Amelioration of Experimental Autoimmune Myasthenia Gravis in

Transcription

Amelioration of Experimental Autoimmune Myasthenia Gravis in
Amelioration of Experimental Autoimmune
Myasthenia Gravis in Rats by Neonatal FcR
Blockade
This information is current as
of February 7, 2017.
Liming Liu, Ana Maria Garcia, Helen Santoro, Yixia Zhang,
Kevin McDonnell, Jennifer Dumont and Alan Bitonti
J Immunol 2007; 178:5390-5398; ;
doi: 10.4049/jimmunol.178.8.5390
http://www.jimmunol.org/content/178/8/5390
Subscriptions
Permissions
Email Alerts
This article cites 57 articles, 18 of which you can access for free at:
http://www.jimmunol.org/content/178/8/5390.full#ref-list-1
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscriptions
Submit copyright permission requests at:
http://www.aai.org/ji/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/cgi/alerts/etoc
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright © 2007 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
References
The Journal of Immunology
Amelioration of Experimental Autoimmune Myasthenia Gravis
in Rats by Neonatal FcR Blockade
Liming Liu,1 Ana Maria Garcia,2 Helen Santoro,3 Yixia Zhang, Kevin McDonnell,
Jennifer Dumont, and Alan Bitonti
M
ost autoimmune diseases involve both cellular and humoral immune responses to self-Ags in disease pathogenesis. However, there are several autoimmune diseases, such as myasthenia gravis (MG),4 that are predominantly
mediated by autoantibodies. MG satisfies the five criteria proposed
by Drachman (1) that characterize Ab-mediated autoimmune diseases: 1) autoantibodies are present in patients with the disease; 2)
Ab interacts with the target Ag; 3) passive transfer of Ab reproduces features of disease; 4) immunization with Ag produces a
model disease; and 5) reduction of Ab levels ameliorates the disease. In fact, MG was the first disease to be identified as being
predominantly Ab mediated (2). The disease symptoms include
muscle weakness and fatigability which are due to autoantibodies
generated against the acetylcholine receptor (AChR) and other
neuromuscular Ags. Up to 90% of MG patients have detectable
Abs against AChR, whereas ⬃10 –15% of the patients are negative
for anti-AChR Abs (seronegative MG; SNMG) (3, 4). However,
Igs from sera of SNMG patients bind to muscle cells that do not
express AChR (5), and they induce reductions in miniature endSyntonix Pharmaceuticals, Waltham, MA 02451
Received for publication October 6, 2006. Accepted for publication January
26, 2007.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
Address correspondence and reprint requests to Dr. Liming Liu, Syntonix Pharmaceuticals, 9 Fourth Avenue, Waltham, MA 02451. E-mail address: [email protected]
2
Current address: Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02472.
3
Current address: Novartis Institutes for BioMedical Research, 100 Technology
Square, Cambridge, MA 02139.
4
Abbreviations used in this paper: MG, myasthenia gravis; AChR, acetylcholine
receptor; EAMG, experimental autoimmune MG; SNMG, seronegative MG; FcRn,
neonatal FcR; SPF, specific pathogen free; CR3, complement receptor 3; Dex, dexamethasone; ␤2m, ␤2-microglobulin; SPR, surface plasmon resonance; mIgG1, murine
IgG1; Ctrl, Control.
Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00
www.jimmunol.org
plate potential amplitudes upon passive transfer to mice (6). Moreover, it has been shown that 47–70% of SNMG patients have serum IgG autoantibodies against the muscle-specific receptor
tyrosine kinase. Muscle-specific receptor tyrosine kinase mediates
the agrin-induced clustering of AChR during synapse formation
and is also expressed at the mature neuromuscular junction (7, 8).
Therefore, in addition to autoantibodies to AChR, Abs to neuromuscular junction components may also be responsible for pathogenesis of MG. The binding of the IgG Abs to AChR and other
muscle Ags recruits complement and triggers inflammatory responses, resulting in damage to the neuromuscular junction which
leads to further weakening of muscle contraction (9). It is logical
that drugs that lower the autoantibody level may have therapeutic
effects on MG. Current therapy involves primarily steroids, immunosuppressant drugs, or cytotoxic drugs which could have severe
side effects (10).
More than 40 years ago, it was hypothesized that there existed
a receptor that was responsible for the regulation of the catabolism
of IgG (11). It was also suggested that these receptors were saturable based on studies that showed increased metabolism of IgG
with hyperimmunization (12). The receptor known as neonatal
FcR (FcRn) was subsequently cloned and was found to be responsible for the protection of IgG from intracellular metabolism (13,
14). FcRn is composed of two subunits: an H chain (a MHC class
I-like molecule); and an L chain, ␤2-microglobulin (␤2m) (13) and
is expressed in many tissues including blood vessels, lung, liver,
intestine, and kidney (15–17). The current understanding of FcRn
function is as follows: IgG is taken up by endothelial cells that line
the vasculature by fluid phase pinocytosis, followed by pH-dependent binding to FcRn (18, 19) in acidic endosomes, thereby protecting IgG from releasing into lysosomal endosomes where protein degradation occurs. The IgG-FcRn complex is then shuttled
back to plasma membrane where the complex dissociates at neutral
pH and IgG is released back to the circulation (20). This protective
pathway is responsible for the long circulating half-life observed
for IgG. Several groups have provided strong evidence supporting
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
The neonatal FcR (FcRn) plays a critical role in IgG homeostasis by protecting it from a lysosomal degradation pathway.
It has been shown that IgG has an abnormally short half-life in FcRn-deficient mice and that FcRn blockade significantly
increases the catabolism of serum IgG in mice. Therefore, reduction of serum IgG half-life may have therapeutic benefits in
Ab-mediated autoimmune diseases. We have studied the therapeutic effects of an anti-rat FcRn mAb, 1G3, in two rat models
of myasthenia gravis, a prototypical Ab-mediated autoimmune disease. Passive experimental autoimmune myasthenia gravis
was induced by administration of an anti-acetylcholine receptor (AChR) mAb, and it was shown that treatment with 1G3
resulted in dose-dependent amelioration of the disease symptoms. In addition, the concentration of pathogenic Ab in the
serum was reduced significantly. The effect of 1G3 was also studied in an active model of experimental autoimmune myasthenia gravis in which rats were immunized with AChR. Treatment with 1G3 significantly reduced the severity of the disease
symptoms as well as the levels of total IgG and anti-AChR IgG relative to untreated animals. These data suggest that FcRn
blockade may be an effective way to treat Ab-mediated autoimmune diseases. The Journal of Immunology, 2007, 178:
5390 –5398.
The Journal of Immunology
Purification of AChR from Torpedo californica electroplax
AChR was purified from T. californica electroplax tissue that was obtained
from Aquatic Research Consultants according to the method described by
Aharonov et al. (30). The presence of four intact subunits was confirmed by
SDS-PAGE, and identity was shown by Western blot analysis using an
anti-AchR Ab (Affinity Bioreagents). The purity of the AChR preparation
was estimated to ⬃90% by SDS-PAGE.
Surface plasmon resonance (SPR) experiments
A Biacore 3000 biosensor system (Biacore) was used to assess the interaction of soluble rat FcRn with 1G3 or its isotype control, mIgG1Ctrl. 1G3
and control mIgG1 were covalently immobilized to a carboxymethyldextran-coated sensor chip (CM5) via primary amines using 1-ethyl-(3dimethylaminopropyl)carbodiimide hydrochloride/N-hydroxysuccinimide
chemistry as recommended by Biacore (BIApplications Handbook, version
AB, section 4.2; Biacore). The IgGs were diluted to 10 –30 ␮g/ml in 50
mM sodium acetate (pH 5) and were immobilized at densities sufficient to
produce 400 – 800 response units. Residual sites were subsequently
blocked by coupling with 1 M ethanolamine hydrochloride, pH 8.5. A
control flow cell was mock coupled with ethanolamine to serve as a blank.
Samples of soluble rat FcRn at different concentrations (in 50 mM sodium
phosphate, 100 mM NaCl (pH 6.0 or pH 7.4) with 0.01% surfactant P20)
were then injected over the biosensor chip for 4 min at 30 ␮l/min followed
by a 10-min dissociation in sample buffer. Between injections, residual
bound FcRn was removed with an 8-s injection of Pierce Ab/Ag gentle
elution buffer followed by a 30-s injection of sample buffer. Kinetic constants were derived from the sensorgram data using the BiaEvaluation software (version 3.1; Biacore). Association and dissociation data were simultaneously fit to models (1:1 Langmuir and heterogeneous ligand-parallel
reactions) supplied in the BiaEvaluation package. Equilibrium dissociation
constants (KD) were determined from the ratio of the kinetic constants (ka
and kd).
FcRn-binding assay
Rat FcRn-expressing cells (5 ⫻ 105/tube/0.1 ml) were incubated on ice
with 100 nM Alexa Fluor-488-labeled 1G3 or control mIgG1 at either pH
6.0 or pH 7.4 for 45 min. After one wash with either pH 6.0 or pH 7.4 PBS
containing 1% BSA, the cells were analyzed in a flow cytometer (Epics
MCL-XL; Beckman Coulter) for fluorescence staining.
FcRn competition assay
Materials and Methods
Reagents
Hybridoma cell lines secreting the mAbs 1G3 (CRL 2434), 1745 (CRL
1745) and mAb35 (HB-8857) were purchased from American Type Culture Collection. The 1745 cell line secretes a mAb against porcine parvovirus (29) and was tested with several cell lines and fresh primary cells
including human 293 cells, U937 cells, rat and mouse fibroblasts, fresh rat
blood leukocytes, and spleen cells; the 1745 mAb did not show staining on
these cells in flow cytometry analysis and therefore was used as an IgG1
isotype control (control mIgG1). The 1G3 hybridoma was cloned twice to
assure stable expression of functional 1G3 mAb. mAb-secreting hybridomas were grown either in roller bottles in DMEM (Invitrogen Life Technologies) supplemented with 10% FBS (Ultra low IgG FBS; Invitrogen
Life Technologies), 2 ⫻ 10⫺5 M 2-ME, 1 mM sodium pyruvate, 100 ␮g/ml
penicillin and streptomycin, 2 mM L-glutamine, 10 mM HEPES, and 0.1
mM nonessential amino acids or in a Bioreactor in protein free CD Hybridoma medium (Invitrogen Life Technologies). mAb was purified from
culture medium using a Hitrap Protein G column (GE Healthcare). Abs
were characterized by SDS-PAGE and isoelectric focusing gel electrophoresis. Affinity for soluble rat FcRn was also measured using surface
plasmon resonance methods (Biacore) as well as competition with IgG for
binding to rFcRn on cells.
Human IgG1 (Synagis; Medimmune) was purchased from a pharmacy.
In some experiments, human IgG1, 1G3, or mIgG1Ctrl were labeled
with Alexa Fluor-488 (Invitrogen Life Technologies) according to the
manufacturer’s instructions. Rat IgG was rat serum IgG supplied by
Sigma-Aldrich.
PE-labeled mouse anti-rat leukocyte surface Ags (OX8, anti-CD8;
OX38, anti-CD4; OX42, anti-CR3), and the isotype controls (IgG1, IgG2a
and IgG2b) were purchased from BD Pharmingen, and PE-labeled His24
(anti-CD45 B cell isoform) was purchased from eBioscience.
A rat fibroblast cell line stably transfected with rat FcRn was kindly
provided by Dr. N. Simister (Brandeis University, Waltham, MA).
Rat FcRn-expressing cells (3–5 ⫻ 105) were incubated at 4°C with 100 nM
Alexa Fluor-488-rat IgG in the presence of various concentrations of 1G3
or mIgG1Ctrl for 45 min in pH 6 binding buffer (calcium and magnesiumfree PBS containing 10 mM EDTA, Invitrogen Life Technologies). After
one wash with binding buffer, the cells were analyzed in a Beckman
Coulter flow cytometer by counting 5000 cells for fluorescence staining.
The background fluorescence was established with cells alone. Data are
expressed as total mean fluorescence intensity, calculated as the fraction of
positive cells multiplied by the mean fluorescence intensity of the positive
cells. The concentration of mAb that inhibits binding by 50% (IC50) was
calculated using SigmaPlot Software 2000 for curve-fitting analysis (Systat
Software).
Flow cytometry analysis of surface marker expression of spleen
cells
Spleens were removed from rats and cell suspensions were prepared by
teasing apart the spleens in a petri dish containing DMEM. The erythrocytes were lysed with ACK lysing buffer (BioSource International), and the
remaining cells were washed three times with PBS containing 1% BSA.
Cells were then incubated for 60 min on ice in a 96-well Costar assay plate
(1 ⫻ 106 cells/well/0.05 ml) with FITC-labeled mouse mAbs against rat
leukocyte surface Ags. After one wash, the cells were analyzed for fluorescence staining in a Beckman Coulter flow cytometer.
Measurement of Abs in serum
To measure total rat serum IgG, 96-well ELISA plates (Costar; Corning) were coated with 2.5 ␮g/ml purified goat anti-rat IgG Fc (Jackson
ImmunoResearch Laboratories) in carbonate-bicarbonate coating buffer
(Sigma-Aldrich) for 60 min at 37°C. After blocking with 0.1% solution
of BSA in PBS (blocking buffer) for 60 min at 37°C, the plates were
washed twice with 0.1% Tween 20 in PBS. Purified rat serum IgG (SigmaAldrich) and test rat sera were diluted in blocking buffer and added to
the plates which were incubated at 37°C for 60 min. After three washes,
goat anti-rat IgG F(ab⬘)2-HRP (Jackson ImmunoResearch Laboratories)
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
the role of FcRn in the regulation of IgG catabolism. IgG in ␤2mdeficient mice have an abnormally short serum half-life compared
with normal mice (21–23). FcRn H chain-deficient mice were also
compromised in their ability to protect IgG and lacked a robust
humoral immune response when immunized with Ag plus adjuvant (14).
Because FcRn has a critical role in maintaining the half-life of
IgG, it has been suggested as an attractive target for therapeutic
intervention in Ab-mediated autoimmune diseases (24, 25).
Akilesh et al. (26) showed that FcRn deficiency conferred protection against arthritis in an FcRn-deficient mice crossed with mice
susceptible to Ab-mediated autoimmune arthritis (K/B ⫻ N mouse
model). High dose i.v. Ig therapy, thought to target FcRn, was also
found to ameliorate arthritis disease symptoms, further suggesting
that FcRn blockade may have anti-inflammatory effects on arthritis. In a rat model of idiopathic thrombocytopenia purpura, another
Ab-mediated autoimmune disease, Getman and Balthasar (27)
demonstrated that 4C9, an anti-rat ␤2m mAb, induced a transient,
dose-dependent increase in the elimination of IgG. However, because ␤2m is also a subunit of MHC class I, the effects of 4C9 on
the IgG catabolism could not be solely attributed to the disruption
of FcRn function. A more specific approach is to directly target the
H chain subunit of FcRn. Recently, it has been shown that a mutant
of human IgG Fc that has greatly enhanced affinity for mouse FcRn
significantly increased the clearance of tracer and endogenous
mouse IgG (28).
Although data are accumulating to suggest that FcRn blockade
enhances the clearance of IgG, there is no direct evidence that
blockade of FcRn H chain could have therapeutic benefits in
Ab-mediated autoimmune diseases. We have investigated the
use of a high affinity anti-rat FcRn H chain mAb, 1G3, in the
treatment of rat models of MG and demonstrated a significant
reduction of disease symptoms for both passive and active models of EAMG.
5391
5392
EFFECTS OF FcRn BLOCKADE ON RAT EAMG
was added to the plates that were then incubated at 37°C for 60 min. All
plates were developed with SureBlue TMB (Kirkegaard & Perry Laboratories), and the reaction was terminated with stop solution (Kirkegaard & Perry
Laboratories). OD450 was determined using a plate reader (Molecular Devices). The concentration of IgG in test sera was calculated from the rat IgG
standard curve, and the limit of detection was ⬃12.5 ng/ml.
The measurement of anti-AChR Ab in serum was the same as that for
total IgG measurement except for some assay-specific reagents. Ninetysix-well ELISA plates were coated with 5 ␮g/ml AChR in carbonate-bicarbonate coating buffer overnight at 4°C. After blocking, either diluted
test rat serum or anti-AChR mAb35 (as a standard) was added to the wells.
The plates were developed with the same protocol described in the measurement of total rat serum IgG, and the limit of detection was ⬃12.5
ng/ml. Measurement of mAb35 concentration in rat serum in the passive
EAMG model followed this same protocol.
To measure total serum IgM, 96-well ELISA plates were coated with
2 ␮g/ml mouse anti-rat IgM mAb in coating buffer (BD Bioscience) and
incubated at 4°C overnight. After blocking, either rat serum or purified
rat serum IgM (Sigma-Aldrich) was added to the plates, and the plates
were incubated at 37°C for 60 min. Then goat anti-rat IgM-HRP (Jackson
ImmunoResearch Laboratories), 1- to 10,000-fold dilutions, was added and
incubated at 37°C for 60 min. The plates were developed with the protocol
described above. The concentration of IgM in test sera was calculated from
the rat IgM standard curve and the limit of detection was ⬃3 ng/ml. The
linear range was 6 –50 ng/ml.
Measurement of serum 1G3 or control mIgG1 levels was performed in
96-well ELISA plates that were coated with 5 ␮g/ml goat anti-mouse IgG
(Fab specific). After blocking, 1G3, control mIgG1, or rat serum was added
to the plates followed by incubation at 37°C for 60 min. After sample
incubation, goat anti-mouse IgG (H ⫹ L)-HRP (Jackson ImmunoResearch
Laboratories) in 1/10,000 dilutions was added to the plates, which were
then incubated at 37°C for 60 min. The plates were developed with the
same protocol described above. The concentration was calculated using a
1G3 standard or control mIgG1 standard curve, and the detection limit was
⬃12 ng/ml.
FIGURE 2. 1G3 Binding to rat FcRn-expressing cells at pH 6.0 and
7.4. Rat FcRn-expressing cells were incubated on ice with Alexa Fluor488-labeled 1G3 or control mIgG1 at either pH 6 (A) or pH 7.4 (B) for
45 min. After one wash, the cells were analyzed by flow cytometry for
fluorescence staining. Cells alone, Cells that were not incubated with
Abs. This experiment was repeated twice with similar results.
Induction and assessment of passive EAMG
All animal work was approved by the Syntonix Institutional Animal Care
and Use Committee. Passive EAMG was induced in female specific pathogen-free (SPF) Lewis rats (Charles River Laboratories) weighing ⬃100 g,
by i.p. administration of mAb35 at 0.75–1 mg/kg. The time of mAb35
administration was defined as time zero. 1G3 or control mIgG1 was administered i.p. at 2 and/or 24 h before disease induction with mAb35.
Disease progression was monitored twice daily by measuring disease score,
body weight, and grip strength. The visual determination of disease symptoms was graded in a scale of 0 – 4 (31), where 0 was no symptoms; 1, weak
grip, fatigability; 2, general weakness, hunched posture at rest, decreased
body weight, tremors; 3, severe weakness, moribund; and 4, death. If
needed, 100 –150 ␮l of blood from a tail clip were collected in a Minicollect serum collection tube (Greiner Bio-one). Grip strength was measured
with a grip meter (Columbus Instruments). PBS was used as the vehicle in
all experiments.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
FIGURE 1. SPR analyses of the interaction of rat-FcRn with immobilized mAb 1G3 or control mIgG1. Results are for a 1 ␮M injection of
rat-FcRn (50 mM sodium phosphate, 100 mM NaCl, 0.01% surfactant P20)
over a surface of 420 response units (RU) of 1G3 or 631 RU control mIgG1
at pH 6.0 or pH 7.4. KD and koff values were determined using BiaEval
software. 1G3 data were fit to a heterogeneous ligand model. This experiment has been repeated twice with similar results.
The Journal of Immunology
FIGURE 5. Effects of 1G3 on serum IgG concentration in rats. Two
groups (four per group) of female Lewis rats were injected i.p. with either
30 mg/kg 1G3 or control mIgG1 at 0 and 24 h. Serum was obtained at 0,
24, 48, 96, 120, and 168 h, and total IgG concentration was measured as
described in Materials and Methods. Arrows, Time of 1G3 injection. This
experiment was repeated once with similar results.
Results
High affinity binding of 1G3 to Rat FcRn and competition for
IgG binding at pH 6.0 and 7.4
Induction and assessment of active EAMG
SPF female Lewis rats, 6 –7 wk old (⬃150 g at the beginning of experiment), were from Charles River Laboratories. Rats were immunized by s.c.
injection at the base of the tail of 50 ␮g/50 ␮l Torpedo AChR emulsified
in CFA containing 10 mg/ml Mycobacterium tuberculosis R37. After immunization, rats were randomized to different groups to receive treatments
(8 –10 rats/group). Both therapeutic and control Abs were administered i.p.
as indicated in the figure legends. The disease progression was monitored
twice weekly (Monday and Friday) by recording body weight, grip
strength, and disease score.
1G3 is a mouse anti-rat FcRn mAb developed by Raghavan et al.
(32). 1G3 and its isotype control mIgG1 were covalently immobilized onto the surface of biosensor chips, and the binding of
soluble rat FcRn to these surfaces was monitored using a Biacore 3000 SPR instrument (Fig. 1). Several methods have been
reported for determining the affinity of the FcRn-IgG interaction using Biacore (33–37), and the particular method chosen
will yield different values for the association and dissociation
constants. Due to the high affinity of the interaction between
Statistic analysis
Data were evaluated for statistical significance using Student’s t test in
Sigmastat provided by Systat Software.
FIGURE 4. Pharmacokinetics of 1G3 in rats. Two groups (four per
group) of SPF female Lewis rats (⬃150 g, 6 –7 wk old) were injected i.p.
with either 1G3 or control mIgG1 (10 mg/kg). Blood was collected, and
serum was prepared at 0.25, 6, 24, 30, 48, 72, 96, 144, 192, and 240 h after
the injection. A specific ELISA was used to determine 1G3 and mIgG1
serum concentration as described in Materials and Methods. This experiment was repeated twice with similar results. ⴱ, The level of 1G3 at 24 h
is ⬍0.01 ␮g/ml.
FIGURE 6. Effects of 1G3 on passive EAMG. SPF female Lewis rats
(⬃100 g, 4 –5 wk old) were divided into five groups (six rats per group).
Animals were treated i.p. with vehicle, 1G3, or mIgG1Ctrl at various
time points before the induction of disease with mAb35. F, Vehicle
treated at ⫺2 h; , 30 mg/kg 1G3 at ⫺24 h; Œ, 30 mg/kg 1G3 at ⫺2
h; ⽧, 30 mg/kg 1G3 at ⫺24 h and ⫺2 h; f, control mIgG1 at ⫺24 and
⫺2 h. Disease symptoms were scored as described in Materials and
Methods. Comparison of disease scores at 24 and 48 h after disease
induction: single-dose 1G3-treated groups vs vehicle- or mIgG1Ctrltreated group, p ⬍ 0.05; two doses of 1G3 vs vehicle or mIgG1Ctrltreated group, p ⬍ 0.001.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
FIGURE 3. Competition of 1G3 for binding of IgG to rat FcRn. Rat
FcRn-expressing cells were incubated with different concentration of competitors in the presence of Alexa Fluor-488-labeled rat IgG for 45 min on
ice. All competition assays were conducted at pH 6.0. After one wash, cells
were analyzed in a Coulter flow cytometer. Results were expressed as total
mean fluorescence intensity (TMFI) calculated as the fraction of positive
cells multiplied by the mean fluorescence intensity. Data are representative
of three separate experiments.
5393
5394
EFFECTS OF FcRn BLOCKADE ON RAT EAMG
1G3 half-life in vivo
The pharmacokinetics of 1G3 and control mIgG1 were studied in rats
after a single i.p. administration. The results showed that 1G3 reached
a maximum serum concentration that was similar to the control
mIgG1 at 6 h, but then the concentration of 1G3 rapidly declined to
unmeasurable levels at 24 h (Fig. 4). In contrast, control mIgG1 had
a much longer half-life of ⬃104 h, similar to the 4- to 5-day half-life
found in normal mice (Ref. 38 and Fig. 4).
1G3 effect on serum IgG concentration in rats
To determine the effect of 1G3 treatments on endogenous serum
IgG concentration, rats were injected with 1G3 at 0 and 24 h, and
the serum IgG concentration was measured. As shown in Fig. 5,
1G3 significantly reduced the serum IgG concentration to ⬃60% of
the starting level, whereas the mIgG Ctrl did not affect endogenous
serum IgG levels. The effect lasted ⬃3 days after the last injection.
Blockade of FcRn inhibits disease symptoms in passive EAMG
FIGURE 7. 1G3 dose response in passive EAMG. SPF female Lewis
rats (⬃100 g, 4 –5 wk old) were treated with various doses of 1G3 at 24
and 2 h before disease induction with mAb35. The effects on disease
symptoms, grip strength, and body weight changes were measured as
described in Materials and Methods. Small arrows, 1G3 treatment;
large arrow, mAb35 injection. F, Vehicle; f, 30 mg/kg 1G3; Œ, 10
mg/kg 1G3; , 3 mg/kg 1G3; ⽧, 1 mg/kg 1G3. A, Disease score; B,
grip strength. – – – –, Average starting grip strength; C, body weight.
– – – –, Average starting body weight. Statistical significance: disease
scores for the 1G3-treated groups compared with the vehicle-treated
group at 24 and 50 h: 10 and 30 mg/kg, p ⬍ 0.001; 3 mg/kg, p ⬍ 0.01;
Passive EAMG was induced by injecting rats with mAb35, an
anti-AChR mAb known to induce the disease (39). We investigated the dose response to mAb35 in this disease model and determined that an i.p. injection of 0.75–1 mg/kg resulted in high
clinical scores (score 3 of a possible 4) without significant lethality
(data not shown). Disease progression was similar to that described
previously, with symptoms typically starting at 12 h, peaking at
48 h and gradually disappearing by 72 h post-mAb35 injection
(39). EAMG symptoms were almost completely prevented when
30 mg/kg 1G3 was administered twice at 24 and 2 h before mAb35
injection (Fig. 6). Single doses of 1G3 (30 mg/kg) administered
1 mg/kg, p ⬍ 0.05. Grip strength for the 1G3-treated group compared with
the vehicle-treated group: 10 mg or 30 mg/kg at 24 and 30 h, p ⬍ 0.001;
3 mg/kg at 24 and 30 h, p ⬍ 0.05; 1 mg/kg at 30 h, p ⬍ 0.05. Body weight
for the 1G3-treated group compared with the vehicle-treated group: 30
mg/kg at 24, 30, and 50 h, p ⬍ 0.01; 10 or 3 mg/kg at 24, 30, and 50 h, p ⬍
0.05; 1 mg/kg at 50 h, p ⬍ 0.05.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
1G3 and the FcRn, it was necessary to perform the analysis with
the IgG immobilized, because the FcRn protein was too sensitive to the conditions needed to regenerate the biosensor chip
surface between injections.
At pH 6.0 and 7.4, the 1G3-FcRn binding data fit best to a
heterogeneous ligand (two-site) model (total response is due to
two independent classes of noninteracting binding sites; the two KD
values each represent some fractional percent, f1% and f2% of the
observed signal). For control mIgG1-FcRn, the binding data were fit
to a simple 1:1 Langmuir model (one site; Refs. 33 and 37).
The equilibrium dissociation constants (KD) are calculated from
the ratios of the kinetic rate constants (KD ⫽ koff/kon). The average
values for three experiments are shown in Fig. 1. 1G3 exhibits a
high affinity for soluble rat FcRn and binds similarly at pH 6.0 and
7.4. By comparison, the control mIgG1 binds weakly to FcRn at
pH 6.0 and there is no apparent binding at pH 7.4. Also, the off-rate
(koff) for the binding of 1G3 to FcRn at pH 6 is two orders of
magnitude slower than for control mIgG1 binding to FcRn.
The binding of 1G3 and control mIgG1 to cells expressing rat
FcRn was also examined. Similar to the results obtained using
Biacore, 1G3 bound to rat FcRn on the cell surface at pH 6 and 7.4,
whereas the control mIgG1 bound only at pH 6 (Fig. 2).
A competition assay using flow cytometry was established to
test whether 1G3 could compete for IgG1 binding to rat FcRnexpressing cells. The results showed that 1G3 competed effectively
with rat IgG for binding to rat FcRn (Fig. 3).
The Journal of Immunology
5395
Table I. Serum mAb35 concentration after 1G3 treatmenta
m〈〉35 (␮g/ml)
Time (h)
Vehicle
1 mg/kg 1G3
3 mg/kg 1G3
10 mg/kg 1G3
30 mg/kg 1G3
3
7
24
50
t1/2
10.9 ⫾ 1.1
8.9 ⫾ 0.8
6.7 ⫾ 0.9
6.7 ⫾ 0.9
114
10.6 ⫾ 2.3
8.6 ⫾ 1.3
5.7 ⫾ 1.6
5.8 ⫾ 0.9
83
10.4 ⫾ 2
8.6 ⫾ 1.8
5.2 ⫾ 0.6*
5.3 ⫾ 0.4*
68
9.2 ⫾ 0.7
8.3 ⫾ 1.1
5.5 ⫾ 0.4**
3.9 ⫾ 0.4***
41
11.4 ⫾ 1.9
8.3 ⫾ 1.5
4.4 ⫾ 0.5***
3.3 ⫾ 0.6***
34
a
Sera were obtained from rats used in the experiment described in Fig. 7. Serum mAb35 concentrations were determined
as described in Materials and Methods. The time indicates hours after mAb35 injection. 1G3-treated groups compared with the
vehicle-treated group: ⴱ, p ⬍ 0.01; ⴱⴱ, p ⬍ 0.03; ⴱⴱⴱ, p ⬍ 0.001.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
either 24 or 2 h before mAb35 injection also significantly suppressed the disease symptoms. Administration of a nonspecific
mIgG1 control with the same dose and schedules (two injections)
failed to protect the animals (Fig. 6). An additional dose-scheduling experiment was performed in which rats were dosed once with
1G3 at 72, 48, or 24 h before disease induction with either 10 or
30 mg/kg mAb35. Only the rats injected 24 h before disease induction were protected (data not shown).
We then tested doses of 1, 3, 10, and 30 mg/kg and found that
1G3 reduced disease symptoms in a dose-dependent manner.
Treatments with 1G3 at either 10 or 30 mg/kg given 24 and 2 h
before disease induction almost completely prevented EAMG disease symptoms whereas 1 and 3 mg/kg 1G3 suppressed the disease
symptoms but to a lesser extent (Fig. 7A). Animals treated with
1G3 gained weight and did not lose grip strength, unlike the control group that exhibited significant declines in both of these parameters (Fig. 7, B and C).
To investigate whether the protective effect of 1G3 on disease
symptoms was reflected in the serum concentration of the diseaseinducing Ab, serum levels of mAb35 were measured. The concentration of mAb35 in serum was significantly reduced in animals
treated with 3, 10, and 30 mg/kg 1G3, and the reduction was dose
dependent (Table I). A dose of 1 mg/kg also reduced serum levels
of mAb35 but did not reach statistical significance.
1G3 reduces the disease severity of active EAMG
Autoimmune diseases such as MG are generally chronic. To investigate whether 1G3 has therapeutic benefit in a chronic model
of EAMG, we immunized rats with AChR in CFA. Treatments
were initiated 21 days after immunization with AChR, when disease symptoms started to appear. A dexamethasone treatment
group was included because steroids such as prednisone or dexamethasone are commonly used in the treatment of MG (10). Treatment with 30 mg/kg 1G3, three times per week, significantly suppressed active EAMG symptoms (Fig. 8A). The 1G3- and
dexamethasone-treated groups retained grip strength, and although
vehicle-treated animals were noticeably weaker in these tests, statistical significance was not reached (Fig. 8B). All three treatment
groups increased in body weight during the first 37 days (Fig. 8C).
After that time, dexamethasone-treated rats experienced a progressive decline in body weight that persisted until the experiment was
terminated 62 days after immunization. In a separate experiment,
mIgG1Ctrl was found to have no significant impact on the disease
progress (data not shown).
To investigate whether the FcRn blockade affected the serum
immunoglobulins in immunized animals, serum levels of total IgG,
IgM and specific anti-AChR IgG were measured (Table II). The baseline level of total serum IgG in the SPF rats was ⬃1.2 ⫾ 0.1 mg/ml,
and this increased markedly over the course of the experiment, reaching a high of 17.1 ⫾ 4.2 mg/ml by day 62. 1G3 suppressed the
FIGURE 8. Effects of 1G3 on active EAMG. Female Lewis rats (⬃150 g,
6 –7 wk old) were immunized with AChR and CFA as described in Materials
and Methods. Twenty-one days after immunization, rats were randomized into
three groups (10 per group) and treated three times per week with 30 mg/kg
1G3, 1 mg/kg dexamethasone, or vehicle. Disease scores, grip strength, and
weight changes were recorded as described in Materials and Methods. (A)
Disease scores; (B) grip strength. – – – –, Average starting grip strength; C,
body weight. – – – –, Average starting body weight. Comparison of disease
scores between the 1G3-treated group and the vehicle-treated group on day 37,
p ⬍ 0.05. These data are representative of three separate experiments.
5396
EFFECTS OF FcRn BLOCKADE ON RAT EAMG
Table II. Total serum IgG and AChR-specific Abs in active EAMGa
Time of
Measurement
Preimmune
Total IgG
Anti-AChR
Day 20
Total IgG
Anti-AChR
Day 34
Total IgG
Anti-AChR
Day 48
Total IgG
Anti-AChR
Day 62
Total IgG
Anti-AChR
Vehicle
(mg/ml)
1G3 (mg/ml)
Dexamethasone
(mg/ml)
1.21 ⫾ 0.70
NDb
1.30 ⫾ 0.20
ND
1.10 ⫾ 0.10
ND
4.90 ⫾ 0.70
0.56 ⫾ 0.17
4.70 ⫾ 1.90
0.45 ⫾ 0.07
6.70 ⫾ 2.40
0.71 ⫾ 0.15
6.40 ⫾ 2.40
0.45 ⫾ 0.16
4.60 ⫾ 1.50
0.20 ⫾ 0.08c
7.50 ⫾ 1.30
0.35 ⫾ 0.12
12.90 ⫾ 3.20
0.29 ⫾ 0.02
7.00 ⫾ 2.50c
0.20 ⫾ 0.02
11.90 ⫾ 5.50
0.12 ⫾ 0.03
17.10 ⫾ 4.20
0.15 ⫾ 0.07
9.20 ⫾ 2.70c
0.15 ⫾ 0.08
11.30 ⫾ 3.90
0.12 ⫾ 0.10
Discussion
increases in IgG, whereas dexamethasone did so less rapidly and to a
lesser extent. Serum AChR-specific IgG were also significantly suppressed after 2 wk of 1G3 treatment, whereas dexamethasone treatment had only minor effects on anti-AChR Abs. Treatment with 1G3
had no effect on serum IgM levels (data not shown).
It is well known that steroid treatment has broad immunosuppressive effects; therefore, we compared the effects of 1G3 to dexamethasone treatment on various indicators of the immune system.
The weights of spleens and draining lymph nodes of animals
treated with 1G3 remained at normal levels, whereas treatment
with dexamethasone reduced the spleen and lymph node weight
significantly, as expected (Table III). It was also of interest to
know whether FcRn blockade affects specific cells of the immune
system. Therefore, we measured a number of major immune cell
populations in the spleen by flow cytometry analysis. Treatment
with 1G3 did not affect the percentages of CD4⫹ and CD8⫹ cells
(primarily T lymphocytes), whereas the His24-expressing population (presumably mostly B cells) was significantly increased. The
number of CR3 (stained with OX42mAB)-expressing cells was
significantly reduced in the 1G3-treated group when compared
with those of the control group. CR3-expressing cells approximately doubled in the dexamethasone-treated group.
Table III. Effects of 1G3 and dexamethasone treatment on body weight and immune cell populations on day 62 after
immunizationa
Weight (mg)
Vehicle
1G3
Dexamethasone
Surface Marker Expression (% total spleen cells)
Spleen
Lymph
nodes
CD4
CD8
His24
OX42
Body
Weight (g)
348 ⫾ 66
347 ⫾ 92
247 ⫾ 32
191 ⫾ 62
191 ⫾ 63
44 ⫾ 25
49.2 ⫾ 3.9
53.8 ⫾ 3.5
25.8 ⫾ 8.1
7.5 ⫾ 1
8.1 ⫾ 1.6
4.3 ⫾ 0.5
28.5 ⫾ 6.0
40.4 ⫾ 5.4
19.5 ⫾ 1.9
42.7 ⫾ 5.8
34.3 ⫾ 5.9
74.5 ⫾ 6.4
171 ⫾ 25
187 ⫾ 24
140 ⫾ 9
a
Spleens and draining inguinal lymph nodes were isolated and weighed when the experiment described in Fig. 8 was completed. Single-cell
suspensions of spleens were also prepared and stained with FITC-labeled mAB recognizing surface CD4, CD8, His24 Ag (a B cell marker), and CR3
(OX42). The results of surface marker staining are expressed as percentage of positive cells of the total spleen cell population. Statistic comparison: spleen
weight, Dex-treated group vs vehicle- or 1G3-treated group, p ⬍ 0.023; lymph node weight, Dex-treated group vs vehicle- or 1G3-treated group, p ⬍
0.002. CD4-expressing cells: Dex-treated group compared with vehicle- or 1G3-treated group, p ⬍ 0.01. CD8-expressing cells: Dex-treated group
compared with vehicle- or 1G3-treated group, p ⬍ 0.01. His24-expressing cells: 1G3-treated group compared with vehicle-treated group, p ⬍ 0.005;
Dex-treated group compared with vehicle- or 1G3-treated group, p ⬍ 0.05. OX42-expressing cells: 1G3 treated group compared with vehicle-treated
group, p ⫽ 0.022; Dex-treated group compared with vehicle- or 1G3-treated group, p ⬍ 0.001. Dex, dexamethasone. The effect of 1G3 on the immune
system was also measured in another active EAMG experiment with similar results.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
a
Sera were obtained from rats used in the experiment described in Fig. 8. Total
serum IgG and anti-AChR IgG were measured with ELISA as described in Materials
and Methods.
b
Not detected.
c
A value of p ⬍ 0.005 when compared with the vehicle-treated group.
Existing evidence shows that FcRn plays a critical role in the homeostasis of serum IgG and that FcRn blockade enhances the
clearance of IgG (14, 28). However, there is no direct evidence
showing that a pH-independent FcRn H chain blocker could ameliorate Ab-mediated autoimmune diseases. In this article, we show
that 1G3, a high affinity and pH-independent blocker of rat FcRn
H chain, enhanced the clearance of pathogenic Abs and had significant therapeutic effects on both passively and actively
induced EAMG.
Depending on the disease severity, MG patients can be categorized into two groups: patients who have developed myasthenic
crisis; and patients who have generalized MG but not in crisis (9).
Passive EAMG induced by mAb35 resembles the disease characteristics of MG crisis, which is severe and has a fast onset. When
given either 24 or 2 h before mAb35 injection, 30 mg/kg 1G3
almost completely prevented the EAMG symptoms. When given
twice, at 3 or 1 mg/kg, 1G3 treatment significantly suppressed the
disease symptoms. The therapeutic effects on disease symptoms
were reflected in the grip strength and body weight changes, which
are a more objective measurement of the disease severity. Importantly, there was a dose-dependent reduction of mAb35 at 48 h in
the serum after 1G3 treatment, which strongly suggests that the
mechanism of 1G3 action was due to the enhanced clearance of
mAb35 by FcRn blockade. The reason why the serum mAb35
reduction lagged behind the disease symptom suppression is unclear. It is known that 50% of IgGs are distributed extravascularly
(38). Therefore, it is possible that 1G3 effects occurred first in the
extravascular space (for which there is no easy and reliable measurement) and manifested in the intravascular space later.
The pathogenic effect of mAb35 may be dependent on its serum
concentration and residence time. Loutrari et al. (40) reported that
mAb35 IgG induced EAMG readily, whereas Fab were not capable of inducing disease symptoms and F(ab⬘)2 induced minimal
disease. This disease induction pattern was attributed to the serum
residence time of these molecules in that Fab, and to a lesser extent
F(ab⬘)2, were rapidly removed from the circulation whereas the
full mAb IgG persisted for a long time (40). Subsequently, Poulas
et al. (41) reported that papain, which cleaves the IgG into free Fab
and Fc fragments, was able to reduce the severity of EAMG when
injected in vivo and that the papain-treated animals had a significant reduction of serum mAb35 concentration. Our data in the
passive EAMG model also show the correlation between therapeutic effects and the reduction of serum mAb35. In this regard,
our data are consistent with prior observations.
The Journal of Immunology
treatment with dexamethasone resulted in a reduction in lymph node
weight, due to the known immunosuppressive effects of the steroid.
CR3 expression is increased in inflammatory situations, and the
expression is reduced when the inflammation wanes (45, 46). It has
also been shown that anti-CR3 mAb treatments (administration of
OX42) significantly reduced the inflammation of acute colitis in rats
(47). In our study, CR3-expressing cells were significantly reduced in
the 1G3-treated group when compared with the vehicle-treated group.
Thus, the reduction of CR3-expressing cells may have contributed to
the therapeutic effects of 1G3. Several mechanisms might be responsible for the reduction of CR3-expressing cells: 1) 1G3 treatment
might have inhibited the function of the major inflammatory mediator,
polymorphonuclear cells. A recent report showed that FcRn may play
a major role in IgG-mediated phagocytosis by polymorphonuclear
cells (48); 2) 1G3 treatment could have suppressed the Ag processing
and presentation by FcRn-expressing monocytes and macrophages,
resulting in reduced inflammation. It has been shown that monocytes
and macrophages express functional FcRn that may positively influence Ag processing and presentation (49); 3) 1G3 treatments suppressed the up-regulation of Ag-specific IgG, resulting in reduced Ag
presentation. Ag-specific IgG complexed with its Ag can potentiate
Ag presentation through Fc␥R binding (50, 51).
Although MG is primarily mediated by autoantibodies, the underlying cellular immune responses also play an important role in autoantibody responses because the anti-AChR Ab response is T cell dependent (44, 52). T cell response to AChR was elevated in MG
patients (52–54). Thus, ideal therapeutics for Ab mediated autoimmune diseases such as MG may be the combination of therapies that
can reduce the autoantibody titer rapidly and, at the same time, control
the underlying cellular immune response. In this regard, FcRn blockade could be used in combination with lower doses of steroids (steroid
sparing). FcRn blockade could potentially lower the autoantibody
level below the threshold that triggers disease symptoms and a lower
dose of steroids or other immunosuppressant could control the underlying cell-mediated response. This combination therapy might
achieve maximum disease control with significantly reduced side effects. There is evidence that i.v. Ig therapy has a steroid sparing effect
in pemphigus patients, reducing the dose of steroid or, in some cases,
eliminating steroids completely (55).
Rituximab, an anti-CD20 mAb, has been tested in some Ab-mediated autoimmune diseases. Although rituximab substantially depleted CD20-expressing B cells in patients with systemic lupus erythematosus, elevated autoantibody titers persisted (56). This may be
due to the presence of low levels of residual autoreactive memory B
cells and/or long-lived autoreactive plasma cells. In agreement with
such observations, it has been shown that rituximab depletes shortlived plasmablasts but not long-lived plasma cells, which continue to
produce autoantibodies (57). In this situation, FcRn blockade may
have an advantage of rapidly controlling autoantibody levels.
In summary, we have shown that 1G3, a high affinity and pHindependent blocker of IgG binding to rat FcRn, substantially ameliorated passive EAMG and significantly reduced the symptoms of
active EAMG in rats without nonspecifically suppressing the immune
system. These results suggest that FcRn blockade may be an effective
and a novel way to treat autoantibody-mediated autoimmune diseases.
Acknowledgments
We are grateful for the technical assistance of Todd Hoehn and Sandy
Walker and helpful discussions from Drs. Adam Mezo and Susan Low.
Disclosures
L. Liu, Y. Zhang, K. McDonell, J. Dumont, and A. Bitonti are current employees, and A. M. Garcia and H. Santoro are former employees of Syntonix
Pharmaceuticals.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
One of the intriguing observations from this study is that 1G3
itself has a very short serum residence time and is completely gone
24 h after injection, whereas the mouse IgG1 control has a long
half-life (104 h). Biacore and flow cytometry experiments showed
that 1G3 bound to rat FcRn with high affinity and had a slow
off-rate at both pH 6 and pH 7.4. This high affinity, pH-independent binding and the rapid disappearance of 1G3 from circulation
suggest that once 1G3 binds to FcRn in vivo, it does not release
rapidly. A similar observation was reported previously in a study
involving human Fc mutants (42). Mutant Fc fragments that were
pH-independent, high affinity binders of mouse FcRn had abnormally short serum residence time when injected into mice (42).
Passive EAMG is a transient disease; therefore, therapeutic effects in this model may suggest a potential treatment option for
myasthenic crisis. To investigate the effects of FcRn blockade on
chronic MG, we used an active EAMG model that involved immunization of rats with AChR and CFA. Administration of 1G3
when disease symptoms appeared significantly suppressed disease
symptoms. The active EAMG model involves both humoral and
cellular arms of the immune response, whereas the passive EAMG
model is a pure Ab-mediated disease. This may explain why the
therapeutic effects on active EAMG are not as striking as in passive EAMG. Dexamethasone, a steroid used in many autoimmune
diseases including MG, also suppressed the disease symptoms but
with significant side effects: a body wasting syndrome; and leukopenia manifested by the significant reduction in weight of the
spleen and lymph nodes.
Immunization with AChR and CFA induced not only an antiAChR response but also a high magnitude immune response to
mycobacterial Ags that are the major components of CFA (43).
Naive rats had a low amount of total IgG in the serum (⬃1 mg/ml).
whereas serum IgG of immunized rats increased substantially (up
to 17 mg/ml). Treatment with 1G3 suppressed the increase in total
serum IgG by ⬃50% when compared with the vehicle-treated
group 48 and 62 days after immunization. It took several weeks of
1G3 treatment to establish IgG reduction, presumably due to the
faster rate of synthesis of IgG compared with its clearance rate in
this hyperimmune situation. Dexamethasone also appeared to suppress the increase in IgG in the serum, although the magnitude of
reduction was less than that induced by 1G3 treatment.
The kinetics of anti-AChR IgG is apparently different from that
of total IgG in that the anti-AChR IgG response appeared to have
reached the peak on day 20 and then declined gradually. Saoudi
et al. (44) reported a similar kinetics of anti-AChR IgG Ab response after immunization with AChR and CFA. Two weeks after
treatment began, serum anti-AChR IgG in the 1G3-treated group
was reduced significantly when compared with vehicle treated
group and this probably contributed to the therapeutic effects observed. It is predicted that FcRn blockade should affect only the
catabolism of IgG and not that of IgM, given that other Ig classes
do not bind to FcRn. Our results show that serum total IgM level
was comparable throughout the study period between the 1G3treated group and the vehicle-treated group.
Roopenian et al. (14) showed that FcRn H chain-deficient mice
are not capable of mounting a robust humoral immune response
but that there is no impairment in the cell-mediated immune response. Our results also suggest that FcRn blockade affects primarily the IgG Ab response and not the overall immune system.
Spleen weights from 1G3-treated and vehicle-treated groups were
comparable, whereas the weights for the dexamethasone-treated
group were significantly reduced. The weights of draining lymph
nodes increased initially after immunization for all animals. After
the treatment began, the lymph node weights were comparable
between the 1G3-treated and the vehicle-treated groups; however,
5397
5398
References
31. Barchan, D., M. C. Souroujon, S. H. Im, C. Antozzi, and S. Fuchs. 1999. Antigenspecific modulation of experimental myasthenia gravis: nasal tolerization with
recombinant fragments of the human acetylcholine receptor ␣-subunit. Proc.
Natl. Acad. Sci. USA 96: 8086 – 8091.
32. Raghavan, M., M. Y. Chen, L. N. Gastinel, and P. J. Bjorkman. 1994. Investigation of the interaction between the class I MHC-related Fc receptor and its
immunoglobulin G ligand. Immunity 1: 303–315.
33. Martin, W. L., and P. J. Bjorkman. 1999. Characterization of the 2:1 complex
between the class I MHC-related Fc receptor and its Fc ligand in solution. Biochemistry 38: 12639 –12647.
34. Medesan, C., P. Cianga, M. Mummert, D. Stanescu, V. Ghetie, and E. S. Ward.
1998. Comparative studies of rat IgG to further delineate the Fc:FcRn interaction
site. Eur. J. Immunol. 28: 2092–2100.
35. Zhou, J., J. E. Johnson, V. Ghetie, R. J. Ober, and E. S. Ward. 2003. Generation
of mutated variants of the human form of the MHC class I-related receptor, FcRn,
with increased affinity for mouse immunoglobulin G. J. Mol. Biol. 332: 901–913.
36. Gurbaxani, B. M., and S. L. Morrison. 2006. Development of new models for the
analysis of Fc-FcRn interactions. Mol. Immunol. 43: 1379 –1389.
37. Vaughn, D. E., and P. J. Bjorkman. 1997. High-affinity binding of the neonatal Fc
receptor to its IgG ligand requires receptor immobilization. Biochemistry 36:
9374 –9380.
38. Waldmann, T. A., and W. Strober. 1969. Metabolism of immunoglobulins. Prog.
Allergy 13: 1–110.
39. Tzartos, S., S. Hochschwender, P. Vasquez, and J. Lindstrom. 1987. Passive
transfer of experimental autoimmune myasthenia gravis by monoclonal antibodies to the main immunogenic region of the acetylcholine receptor. J. Neuroimmunol. 15: 185–194.
40. Loutrari, H., A. Kokla, and S. J. Tzartos. 1992. Passive transfer of experimental
myasthenia gravis via antigenic modulation of acetylcholine receptor. Eur. J. Immunol. 22: 2449 –2452.
41. Poulas, K., T. Tsouloufis, and S. J. Tzartos. 2000. Treatment of passively transferred experimental autoimmune myasthenia gravis using papain. Clin. Exp. Immunol. 120: 363–368.
42. Dall’Acqua, W. F., R. M. Woods, E. S. Ward, S. R. Palaszynski, N. K. Patel,
Y. A. Brewah, H. Wu, P. A. Kiener, and S. Langermann. 2002. Increasing the
affinity of a human IgG1 for the neonatal Fc receptor: biological consequences.
J. Immunol. 169: 5171–5180.
43. Ljungqvist, L., A. Worsaae, and I. Heron. 1988. Antibody responses against
Mycobacterium tuberculosis in 11 strains of inbred mice: novel monoclonal antibody specificities generated by fusions, using spleens from BALB.B10 and
CBA/J mice. Infect. Immun. 56: 1994 –1998.
44. Saoudi, A., I. Bernard, A. Hoedemaekers, B. Cautain, K. Martinez, P. Druet,
M. De Baets, and J. C. Guery. 1999. Experimental autoimmune myasthenia gravis may occur in the context of a polarized Th1- or Th2-type immune response
in rats. J. Immunol. 162: 7189 –7197.
45. Popovich, P. G., P. Wei, and B. T. Stokes. 1997. Cellular inflammatory response
after spinal cord injury in Sprague-Dawley and Lewis rats. J. Comp. Neurol. 377:
443– 464.
46. Lemstrom, K. B., A. K. Raisanen-Sokolowski, P. J. Hayry, and P. K. Koskinen.
1996. Triple drug immunosuppression significantly reduces aortic allograft arteriosclerosis in the rat. Arterioscler. Thromb. Vasc. Biol. 16: 553–564.
47. Palmen, M. J., C. D. Dijkstra, M. B. van der Ende, A. S. Pena, and E. P. van Rees.
1995. Anti-CD11b/CD18 antibodies reduce inflammation in acute colitis in rats.
Clin. Exp. Immunol. 101: 351–356.
48. Vidarsson, G., A. M. Stemerding, N. M. Stapleton, S. E. Spliethoff, H. Janssen,
F. E. Rebers, M. de Haas, and J. G. van de Winkel. 2006. FcRn: an IgG receptor
on phagocytes with a novel role in phagocytosis. Blood 108: 3573–3579.
49. Zhu, X., G. Meng, B. L. Dickinson, X. Li, E. Mizoguchi, L. Miao, Y. Wang,
C. Robert, B. Wu, P. D. Smith, W. I. Lencer, and R. S. Blumberg. 2001. MHC
class I-related neonatal Fc receptor for IgG is functionally expressed in monocytes, intestinal macrophages, and dendritic cells. J. Immunol. 166: 3266 –3276.
50. Hamano, Y., H. Arase, H. Saisho, and T. Saito. 2000. Immune complex and Fc
receptor-mediated augmentation of antigen presentation for in vivo Th cell responses. J. Immunol. 164: 6113– 6119.
51. Yada, A., S. Ebihara, K. Matsumura, S. Endo, T. Maeda, A. Nakamura,
K. Akiyama, S. Aiba, and T. Takai. 2003. Accelerated antigen presentation and
elicitation of humoral response in vivo by Fc␥RIIB- and Fc␥RI/III-mediated
immune complex uptake. Cell. Immunol. 225: 21–32.
52. Saito, R., H. Onodera, H. Tago, Y. Suzuki, M. Shimizu, Y. Matsumura, T. Kondo,
and Y. Itoyama. 2005. Altered expression of chemokine receptor CXCR5 on T cells
of myasthenia gravis patients. J. Neuroimmunol. 170: 172–178.
53. Richman, D. P., J. Patrick, and B. G. Arnason. 1976. Cellular immunity in myasthenia gravis: response to purified acetylcholine receptor and autologous thymocytes. N. Engl. J. Med. 294: 694 – 698.
54. McQuillen, D. P., S. M. Koethe, and M. P. McQuillen. 1983. Cellular response
to human acetylcholine receptor in patients with myasthenia gravis. J. Neuroimmunol. 5: 59 – 65.
55. Yeh, S. W., N. Sami, and R. A. Ahmed. 2005. Treatment of pemphigus vulgaris:
current and emerging options. Am. J. Clin. Dermatol. 6: 327–342.
56. Anolik, J. H., J. Barnard, A. Cappione, A. E. Pugh-Bernard, R. E. Felgar,
R. J. Looney, and I. Sanz. 2004. Rituximab improves peripheral B cell abnormalities
in human systemic lupus erythematosus. Arthritis Rheum. 50: 3580 –3590.
57. Hoyer, B. F., R. A. Manz, A. Radbruch, and F. Hiepe. 2005. Long-lived plasma cells
and their contribution to autoimmunity. Ann. NY Acad. Sci. 1050: 124 –133.
Downloaded from http://www.jimmunol.org/ by guest on February 7, 2017
1. Drachman, D. B. 1990. How to recognize an antibody-mediated autoimmune
disease: criteria. Res. Publ. Assoc. Res. Nerv. Ment. Dis. 68: 183–186.
2. Vincent, A. 2002. Unravelling the pathogenesis of myasthenia gravis. Nat. Rev.
Immunol. 2: 797– 804.
3. Lindstrom, J. M., M. E. Seybold, V. A. Lennon, S. Whittingham, and
D. D. Duane. 1976. Antibody to acetylcholine receptor in myasthenia gravis:
prevalence, clinical correlates, and diagnostic value. Neurology 26: 1054 –1059.
4. Evoli, A., E. Bartoccioni, A. P. Batocchi, F. Scuderi, and P. Tonali. 1989. AntiAChR-negative myasthenia gravis: clinical and immunological features. Clin.
Invest. Med. 12: 104 –109.
5. Blaes, F., D. Beeson, P. Plested, B. Lang, and A. Vincent. 2000. IgG from
“seronegative” myasthenia gravis patients binds to a muscle cell line, TE671, but
not to human acetylcholine receptor. Ann. Neurol. 47: 504 –510.
6. Burges, J., A. Vincent, P. C. Molenaar, J. Newsom-Davis, C. Peers, and D. Wray.
1994. Passive transfer of seronegative myasthenia gravis to mice. Muscle Nerve
17: 1393–1400.
7. Hoch, W., J. McConville, S. Helms, J. Newsom-Davis, A. Melms, and
A. Vincent. 2001. Auto-antibodies to the receptor tyrosine kinase MuSK in patients with myasthenia gravis without acetylcholine receptor antibodies. Nat.
Med. 7: 365–368.
8. Evoli, A., P. A. Tonali, L. Padua, M. L. Monaco, F. Scuderi, A. P. Batocchi,
M. Marino, and E. Bartoccioni. 2003. Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. Brain 126: 2304 –2311.
9. Nicolle, M. W. 2002. Myasthenia gravis. Neurologist 8: 2–21.
10. Schwendimann, R. N., E. Burton, and A. Minagar. 2005. Management of myasthenia gravis. Am. J. Ther. 12: 262–268.
11. Brambell, F. W., W. A. Hemmings, and I. G. Morris. 1964. A theoretical model
of gamma-globulin catabolism. Nature 203: 1352–1354.
12. Fahey, J. L., and A. G. Robinson. 1963. Factors controlling serum gamma-globulin concentration. J. Exp. Med. 118: 845– 868.
13. Simister, N. E., and K. E. Mostov. 1989. An Fc receptor structurally related to
MHC class I antigens. Nature 337: 184 –187.
14. Roopenian, D. C., G. J. Christianson, T. J. Sproule, A. C. Brown, S. Akilesh,
N. Jung, S. Petkova, L. Avanessian, E. Y. Choi, D. J. Shaffer, et al. 2003. The
MHC class I-like IgG receptor controls perinatal IgG transport, IgG homeostasis,
and fate of IgG-Fc-coupled drugs. J. Immunol. 170: 3528 –3533.
15. Israel, E. J., S. Taylor, Z. Wu, E. Mizoguchi, R. S. Blumberg, A. Bhan, and
N. E. Simister. 1997. Expression of the neonatal Fc receptor, FcRn, on human
intestinal epithelial cells. Immunology 92: 69 –74.
16. Borvak, J., J. Richardson, C. Medesan, F. Antohe, C. Radu, M. Simionescu,
V. Ghetie, and E. S. Ward. 1998. Functional expression of the MHC class I-related receptor, FcRn, in endothelial cells of mice. Int. Immunol. 10: 1289 –1298.
17. Antohe, F., L. Radulescu, A. Gafencu, V. Ghetie, and M. Simionescu. 2001.
Expression of functionally active FcRn and the differentiated bidirectional transport of IgG in human placental endothelial cells. Hum. Immunol. 62: 93–105.
18. Rodewald, R. 1976. pH-dependent binding of immunoglobulins to intestinal cells
of the neonatal rat. J. Cell Biol. 71: 666 – 669.
19. Raghavan, M., V. R. Bonagura, S. L. Morrison, and P. J. Bjorkman. 1995. Analysis of the pH dependence of the neonatal Fc receptor/immunoglobulin G interaction using antibody and receptor variants. Biochemistry 34: 14649 –14657.
20. Ober, R. J., C. Martinez, X. Lai, J. Zhou, and E. S. Ward. 2004. Exocytosis of IgG
as mediated by the receptor, FcRn: an analysis at the single-molecule level. Proc.
Natl. Acad. Sci. USA 101: 11076 –11081.
21. Ghetie, V., J. G. Hubbard, J. K. Kim, M. F. Tsen, Y. Lee, and E. S. Ward. 1996.
Abnormally short serum half-lives of IgG in ␤2-microglobulin-deficient mice.
Eur. J. Immunol. 26: 690 – 696.
22. Israel, E. J., D. F. Wilsker, K. C. Hayes, D. Schoenfeld, and N. E. Simister. 1996.
Increased clearance of IgG in mice that lack ␤2-microglobulin: possible protective role of FcRn. Immunology 89: 573–578.
23. Junghans, R. P., and C. L. Anderson. 1996. The protection receptor for IgG
catabolism is the ␤2-microglobulin-containing neonatal intestinal transport receptor. Proc. Natl. Acad. Sci. USA 93: 5512.
24. Christianson, G. J., W. Brooks, S. Vekasi, E. A. Manolfi, J. Niles, S. L. Roopenian,
J. B. Roths, R. Rothlein, and D. C. Roopenian. 1997. ␤2-Microglobulin-deficient
mice are protected from hypergammaglobulinemia and have defective antibody responses because of increased IgG catabolism. J. Immunol. 159: 4781– 4792.
25. Liu, Z., D. C. Roopenian, X. Zhou, G. J. Christianson, L. A. Diaz, D. D. Sedmak,
and C. L. Anderson. 1997. ␤2-Microglobulin-deficient mice are resistant to bullous pemphigoid. J. Exp. Med. 186: 777–783.
26. Akilesh, S., S. Petkova, T. J. Sproule, D. J. Shaffer, G. J. Christianson, and
D. Roopenian. 2004. The MHC class I-like Fc receptor promotes humorally mediated autoimmune disease. J. Clin. Invest. 113: 1328 –1333.
27. Getman, K. E., and J. P. Balthasar. 2005. Pharmacokinetic effects of 4C9, an antiFcRn antibody, in rats: implications for the use of FcRn inhibitors for the treatment
of humoral autoimmune and alloimmune conditions. J. Pharm. Sci. 94: 718 –729.
28. Vaccaro, C., J. Zhou, R. J. Ober, and E. S. Ward. 2005. Engineering the Fc region
of immunoglobulin G to modulate in vivo antibody levels. Nat. Biotechnol. 23:
1283–1288.
29. Katz, J. B., and R. A. van Deusen. 1985. Radioimmunoassay of adjuvant-associated porcine parvovirus using a monoclonal antibody in a nitrocellulose membrane system. J. Virol. Methods 12: 193–198.
30. Chak, A., and A. Karlin. 1992. Purification and reconstitution of nicotinic acetylcholine receptor. Methods Enzymol. 207: 546 –555.
EFFECTS OF FcRn BLOCKADE ON RAT EAMG