ArticlePDF Available

Recent advances in the understanding and therapy of myasthenia gravis

Authors:

Abstract and Figures

Myasthenia gravis (MG) is a T-cell dependent autoimmune disease mediated by autoantibodies, which mainly target muscle nicotinic acetylcholine receptors (AChR) and cause loss of functional AChRs in the neuromuscular junction. Both MG and its major autoantigen are studied extensively, yet the etiology of the disease remains unclear, although it is known to be associated with the thymus. A genetic predisposition, combined with several unidentified environmental stimuli, likely creates a favorable milieu in which the disease can appear. Current research focusses on elucidating the cellular and molecular pathways of immune dysregulation, which underly MG outburst and progression. Considerable progress has been made concerning the involvement of the thymus, the identification of impaired mechanisms of immune control and the B–T-cell interaction in MG pathogenesis, while the role of chemokines arises as an intriguing new puzzle. Recent findings fueled the development of novel therapeutic approaches with some encouraging, although preliminary, results. This review summarizes recent achievements in the fields of both basic research and therapeutics.
Content may be subject to copyright.
10.2217/14796708.1.6.799 © 2006 Future Medicine Ltd ISSN 1479-6708 Future Neurol. (2006) 1(6), 799–801 799
REVIEW
Recent advances in the understanding and
therapy of myasthenia gravis
Efrosini Fostieri,
Kalliopi Kostelidou,
Konstantinos Poulas &
Socrates J Tzartos
Author for correspondence
Hellenic Pasteur Institute,
Department of Biochemistry,
127 Vas. Sofias Avenue,
11521 Athens, Greece
and
Department of Pharmacy,
University of Patras,
26504 Patras, Greece
Tel.: +30 210 647 8842;
Fax: +30 210 647 8842;
tzartos@pasteur.gr
Keywords: autoantibodies,
autoimmunity, autoreactive
T cells, myasthenia gravis,
neuromuscular junction,
nicotinic acetylcholine
receptor, specific
immunotherapy, thymus
Myasthenia gravis (MG) is a T-cell dependent autoimmune disease mediated by
autoantibodies, which mainly target muscle nicotinic acetylcholine receptors (AChR) and
cause loss of functional AChRs in the neuromuscular junction. Both MG and its major
autoantigen are studied extensively, yet the etiology of the disease remains unclear,
although it is known to be associated with the thymus. A genetic predisposition, combined
with several unidentified environmental stimuli, likely creates a favorable milieu in which the
disease can appear. Current research focusses on elucidating the cellular and molecular
pathways of immune dysregulation, which underly MG outburst and progression.
Considerable progress has been made concerning the involvement of the thymus, the
identification of impaired mechanisms of immune control and the B–T-cell interaction in MG
pathogenesis, while the role of chemokines arises as an intriguing new puzzle. Recent
findings fueled the development of novel therapeutic approaches with some encouraging,
although preliminary, results. This review summarizes recent achievements in the fields of
both basic research and therapeutics.
In the autoimmune disease myasthenia gravis
(MG), the major autoantigen is the muscle nico-
tinic acetylcholine receptor (AChR), a postsynap-
tic ligand-gated ion channel of the
neuromuscular junction (NMJ) with the stoichi-
ometry
α
2
βεδ (adult muscle) or α
2
βγδ (fetal and
denervated adult muscle). The anti-AChR
autoantibodies cause the loss of functional AChR
molecules resulting in impaired neuromuscular
transmission, which is reflected clinically with
weakness and fatigability of voluntary muscles.
The clinical and immunopathological features of
MG can be reproduced effectively in an animal
model. Experimental autoimmune MG (EAMG)
is induced actively in animals, most commonly
mice and rats, by immunization with hetero-
logous AChR and is due to antibodies (Abs) raised
against the heterologous AChR, which cross-react
with the autologous receptor molecule.
Experimental data regarding MG is relatively
vast, yet the etiology of the disease remains elu-
sive, although related with thymic abnormali-
ties. Current treatment lacks specificity and is
often accompanied by various side effects.
Hence, ongoing research is focussed on
unraveling the fine cellular and molecular path-
ways underlying the outburst and progression
of the disease. A more comprehensive view of
MG would give rise to specific and sophisti-
cated therapeutic approaches targeted at differ-
ent levels of immune modulation. This review
on MG presents recent achievements in the
fields of basic research and therapeutics.
The mosaic of different myasthenia
gravis phenotypes
MG is a remarkably heterogeneous disease. It can
either remain localized to a single muscle group
(usually, but not necessarily, the extraocular mus-
cles – named ocular MG) or it can spread to other
skeletal muscles (termed generalized). A major
classification of generalized MG is made on the
basis of the presence or absence of anti-AChR Abs
(seropositive or seronegative, respectively).
Seropositive myasthenia gravis
This group, representing approximately 85% of
MG patients, comprises three distinct disease
entities
(Table 1):
Early-onset MG appears before 40 years of
age, affects mostly women and is commonly
related with thymic follicular hyperplasia;
• Late-onset MG affects people older than
40 years of age with normal or atrophic
thymus and shows a bias for men
[1];
Paraneoplastic MG associated with thymoma
affects men and women of any age (mostly
between 40 and 60 years of age) equally and is
characterized by the presence of Abs to other
striated muscle antigens, mainly titin and the
ryanodine receptor
[2].
Seronegative MG: the discovery of
anti-muscle specific kinase autoantibodies
Approximately 15% of patients with generalized
MG symptoms have no detectable anti-AChR
Abs in their serum. Despite various evidence
For reprint orders, please contact:
reprints@futuremedicine.com
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
800 Future Neurol. (2006) 1(6)
pointing to an autoantibody-mediated disease, the
autoantigen in seronegative MG had long
remained unknown. Vincent and colleagues
were the first to identify the muscle-specific
kinase (MuSK), a key player in postsynaptic
NMJ differentiation and AChR clustering
(Figure 1), as the target of the autoimmune
response in a proportion of seronegative MG
patients (
40% [3–5]). The presence of anti-MuSK
Abs appears to define a subgroup of seronegative
MG patients with distinct clinical characteristics:
predominantly localized, commonly bulbar, mus-
cle weakness and muscle atrophy, which often
restricts complete response to commonly used
therapies (acetylcholinesterase inhibitors and con-
ventional immunosuppresants)
[6]. The patho-
genic potential of anti-MuSK autoantibodies has
recently been demonstrated by their ability to
induce an MG-like muscle weakness in MuSK-
immunized rats or rabbits
[7,8] by severely hamper-
ing AChR clustering
[8]. Interestingly, a nonim-
munoglobulin (Ig)G plasma factor, which
transiently inhibits the function of AChR mole-
cules without reducing their number, has been
described for seronegative MG patients
[9]. The
unknown factor likely induces a desensitization-
like state acting directly on human AChR,
mimicking an allosteric modulator
[10].
Genetic predisposition to
myasthenia gravis
MG onset involves a genetically favorable back-
ground combined with several unidentified
environmental stimuli. Strong evidence for an
Table 1. Subgroups of seropostive myasthenia gravis patients.
Subtype
of MG
Age of onset Sex Thymic
pathology
HLA
association
Related autoantibodies
Early-onset < 40 years Mostly women Follicular
hyperplasia
B8, DR3 Possibly other tissue-specific Abs.
Late-onset > 40 years A bias for men Normal or
atrophic
DR7 Abs against muscle antigens, mainly titin
and the ryanodine receptor.
Paraneoplastic Usually 40–60
years
Equally men
and women
Thymoma No clear
association
Abs against muscle antigens and
cytokines (e.g., IFN-α and IL-12).
Abs: Antibodies; HLA: Human leukocyte antigen; IFN: Interferon; IL: Interleukin; MG: Myasthenia gravis.
Figure 1. Model showing part of the postsynaptic apparatus, including the
acetycholine receptor, in the neuromuscular junction.
Binding of neuronal agrin to its receptor complex initiates the signaling scaffold leading to AChR clustering
at the neuromuscular junction. MASK and MuSK may form the receptor complex of agrin. RATL remains an
unidentified molecule that likely links MuSK to rapsyn. Rapsyn is required for cross-linking of AChRs and
other post-synaptic components into large aggregates.
AChR: Acetycholine receptor; MASK: Myotube-associated specificity component; MuSK: Muscle-specific
kinase; RATL: Rapsyn-associated transmembrane linker.
Rapsyn
Extracellular
Intracellular
Channel pore
AChR
MuSK
Agrin
MASK
RATL
D
E
G
J
H
D
www.futuremedicine.com 801
Recent advances myasthenia gravis research – REVIEW
immunogenetic predisposition to the disease
was provided by the early implication of the
human leukocyte antigen (HLA) complex in
early-onset MG with thymic hyperplasia
(HLA-DR3 and B8) and late-onset MG
patients with anti-titin Abs (HLA-DR7),
whereas for the paraneoplastic MG subset, no
clear HLA association has currently been
described
[11].
The MYAS1 locus
Although the 8.1 ancestral haplotype, including
HLA-A1 and B8 alleles for class I and DR3 for
class II, has been reproducibly associated with
early-onset MG, the exact disease loci remained
unidentified. Garchon and colleagues initially
established a genetic linkage between MG and
an HLA locus, termed MYAS1
[11]. The MYAS1
locus was then mapped to a segment encompass-
ing the distal part of class III and the proximal
part of class I genes, containing the tumor
necrosis factor (TNF)/Lymphotoxin genes but
excluding the cluster of complement factor
genes
(Figure 2) [12]. Interestingly, the MYAS1
interval overlaps with a region implicated
recently in rheumatoid arthritis suggesting a
nonantigen-specific immunoregulatory function
for the 8.1 haplotype
[13].
Three-gene model
In a previous study, MG was associated with a
class II allele (unrelated to the 8.1 haplotype),
DQA1*0101, which encodes the
α-chain of the
class II molecule DQ
[14]. This allele was shown
to interact with a polymorphism of the
CHRNA1 gene, encoding the AChR
α-subunit [14]. Recent evidence supported the
HLA-dependent control of anti-AChR Ab titers
in MG patients with hyperplastic or normal thy-
mus. According to this three-locus model, two
loci, CHRNA1 and HLA-DQA1 interact to
result in high autoantibody titers, whereas the
third locus associated with the 8.1 haplotype
exerts an additive effect, possibly through non-
antigen-specific immune dysregulation
[15].
Whether this third locus is MYAS1 requires
further investigation.
Humoral & cellular immunity &
experimental autoimmune MG
Antigenic structure of acetylcholine receptor
In the 1980s, the identification of the main
immunogenic region (MIR) as the target for the
majority of anti-AChR monoclonal Abs (mAbs)
isolated from AChR-immunized experimental ani-
mals
[16], orientated the immunological research
towards the study of anti-AChR Abs and their
pathogenic mechanisms. It is now well-docu-
mented that the MIR is not a single epitope but
rather comprises a cluster of overlapping confor-
mation-dependent epitopes, with the core
sequence
α67–76, located extracellularly on the
α-subunit of AChR [17], without excluding that
neighboring AChR subunits also contribute to the
region
[18]. The MIR is of major pathological
importance, since anti-MIR mAbs very effectively
induce passively transferred EAMG and cause
AChR loss
[19]. In MG patient sera, approximately
50% of anti-AChR Abs recognize the
α-subunit
[20], while the content of serum anti-AChR Abs in
anti-MIR Abs has been determined only indi-
rectly, through competition studies, as 50–60%
[21]. Considerable serum Ab fractions recognize
various non-
α AChR subunits (at least β, γ and ε),
Figure 2. Schematic map of the human leukocyte antigen complex and localization of
the MYAS1 locus.
Bars at the top indicate the extent of the three main HLA classes. Landmark genes and the two microsatellite
markers that delimit the MYAS1 interval, BAT3 and C3.2.11, are shown.
Cen: Centromere; HLA: Human leukocyte antigen; Tel: Telomere; TNF: Tumor necrosis factor.
(Reproduced from Vandiedonck C, Giraud M, Garchon HJ: Genetics of autoimmune myasthenia gravis: the
multifaceted contribution of the HLA. 25(Suppl.), 6–11 J. Autoimmun. (© 2005) with permission
from Elsevier.
II III I
HLA class
DRB1 C4B TNF B C E A
BAT3 C3-2-11
Loci
cen tel
II III I
HLA class
DRB1 C4B TNF B C E A
BAT3 C3-2-11
Loci
cen tel
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
802 Future Neurol. (2006) 1(6)
as demonstrated directly through the binding of
MG Abs to recombinant extracellular domains
of these subunits expressed in a yeast expression
system
[Kostelidou K et al., Unpublished Results] [22]. Inter-
estingly, in a small portion of MG sera, Abs
against cytoplasmic AChR epitopes are also
detected
[23]; however, their significance
is unknown.
Effector humoral response
In both MG and EAMG, the anti-AChR Ab
response is polyclonal and heterogenic. The anti-
AChR autoantibodies destroy functional AChRs
by complement-dependent lysis of the postsynap-
tic membrane, increased AChR degradation via
antigenic modulation, or less commonly, by direct
inhibition of neurotransmitter binding. However,
the relative significance of these mechanisms in
individual patients has not yet been well-estab-
lished. The key role of complement in MG and
EAMG pathogenesis is corroborated by early and
recent studies demonstrating that complement
blockade/deficiency can protect animals against
induction of EAMG
[24–26]. Alternatively, the
absence of complement regulatory molecules,
such as the decay-accelerating factor (DAF), sig-
nificantly enhances susceptibility to EAMG, since
DAF
-/-
transgenic mice were shown to be signifi-
cantly more prone to EAMG induction than their
DAF
+/+
littermates [27]. Recently, Christadoss and
colleagues provided direct evidence for the impli-
cation of the classical (and not the alternative)
complement pathway in the elicitation of the
actively induced EAMG
[28]. The same group
associated EAMG severity with the level of C1q
conjugated-circulating immune complexes, which
are known to trigger the classical complement cas-
cade
[29], and further demonstrated an involve-
ment of the immune complex receptor Fc
γ
receptor III in EAMG pathogenesis [30].
B & T cell interaction through
costimulation molecules
Autoreactive CD4
+
T cells from MG patients
respond to various synthetic AChR peptides
from different AChR subunits in proliferation
or enzyme-linked immuno-spot (ELISPOT)
assays in the context of major histocompatibil-
ity complex (MHC) class II molecules
[31–33]
and provide help to B cells, thus becoming
essential for anti-AChR Ab production
[34].
Current immunological research addresses the
question of costimulation requirements for the
activation of AChR-specific T cells and
initiation of pathogenic Ab production.
Binding of the CD40 ligand (CD40L) to
CD40 triggers B cells to function as antigen pre-
senting cells (APC) and upregulates expression
of the costimulatory molecules B7-1 and -2,
which interact with CD28 and the cytotoxic
T lymphocyte-associated antigen (CTLA)-4. In
EAMG, CD40–CD40L interaction was found
to be essential for both disease induction and
progression, since CD40L
-/-
mice were com-
pletely resistant to EAMG demonstrating
severely compromised AChR-reactive B-cell and
T-helper (Th)1/Th2 responses
[35] and anti-
CD40L treatment during the course of an ongo-
ing EAMG still suppressed the disease, involving
a downregulation of B7-2 and the Th1 cytokines
interleukin (IL)-12 and interferon (IFN)-
γ and
an upregulation of CTLA-4
[36].
Concerning the B7-mediated signaling,
CD28
-/-
mice were less susceptible, but not com-
pletely resistant, to EAMG induction compared
with wild-type controls
[35], while in vitro blockade
of B7 signaling induced an anergy-like state in
AChR-specific T-cell lines
[37]. Christadoss and
colleagues demonstrated, using knockout mice,
that B7-1 (but not B7-2) deficiency significantly
decreased the anti-AChR Ab-production following
AChR immunization, likely perturbing T- and B-
cell communication rather than suppressing the
primary response to AChR
(Table 2) [38].
Hence, the B7-1 molecule, as well as the
CD40–CD40L interaction, appear to be sine qua
non for the costimulation events leading to the
production of anti-AChR Abs by B cells.
Recently, another member of the CD28 family,
the inducible costimulatory molecule (ICOS) was
implicated in EAMG pathogenesis. ICOS is likely
to be important for downstream stimulation of
the effector T-cell functions (cytokine production
and CD40L expression) that are crucial for the
initiation and maintenance of the B-cell response.
Genetic disruption of ICOS was shown to confer
resistance to actively induced EAMG
[39].
Effect of cytokines on
experimental autoimmune
myasthenia gravis pathogenesis
Cytokines have pleiotropic functions. Depend-
ing on the time, and place, of action they may
exert different effects on immune responses.
Various studies have explored the role of
cytokines on EAMG pathogenesis
(Table 3).
The cytokine IL-12 facilitates EAMG develop-
ment. Resistance of IL-12
-/-
mice to EAMG was
related to both decreased production of comple-
ment fixating anti-AChR Abs and elicitation of a
www.futuremedicine.com 803
Recent advances myasthenia gravis research – REVIEW
stronger anti-AChR Th2 response [40]. Unlike
IL-12, the role of IFN-
γ is less clear – IFN-γ recep-
tor knockouts exhibited significantly lower inci-
dence and disease severity than their wild-type
controls
[41] and IFN-γ deficiency conferred resist-
ance to IFN-
γ
-/-
mice [42]. However, in another
transgenic model of different genetic background,
IFN-
γ disruption did not affect EAMG induction
[40]. The proinflammatory cytokine IL-18 is also
involved in EAMG pathogenesis. IL-18
-/-
mice
were resistant to EAMG
[43], while treatment with
an anti-IL-18 Ab suppressed ongoing EAMG by
upregulating the immunosuppressive Th3
cytokine transforming growth factor
β [44].
The Th2 cytokines IL-4, -6 and -10 have also
been studied. Genetic disruption of IL-10 con-
ferred EAMG resistance to IL-10
-/-
mice [45], while
simultaneous transgenic expression of IL-2 and
-10 increased susceptibility to EAMG
[46]. These
findings suggest that IL-10 facilitates EAMG,
potentially by stimulating B-cell differentia-
tion/proliferation and Ig class switch. IL-6
-/-
mice,
as with the IL-10 knockouts, were significantly less
susceptible to EAMG induction than their wild-
type littermates, demonstrating a significantly
compromised germinal center formation,
decreased serum complement levels and impaired
class switching
[47]. A similarly defective class
switching was observed in AChR-immunized
knockout mice genetically deficient for TNF
receptor p55 and p75
[48] or for TNF-β [49]. These
animals were also resistant to EAMG. Christadoss
and colleagues proposed that a cytokine hierarchy
exists in the development of EAMG, with IL-6,
TNF and IL-18 playing the frontline role
[47]. It
remains to be clarified whether IL-6 and TNF act
in concert or whether one regulates the production
of the other. Genetically-deficient mice were also
used to investigate the role of IL-5 in EAMG.
Upon immunization with AChR, IL-5
-/-
mice
exhibited increased resistance to EAMG, com-
pared with wild-type controls, which was related
with reduced primary anti-AChR lymphocyte
response and reduced C3 complement levels in
muscle extracts
[50].
Unlike the other Th2 cytokines, IL-4 is not a
prerequisite for EAMG induction
[51]. After a sin-
gle immunization with To rp e d o AChR, IL-4
-/-
mice develop a chronic form of EAMG marked by
the persisting presence of anti-mouse AChR Abs
and mouse AChR-responsive CD4
+
T cells [52].
These findings underlie the protective role that
IL-4 possibly exerts by modulating/intercepting
anti-AChR Ab production and EAMG symptoms.
Signal transducer and activator of transcription
(STAT)4 and 6 molecules mediate the differentia-
tion of naive CD4
+
T cells into Th1 and 2 cells,
after interaction with IL-12 or -4, respectively. In
Table 2. The implication of costimulation molecules in experimental autoimmune myasthenia
gravis pathogenesis.
Interaction Experimental approach Mechanism of action Effects observed Ref.
CD40L–CD40
CD40L
-/-
mice
Severely compromised AChR-
reactive B-cell responses,
reduced Th1/Th2 responses
Complete resistance
[35]
Anti-CD40L Ab to rats Downregulation of B7–2, IL-12
and IFN-γ and upregulation
of CTLA-4
Suppression of
ongoing disease
[36]
B7–CD28
CD28
-/-
mice
Decreased AChR-specific Abs
of the IgG1 isotype, switch to
Th1 profile
Reduced susceptibility [35]
CD28 analog in AChR-specific
T-cell lines
Decreased IL-2 production and
lymphoproliferative responses
Anergy [37]
B7–1
-/-
and B7–2
-/-
mice
Significantly decreased anti-
AChR Ab production, likely due
to perturbed T- and B-cell
communication
Resistance [38]
ICOSL–ICOS
ICOS
-/-
Decreased anti-AChR Ab
production, inhibition of
Th1/Th2 differentiation in
response to AChR
Resistance
[39]
Abs: Antibodies; AChR: Acetycholine receptor; CTLA: Cytotoxic T-lymphocyte antigen; ICOS: Inducible costimulatory molecule; Ig: Immunoglobulin;
IL: Interleukin; IFN: Interferon; Th: T helper.
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
804 Future Neurol. (2006) 1(6)
STAT4
-/-
mice, all functions stimulated by IL-12
are impaired, including IFN-
γ production by Th1
cells, whereas STAT6
-/-
mice have defective IL-4-
mediated functions, such as B-cell proliferation
and Th2 cell development. A recent study, using
STAT4- or -6-deficient mice, demonstrated that
after AChR immunization, STAT6
-/-
mice were
significantly more susceptible to EAMG than the
STAT4
-/-
and wild-type animals [53]. The latter
suggests a prevalent role for a Th1 cell response in
EAMG pathogenesis, yet the implication of IL-10
and -6 (discussed above) denotes that the anti-
AChR autoimmune response likely involves a
hybrid Th1/Th2 polarization.
Role of the thymus in myasthenia
gravis pathogenesis
As mentioned previously, the thymus has been
implicated in the onset and/or maintenance of
MG due to the frequent coappearance of thymic
abnormalities with MG (50–60% of patients
present with thymic hyperplasia and 10% with
thymoma), and to the generally beneficial effect
of thymectomy on patients’ clinical profile
[54].
The phenotypically distinct entities (early-onset,
late-onset and paraneoplastic MG) may underlie
the existence of several etiological routes for
AChR autoimmunity, manifested with uniform
clinical symptoms. Therefore, hyperplastic thy-
mus and thymoma are generally thought to
contribute differently to disease pathogenesis.
Hyperplastic thymus
A predominant characteristic of thymic hyper-
plasia is the massive B- and T-lymphocyte infil-
tration in the thymic medulla, where both
single AChR subunits and intact AChR mole-
cules are normally expressed by thymic epithe-
lial cells (TEC)
[55] and myoid cells [56]. Plasma
cells, which spontaneously produce anti-AChR
Table 3. The effect of cytokines in experimental autoimmune myasthenia gravis pathogenesis.
Cytokine Experimental approach Mechanism of action Effects observed Ref.
IL-12
IL-12
-/-
mice
Decreased production of
complement fixating anti-
AChR Abs, elicitation of a
stronger anti-AChR
Th2 response
Resistance
[40]
IFN-γ
IFN-γ R
-/-
mice
Low anti-AChR Abs Reduced
susceptibility
[41]
IFN-γ
-/-
mice
(of H-2[b] haplotype)
Reduction of anti-AChR Abs Resistance [42]
IFN-γ
-/-
mice (C57BL6)
No effect [40]
IL-18
IL-18
-/-
mice
Reduction of anti-AChR Abs
and Th1 response
Resistance
[43]
Anti-IL-18 Ab to rats Upregulation of TGF-β,
decreased Th1 response
Suppression of
ongoing disease
[44]
IL-10
IL-10
-/-
mice
Stimulation of B-cell
differentiation/proliferation
and Ig class switch
Resistance [45]
Transgenic mouse model
expressing IL-10 under the
IL-2 promoter
Stimulation of B-cell
differentiation/proliferation
and Ig class switch
Increased
susceptibility
[46]
IL-6
IL-6
-/-
mice
Significantly compromized
germinal center formation,
decreased complement levels,
and impaired class switching
Reduced
susceptibility
[47]
IL-5
IL-5
-/-
mice
Reduced primary anti-AChR
lymphocyte response and C3
complement levels in
muscle extracts
Reduced
susceptibility
[50]
IL-4
IL-4
-/-
mice
No effect
[51]
Abs: Antibodies; AChR: Acetylcholine receptor; IFN: Interferon; Ig: Immunoglobulin; IL: Interleukin; TGF: Transforming growth factor;
Th: T-helper cell.
www.futuremedicine.com 805
Recent advances myasthenia gravis research – REVIEW
Abs, have been isolated from thymic germinal
centers derived from early-onset MG patients
[57], in addition to AChR responsive CD4
+
T
cells, which were shown to contain a poten-
tially auto-reactive subpopulation displaying
increased Fas expression
[58]. Hence, molecules
promoting T- and B-cell survival and migra-
tion are likely related with pathogenesis of
thymic hyperplasia. In fact, the B-cell differenti-
ation/antiapoptotic cytokine IL-6 and the regu-
lator of transepithelial T-cell migration
RANTES, are over-expressed in MG thymic
epithelium. This results from the constitutive
over-expression (by TEC) and activation of the
p38 and extracellular regulated kinase 1/2
mitogen-activated protein kinases (MAPK),
suggesting that a dysfunction in MAPK tran-
scriptional or post-transcriptional control may
initiate alterations in myasthenic thymus
morphology with pathological impact
[59].
TEC and myoid cells may also be implicated in
the primary autosensitization step in MG. Levin-
son and collaborators proposed that an inflamma-
tory reaction in the hyperplastic thymus
modulates the expression of AChR and that of
several immune components (MHC and costim-
ulatory molecules), and facilitates the re-entry
into the thymus of autoreactive CD4
+
Tcells that
escaped central deletion
(Figure 3) [60]. AChR
expression and presentation within this activated
environment, combined with a genetically suscep-
tible background, could be adequate to prime the
AChR-responsive CD4
+
T cells, leading to
autoimmunity
[60]. Evidence provided by Berrih-
Aknin and colleagues supports this hypothesis:
firstly a large number of IFN-
γ- and TNF-α-regu-
lated genes, including MHC II genes, are highly
expressed in the myasthenic thymus, suggesting
the existence of a proinflammatory environment;
and, secondly, AChR expression in the thymus is
significantly susceptible to IFN-γ and TNF-
α
regulation [61]. However, the initial stimulus
inducing the inflammatory environment remains
unidentified, although bacterial and viral agents
are likely candidates.
Apart from the autoimmunity-favorable micro-
environment in the hyperplastic MG thymus,
additional control mechanisms must fail in order
for the disease to develop. Indeed, the regulatory
CD4
+
CD25
+
Tcells, although normal in
number, were found to be severely defective in
their ability to suppress autoreactive CD4
+
Tcells,
and demonstrated a significantly decreased
expression of the Foxp3 transcription factor that
is essential for the regulatory T-cell function
[62].
Interestingly, in the case of thymic hyperplasia
with diffuse B-cell infiltration (thymitis), as well as
in involuted thymus, Toll-like receptor (TLR)4
mRNA levels were shown to be significantly up-
regulated when compared with expression in ger-
minal center hyperplastic thymus and thymoma,
suggesting the existence of a relationship between
innate immunity and MG. An attractive theory
would be that TLR4-mediated signaling is initiated
in response to an exogenous or intrinsic alarm stim-
ulus, which activates the innate immune system,
eventually contributing to autoimmunity
[63].
Thymoma
Thymomas are thymic epithelial tumors fre-
quently associated with paraneoplastic auto-
immunity. Approximately 30% of all
thymoma patients develop MG, their auto-
antibody repertoire comprising Abs against
AChR and other muscle antigens, including
actin, myosin, the ryanodine receptor and
especially titin
[64]. MG-associated thymomas
share several common features:
Intratumorous thymopoiesis and maturation
of potentially autoreactive T cells
[65];
Reduced expression of MHC class II
molecules
[66–68];
Intratumorous enrichment in AChR-respo-
sive T cells
[69] restricted to minority HLA
isotypes, not commonly found in non-
thymomatous MG patients
[70];
Export of mature T cells to the periphery
[71].
In fact, a strong correlation exists between para-
neoplastic MG and the ability of thymomas to
produce and export mature naive CD4
+
T cells,
given the significantly increased levels of this
T-cell subset in MG-positive compared with MG-
negative thymomas
[72].
Although no myoid cells or intact AChR are
present in thymomas, single receptor subunits have
been detected. However, it is disputable whether
thymomas actively sensitize
[70,71] or just fail to tol-
eralize
[67] the newly-produced auto-reactive
thymocytes against AChR. In any case, given that
B cells are generally rare in thymomas, a humoral
immune response should occur in the periphery.
Interestingly, neutralizing autoantibodies against
IFN-
α and IL-12 are found commonly in paraneo-
plastic MG, potentially affecting Th1 polarization
of the autoimmune response. Anti-IFN-
α Abs
have been isolated from MG thymomas, suggest-
ing that active priming against these cytokines
actually occurs within the thymoma
[73].
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
806 Future Neurol. (2006) 1(6)
According to a model for paraneoplastic MG
pathogenesis proposed by Marx and colleagues,
thymomas produce and export incompletely
toleralized, and thus potentially auto-reactive,
mature naive T cells
[74]. When exported to the
periphery, the thymoma-derived nontolero-
genic T cells may gradually convert the physio-
logically tolerant T-cell subset into a mixed
autoimmunity-susceptible T-cell repertoire origi-
nating from both the thymus and the thymoma
(Figure 4). In this case also, an as yet unidentified
stimulus must occur to activate the potentially
autoreactive Tcells in order for them to provide
help to autoantibody-producing B cells outside the
thymoma. Interestingly, the anti-IFN-
α and
anti-IL-12 autoantibodies discussed above
could act to facilitate the T-cell dependent
production of anti-AChR autoantibodies
[74].
Role of muscle in myasthenia
gravis pathogenesis
Muscle is the target organ of the autoimmune
response in MG. Numerous studies indicate that
far from staying inert before autoimmune attack,
the muscle actively participates in shaping the
eventual disease symptoms by appropriately adapt-
ing AChR expression and secreting/receiving
immunomodulating messages.
Figure 3. New model for intrathymic pathogenesis of myasthenia gravis related with thymus hyperplasia.
An inflammatory reaction in the hyperplastic thymus modulates the expression of AChR and several other immune components, mostly
MHC and costimulatory molecules, (2) and facilitates re-entry into the thymus of auto-reactive CD4
+
T cells (3) that have escaped central
deletion (1). Enhanced AChR expression and presentation within this activated environment, combined with a genetically susceptible
background, could be adequate to prime the AChR-responsive CD4
+
T cells (4), leading to autoimmunity.
AChR: Acetylcholine receptor; MHC: Major histocompatibility complex; TEC: Thymic epithelial cell.
Based on
[60].
Thymic medulla
P
2. Inflammatory e
4.
AChR
the thymus.
www.futuremedicine.com 807
Recent advances myasthenia gravis research – REVIEW
In the case of antigenic modulation, mRNA lev-
els of muscle AChR subunits increase, as demo-
nstrated both in cell culture and MG patient
muscle biopsies, likely following a mechanism
attempting to counterbalance AChR loss
[75]. In
passively transferred EAMG, muscle AChR
expression is upregulated after the administration
of TNF-
α (but not IFN-γ) [61]. The differential
effect of IFN-
γ on thymic and muscle AChR tran-
scripts likely suggests a different pathway regulat-
ing AChR expression in both the thymus and
muscle. Additionally, upon in vivo or in vitro expo-
sure to anti-AChR Abs, rat skeletal muscle cells
respond by producing several immunomodulating
factors such as the disease-promoting molecules
IFN-γ, IL-1, -15, the monocyte chemoattractant
protein-1 chemokine
[76,77] and nitric oxide that
acts protectively
[78]. A recent study demonstrated
that muscle in EAMG and MG also over-pro-
duces the IFN-γ-inducible protein (IP)-10 chemo-
kine and its receptor, CXCR3. Expression of these
molecules is suppressed in experimental animals
after induction of mucosal tolerance using an
AChR fragment
[79]. Since IP-10 and CXCR3
mediate inflammatory cell recruitment, yet no
signs of inflammation are observed in myasthenic
muscle, their role in disease pathogenesis remains
enigmatic
[79].
Figure 4. Proposed model for pathogenesis of paraneoplastic myasthenia gravis.
Thymomas produce and export nontoleralized, potentially autoreactive mature naive CD4
+
T cells that, upon
entering the periphery, gradually convert the physiologically tolerant T-cell subset into a mixed
autoimmunity-susceptible T-cell repertoire. Within this context, thymoma-derived mature naive CD4
+
T cells
are primed against AChR presented by APCs or DCs (although the site of sensitization or the initiating
stimulus remain unknown) and help autoantibody-producing B cells outside the thymoma.
AChR: Acetylcholine receptor; APC: Antigen presenting cell; DC: Dendritic cell.
Based on
[74].
Thymoma
Periphery
Reduced MHC II expression
Altered MHC II restriction
Impaired negative selection
Immature
Tcell
Mature naive
CD4
+
Tcell
Mature naive
CD4
+
T cells
(from thymoma)
+
Unidentified
stimulus
Activatio
Autoantibody-s
Bcell
Export to
the periphery
Abnormal selection
Mixed autoimmunity-prone
CD4
+
population
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
808 Future Neurol. (2006) 1(6)
An emerging new scenery for
myasthenia gravis therapeutics
A greater insight into the mechanisms governing
MG autoimmunity lead to the design of novel,
immune-directed therapeutic strategies. These
include both new-generation immunosuppres-
sive agents and purely innovative approaches cre-
ated upon the MG-specific demands.
Current treatment
Current medications control MG by improving
neuromuscular transmission or downmodulating
immune function. Acetylcholinesterase inhibi-
tors, most commonly pyridostigmine (Mesti-
non
®
) and neostigmine (Prostigmin
®
) remain
the first-line of MG treatment. Both enhance
muscle strength by increasing acetylcholine levels
in the NMJ, but their effect often diminishes
after some months of administration.
Immunosuppressive treatment includes corti-
costeroids (mainly prednisolone), which are
usually effective in reducing anti-AChR anti-
body titer, but induce serious side effects (i.e.,
weight gain, hypertension osteoporosis) and
nonsteroidal immunosupressants, usually aza-
thioprine, which inhibit lymphocyte prolifera-
tion, have fewer side effects, but require a
longer period to act
[80]. Azathioprine is most
often used together with corticosteroids. Myco-
phenolate mofetil (CellCept), a potent inhibi-
tor of lymphocyte proliferation, has recently
been introduced in MG treatment and gener-
ally proved effective and well-tolerated
[81]. Tac-
rolimus (IL-2-suppressor) and Rituximab
®
(Ab
against the B-cell surface-marker CD20), which
are widely used in transplantation medicine and
lymphoma treatment, respectively, are also can-
didates for study in MG patients, although
there is some skepticism on their usefulness in
view of their toxicity.
Surgical treatment of MG involves thymec-
tomy. The effectiveness of thymecomy is well-
established for early-onset MG, but its thera-
peutic value is less obvious for late-onset MG
patients, while postsurgical clinical improve-
ment is even less common for thymomatous
MG patients. Controlled prospective studies on
this issue would provide evidence clarifying the
therapeutic benefits of thymectomy for the
three subsets of MG patients
[82].
In the case of severely affected MG patients,
plasmapheresis and administration of intrave-
nous immunoglobulins (IVIGs) produce a
direct, albeit short-term, improvement. Addi-
tionally plasmapheresis presents the severe
drawback of removing protective Abs and
plasma components, thus necessitating the use
of replacement fluids, with the risk of infection
and allergic reactions
[83]. Due to these disad-
vantages, plasmapheresis is mostly considered as
a second-line treatment and IVIG, which pro-
duces significantly fewer adverse effects, is gen-
erally preferred. Although not yet clearly
understood, IVIG action lies on different levels
of the autoimmune response: pathogenic
autoantibody production, complement activa-
tion, cytokine profile and activation/effector
functions of T and B cells
[84].
Experimental approaches
The experimental therapeutic strategies currently
under investigation aim to eliminate the auto-
immune response, whilst bypassing generalized
immunosuppression.
Phospodiesterase inhibitors
Recently, Fuchs and colleagues revealed an
involvement of phosphodiesterases in EAMG
pathogenesis and tested the effect of the phos-
phodiesterase inhibitor pentoxifylline on
EAMG progression. Pentoxifylline administra-
tion during the acute or chronic stage of the dis-
ease repressed EAMG progression. Humoral and
cellular anti-AChR responses were down regu-
lated, phosphodiesterase and TNF-
α expression
were decreased in lymph node cells and muscle,
as well as IL-18, -12 and -10 (in lymph nodes).
Remarkably, expression of the Foxp3 transcrip-
tion factor was upregulated, suggesting an
enhanced function of the CD4
+
CD25
+
regula-
tory T cells. These findings, together with the
fact that phosphodiesterase inhibitors are
already an approved treatment for other disor-
ders, makes them very promising candidates for
MG immunotherapy
[85].
Tolerance induction
Induction of mucosal (oral or nasal) tolerance
using various AChR-derived molecules, (native or
denatured AChR, recombinant AChR fragments,
AChR peptides or altered peptides) has been tested
with some encouraging results. This approach
appears to suppress animal disease by altering the
polarization of the anti-AChR autoimmune
response. The shift in Th-cell subsets greatly
depends on the tolerogen (source, conformation
and size) and the route of administration (mucosal
exposure rather than injection). Fuchs and col-
leagues have demonstrated that mucosal adminis-
tration of recombinant fragments of the human
www.futuremedicine.com 809
Recent advances myasthenia gravis research – REVIEW
AChR α-subunit extracellular domain suppressed
ongoing EAMG at the acute and even chronic
phases of the disease, as assessed by clinical score,
decreased anti-AChR Ab titers and reduced lym-
phoproliferation in response to AChR
[86,87],
while Link and colleagues used To r pe do AChR as
the tolerogen
[88]. Mucosal tolerance likely acts by
introducing a transmission from Th1 to a mixed
Th2/Th3 regulation
[86,87]. Alternatively, oral
administration of a syngeneic recombinant frag-
ment of the rat AChR
α-subunit also sup-
pressed ongoing EAMG in rats, although via a
different mechanism (a shift from Th1 to Th2
regulation)
[89]. Regarding the syngeneic frag-
ment use for tolerance induction, Baggi and
colleagues reported a breakdown of tolerance to
a self-rat AChR
α-subunit peptide, leading to
EAMG in rats
[90]. However, this pathogenic
effect is likely to be related to the route of pep-
tide administration, which was injection
instead of mucosal treatment.
T-cell receptor vaccination
As discussed previously, early-onset MG mostly
comprises female patients with thymic hyper-
plasia displaying the HLA-DR3 haplotype. Inter-
estingly, HLA-DR3 positive MG patients were
reported to exhibit a bias for the V
β5.1 TCR in
their thymic CD4
+
T cells and to have elevated
anti-V
β5.1 Abs, which tend to act regulatorily
and are related with low disease severity and posi-
tive prognosis
[91]. These data could provide two
alternatives for TCR vaccination in MG:
•Use of Vβ5.1-derived peptides to boost the
spontaneously-produced response against
autoreactive T cells;
Administration of anti-Vβ5.1 Abs to directly
suppress autoreactive T-cell function.
In fact, an anti-Vβ5.1 Ab has already been
tested in rat EAMG with positive results
[92],
although application to humans would require
human or humanized anti-Vβ5.1 Abs. However,
there is some skepticism regarding TCR vaccina-
tion for MG treatment, which derives mainly
from the issue of TCR usage restriction in MG
per se, since there are studies challenging the
Vβ5.1 T-cell ‘prevalence’ in the thymus of MG
patients
[93].
Manipulated antigen presenting cells
Different strategies involving manipulated
APCs are currently under investigation. Drach-
man and colleagues used viral vectors to convert
APCs into ‘guided missiles’ that specifically
eliminated AChR-specific T cells. The geneti-
cally engineered APCs effectively present the
α-subunit extracellular domain of AChR and
express Fas ligand to selectively interact with
activated AChR-specific T cells and destroy
them via apoptosis
[94]. Alternatively, dendritic
cells (DCs) from EAMG rats, upon in vitro
exposure to IL-10 and subsequent injection,
hindered ongoing EAMG
[95]. Their action was
related with the downregulation of costimula-
tory molecules and decreased anti-AChR Ab
production, likely through blockade of IL-10
expression. These data create expectations for
trials of modified autologous DCs in humans,
given that DC vaccination is already applied for
other conditions, such as cancers.
Acetylcholinesterase gene silencing using
antisense oligonucleotides
In a recent study, acetylcholinesterase expression
was abrogated using antisense oligonucleotides
targeting the acetylcholinesterase mRNA gene
[96].
After treatment with the acetylcholinesterase gene
antisense oligonucleotides, the clinical profile of
rats with EAMG was significantly improved. In
fact, the antisense approach was shown to be more
effective than pyridostigmine, presenting the addi-
tional advantage of selectively blocking alterna-
tively spliced acetylcholinesterase variants.
Nevertheless, an issue arises of how this method
could be applied in humans.
Protective antiacetylcholine receptor
antibody fragments
The use of nonpathogenic univalent human
anti-AChR Ab fragments that bind to AChR,
thereby preventing the binding of pathogenic
autoantibodies, offers another option for MG
treatment. A number of human or humanized
anti-AChR Ab fragments have already been
developed
[97–100]. Among them, two sets of
human anti-AChR Fabs, one against the MIR
and the other against an immunodominant
region close to the MIR, effectively compete
with a great majority of MG patients sera for
binding to AChR
[97,99]. Their combination
could prove useful for short-term therapy, for
example, in life-threatening crises. Attempts to
prolong their in vivo half-life are in progress.
Anti-AChR Ab-specific plasma clearance
An affinity column bearing the AChR or appro-
priate AChR fragments to selectively withhold
the anti-AChR Abs allowing the return of ‘anti-
AChR Ab-cleared’ plasma to the patient would
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
810 Future Neurol. (2006) 1(6)
overcome the major disadvantage of plasma
exchange, namely the indiscriminate removal of
protective Abs together with the pathogenic
ones. Such a column was introduced by
Takamori and colleagues who used the To rp ed o
AChR
α-subunit synthetic peptide 183–200,
containing part of the acetylcholine binding site
[101]. However, the immunoadsorption efficiency
of such columns bearing small, nonhuman, syn-
thetic peptides, is likely inadequate, as the
majority of anti-AChR Abs in MG patients are
highly conformation dependent.
Our group has developed a specific Ab-
apheresis therapy for MG, through the con-
struction of ‘immunoadsorbent’ columns car-
rying AChR fragments of native-like
conformational features. We have successfully
expressed the extracellular domains (ECDs,
amino acids 1–210) of the human AChR
α, β,
γ and ε subunits in water-soluble form using
the yeast Pichia pastoris expression system
[22,102]. Subsequent immobilization of each
ECD on an insoluble carrier (CNBr-Sepha-
rose) led to the construction of ‘immuno-
adsorbent’ columns, which were tested for the
feasibility of selective ex vivo elimination of
patients’ anti-AChR Abs. Immunoadsorption
experiments performed with
α-ECD-Sepha-
rose demonstrated that, on average, 35% of
autoantibodies were adsorbed from a set of 64
random MG positive sera
[22,103]. Preliminary
experiments with each of the non-
α-ECD-
Sepharoses and the same set of sera yielded dif-
ferent average immunoadsorption percentages
of autoantibodies by each ECD as follows:
22% by
β-ECD, 21% by γ-ECD and 18% by
ε-ECD, while the combination of all four sub-
units (
α, β, γ, ε) demonstrated that they could
act in an additive fashion or could at least
remove a considerably higher percentage of
autoantibodies than that of any individual
immunoadsorbent
(Figure 5) [Kostelidou et al., Unpub-
lished Data]
. These results suggest that the com-
bined use of all subunits for the preparation of
an immunoadsorbent column could indeed
result in the specific removal of the majority of
autoantibodies from a large portion of MG
sera. A drawback of this approach is that elimi-
nation of the anti-AChR Abs may result in
compensatory overproduction of novel Abs.
Yet such overproduction should be milder than
that occurring in plasmapheresis, where most
Igs are removed, including possible T-cell
inhibitory Igs. The characterization of the
properties of the
α-ECD-immunoadsorbent
column (which are currenly being evaluated on
other ECD-Sepharose matrices and appear to be
consistently preserved) suggested that a matrix
(Sepharose) column with approximately 5 mg of
immobilized polypeptides would suffice to
immunoadsorb most of the anti-AChR autoanti-
bodies from a typical MG serum in a short period
of time. The column could be recycled, thus ren-
dering this technique a feasible, antigen-specific,
therapeutic approach
[22,103].
Conclusion
During the previous decade, we witnessed a turn
of the immunological research on MG towards
the study of the immunological mechanisms,
cellular and molecular, which trigger/maintain
the disease and determine its severity.
Overall, significant progress has been made,
especially regarding MG etiology. A severely
impaired function of the CD4
+
CD25
+
regulatory
T cells to suppress autoreactive CD4
+
T cells was
identified, while the hyperplastic thymus appears
to be involved in MG pathogenesis providing a
hyperactive proinflammatory microenvironment
that is prone to breakage of tolerance to AChR.
What is now greatly anticipated is the identifica-
tion of the stimulus creating this autoimmunity-
nesting milieu. Interestingly, muscle, the target-
organ of the anti-AChR response, appears to
actively contribute in shaping the final MG
symptoms by appropriately adapting AChR
expression and secreting/receiving immuno-
modulating messages, such as chemokines. The
role of these molecules in MG is puzzling, since
chemokines are mainly recruiters of inflammatory
cells, yet no signs of inflammatory cell infiltration
are observed in myasthenic muscle.
Based on recent findings, several approaches
that aim to selectively control the anti-AChR
response without suppressing the overall
immune system, have been rationally designed
Although still experimental, encouraging results
for some of them (phosphodiesterase inhibitors,
manipulated autologous DCs and anti-AChR
Ab-specific apheresis) create expectations for
future trials in humans.
Future perspective
Several issues regarding MG etiology remain
obscure, most importantly the triggering agent.
Although viral or bacterial antigens have been
implicated occasionally, no definite associations
were established. Additionally, muscle-secreted
chemokines have appeared recently in MG
bibliography, but their role is unidentified. The
www.futuremedicine.com 811
Recent advances myasthenia gravis research – REVIEW
involvement of chemokine signaling in MG is a
new, open field, which may soon reveal novel
aspects of disease pathogenesis.
It is obvious that not every therapeutic stra-
tegy that has proven successful in experimental
models will necessarily be effective in humans.
However, recent data from several experimen-
tal approaches likely justifies the initiation of
carefully designed clinical trials in human
patients. Given the low prevalence of the
disease, joint approaches involving patients
from multiple countries will likely be required
and the creation of an international MG registry
could prove very useful to promote this
move. Pentoxifylline seems a strong candidate
for clinical trials, given its encouraging results in
EAMG animals, and already being an approved
medication for several other conditions (mostly
circulatory disorders).
Additionally, our studies demonstrate that
ex vivo serum clearance from anti-AChR Abs
using specific plasma exchange is an effective and
feasible approach that could be applied relatively
easily in clinical practice. Further optimization
of ongoing experimental approaches will provide
new options for specific MG treatment.
Figure 5. Immunoadsorption of anti-acetycholine receptor antibodies from myasthenia gravis sera using
CNBr-sepharose immobilized extracellular domains.
Samples from myasthenia gravis sera (20 fmoles) were incubated with excess of Sepharose-immobilized extracelllular domains (ECDs)
(1 g). The unbound anti-acetylcholine receptor antibodies present in the supernatants were measured by RIA and the percentage of ECD-
absorbed anti-AChR Abs was calculated. Each bar shows the percentage of absorbed Abs by the specified ECD. (A) Immunoadsorption
of anti-AChR Abs from 64 MG sera using Sepharose-ECD. Sera are sorted in decreasing percentage of immunoadsorption
[103]. (B)
Immunoadsorption of anti-AChR autoantibodies from 7 MG sera, using immobilized
α, β, γ and ε-ECDs (first four columns) or a
combination of all the four subunits (last column). The combined use of all ECDs consistently removes more Abs than any individual ECD,
although this is often lower than the sum of individual ECD columns (derived from
[22]).
0
10
20
30
40
50
60
70
80
90
100
58
147
10 13 16 19 22 25 28 31 34 37 40 43 46 49 52 55 61 64
Sera
A
0
10
20
30
40
50
60
70
80
1234567
B
Sera
% Immunoadsorption
% Immunoadsorption
D
E
J
H
Total
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
812 Future Neurol. (2006) 1(6)
Acknowledgements
Original work of the authors was supported by grants from the
QoL program of the European Commission, the Muscular
Dystrophy Association, the Association Française contre
les Myopathies and the Greek General Secretariat of Research
and Technology.
Executive summary
Background
Myasthenia gravis (MG) is a T-cell-dependent autoimmune disease-mediated by autoantibodies, mostly against muscle nicotinic
acetylcholine receptor (AChR), a ligand-gated ion-channel of the neuromuscular junction (NMJ).
The anti-AChR autoantibodies cause loss of functional AChR molecules at the NMJ, resulting in weakness and fatigability of
voluntary muscles.
A small fraction of MG patients have antibodies (Abs) to the muscle-specific kinase (MuSK), a key-player molecule in NMJ
differentiation, rather than to the AChR.
The phenotypic heterogeneity of myasthenia gravis
Ocular MG: where the disease remains localized to the extraocular muscles.
Generalized MG: where the disease spreads to other skeletal muscles. Depending on the presence or absence of anti-AChR Abs,
it is subdivided in seropositive or seronegative MG, respectively.
Seropositive MG (~85% of MG patients) comprises: (i) early-onset MG (age <40 years) affects mostly women and is commonly
related with thymic hyperplasia; (ii) late-onset MG (age >40 years) affects people with normal or atrophic thymus; and (iii)
paraneoplastic MG associated with thymoma (age 40–60 years old) characterized by the presence of Abs to other striated muscle
antigens, mainly titin.
Seronegative MG represents approximately 15% of patients, approximately 40% of which have autoantibodies against MuSK of
probable pathogenic significance.
Genetic predisposition to MG
Early-onset MG has been associated with the human leukocyte antigen (HLA)-DR3 and B8, and late-onset MG with the HLA-DR7
(for paraneoplastic MG, no clear HLA association has been described).
Recently, a genetic linkage was established between MG and the MYAS1 HLA locus. Additionally, an HLA-dependent control of
anti-AChR Ab titers in MG patients with hyperplastic or normal thymus was proposed.
Antigenic structure of acetylcholine receptor
The main immunogenic region (MIR) of the AChR is a major target of anti-AChR Abs from experimental animals and MG patients.
It comprises a cluster of overlapping conformation-dependent epitopes, with the core sequence α67–76, located extracellularly
on the α-subunit of AChR.
The MIR is of major pathological importance in experimental autoimmune MG, since anti-MIR monoclonal Abs very effectively
induce passively transferred experimental autoimmune myasthenia gravis and cause AChR loss. A major pathological significance
of MIR is also suggested in MG.
Antibodies to other AChR subunits are also present in MG sera.
Effector humoral response in MG
Among the three Ab-mediated pathogenic mechanisms of AChR loss in NMJ, namely complement-dependent lysis of the
postsynaptic membrane, increased AChR degradation via antigenic modulation and direct inhibition of neurotransmitter binding,
the classical complement pathway likely plays the major pathogenic role.
B- & T-cell interaction through costimulation molecules
Autoreactive CD4
+
T cells from MG patients recognize various AChR epitopes in the context of major histocompatibility complex
(MHC) class II molecules and help B cells, becoming essential for anti-AChR Ab production.
CD40–CD40 ligand interaction and B7-mediated signaling, especially the B7-1–CD28 interaction, are essential both for EAMG
induction and progression. The inducible costimulatory molecule is also implicated in EAMG pathogenesis.
The effect of cytokines on EAMG pathogenesis
Interleukin (IL)-12 and -18 facilitate EAMG development, while the role of interferon (IFN)-γ is less clear. The T-helper (Th)2
cytokines IL-10, -6 and -5 also promote EAMG pathogenesis, while IL-4 likely acts protectively.
Overall, the Th1 cell response has a predominant pathogenic role in EAMG (and possibly MG), yet the involvement of IL-10 and -6
in disease pathogenesis may denote the implication of a mixed Th1/Th2 anti-AChR autoimmune response.
www.futuremedicine.com 813
Recent advances myasthenia gravis research – REVIEW
Hyperplastic thymus & MG pathogenesis
Hyperplastic thymus is characterized by: (a) massive B- and T-lymphocyte infiltration in the thymic medulla, where AChR is
normally expressed by thymic epithelial cells and myoid cells, and (b) the presence of AChR-responsive CD4
+
T cells and plasma
cells spontaneously producing anti-AChR Abs.
It likely offers an autoimmunity-susceptible hyperactive inflammatory environment over-expressing IFN-γ- and tumor necrosis
factor (TNF)-α-regulated molecules (AChR, MHC and costimulatory molecules) and facilitating the return of autoreactive CD4
+
Tcells that escaped central deletion. The initial stimulus inducing this inflammatory environment remains unidentified.
Regulatory CD4
+
CD25
+
T cells in hyperplastic thymus, although normal in number, are severely defective in their ability to
suppress autoreactive CD4
+
T cells, as evaluated with a specific mitogenic assay.
Thymoma & MG pathogenesis
MG-associated thymomas are thymic epithelial tumors characterized by: (a) intratumorous enrichment in AChR-resposive T cells;
(b) intratumorous thymopoiesis and maturation of potentially autoreactive T cells; and (c) ability to export mature T cells to
the periphery.
It is still unclear whether thymomas actively sensitize the newly-produced autoreactive thymocytes against AChR or just fail to
effectively toleralize them.
Neutralizing autoantibodies against IFN-
α and IL-12 commonly found in paraneoplastic MG suggest active priming against these
cytokines within the thymoma.
The role of muscle in myasthenia gravis pathogenesis
Muscle actively participates in shaping the eventual disease symptoms by adapting AChR expression and secreting/receiving
immunomodulating messages.
Muscle AChR mRNA levels are upregulated in cell cultures and MG patients as a result of AChR loss due to antigenic modulation,
and in passively transferred EAMG after administration of TNF-
α (but not IFN-γ).
Upon in vivo or in vitro exposure to anti-AChR Abs, rat skeletal muscles respond by producing several immunomodulating factors,
such as the disease-promoting molecules IFN-
γ, IL-1, -15 and several chemokines, or nitric oxide that acts protectively.
Current myasthenia gravis treatment
Current medications control MG by improving neuromuscular transmission or downmodulating the immune system
function. They include acetylcholinesterase inhibitors and steroid or nonsteroid immunosuppressants, while in severely
affected patients, administration of intravenous immunoglobulins or plasmapheresis are recommended. Surgical MG treatment
consists in thymectomy.
Experimental therapeutic approaches
The aim of experimental therapeutic strategies is to eliminate the autoimmune response without suppressing the overall immune
system.
Administration of phosphodiesterase inhibitors, induction of oral or nasal tolerance using AChR fragments, vaccination with T-cell
receptor peptides or manipulated autologous dendritic cells, administration of AChR-protective Ab fragments and use of a plasma
exchange approach for selective anti-AChR Ab clearance provide encouraging results.
Conclusion
During the last decade, immunological research on MG has focused on studying the immunological mechanisms, cellular and
molecular, which trigger/maintain the disease and determine its severity.
Although significant progress has been made regarding MG etiology, the initial disease-inducing signal remains unidentified,
while several other issues, for example the role of chemokines in MG pathogenesis, are open for investigation.
Promising results from several experimental therapeutic approaches create expectations for future trials in humans.
Executive summary
Bibliography
Papers of special note have been highlighted as
either of interest (•) or of considerable interest (••)
to readers.
1. Poulas K, Tsibri E, Kokla A et al.:
Epidemiology of seropositive myasthenia
gravis in Greece. J. Neurol. Neurosurg.
Psychiatry 71, 352–356 (2001).
2. Lindstrom JM: Acetylcholine receptors and
myasthenia. Muscle Nerve 23, 453–477
(2000).
•• Excellent overview of myasthenia
gravis (MG).
3. Hoch W, McConville J, Helms S et al.:
Auto-antibodies to the receptor tyrosine
kinase MuSK in patients with myasthenia
gravis without acetylcholine receptor
antibodies. Nat. Med. 7, 365–368 (2001).
•• Identifies muscle-specific kinase (MuSK) as
the autoantigen in a significant portion of
seronegative MG patients.
4. Evoli A, Tonali PA, Padua L et al.:
Clinical correlates with anti-MuSK
antibodies in generalized seronegative
myasthenia gravis. Brain 126, 2304–2311
(2003).
5. Zhou L, McConville J, Chaudhry V et al.:
Clinical comparison of muscle-specific
tyrosine kinase (MuSK) antibody-positive
and -negative myasthenic patients.
Muscle Nerve 30, 55–60
(2004).
6. Vincent A, Bowen J, Newsom-Davis J,
McConville J: Seronegative generalised
myasthenia gravis: clinical features,
antibodies, and their targets.
Lancet Neurol. 2, 99–106
(2003).
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
814 Future Neurol. (2006) 1(6)
7. Jha S, Xu K, Maruta T et al.:
Myasthenia gravis induced in mice by
immunization with the recombinant
extracellular domain of rat muscle-specific
kinase (MuSK). J. Neuroimmunol. 175,
107–117 (2006).
•• Demonstrates the pathogenic role of anti-
MuSK antibodies (Abs).
8. Shigemoto K, Kubo S, Maruyama N et al.:
Induction of myasthenia by immunization
against muscle-specific kinase. J. Clin. Invest.
116(4), 1016–1024 (2006).
•• Demonstrates the pathogenic role of anti-
MuSK Abs.
9. Plested CP, Tang T, Spreadbury I et al.: AChR
phosphorylation and indirect inhibition of
AChR function in
seronegative MG. Neurology 59, 1682–1688
(2002).
10. Spreadbury I, Kishore U, Beeson D,
Vincent A: Inhibition of acetylcholine receptor
function by seronegative myasthenia gravis
non-IgG factor correlates with desensitisation.
J. Neuroimmunol. 162, 149–156
(2005).
11. Giraud M, Beaurain G, Yamamoto AM et al.:
Linkage of HLA to myasthenia gravis and
genetic heterogeneity depending on anti-titin
antibodies. Neurology 57, 1555–1560 (2001).
12. Vandiedonck C, Beaurain G, Giraud M etal.:
Pleiotropic effects of the 8.1 HLA haplotype in
patients with autoimmune myasthenia gravis
and thymus hyperplasia. Proc. Natl Acad. Sci.
USA 101, 15464–15469 (2004).
Identifies the MYAS1 locus.
13. Vandiedonck C, Giraud M, Garchon HJ:
Genetics of autoimmune myasthenia gravis:
the multifaceted contribution of the HLA
complex. J. Autoimmun. 25(Suppl.) 6–11
(2005).
14. Djabiri F, Gomez L, Bach JF, Garchon HJ:
A dinucleotide repeat and a Hae III
polymorphism in the human nicotinic
acetylcholine receptor α-subunit gene
(CHRNA1). Clin. Genet. 52, 130–132
(1997).
15. Giraud M, Beaurain G, Eymard B et al.:
Genetic control of autoantibody expression in
autoimmune myasthenia gravis: role of the
self-antigen and of HLA-linked loci. Genes
Immun. 5, 398–404 (2004).
•• Model of a three-gene combination
controlling antiacetycholine receptor (AChR)
Ab titer.
16. Tzartos SJ, Lindstrom JM: Monoclonal
antibodies used to probe acetylcholine receptor
structure: localization of the main
immunogenic region and detection of
similarities between subunits. Proc. Natl Acad.
Sci. USA 77, 755–759 (1980).
17. Papadouli I, Sakarellos C, Tzartos SJ:
High-resolution epitope mapping and
fine antigenic characterization of the main
immunogenic region of the acetylcholine
receptor. Improving the binding
activity of synthetic analogues of the
region. Eur. J. Biochem. 211, 227–234
(1993).
18. Fostieri E, Beeson D, Tzartos SJ:
The conformation of the main
immunogenic region on the
α-subunit of
muscle acetylcholine receptor is affected by
neighboring receptor subunits. FEBS Lett.
481, 127–130 (2000).
19. Tzartos SJ, Barkas T, Cung MT et al.:
Anatomy of the antigenic structure of
a large membrane autoantigen, the
muscle-type nicotinic acetylcholine
receptor. Immunol. Rev. 163, 89–120
(1998).
20. Loutrari H, Kokla A, Trakas N, Tzartos SJ:
Expression of human–Torpedo hybrid
acetylcholine receptor (AChR) for
analysing the subunit specificity of
antibodies in sera from patients with
myasthenia gravis (MG). Clin. Exp.
Immunol. 109, 538–546 (1997).
21. Tzartos SJ, Seybold ME, Lindstrom JM:
Specificities of antibodies to acetylcholine
receptors in sera from myasthenia gravis
patients measured by monoclonal
antibodies. Proc. Natl Acad. Sci. USA 79,
188–192 (1982).
22. Kostelidou K, Trakas N, Zouridakis M et al.:
Expression and characterization of
soluble forms of the extracellular
domains of the β, γ and ε subunits
of the human muscle acetylcholine
receptor. FEBS J. 273, 3557–3568
(2006).
23. Tzartos SJ, Remoundos M: Detection
of antibodies directed against the
cytoplasmic region of the human
acetylcholine receptor in sera from
myasthenia gravis patients. Clin. Exp.
Immunol. 116, 146–152 (1999).
24. Christadoss P: C5 gene influences
the development of murine myasthenia
gravis. J. Immunol. 140, 2589–2592
(1988).
25. Biesecker G, Gomez CM: Inhibition of
acute passive transfer experimental
autoimmune myasthenia gravis with
Fab antibody to complement C6.
J. Immunol. 142, 2654–2659 (1989).
26. Piddlesden SJ, Jiang S, Levin JL, Vincent A,
Morgan BP: Soluble complement receptor 1
(sCR1) protects against experimental
autoimmune myasthenia gravis.
J. Neuroimmunol. 71, 173–177 (1996).
27. Lin F, Kaminski HJ, Conti-Fine BM et al.:
Markedly enhanced susceptibility to
experimental autoimmune myasthenia
gravis in the absence of decay-accelerating
factor protection. J. Clin. Invest. 110,
1269–1274 (2002).
Complement regulatory molecules
protect against experimental
autoimmune (EA)MG.
28. Tuzun E, Scott BG, Goluszko E, Higgs S,
Christadoss P: Genetic evidence for
involvement of classical complement
pathway in induction of experimental
autoimmune myasthenia gravis. J. Immunol.
171, 3847–3854 (2003).
Provides evidence for the major role of
the classical complement pathway in
MG pathogenesis.
29. Tuzun E, Scott BG, Yang H et al.:
Circulating immune complexes augment
severity of antibody-mediated myasthenia
gravis in hypogammaglobulinemic RIIIS/J
mice. J. Immunol. 172, 5743–5752
(2004).
30. Tuzun E, Saini SS, Yang H et al.:
Genetic evidence for the involvement
of Fc
γ receptor III in experimental
autoimmune myasthenia gravis
pathogenesis. J. Neuroimmunol. 174(1–2),
157–167 (2006).
31. Link H, Olsson O, Sun J et al.: Acetylcholine
receptor-reactive T and B cells in myasthenia
gravis and controls. J. Clin. Invest. 87,
2191–2195 (1991).
32. Protti MP, Manfredi AA, Horton RM,
Bellone M, Conti-Tronconi BM:
Myasthenia gravis: recognition of a
human autoantigen at the molecular level.
Immunol. Today 14, 363–368
(1993).
33. Wang ZY, Okita DK, Howard J Jr,
Conti-Fine BM: T-cell recognition of
muscle acetylcholine receptor subunits in
generalized and ocular myasthenia
gravis. Neurology 50, 1045–1054
(1998).
34. Rosenberg JS, Oshima M, Atassi MZ:
B-cell activation in vitro by helper
T cells specific to region
α146–162 of
Torpedo californica nicotinic a
cetylcholine receptor. J. Immunol. 157,
3192–3199 (1996).
35. Shi FD, He B, Li H et al.: Differential
requirements for CD28 and CD40
ligand in the induction of experimental
autoimmune myasthenia gravis.
Eur. J. Immunol. 28, 3587–3593
(1998).
Delineation of the role of CD40 ligand and
CD28 in EAMG induction.
www.futuremedicine.com 815
Recent advances myasthenia gravis research – REVIEW
36. Im SH, Barchan D, Maiti PK, Fuchs S,
Souroujon MC: Blockade of CD40 ligand
suppresses chronic experimental myasthenia
gravis by down-regulation of Th1
differentiation and up-regulation of CTLA-4.
J. Immunol. 166, 6893–6898 (2001).
CD40 ligand is essential not only for EAMG
induction, but also for EAMG progression.
37. McIntosh KR, Linsley PS, Drachman DB:
Immunosuppression and induction of anergy
by CTLA4Ig in vitro: effects on cellular and
antibody responses of lymphocytes from rats
with experimental autoimmune myasthenia
gravis. Cell Immunol. 166, 103–112 (1995).
38. Poussin MA, Tuzun E, Goluszko E et al.: B7-1
costimulatory molecule is critical for the
development of experimental autoimmune
myasthenia gravis. J. Immunol. 170,
4389–4396 (2003).
Demonstrates the role of B7 signaling in
EAMG induction.
39. Scott BG, Yang H, Tuzun E et al.: ICOS is
essential for the development of experimental
autoimmune myasthenia gravis.
J. Neuroimmunol. 153, 16–25 (2004).
40. Karachunski PI, Ostlie NS, Monfardini C,
Conti-Fine BM: Absence of IFN-γ or IL-12
has different effects on experimental
myasthenia gravis in C57BL/6 mice.
J. Immunol. 164, 5236–5244
(2000).
Interleukin (IL)-12, but probably not
interferon (IFN)-γ, is essential for
EAMG induction.
41. Zhang GX, Xiao BG, Bai XF et al.: Mice with
IFN-
γ receptor deficiency are less susceptible to
experimental autoimmune myasthenia gravis.
J. Immunol. 162, 3775–3781 (1999).
42. Balasa B, Deng C, Lee J et al.: Interferon-γ
(IFN-γ) is necessary for the genesis of
acetylcholine receptor-induced clinical
experimental autoimmune myasthenia gravis
in mice. J. Exp. Med. 186, 385–391 (1997).
Study supporting the pathogenic role of
IFN-γ in EAMG pathogenesis using a
transgenic model of different genetic
background compared with
[40].
43. Shi FD, Wang HB, Li H et al.: Natural killer
cells determine the outcome of
B-cell-mediated autoimmunity. Nat. Immunol.
1, 245–251 (2000).
Evidence for the essential role of IL-18
in EAMG induction.
44. Im SH, Barchan D, Maiti PK et al.:
Suppression of experimental myasthenia
gravis, a B cell-mediated autoimmune disease,
by blockade of IL-18. Faseb J. 15, 2140–2148
(2001).
Demonstrates that IL-18 inhibition
suppresses ongoing EAMG.
45. Poussin MA, Goluszko E, Hughes TK,
Duchicella SI, Christadoss P: Suppression of
experimental autoimmune myasthenia gravis
in IL-10 gene-disrupted mice is associated with
reduced B cells and serum cytotoxicity on
mouse cell line expressing AChR.
J. Neuroimmunol. 111, 152–160 (2000).
46. Ostlie NS, Karachunski PI, Wang W et al.:
Transgenic expression of IL-10 in T cells
facilitates development of experimental
myasthenia gravis. J. Immunol. 166,
4853–4862 (2001).
Role of IL-10 in MG pathogenesis.
47. Deng C, Goluszko E, Tuzun E, Yang H,
Christadoss P: Resistance to experimental
autoimmune myasthenia gravis in
IL-6-deficient mice is associated with reduced
germinal center formation and C3 production.
J. Immunol. 169, 1077–1083 (2002).
Role of IL-6 in MG pathogenesis.
48. Goluszko E, Deng C, Poussin MA,
Christadoss P: Tumor necrosis factor receptor
p55 and p75 deficiency protects mice from
developing experimental autoimmune
myasthenia gravis. J. Neuroimmunol.
122, 85–93 (2002).
49. Goluszko E, Hjelmstrom P, Deng C et al.:
Lymphotoxin-α deficiency completely protects
C57BL/6 mice from developing clinical
experimental autoimmune myasthenia gravis.
J. Neuroimmunol. 113, 109–118 (2001).
50. Poussin MA, Goluszko E, Franco JU,
Christadoss P: Role of IL-5 during primary
and secondary immune response to
acetylcholine receptor. J. Neuroimmunol. 125,
51–58 (2002).
51. Balasa B, Deng C, Lee J, Christadoss P,
Sarvetnick N: The Th2 cytokine IL-4 is not
required for the progression of
antibody-dependent autoimmune myasthenia
gravis. J. Immunol. 161, 2856–2862
(1998).
52. Ostlie N, Milani M, Wang W, Okita D,
Conti-Fine BM: Absence of IL-4 facilitates
the development of chronic autoimmune
myasthenia gravis in C57BL/6 mice.
J. Immunol. 170, 604–612 (2003).
Induction of an alternative chronic form of
EAMG in IL-4-deficient mice.
53. Wang W, Ostlie NS, Conti-Fine BM,
Milani M: The susceptibility to
experimental myasthenia gravis of STAT6
-/-
and STAT4
-/-
BALB/c mice suggests a
pathogenic role of Th1 cells. J. Immunol.
172, 97–103 (2004).
54. Mantegazza R, Baggi F, Bernasconi P et al.:
Video-assisted thoracoscopic extended
thymectomy and extended transsternal
thymectomy (T-3b) in non-thymomatous
myasthenia gravis patients: remission after
6years of follow-up. J. Neurol. Sci. 212,
31–36 (2003).
55. Wakkach A, Guyon T, Bruand C et al.:
Expression of acetylcholine receptor genes
in human thymic epithelial cells: implications
for myasthenia gravis. J. Immunol. 157,
3752–3760 (1996).
•• Demonstrates that acetychline receptor
(AChR) is normally expressed in the thymus.
56. Mesnard-Rouiller L, Bismuth J, Wakkach A,
Poea-Guyon S, Berrih-Aknin S: Thymic
myoid cells express high levels of muscle
genes. J. Neuroimmunol. 148, 97–105 (2004).
57. Sims GP, Shiono H, Willcox N, Stott DI:
Somatic hypermutation and selection
of B cells in thymic germinal centers
responding to acetylcholine receptor
in myasthenia gravis. J. Immunol. 167,
1935–1944 (2001).
58. Moulian N, Bidault J, Truffault F et al.:
Thymocyte Fas expression is dysregulated
in myasthenia gravis patients with
anti-acetylcholine receptor antibody.
Blood 89, 3287–3295 (1997).
•Thymic CD4
+
and CD4
+
CD8
+
Tcells from
MG patients include a hyperactivated subset
over-expressing Fas.
59. Colombara M, Antonini V, Riviera AP et al.:
Constitutive activation of p38 and ERK1/2
MAPKs in epithelial cells of myasthenic
thymus leads to IL-6 and RANTES
overexpression: effects on survival and
migration of peripheral T and B cells.
J. Immunol. 175, 7021–7028 (2005).
60. Levinson AI, Song D, Gaulton G, Zheng Y:
The intrathymic pathogenesis of myasthenia
gravis. Clin. Dev. Immunol. 11, 215–220
(2004).
New model relating MG pathogenesis with
an inflammatory environment in
hyperplastic thymus.
61. Poea-Guyon S, Christadoss P, Le Panse R
et al.: Effects of cytokines on acetylcholine
receptor expression: implications for
myasthenia gravis. J. Immunol.
174, 5941–5949 (2005).
Evidence for the existence of an
inflammatory environment in hyperplastic
thymus upregulating the expression of
AChR and major histocompatibility
complex molecules.
62. Balandina A, Lecart S, Dartevelle P,
Saoudi A, Berrih-Aknin S: Functional defect
of regulatory CD4
+
CD25
+
T cells in the
thymus of patients with autoimmune
myasthenia gravis. Blood 105, 735–741
(2005).
•• Identifies a functional defect in
CD4
+
CD25
+
regulatory T cells in
myasthenic thymus.
REVIEW – Fostieri, Kostelidou, Poulas & Tzartos
816 Future Neurol. (2006) 1(6)
63. Bernasconi P, Barberis M, Baggi F et al.:
Increased toll-like receptor 4 expression in
thymus of myasthenic patients with
thymitis and thymic involution. Am. J.
Pathol. 167, 129–139 (2005).
64. Romi F, Skeie GO, Gilhus NE, Aarli JA:
Striational antibodies in myasthenia
gravis: reactivity and possible clinical
significance. Arch. Neurol. 62, 442–446
(2005).
65. Vincent A, Willcox N: The role of T-cells
in the initiation of autoantibody responses
in thymoma patients. Pathol. Res. Pract.
195, 535–540 (1999).
66. Inoue M, Okumura M, Miyoshi S et al.:
Impaired expression of MHC class II
molecules in response to interferon
(IFN)-
γ on human thymoma neoplastic
epithelial cells. Clin. Exp. Immunol. 117,
1–7 (1999).
67. Kadota Y, Okumura M, Miyoshi S et al.:
Altered T cell development in human
thymoma is related to impairment of
MHC class II transactivator expression
induced by interferon-
γ (IFN-γ). Clin.
Exp. Immunol. 121, 59–68 (2000).
68. Strobel P, Helmreich M, Kalbacher H,
Muller-Hermelink HK, Marx A: Evidence
for distinct mechanisms in the shaping of
the CD4 T cell repertoire in histologically
distinct myasthenia gravis-associated
thymomas. Dev. Immunol. 8, 279–290
(2001).
69. Sommer N, Willcox N, Harcourt GC,
Newsom-Davis J: Myasthenic thymus and
thymoma are selectively enriched in
acetylcholine receptor-reactive T cells.
Ann. Neurol. 28, 312–319 (1990).
70. Nagvekar N, Moody AM, Moss P et al.:
A pathogenetic role for the thymoma in
myasthenia gravis. Autosensitization of IL-4-
producing T cell clones recognizing
extracellular acetylcholine receptor epitopes
presented by minority class II isotypes. J. Clin.
Invest. 101, 2268–2277 (1998).
Evidence for active priming of maturing
AChR-responsive thymocytes within
the thymoma.
71. Hoffacker V, Schultz A, Tiesinga JJ et al.:
Thymomas alter the T-cell subset composition
in the blood: a potential mechanism for
thymoma-associated autoimmune disease.
Blood 96, 3872–3879 (2000).
•• Thymopoiesis within the thymoma confers
immunological alterations in the blood.
72. Strobel P, Helmreich M, Menioudakis G
et al.: Paraneoplastic myasthenia gravis
correlates with generation of mature naive
CD4
+
T cells in thymomas. Blood 100,
159–166 (2002).
Evidence for a correlation between
thymoma-associated MG and the ability
of thymomas to export mature naive
CD4
+
Tcells.
73. Shiono H, Wong YL, Matthews I et al.:
Spontaneous production of anti-IFN-α and
anti-IL-12 autoantibodies by thymoma cells
from myasthenia gravis patients suggests
autoimmunization in the tumor.
Int. Immunol. 15, 903–913 (2003).
74. Marx A, Muller-Hermelink HK, Strobel P:
The role of thymomas in the development
of myasthenia gravis. Ann. NY Acad. Sci.
998, 223–236 (2003).
75. Guyon T, Wakkach A, Poea S et al.:
Regulation of acetylcholine receptor gene
expression in human myasthenia gravis
muscles. Evidences for a compensatory
mechanism triggered by receptor loss.
J. Clin. Invest. 102, 249–263
(1998).
AChR subunit mRNAs are upregulated in
response to AChR loss due to antigenic
modulation.
76. Stegall T, Krolick KA: Myocytes respond
in vivo to an antibody reactive with the
acetylcholine receptor by upregulating
interleukin-15: an interferon-
γ activator
with the potential to influence the severity
and course of experimental myasthenia
gravis. J. Neuroimmunol. 119, 377–386
(2001).
77. Reyes-Reyna S, Stegall T, Krolick KA: Muscle
responds to an antibody reactive with the
acetylcholine receptor by upregulating
monocyte chemoattractant protein 1:
a chemokine with the potential to influence
the severity and course of experimental
myasthenia gravis. J. Immunol. 169,
1579–1586 (2002).
Chemokine upregulation in muscle as a result
of antigenic modulation.
78. Garcia YR, Krolick KA: Short-circuiting
autoimmune disease by target-tissue-derived
nitric oxide. Clin. Immunol. 113, 74–80
(2004).
79. Feferman T, Maiti PK, Berrih-Aknin S et al.:
Overexpression of IFN-induced protein 10
and its receptor CXCR3 in myasthenia gravis.
J. Immunol. 174, 5324–5331 (2005).
Chemokine upregulation in muscle as a result
of antigenic modulation.
80. Sieb JP: Myasthenia gravis: emerging new
therapy options. Curr. Opin. Pharmacol. 5,
303–307 (2005).
81. Meriggioli MN, Ciafaloni E, Al-Hayk KA
et al.: Mycophenolate mofetil for
myasthenia gravis: an analysis of efficacy,
safety, and tolerability. Neurology 61,
1438–1440 (2003).
82. Wolfe GI, Kaminski HJ, Jaretzki A 3rd, Swan
A, Newsom-Davis J: Development of a
thymectomy trial in nonthymomatous
myasthenia gravis patients receiving
immunosuppressive therapy. Ann. NY Acad.
Sci. 998, 473–480 (2003).
83. Batocchi AP, Evoli A, Di Schino C, Tonali P:
Therapeutic apheresis in myasthenia gravis.
Ther. Apher. 4, 275–279 (2000).
84. Dalakas MC: The use of intravenous
immunoglobulin in the treatment of
autoimmune neuromuscular diseases:
evidence-based indications and safety profile.
Pharmacol. Ther. 102, 177–193 (2004).
85. Aricha R, Feferman T, Souroujon MC, Fuchs
S: Overexpression of phosphodiesterases in
experimental autoimmune myasthenia gravis:
suppression of disease by a phosphodiesterase
inhibitor. Faseb J. 20, 374–376 (2006).
Implication of phosphodiesterases in EAMG
pathogenesis, evidence for the therapeutic
potential of phosphodiesterase inhibitors in
MG.
86. Barchan D, Souroujon MC, Im SH,
Antozzi C, Fuchs S: Antigen-specific
modulation of experimental myasthenia gravis:
nasal tolerization with recombinant fragments
of the human acetylcholine receptor
α-subunit. Proc. Natl Acad. Sci. USA 96,
8086–8091 (1999).
87. Im SH, Barchan D, Fuchs S, Souroujon MC:
Suppression of ongoing experimental
myasthenia by oral treatment with an
acetylcholine receptor recombinant fragment.
J. Clin. Invest. 104, 1723–1730 (1999).
Oral treatment with AChR fragments as a
specific therapeutic strategy for MG.
88. Ma CG, Zhang GX, Xiao BG et al.:
Suppression of experimental autoimmune
myasthenia gravis by nasal administration of
acetylcholine receptor. J. Neuroimmunol. 58,
51–60 (1995).
89. Maiti PK, Feferman T, Im SH, Souroujon
MC, Fuchs S: Immunosuppression of rat
myasthenia gravis by oral administration of a
syngeneic acetylcholine receptor fragment.
J. Neuroimmunol. 152, 112–120 (2004).
90. Baggi F, Annoni A, Ubiali F et al.:
Breakdown of tolerance to a self-peptide of
acetylcholine receptor
α-subunit induces
experimental myasthenia gravis in rats.
J. Immunol. 172, 2697–2703 (2004).
91. Jambou F, Zhang W, Menestrier M et al.:
Circulating regulatory anti-T cell receptor
antibodies in patients with myasthenia
gravis. J. Clin. Invest. 112, 265–274
(2003).
Evidence for the existence of regulatory anti-
T-cell receptor Abs in MG; prospects for T-
cell receptor vaccination in MG.
www.futuremedicine.com 817
Recent advances myasthenia gravis research – REVIEW
92. Xu L, Villain M, Galin FS, Araga S, Blalock
JE: Prevention and reversal of experimental
autoimmune myasthenia gravis by a
monoclonal antibody against acetylcholine
receptor-specific T cells. Cell Immunol. 208,
107–114 (2001).
93. Matsumoto Y, Matsuo H, Sakuma H et al.:
CDR3 spectratyping analysis of the TCR
repertoire in myasthenia gravis. J. Immunol.
176, 5100–5107 (2006).
94. Drachman DB, Wu JM, Miagkov A et al.:
Specific immunotherapy of experimental
myasthenia by genetically engineered APCs:
the ‘guided missile’ strategy. Ann. NY Acad.
Sci. 998, 520–532 (2003).
95. Duan RS, Adikari SB, Huang YM, Link H,
Xiao BG: Protective potential of
experimental autoimmune myasthenia
gravis in Lewis rats by IL-10-modified
dendritic cells. Neurobiol. Dis. 16, 461–467
(2004).
96. Boneva N, Hamra-Amitay Y, Wirguin I,
Brenner T: Stimulated-single fiber
electromyography monitoring of anti-sense
induced changes in experimental
autoimmune myasthenia gravis. Neurosci.
Res. 55, 40–44 (2006).
97. Graus YF, de Baets MH, Parren PW et al.:
Human anti-nicotinic acetylcholine receptor
recombinant Fab fragments isolated from
thymus-derived phage display libraries from
myasthenia gravis patients reflect
predominant specificities in serum and
block the action of pathogenic serum
antibodies. J. Immunol. 158, 1919–1929
(1997).
98. Papanastasiou D, Mamalaki A, Eliopoulos E
et al.: Construction and characterization of a
humanized single chain Fv antibody
fragment against the main immunogenic
region of the acetylcholine receptor.
J. Neuroimmunol. 94, 182–195 (1999).
99. Fostieri E, Tzartos SJ, Berrih-Aknin S,
Beeson D, Mamalaki A: Isolation of potent
human Fab fragments against a novel highly
immunogenic region on human muscle
acetylcholine receptor which protect the
receptor from myasthenic autoantibodies.
Eur. J. Immunol. 35, 632–643 (2005).
100. Protopapadakis E, Kokla A, Tzartos SJ,
Mamalaki A: Isolation and characterization
of human anti-acetylcholine receptor
monoclonal antibodies from transgenic mice
expressing human immunoglobulin loci.
Eur. J. Immunol. 35, 1960–1968 (2005).
101. Takamori M, Maruta T: Immunoadsorption
in myasthenia gravis based on specific
ligands mimicking the immunogenic sites of
the acetylcholine receptor. Ther. Apher. 5,
340–350 (2001).
102. Psaridi-Linardaki L, Mamalaki A,
Remoundos M, Tzartos SJ: Expression of
soluble ligand- and antibody-binding
extracellular domain of human muscle
acetylcholine receptor α subunit in yeast
Pichia pastoris. Role of glycosylation in
α-bungarotoxin binding. J. Biol. Chem. 277,
26980–26986 (2002).
103. Psaridi-Linardaki L, Trakas N, Mamalaki A,
Tzartos SJ: Specific immunoadsorption of
the autoantibodies from myasthenic patients
using the extracellular domain of the human
muscle acetylcholine receptor α-subunit.
Development of an antigen-specific
therapeutic strategy. J. Neuroimmunol. 159,
183–191 (2005).
Affiliations
Efrosini Fostieri, PhD
Hellenic Pasteur Institute, Department of
Biochemistry, 127 Vas. Sofias Avenue,
11521 Athens, Greece
fostieri@pasteur.gr
Kalliopi Kostelidou,
PhD
Hellenic Pasteur Institute, Department of
Biochemistry, 127 Vas. Sofias Avenue
11521 Athens, Greece
k.kostelidou@pasteur.gr
Konstantinos Poulas,
PhD
Department of Pharmacy, University of Patras,
26504 Patras, Greece
kpoulas@upatras.gr
Socrates J Tzartos,
PhD
Hellenic Pasteur Institute, Department of
Biochemistry, 127 Vas. Sofias Avenue, 11521
Athens, Greece
and,
Department of Pharmacy, University of Patras,
26504 Patras, Greece
Tel.: +30 210 647 8842;
Fax: +30 210 647 8842;
tzartos@pasteur.gr
... In terms of immunomodulatory treatments, thymectomy is used for early onset and thymoma MG, whereas immunosuppressants, usually glucocorticoids with or without azathioprine, are used for all patients with MG. Other immunosuppressive drugs, such as mycophenolate, have been used in limited studies as adjunctive immunosuppressants for the treatment of severe, refractory and high dose steroid-dependent MG [12,13]. Some of the major adverse effects linked to immunosuppression are osteoporosis, hypertension , gastrointestinal discomfort and psychiatric changes. ...
... Interestingly , these two sets of anti-AChR Fabs seem to compete with distinct anti-AChR auto-Ab subpopu- lations [30]. This suggests that the combination of a small number of protective anti-AChR Ab fragments to create a panel of AChR protectors shielding the most important immunogenic AChR epitopes may be a requirement for the development of an efficient therapeutic Ab set [13]. The problem of the short in vivo half-life of human Fab and svFv fragments has been addressed by a more elegant approach, i.e. by transferring the human Fab into an IgG4 mAb. ...
Article
Full-text available
Acquired autoimmune myasthenia gravis (MG) is the most common disease that affects the neuromuscular junction (NMJ). MG is associated with autoantibodies (auto-Abs) to components of the NMJ. About 85-90% of MG patients have auto-Abs against the muscle nicotinic acetylcholine receptor (AChR), while about half of the remaining patients have auto-Abs against muscle-specific kinase. Auto-Abs, in combination with local deposition of complement, reduce the number of available post-synaptic nicotinic AChRs and thereby impair neuromuscular transmission. Current medications for MG are non-specific and include acetylcholinesterase inhibitors, immunosuppressants, plasma exchange, intravenous Ig administration and thymectomy. Treatments that selectively target the anti-AChR auto-Abs may prove to be more effective and free of side-effects. We here review two approaches aimed at the development of antigen-specific therapies for MG. The first is specific apheresis of Abs from patients' sera using immobilised recombinant AChR domains as immunoadsorbents. Indeed, we have recently shown that the combined recombinant extracellular domains of all human AChR subunits are capable of specifically immunoadsorbing the majority of pathogenic auto-Abs from several MG sera. The second therapeutic approach is the development of non-pathogenic anti-AChR monoclonal Abs that could potentially be used as protective agents by blocking the binding of patients' auto-Abs to the AChR.
... Antigen-specific treatment of MG should be promising, and such strategies work in animal models. Induction of specific immunological tolerance to AChR or MuSK, shifting the immune response from harmful to nonharmful is theoretically possible [3, 30, 31]. Strategies involving antigen-presenting cells are considered for treatment of autoimmune disorders, and manipulation of this process could be antigen-specific. ...
Article
Full-text available
Patients with autoimmune myasthenia gravis (MG) should be further classified before initiating therapy, as treatment response varies for ocular versus generalised, early onset versus late onset, and acetylcholine receptor antibody positive versus MuSK antibody positive disease. Most patients need immunosuppression in addition to symptomatic therapy. Prednisolone and azathioprine represent first choice drugs, whereas several second choice options are recommended and should be considered. Thymectomy should be undertaken in MG with thymoma and in generalised, early-onset MG. For MG crises and other acute exacerbations, intravenous immunoglobulin (IvIg) and plasma exchange are equally effective and safe treatments. Children and females in child bearing age need special attention regarding potential side effects of immunosuppressive therapy. MG pathogenesis is known in detail, but the immune therapy is still surprisingly unspecific, without a pin-pointed attack on the defined disease-inducing antigen-antibody reaction being available.
... Other commonly used immunomodulating drugs primarily interfere with T and B cell proliferation and/or function. Some of the major adverse effects linked to immunosuppression are osteoporosis, hypertension, gastrointestinal discomfort , and psychiatric changes (Kaminski, 2003; Fostieri et al., 2006). A different therapeutic approach, plasma exchange (plasmapheresis ), is used mostly in severe MG symptoms or in myasthenic crisis and also in cases where muscle function must be optimized, such as before surgery (including thymectomy). ...
Article
Myasthenia gravis (MG), a prototypic antibody-mediated autoimmune disease, presents an excellent target for scientific research aimed at a better understanding of the disease itself and the source that triggers an autoimmune reaction in an organism. MG is a neuromuscular disease caused mainly by an autoimmune response against the nicotinic acetylcholine receptor (AChR) which interferes with neuromuscular transmission. This review focuses on our studies on the extracellular domains of human muscle AChR subunits in an effort to develop an approach for the specific therapeutic apheresis of autoantibodies from patients' sera using the immobilized subunits as immunoadsorbents. The ability of the anti-AChR antibodies isolated by this technique, but not of the depleted sera, to induce disease is also described. This review is dedicated to the late Prof. John Newsom-Davis, who was the first to introduce the use of plasmapheresis for MG.
... MG is characterized by the presence of circulating autoantibodies usually directed against muscle nAChR. Although a large fraction of the serum autoantibodies against the nAChR target the α subunit, autoantibodies directed against other subunits may also contribute to the pathology of MG ( Tzartos et al., 1998, Fostieri et al., 2006). Despite direct evidence for the role of anti-nAChR autoantibody in experimental MG, their role in the pathology of human MG has only been previously demonstrated indirectly (reviewed in Berrih-Aknin, 1995;De Baets and Stassen, 2002). ...
Article
The muscle nicotinic acetylcholine receptor (nAChR) is the major autoantigen in the autoimmune disease myasthenia gravis (MG), in which autoantibodies bind to, and cause loss of, nAChRs. Antibody-mediated nAChR loss is caused by the action of complement and by the acceleration of nAChR internalization caused by antibody-induced cross-linking of nAChR molecules (antigenic modulation). To obtain an insight into the role of the various anti-nAChR antibody specificities in MG, we have studied nAChR antigenic modulation caused by isolated anti-subunit autoantibodies. Autoantibodies against the nAChR alpha or beta subunits were isolated from four MG sera by affinity chromatography on columns carrying immobilized recombinant extracellular domains of human nAChR expressed in the yeast Pichia pastoris. The isolated anti-alpha and anti-beta autoantibodies, as well as untreated MG sera, induced nAChR antigenic modulation in TE671 cells. Partially antibody-depleted sera exhibited reduced modulating activity, whereas a serum completely depleted of anti-nAChR antibodies exhibited no nAChR modulation. Interestingly, the anti-alpha autoantibodies were, on average, approximately 4.3 times more effective than the anti-beta autoantibodies. The present work supports the notion that anti-nAChR autoantibodies may be the sole nAChR-reducing factor in anti-nAChR antibody-seropositive MG, and that anti-alpha-subunit autoantibodies are the dominant pathogenic autoantibody specificity.
... j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / b b a p a p AChR cause loss of functional AChRs in the neuromuscular junction. Antibodies against AChR are found in nearly 80%–90% of patients with generalized MG and are considered the main pathogenic factor in MG [18,19]. Driven by the important pathophysiological roles of the AChRs, many efforts have been made to obtain detailed information on their structure. ...
Article
The muscle nicotinic acetylcholine receptor (AChR) is the prototype of the ligand-gated ion channels (or Cys-loop receptors), formed by 5 homologous subunits (alpha2betagammadelta or alpha2betagammaepsilon), and is the major autoantigen in the autoimmune disease, myasthenia gravis. Previously, we expressed the wild-type extracellular domain (ECD) of the gamma-subunit (gammaECD) of the AChR in yeast Pichia pastoris at 0.3-0.8 mg/L, in soluble but microaggregate form, to use as starting material for structural and antigenicity studies. To optimize these characteristics, we constructed and characterized four gammaECD variants: (a) mutants-1 (gammaC61S) and -2 (gammaC106S-C115S), where the non-conserved Cys of gammaECD were replaced by serines, (b) mutant-3 (gammaCysLoop), where the gamma Cys-loop region was substituted by the cognate region of the acetylcholine binding protein (AChBP) and (c) mutant-4 (gammaCysLoop-C106S-C115S), where both the C106S-C115S and Cys-loop mutations were combined. None of mutants-1 and -2 displayed any improvement, while mutant-3 and -4 were mostly in dimeric form and expressed at much higher levels (2.5 mg/L and 3.5 mg/L respectively). All four mutants and wild-type gammaECD were recognized by sera from myasthenic patients, but mutants-3 and -4 exhibited higher efficiency, compared to wild-type or mutants-1 and -2. These results suggest that the substitution of the Cys-loop region of any AChR ECD with the AChBP counterpart leads to AChR ECD of improved conformation, more suitable for structural and therapeutic studies.
... 7 Current treatments used for MG include the use of cholinesterase inhibitors, immunosuppressive drugs, plasmapheresis, infusion of intravenous immunoglobulin (IVIg), and thymectomy. 8,9 Plasmapheresis, a short-term immunomodulatory treatment, is based on the removal of plasma and circulating factors therein, including pathogenic anti-AChR autoantibodies, thus reducing autoimmune attack at the neuromuscular junction. 10 One disadvantage of the treatment is the concomitant removal of indispensable components of the plasma which must be replaced, 11 thus increasing the cost and entailing the risk of anaphylactic reactions and transfusion-related infections. ...
Article
Myasthenia gravis (MG) is usually caused by autoantibodies against muscle nicotinic acetylcholine receptor (AChR), which is composed of five subunits (alpha(2)betagammadelta or alpha(2)betaepsilondelta). Current treatments, including plasmapheresis, are nonspecific, causing several side effects. We aim to develop an antigen-specific alternative to plasmapheresis, since the latter removes indispensable plasma components in addition to anti-AChR antibodies. We are developing a method for the selective depletion of the anti-AChR autoantibodies from patients' plasma through the construction of "immunoadsorbent" columns carrying AChR domains. We have expressed the extracellular domains (ECDs, amino acids approximately 1-210/220) of all human muscle AChR subunits in Pichia pastoris and, in preliminary experiments, in E. coli. The ECDs were immobilized (individually or mixed) on Sepharose beads, producing Sepharose-ECD columns, which were tested for their immunoadsorbing capacity on MG sera and shown to specifically eliminate major autoantibody fractions from several MG sera. The immobilized ECDs remained stable and did not dissociate from their matrix after incubation with serum, whereas the procedure was neither toxic nor immunogenic in two experimental rabbits. Testing the intact or antibody-depleted MG sera and the affinity purified autoantibodies showed that both the intact sera and the purified autoantibodies, but not the antibody-depleted sera, could induce AChR loss in cell cultures and experimental MG in rats. This preliminary study suggests that the myasthenic potency of MG sera is entirely due to their anti-AChR antibodies and therefore their depletion should be of therapeutic value. We conclude that ECD-mediated immunoadsorption can be used as an efficient, antigen-specific therapy for MG.
Chapter
Myasthenia gravis (MG) is an autoimmune disease mediated by autoantibodies that mainly target muscle nicotinic acetylcholine receptor (AChR) and cause loss of functional AChRs in the neuromuscular junction. Despite extensive knowledge from studies on MG and its major autoantigen, the aetiology of the disease remains unclear, thus rendering therapeutic options unable to target the causative agent. Latest progress on recombinant expression of the AChR subunits has allowed for some alternative, though promising, therapeutic approaches. The scope of this chapter is to provide an overview of the recent achievements in the field of emerging therapeutics for MG, including antigen-specific therapies, which are directed at the autoimmune response. KeywordsAutoimmunity–Myasthenia gravis–Nicotinic acetylcholine receptor–Neuromuscular junction–Autoantibodies–Specific immunotherapy
Article
Full-text available
Based on the crystal structure of the extracellular domain (ECD) of the mouse nicotinic acetylcholine receptor (nAChR) alpha1 subunit bound to α-bungarotoxin (α-Btx) we have generated in silico models of the human nAChR α1 bound to α-Btx and α-cobratoxin (α-Cbtx), both in the presence and in the absence of the N-linked carbohydrate chain. To gain further insight into the structural role of glycosylation molecular dynamics (MD) simulations were carried out in explicit solvent so as to compare the conformational dynamics of the binding interface between nAChR α1 and the two toxins. An interesting observation during the course of the MD simulations is the strengthening of the receptor-toxin interaction in the presence of the carbohydrate chain, mediated through a shift in the position of the sugars towards the bound toxin. Critical protein-sugar interactions implicate residues Ser187 and Trp184 of nAChR and Thr6, Ser9, and Thr15 of α-Btx, as well as Thr6 and Pro7 of α-Cbtx. Analysis of the predicted residue-specific intermolecular interactions is intended to inspire biophysical studies on the functional role of glycosylation in the gating mechanism.
Article
Myasthenia gravis (MG) is an autoimmune neuromuscular transmission disorder where well-defined autoantibodies against muscle and muscle cell membrane molecules are directly pathogenetic. All MG patients should be defined for subtype, as such a subclassification has treatment consequences. Ocular MG, early-onset MG, late-onset MG, MG with thymoma, MG with anti-muscle-specific tyrosine kinase antibodies and MG with no defined antibodies constitute the six MG categories. The MG diagnostic process includes neurophysiology, neuroimmunology, neuropharmacology and imaging. In addition to symptomatic therapy with acetylcholine esterase inhibitors, most patients need thymectomy and/or immunosuppressive drugs. Today's treatment is not immunospecific and far from antigen-specific, even if the pathogenesis is known in detail. Strategies for acetylcholine receptor tolerance induction, manipulating acetylcholine receptor antigen presentation or suppressing acetylcholine receptor-specific B-cells or plasma cells work in experimental MG, but have not yet been attempted properly for the human disease, or they do not work. Apart from the 10-15% of patients with paraneoplastic MG, the cause of the disease is not known. Until curative or antigen-specific therapy become available, the well-established treatment gives good-to-excellent results in most patients, with acceptable quality of life and no increased mortality. Acute and intensive care treatment during MG exacerbation is a cornerstone in the treatment.
Article
Myasthenia gravis (MG) is usually caused by autoantibodies against the human muscle acetylcholine receptor (AChR). Plasmapheresis offers a therapeutic option, but, as well as removing the pathogenic anti-AChR autoantibodies, it non-specifically removes indispensable immunoglobulins. An attractive alternative to plasmapheresis would be the extracorporeal specific removal of the autoantibodies using AChR-based immunoadsorbents. Previously, we used the N-terminal extracellular domain (ECD) of the AChR alpha subunit to immunoadsorb anti-alpha subunit autoantibodies from MG sera. In this study, we immobilised the beta -, gamma- and epsilon-AChR ECDs on Sepharose and tested them as immunoadsorbents on 50 MG sera. A given ECD removed a different percentage of autoantibodies from different sera and different ECDs removed different percentages from the same serum; on average, the beta-, gamma- and epsilon-ECDs removed 22%, 20% and 15.5% of the autoantibodies, respectively. Immunoadsorption was completed in 3 min, 1 mug of ECD removed approximately 2 pmol of autoantibodies, and the immunoadsorbent could be recycled approximately 4 times. The combined use of two (alpha+gamma) or four (alpha+beta+gamma+epsilon) ECDs in a single immunoadsorbent resulted in much higher (often additive) immunoadsorption. These results show that MG sera have autoantibodies against several AChR subunits, and suggest that the combined use of all AChR ECDs could provide the basis for a novel, antigen-specific therapy for MG.
Article
Full-text available
The muscle weakness in myasthenia gravis (MG) is mediated by autoantibodies against the nicotinic acetylcholine receptor (AChR) at the neuromuscular junction. Production of these pathogenic autoantibodies is believed to be associated with germinal centers (GC) and anti-AChR-secreting plasma cells in the hyperplastic thymus of patients with early onset MG (EOMG). Here, we describe the repertoire of rearranged heavy chain V genes and their clonal origins in GC from a typical EOMG patient. Three hundred fifteen rearranged Ig V H genes were amplified, cloned, and sequenced from sections of four thymic GC containing AChR-specific B cells. We found that thymic GC contain a remarkably heterogeneous population of B cells. Both naive and circulating memory B cells undergo Ag-driven clonal proliferation, somatic hypermutation, and selection. Numerous B cell clones were present, with no individual clone dominating the response. Comparisons of B cell clonal sequences from different GC and known anti-AChR Abs from other patients showed convergent mutations in the complementarity determining regions. These results are consistent with AChR driving an ongoing GC response in the thymus of EOMG patients. This is the first detailed analysis of B cell clones in human GC responding to a defined protein Ag, and the response we observed may reflect the effects of chronic stimulation by autoantigen. The Journal of Immunology, 2001, 167: 1935-1944.
Article
Full-text available
Myasthenia gravis (MG) is an autoimmune disorder in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. In an attempt to develop an antigen-specific therapy for MG, we administered a nonmyasthenogenic recombinant fragment of AChR orally to rats. This fragment, corresponding to the extracellular domain of the human AChR alpha-subunit (Halpha1-205), protected rats from subsequently induced experimental autoimmune myasthenia gravis (EAMG) and suppressed ongoing EAMG when treatment was initiated during either the acute or chronic phases of disease. Prevention and suppression of EAMG were accompanied by a significant decrease in AChR-specific humoral and cellular responses. The underlying mechanism for the Halpha1-205-induced oral tolerance seems to be active suppression, mediated by a shift from a T-helper 1 (Th1) to a Th2/Th3 response. This shift was assessed by changes in the cytokine profile, a deviation of anti-AChR IgG isotypes from IgG2 to IgG1, and a suppressed AChR-specific delayed-type hypersensitivity response. Our results in experimental myasthenia suggest that oral administration of AChR-specific recombinant fragments may be considered for antigen-specific immunotherapy of myasthenia gravis.
Article
Full-text available
Myasthenia gravis (MG) is strongly associated with antibodies to acetylcholine receptor (AChR), whereas the extent of T cell involvement is not settled. The number of cells secreting interferon-gamma (IFN-gamma) in response to AChR during 48 h culture of blood mononuclear cells (PBL) may reflect AChR-reactive T cells. Using an immunospot assay, we detected such cells in 23 of 30 patients with MG at a mean number of 1 per 33.333 PBL. AChR-reactive T cells were also found in patients with other neurological diseases (OND) and in healthy subjects but at lower frequencies and numbers. The T cell response to purified protein derivative and to PHA, and also to two major myelin proteins (basic protein and proteolipid protein) did not differ between MG and the two control groups, underlining the specificity of an augmented T cell reactivity to AChR in MG. Evaluation of the B cell response by enumerating anti-AChR IgG antibody secreting cells revealed such cells in 27 of 28 patients with MG at a mean value of 1 per 14,085 PBL. Cells secreting anti-AChR antibodies of the IgA and IgM isotypes were also detected in MG, but less frequently, at lower numbers, and only in conjunction with IgG antibody secreting cells. Anti-AChR antibody secreting cells were also found among patient with OND and in healthy controls, but at lower frequencies and numbers. These data confirm that AChR is a major target for autoimmune response in MG.
Article
Thymomas are the only tumors that are proven to generate mature T cells from immature precursors. It is unknown, however, whether intratumorous thymopoiesis has an impact on the peripheral T-cell pool and might thus be related to the high frequency of thymoma-associated myasthenia gravis. This study shows, using fluorescence-activated cell sorting-based analyses and T-cell proliferation assays, that thymopoiesis and T-cell function in thymomas correspond with immunologic alterations in the blood. Specifically, the proportion of circulating CD45RA+CD8+ T cells is significantly increased in patients with thymoma compared with normal controls, in accordance with intratumorous T-cell development that is abnormally skewed toward the CD8+ phenotype. Moreover, it is primarily the proportion of circulating CD45RA+CD8+ T cells that decreases after thymectomy. The results also demonstrate that T cells reactive toward recombinant autoantigens are distributed equally between thymomas and blood, whereas T-cell responses to foreign antigen (ie, tetanus toxoid) are seen only among circulating T cells and not among thymoma-derived T cells. These functional studies support the hypothesis that thymopoiesis occurring within thymomas alters the peripheral T-cell repertoire. Because many thymomas are enriched with autoantigen-specific T cells, a disturbance of circulating T-cell subset composition by export of intratumorous T cells may contribute to paraneoplastic autoimmune disease arising in patients with thymoma.
Article
We assayed cryopreserved sera from 38 acetylcholine receptor (AChR) antibody-negative patients with myasthenia gravis (MG) who were followed clinically for muscle-specific tyrosine kinase (MuSK) antibodies and analyzed and compared their clinical characteristics. None of 13 sera from patients with purely ocular MG were positive. Sera from 10 of 25 patients (40%) with generalized MG were positive for MuSK antibodies. The age at onset of myasthenic symptoms was significantly earlier in MuSK antibody-positive patients (P = 0.02). MuSK antibodies were present in AChR antibody-negative patients of either gender, with virtually identical prevalence in women (41.2%) and men (37.5%). The distribution of weakness more commonly involved neck muscles in MuSK antibody-positive patients, and limb muscles in MuSK antibody-negative patients. Patients responded to immunosuppressive treatment regardless of whether MuSK antibody was present. We conclude that MuSK antibodies are present and diagnostically useful in a subset of myasthenic patients without AChR antibodies. Although the distribution of weakness differs somewhat depending on whether MuSK antibodies are present, responses to anticholinesterase and immunosuppressive treatments are similar. © 2004 Wiley Periodicals, Inc. Muscle Nerve 30: 55–60, 2004
Article
Although treatment of MG with general immunosuppressive agents is often effective, it has important drawbacks, including suppression of the immune system as a whole, with the risks of infection and neoplasia, and numerous other adverse side effects. Ideally, treatment of MG should eliminate the specific pathogenic autoimmune response to AChR, without otherwise suppressing the immune system or producing other adverse side effects. Although antibodies to AChR are directly responsible for the loss of AChRs at neuromuscular junctions in MG, the AChR antibody response is T cell-dependent, and immunotherapy directed at T cells can abrogate the autoantibody response, with resulting benefit. As in other autoimmune diseases, the T cell response in MG is highly heterogeneous. The design of specific immunotherapy must take this heterogeneity into account and target the entire repertoire of AChR-specific T cells. We describe our investigation of a novel strategy for specific immunotherapy of MG, involving gene transfer to convert antigen-presenting cells (APCs) to “guided missiles” that target AChR-specific T cells, and that induce apoptosis and elimination of those T cells. This strategy uses the ability of APCs from a given individual to present the entire spectrum of AChR epitopes unique for that individual, and thereby to target the entire repertoire of antigen-specific T cells of the same individual. Using viral vectors, we have genetically engineered the APCs to process and present the most important domain of the AChR molecule, and to express a “warhead” of Fas ligand (FasL) to eliminate the activated AChR-specific T cells with which they interact. Our results show that the APCs express the appropriate gene products, and effectively and specifically eliminate AChR-specific T cells by the Fas/FasL pathway, while sparing T cells of other specificities.
Article
Much progress has been made in the 26 years since initial studies of the first purified acetylcholine receptors (AChRs) led to the discovery that an antibody-mediated autoimmune response to AChRs causes the muscular weakness and fatigability characteristic of myasthenia gravis (MG) and its animal model, experimental autoimmune myasthenia gravis (EAMG). Now, the structure of muscle AChRs is much better known. Monoclonal antibodies to muscle AChRs, developed as model autoantibodies for studies of EAMG, were used for initial purifications of neuronal AChRs, and now many homologous subunits of neuronal nicotinic AChRs have been cloned. There is a basic understanding of the pathological mechanisms by which autoantibodies to AChRs impair neuromuscular transmission. Immunodiagnostic assays for MG are used routinely. Nonspecific approaches to immunosuppressive therapy have been refined. However, fundamental mysteries remain regarding what initiates and sustains the autoimmune response to muscle AChRs and how to specifically suppress this autoimmune response using a practical therapy. Many rare congenital myasthenic syndromes have been elegantly shown to result from mutations in muscle AChRs. These studies have provided insights into AChR structure and function as well as into the pathological mechanisms of these diseases. Evidence has been found for autoimmune responses even to some central nervous system neurotransmitter receptors, but only one neuronal AChR has so far been implicated in an autoimmune disease. Thus far, only two neuronal AChR mutations have been found to be associated with a rare form of epilepsy, but many more neuronal AChR mutations will probably be found to be associated with disease in the years ahead. © 2000 John Wiley & Sons, Inc. Muscle Nerve 23: 453–477, 2000.
Article
Myasthenia gravis (MG) is caused by autoantibodies to the acetylcholine receptor (AChR). Experiments with fetal (α2βγδ) and adult (α2βϵδ) AChR and with recombinant subunit dimers showed that some monoclonal antibodies (mAbs) against the main immunogenic region (MIR), located on the α-subunit of the AChR, bind better to fetal AChR and to αγ subunit dimer than to adult AChR and αϵ dimer and equally to both αβ and αδ. However, other anti-MIR mAbs prefer adult AChR and αϵ dimer, bind well to αβ but weakly to αδ. These results suggest that the MIR conformation is affected by the neighboring γ/ϵ- and δ-subunits and may contribute to understanding the antibody specificities in MG.
Article
We compared T-cell proliferative responses to acetylcholine receptor (AChR) and to purified protein derivative (PPD) (of tuberculin) of hyperplastic thymus, thymoma, and blood cells from patients with myasthenia gravis (MG). Hyperplastic MG thymus cells gave significantly higher and more consistent responses to AChR than parallel cultures of autologous blood cells, whereas responses to PPD showed an opposite trend. Thus there was a preferential localization of AChR-reactive T cells in the hyperplastic MG thymus. Furthermore, there was a strong correlation between blood and thymus cell responses to PPD (but not to AChR), arguing that the hyperplastic MG thymus contains a sample of sensitized peripheral T cells. By contrast, both AChR- and PPD-responsive T cells were almost undetectable in thymus from nonmyasthenic patients, which is evidently much less receptive to circulating T cells. Cells from MG thymomas showed the highest stimulations by AChR but did not consistently react to PPD. However, the uninvolved thymus adjacent to these thymomas behaved almost identically to the hyperplastic samples described above. Our interpretation is that AChR-specific T cells are initially sensitized in the MG thymoma but are selectively trapped in the hyperplastic thymus after being primed elsewhere.
Article
The role of the lytic complement C5b-9 membrane attack complex (MAC) in acute passive transfer experimental autoimmune myasthenia gravis (EAMG) produced in rats was investigated by in vivo inhibition of MAC formation with anti-C6 Fab. Anti-C6 Fab totally inhibited in vitro serum hemolytic activity, but did not consume or inhibit early complement pathways. Injection of rats with 0.12 mg/ml anti-C6 Fab reduced serum C6 to 8% and inhibited the muscle weakness, electrophysiologic abnormalities and loss of acetylcholine receptor (AChR) associated with acute EAMG. This level of C6 inhibition reduced the total serum complement hemolytic activity to 29% of normal but did not reduce the serum levels of complement components C3, C5, or C7. Treatment of rats with lower amounts of anti-C6 Fab (0.08 mg/ml) also inhibited clinical and electrophysiologic signs of EAMG, however, the lower amount of anti-C6 did not prevent the loss of muscle AChR. Both the higher and the lower amount of anti-C6 Fab inhibited the accumulation of macrophages at muscle motor end-plates. The inhibition by anti-C6 indicates that muscle weakness and electrophysiologic abnormalities associated with EAMG are dependent on the complement MAC, and that muscle weakness results from tissue injury in addition to loss of muscle membrane and AChR.