Clairac G, Weiss JM, Cufi P, Dartevelle P, et al. Thymic motor end-plate in myasthenia gravis: ultrastructural and light microscopic lo-
remodeling associated with hyperplasia in myasthenia gravis. Autoimmunity calization and electrophysiologic correlations. Mayo Clin Proc 1977;52:267–80.
2010;43:401–12. [46] Chamberlain-Banoub J, Neal JW, Mizuno M, Harris CL, Morgan BP. Complement
[14] Alpert LI, Papatestas A, Kark A, Osserman RS, Osserman K. A histologic reappraisal membrane attack is required for endplate damage and clinical disease in passive
of the thymus in myasthenia gravis. A correlative study of thymic pathology and experimental myasthenia gravis in Lewis rats. Clin Exp Immunol 2006;146:
response to thymectomy. Arch Pathol 1971;91:55–61. 278–86.
[15] Myking A, Skeie GO, Varhaug JE, Andersen KS, Gilhus NE, Aarli JA. The [47] Loutrari H, Kokla A, Tzartos SJ. Passive transfer of experimental myasthenia
histomorphology of the thymus in late onset, non-thymoma myasthenia gravis. gravis via antigenic modulation of acetylcholine receptor. Eur J Immunol
Eur J Neurol 1998;5:401–5. 1992;22:2449–52.
[16] Maniaol AH, Elsais A, Lorentzen AR, Owe JF, Viken MK, Saether H, et al. Late onset [48] Losen M, Martinez-Martinez P, Phernambucq M, Schuurman J, Parren PW, De
myasthenia gravis is associated with HLA DRB1*15:01 in the Norwegian popula- Baets MH. Treatment of myasthenia gravis by preventing acetylcholine receptor
tion. PLoS One 2012;7:e36603. modulation. Ann N Y Acad Sci 2008;1132:174–9.
[17] Cavalcante P, Cufi P, Mantegazza R, Berrih-Aknin S, Bernasconi P, Le Panse R. Etiology [49] Heinemann S, Bevan S, Kullberg R, Lindstrom J, Rice J. Modulation of acetylcho-
of myasthenia gravis: innate immunity signature in pathological thymus. Autoimmun line receptor by antibody against the receptor. Proc Natl Acad Sci U S A 1977;74:
Rev 2013;12(9):863–74 (this issue). 3090–4.
[18] Marx A, Pfister F, Schalke B, Saruhan-Direskeneli G, Melms A, Ströbel P. The dif- [50] Drachman DB, Angus CW, Adams RN, Michelson JD, Hoffman GJ. Myasthenic anti-
ferent roles of the thymus in the pathogenesis of the various myasthenia gravis bodies cross-link acetylcholine receptors to accelerate degradation. N Engl J Med
subtypes. Autoimmun Rev 2013;12(9):875–84 (this issue). 1978;298:1116–22.
[19] Gilhus NE. Autoimmune myasthenia gravis. Expert Rev Neurother 2009;9:351–8. [51] Gomez CM, Richman DP. Anti-acetylcholine receptor antibodies directed against
[20] Vincent A. Unravelling the pathogenesis of myasthenia gravis. Nat Rev Immunol the alpha-bungarotoxin binding site induce a unique form of experimental
2002;2:797–804. myasthenia. Proc Natl Acad Sci U S A 1983;80:4089–93.
[21] Romi F, Gilhus NE, Aarli JA. Myasthenia gravis: clinical, immunological, and ther- [52] Almon RR, Andrew CG, Appel SH. Serum globulin in myasthenia gravis: inhibition
apeutic advances. Acta Neurol Scand 2005;111:134–41. of alpha-bungarotoxin binding to acetylcholine receptors. Science 1974;186:55–7.
[22] Vincent A, Palace J, Hilton-Jones D. Myasthenia gravis. Lancet 2001;357:2122–8. [53] Vernet-der Garabedian B, Morel E, Bach JF. Heterogeneity of antibodies directed
[23] Scadding GK, Vincent A, Newsom-Davis J, Henry K. Acetylcholine receptor anti- against the alpha-bungarotoxin binding site on human acetylcholine receptor
body synthesis by thymic lymphocytes: correlation with thymic histology. and severity of myasthenia gravis. J Neuroimmunol 1986;12:65–74.
Neurology 1981;31:935–43. [54] Whiting PJ, Vincent A, Newsom-Davis J. Acetylcholine receptor antibody charac-
[24] Kuks JB, Oosterhuis HJ, Limburg PC, The TH. Anti-acetylcholine receptor antibodies teristics in myasthenia gravis. Fractionation of alpha-bungarotoxin binding site
decrease after thymectomy in patients with myasthenia gravis. Clinical correla- antibodies and their relationship to IgG subclass. J Neuroimmunol 1983;5:1–9.
tions. J Autoimmun 1991;4:197–211. [55] Hara H, Hayashi K, Ohta K, Itoh N, Nishitani H, Ohta M. Detection and character-
[25] Leprince C, Cohen-Kaminsky S, Berrih-Aknin S, Vernet-Der Garabedian B, Treton D, ization of blocking-type anti-acetylcholine receptor antibodies in sera from
Galanaud P, et al. Thymic B cells from myasthenia gravis patients are activated B patients with myasthenia gravis. Clin Chem 1993;39:2053–7.
cells. Phenotypic and functional analysis. J Immunol 1990;145:2115–22. [56] Lang B, Richardson G, Rees J, Vincent A, Newsom-Davis J. Plasma from myasthe-
[26] Andrews PI, Massey JM, Sanders DB. Acetylcholine receptor antibodies in juvenile nia gravis patients reduces acetylcholine receptor agonist-induced Na + flux
myasthenia gravis. Neurology 1993;43:977–82. into TE671 cell line. J Neuroimmunol 1988;19:141–8.
[27] Andrews PI, Massey JM, Howard Jr JF, Sanders DB. Race, sex, and puberty influence [57] Wintzen AR, Plomp JJ, Molenaar PC, van Dijk JG, van Kempen GT, Vos RM, et al.
onset, severity, and outcome in juvenile myasthenia gravis. Neurology 1994;44: Acquired slow-channel syndrome: a form of myasthenia gravis with prolonged
1208–14. open time of the acetylcholine receptor channel. Ann Neurol 1998;44:657–64.
[28] Chiu HC, Vincent A, Newsom-Davis J, Hsieh KH, Hung T. Myasthenia gravis: [58] Martinez-Martinez P, Losen M, Duimel H, Frederik P, Spaans F, Molenaar P, et al.
population differences in disease expression and acetylcholine receptor anti- Overexpression of rapsyn in rat muscle increases acetylcholine receptor levels in
body titers between Chinese and Caucasians. Neurology 1987;37:1854–7. chronic experimental autoimmune myasthenia gravis. Am J Pathol 2007;170:
[29] Tindall RS. Humoral immunity in myasthenia gravis: biochemical characteriza- 644–57.
tion of acquired antireceptor antibodies and clinical correlations. Ann Neurol [59] Ito H, Yoshimura T, Satoh A, Takino H, Tsujihata M, Nagataki S. Immunohisto-
1981;10:437–47. chemical study of utrophin and dystrophin at the motor end-plate in myasthenia
[30] Tindall RS. Humoral immunity in myasthenia gravis: clinical correlations of gravis. Acta Neuropathol 1996;92:14–8.
anti-receptor antibody avidity and titer. Ann N Y Acad Sci 1981;377:316–31. [60] Slater CR, Young C, Wood SJ, Bewick GS, Anderson LV, Baxter P, et al. Utrophin
[31] Demaine A, Willcox N, Janer M, Welsh K, Newsom-Davis J. Immunoglobulin abundance is reduced at neuromuscular junctions of patients with both inherited
heavy chain gene associations in myasthenia gravis: new evidence for disease and acquired acetylcholine receptor deficiencies. Brain 1997;120(Pt 9):1513–31.
heterogeneity. J Neurol 1992;239:53–6. [61] Losen M, Stassen MH, Martinez-Martinez P, Machiels BM, Duimel H, Frederik P,
[32] Oosterhuis HJ, Limburg PC, Hummel-Tappel E, The TH. Anti-acetylcholine recep- et al. Increased expression of rapsyn in muscles prevents acetylcholine receptor
tor antibodies in myasthenia gravis. Part 2. Clinical and serological follow-up of loss in experimental autoimmune myasthenia gravis. Brain 2005;128:2327–37.
individual patients. J Neurol Sci 1983;58:371–85. [62] Martinez-Martinez P, Phernambucq M, Steinbusch L, Schaeffer L, Berrih-Aknin S,
[33] Rodgaard A, Nielsen FC, Djurup R, Somnier F, Gammeltoft S. Acetylcholine recep- Duimel H, et al. Silencing rapsyn in vivo decreases acetylcholine receptors and
tor antibody in myasthenia gravis: predominance of IgG subclasses 1 and 3. Clin augments sodium channels and secondary postsynaptic membrane folding.
Exp Immunol 1987;67:82–8. Neurobiol Dis 2009;35:14–23.
[34] Vincent A, Newsom-Davis J. Acetylcholine receptor antibody characteristics in [63] De Baets M, Stassen M, Losen M, Zhang X, Machiels B. Immunoregulation in ex-
myasthenia gravis. I. Patients with generalized myasthenia or disease restricted perimental autoimmune myasthenia gravis — about T cells, antibodies, and
to ocular muscles. Clin Exp Immunol 1982;49:257–65. endplates. Ann N Y Acad Sci 2003;998:308–17.
[35] Lefvert AK, Cuenoud S, Fulpius BW. Binding properties and subclass distribution [64] Guyon T, Levasseur P, Truffault F, Cottin C, Gaud C, Berrih-Aknin S. Regulation of
of anti-acetylcholine receptor antibodies in myasthenia gravis. J Neuroimmunol acetylcholine receptor alpha subunit variants in human myasthenia gravis.
1981;1:125–35. Quantification of steady-state levels of messenger RNA in muscle biopsy using
[36] Lefvert AK, Bergstrom K, Matell G, Osterman PO, Pirskanen R. Determination of the polymerase chain reaction. J Clin Invest 1994;94:16–24.
acetylcholine receptor antibody in myasthenia gravis: clinical usefulness and [65] Moulian N, Wakkach A, Guyon T, Poea S, Aissaoui A, Levasseur P, et al. Respective
pathogenetic implications. J Neurol Neurosurg Psychiatry 1978;41:394–403. role of thymus and muscle in autoimmune myasthenia gravis. Ann N Y Acad Sci
[37] Lindstrom JM, Seybold ME, Lennon VA, Whittingham S, Duane DD. Antibody to 1998;841:397–406.
acetylcholine receptor in myasthenia gravis. Prevalence, clinical correlates, and [66] Ito Y, Miledi R, Molenaar PC, Vincent A, Polak RL, van Gelder M, et al. Acetylcho-
diagnostic value. Neurology 1976;26:1054–9. line in human muscle. Proc R Soc Lond B Biol Sci 1976;192:475–80.
[38] Vincent A, Newsom Davis J. Anti-acetylcholine receptor antibodies. J Neurol [67] Molenaar PC, Newsom-Davis J, Polak RL, Vincent A. Choline acetyltransferase in
Neurosurg Psychiatry 1980;43:590–600. skeletal muscle from patients with myasthenia gravis. J Neurochem 1981;37:
[39] Takeo G, Motomura M, Mats H, Ohishi K, Yoshimura T, Tsujihata M, et al. Effect of 1081–8.
myasthenic IgG on degradation of junctional acetylcholine receptor. Muscle [68] Molenaar PC, Polak RL, Miledi R, Alema S, Vincent A, Newsom-Davis J. Acetylcho-
Nerve 1993;16:840–8. line in intercostal muscle from myasthenia gravis patients and in rat diaphragm
[40] Newsom-Davis J, Wilson SG, Vincent A, Ward CD. Long-term effects of repeated after blockade of acetylcholine receptors. Prog Brain Res 1979;49:449–58.
plasma exchange in myasthenia gravis. Lancet 1979;1:464–8. [69] Cull-Candy SG, Miledi R, Trautmann A, Uchitel OD. On the release of transmitter
[41] Newsom-Davis J, Vincent A. Combined plasma exchange and immunosuppression at normal, myasthenia gravis and myasthenic syndrome affected human
in myasthenia gravis. Lancet 1979;2:688. end-plates. J Physiol 1980;299:621–38.
[42] Janssen SP, Phernambucq M, Martinez-Martinez P, De Baets MH, Losen M. Immu- [70] Plomp JJ, Van Kempen GT, De Baets MB, Graus YM, Kuks JB, Molenaar PC. Acetylcho-
nosuppression of experimental autoimmune myasthenia gravis by mycophenolate line release in myasthenia gravis: regulation at single end-plate level. Ann Neurol
mofetil. J Neuroimmunol 2008;201–202:111–20. 1995;37:627–36.
J.J.G.M. Verschuuren et al. / Autoimmunity Reviews 12 (2013) 918–923 923

[71] Miledi R, Molenaar PC, Polak RL. Alpha-Bungarotoxin enhances transmitter [94] Niks EH, van Leeuwen Y, Leite MI, Dekker FW, Wintzen AR, Wirtz PW, et al. Clin-
“released” at the neuromuscular junction. Nature 1978;272:641–3. ical fluctuations in MuSK myasthenia gravis are related to antigen-specific IgG4
[72] Masuda T, Motomura M, Utsugisawa K, Nagane Y, Nakata R, Tokuda M, et al. Anti- instead of IgG1. J Neuroimmunol 2008;195:151–6.
bodies against the main immunogenic region of the acetylcholine receptor corre- [95] Aalberse RC, Stapel SO, Schuurman J, Rispens T. Immunoglobulin G4: an odd
late with disease severity in myasthenia gravis. J Neurol Neurosurg Psychiatry antibody. Clin Exp Allergy 2009;39:469–77.
2012;83:935–40. [96] Schroeder Jr HW, Cavacini L. Structure and function of immunoglobulins. J Allergy
[73] Luo J, Taylor P, Losen M, de Baets MH, Shelton GD, Lindstrom J. Main immunogenic Clin Immunol 2010;125:S41–52.
region structure promotes binding of conformation-dependent myasthenia gravis [97] Daha NA, Banda NK, Roos A, Beurskens FJ, Bakker JM, Daha MR, et al. Complement
autoantibodies, nicotinic acetylcholine receptor conformation maturation, and activation by (auto-) antibodies. Mol Immunol 2011;48:1656–65.
agonist sensitivity. J Neurosci 2009;29:13898–908. [98] Aalberse RC, Schuurman J. IgG4 breaking the rules. Immunology 2002;105:9–19.
[74] Vrolix K, Fraussen J, Molenaar PC, Losen M, Somers V, Stinissen P, et al. The [99] Sitaru C, Mihai S, Zillikens D. The relevance of the IgG subclass of autoantibodies
auto-antigen repertoire in myasthenia gravis. Autoimmunity 2010;43:380–400. for blister induction in autoimmune bullous skin diseases. Arch Dermatol Res
[75] Marx A, Willcox N, Leite MI, Chuang WY, Schalke B, Nix W, et al. Thymoma and 2007;299:1–8.
paraneoplastic myasthenia gravis. Autoimmunity 2010;43:413–27. [100] Beck Jr LH, Bonegio RG, Lambeau G, Beck DM, Powell DW, Cummins TD, et al.
[76] Mossman S, Vincent A, Newsom-Davis J. Myasthenia gravis without acetylcholine- M-type phospholipase A2 receptor as target antigen in idiopathic membranous
receptor antibody: a distinct disease entity. Lancet 1986;1:116–9. nephropathy. N Engl J Med 2009;361:11–21.
[77] Burges J, Vincent A, Molenaar PC, Newsom-Davis J, Peers C, Wray D. Passive transfer [101] Ferrari S, Mudde GC, Rieger M, Veyradier A, Kremer Hovinga JA, Scheiflinger F.
of seronegative myasthenia gravis to mice. Muscle Nerve 1994;17:1393–400. IgG subclass distribution of anti-ADAMTS13 antibodies in patients with acquired
[78] Vincent A, Leite MI, Farrugia ME, Jacob S, Viegas S, Shiraishi H, et al. Myasthenia thrombotic thrombocytopenic purpura. J Thromb Haemost 2009;7:1703–10.
gravis seronegative for acetylcholine receptor antibodies. Ann N Y Acad Sci [102] Rock B, Martins CR, Theofilopoulos AN, Balderas RS, Anhalt GJ, Labib RS, et al. The
2008;1132:84–92. pathogenic effect of IgG4 autoantibodies in endemic pemphigus foliaceus (fogo
[79] Hoch W, McConville J, Helms S, Newsom-Davis J, Melms A, Vincent A. Auto- selvagem). N Engl J Med 1989;320:1463–9.
antibodies to the receptor tyrosine kinase MuSK in patients with myasthenia [103] Lindstrom J. Is “seronegative” MG explained by autoantibodies to MuSK?
gravis without acetylcholine receptor antibodies. Nat Med 2001;7:365–8. Neurology 2004;62:1920–1.
[80] Leite MI, Jacob S, Viegas S, Cossins J, Clover L, Morgan BP, et al. IgG1 antibodies to [104] Selcen D, Fukuda T, Shen XM, Engel AG. Are MuSK antibodies the primary cause
acetylcholine receptors in ‘seronegative’ myasthenia gravis. Brain 2008;131: of myasthenic symptoms? Neurology 2004;62:1945–50.
1940–52. [105] Shiraishi H, Motomura M, Yoshimura T, Fukudome T, Fukuda T, Nakao Y, et al. Ace-
[81] Higuchi O, Hamuro J, Motomura M, Yamanashi Y. Autoantibodies to low-density li- tylcholine receptors loss and postsynaptic damage in MuSK antibody-positive
poprotein receptor-related protein 4 in myasthenia gravis. Ann Neurol 2011;69: myasthenia gravis. Ann Neurol 2005;57:289–93.
418–22. [106] Cole RN, Reddel SW, Gervasio OL, Phillips WD. Anti-MuSK patient antibodies
[82] Pevzner A, Schoser B, Peters K, Cosma NC, Karakatsani A, Schalke B, et al. disrupt the mouse neuromuscular junction. Ann Neurol 2008;63:782–9.
Anti-LRP4 autoantibodies in AChR- and MuSK-antibody-negative myasthenia [107] ter Beek WP, Martinez-Martinez P, Losen M, de Baets MH, Wintzen AR,
gravis. J Neurol 2012;259:427–35. Verschuuren JJ, et al. The effect of plasma from muscle-specific tyrosine kinase
[83] Zhang B, Tzartos JS, Belimezi M, Ragheb S, Bealmear B, Lewis RA, et al. Autoanti- myasthenia patients on regenerating endplates. Am J Pathol 2009;175:1536–44.
bodies to lipoprotein-related protein 4 in patients with double-seronegative [108] Cole RN, Ghazanfari N, Ngo ST, Gervasio OL, Reddel SW, Phillips WD. Patient au-
myasthenia gravis. Arch Neurol 2012;69:445–51. toantibodies deplete postsynaptic muscle-specific kinase leading to disassembly
[84] Lavrnic D, Losen M, Vujic A, De Baets M, Hajdukovic LJ, Stojanovic V, et al. The of the ACh receptor scaffold and myasthenia gravis in mice. J Physiol 2010;588:
features of myasthenia gravis with autoantibodies to MuSK. J Neurol Neurosurg 3217–29.
Psychiatry 2005;76:1099–102. [109] Kawakami Y, Ito M, Hirayama M, Sahashi K, Ohkawara B, Masuda A, et al. Anti-
[85] Evoli A, Tonali PA, Padua L, Monaco ML, Scuderi F, Batocchi AP, et al. Clinical cor- MuSK autoantibodies block binding of collagen Q to MuSK. Neurology 2011;77:
relates with anti-MuSK antibodies in generalized seronegative myasthenia 1819–26.
gravis. Brain 2003;126:2304–11. [110] Klooster R, Plomp JJ, Huijbers MG, Niks EH, Straasheijm KR, Detmers FJ, et al.
[86] Guptill JT, Sanders DB, Evoli A. Anti-MuSK antibody myasthenia gravis: clinical find- Muscle-specific kinase myasthenia gravis IgG4 autoantibodies cause severe neu-
ings and response to treatment in two large cohorts. Muscle Nerve 2011;44:36–40. romuscular junction dysfunction in mice. Brain 2012;135:1081–101.
[87] Hatanaka Y, Hemmi S, Morgan MB, Scheufele ML, Claussen GC, Wolfe GI, et al. [111] Viegas S, Jacobson L, Waters P, Cossins J, Jacob S, Leite MI, et al. Passive and active
Nonresponsiveness to anticholinesterase agents in patients with MuSK-antibody- immunization models of MuSK-Ab positive myasthenia: electrophysiological ev-
positive MG. Neurology 2005;65:1508–9. idence for pre and postsynaptic defects. Exp Neurol 2012;234:506–12.
[88] Diaz-Manera J, Martinez-Hernandez E, Querol L, Klooster R, Rojas-Garcia R, [112] Morsch M, Reddel SW, Ghazanfari N, Toyka KV, Phillips WD. Muscle specific ki-
Suarez-Calvet X, et al. Long-lasting treatment effect of rituximab in MuSK myas- nase autoantibodies cause synaptic failure through progressive wastage of post-
thenia. Neurology 2012;78:189–93. synaptic acetylcholine receptors. Exp Neurol 2012;237:286–95.
[89] Horvath B, Huizinga J, Pas HH, Mulder AB, Jonkman MF. Low-dose rituximab is [113] Mori S, Kubo S, Akiyoshi T, Yamada S, Miyazaki T, Hotta H, et al. Antibodies
effective in pemphigus. Br J Dermatol 2012;166:405–12. against muscle-specific kinase impair both presynaptic and postsynaptic func-
[90] Bhol K, Natarajan K, Nagarwalla N, Mohimen A, Aoki V, Ahmed AR. Correlation of tions in a murine model of myasthenia gravis. Am J Pathol 2012;180:798–810.
peptide specificity and IgG subclass with pathogenic and nonpathogenic autoan- [114] Kim N, Stiegler AL, Cameron TO, Hallock PT, Gomez AM, Huang JH, et al. Lrp4 is a
tibodies in pemphigus vulgaris: a model for autoimmunity. Proc Natl Acad Sci U receptor for Agrin and forms a complex with MuSK. Cell 2008;135:334–42.
S A 1995;92:5239–43. [115] Yumoto N, Kim N, Burden SJ. Lrp4 is a retrograde signal for presynaptic differen-
[91] Leite MI, Strobel P, Jones M, Micklem K, Moritz R, Gold R, et al. Fewer thymic tiation at neuromuscular synapses. Nature 2012;489:438–42.
changes in MuSK antibody-positive than in MuSK antibody-negative MG. Ann [116] Kummer TT, Misgeld T, Sanes JR. Assembly of the postsynaptic membrane at the
Neurol 2005;57:444–8. neuromuscular junction: paradigm lost. Curr Opin Neurobiol 2006;16:74–82.
[92] Bartoccioni E, Scuderi F, Augugliaro A, Chiatamone Ranieri S, Sauchelli D, Alboino [117] Huze C, Bauche S, Richard P, Chevessier F, Goillot E, Gaudon K, et al. Identifica-
P, et al. HLA class II allele analysis in MuSK-positive myasthenia gravis suggests a tion of an agrin mutation that causes congenital myasthenia and affects synapse
role for DQ5. Neurology 2009;72:195–7. function. Am J Hum Genet 2009;85:155–67.
[93] Niks EH, Kuks JB, Roep BO, Haasnoot GW, Verduijn W, Ballieux BE, et al. Strong [118] Gautam M, Noakes PG, Moscoso L, Rupp F, Scheller RH, Merlie JP, et al. Defective
association of MuSK antibody-positive myasthenia gravis and HLA-DR14-DQ5. neuromuscular synaptogenesis in agrin-deficient mutant mice. Cell 1996;85:
Neurology 2006;66:1772–4. 525–35.

You might also like