ArticlePDF Available

Demonstration of Immunoglobulin G, A, and E Autoantibodies to the Human Thyrotropin Receptor Using Flow Cytometry

Authors:

Abstract and Figures

Human TSH receptor (TSHR) autoantibodies with biological activity result in thyroid dysfunction, but antibodies that simply bind do not. We have applied flow cytometry to the measurements of IgG, IgA, and IgE immunoreactivity to the TSHR in patients with Graves' disease (GD) and thyroid eye disease (TED) and in normal controls. CHO cells stably expressing the extracellular domain of the TSHR with a glycophosphatidylinositol anchor were produced and found to express approximately 4 times as many receptors, but of similar affinity, as JP09 in TSH binding studies. Substantial increases in median fluorescence and peak channel fluorescence were obtained by flow cytometry using TSHR monoclonal antibodies on the glycophosphatidylinositol cells. IgG autoantibodies were demonstrated in 55 of 65 untreated GD patients, 3 of 25 normal subjects, and 4 of 8 atypical TED sera (negative for TSHR autoantibodies with biological activity) by flow cytometry and correlated poorly with thyroid-stimulating antibodies. IgA antibodies were present in 1 of 12 normal, 1 of 7 treated GD with TED, and 3 of 8 atypical TED sera. IgE binding was observed in 1 of 12 normal, 2 of 8 treated GD without TED, 1 of 6 treated GD with TED, and 0 of 8 atypical TED sera. In conclusion, we have demonstrated autoantibodies that bind directly to the TSHR in the majority of GD patients and in 50% of patients with atypical TED and a small number of normal controls lacking TSHR antibodies that affect function. Although predominantly IgG lambda, TSHR autoantibodies of the IgA and IgE isotypes are also detectable.
Content may be subject to copyright.
Demonstration of Immunoglobulin G, A, and E
Autoantibodies to the Human Thyrotropin Receptor
Using Flow Cytometry
RUSSELL METCALFE, NICOLA JORDAN, PHILIP WATSON, SEVIM GULLU, MARIE WILTSHIRE,
MICHELE CRISP, CAROL EVANS, ANTHONY WEETMAN, AND MARIAN LUDGATE
Department of Medicine, Clinical Sciences Center, Northern General Hospital (R.M., P.W., A.W.), Sheffield, United Kingdom
S5 7AU; and Departments of Medicine (N.J., S.G., M.C., M.L.) and Pathology (M.W.), University of Wales College of
Medicine, and Department of Medical Biochemistry, University Hospital of Wales National Health Service Trust (N.J.,
C.E.), Heath Park, Cardiff, United Kingdom CF14 4XN
Human TSH receptor (TSHR) autoantibodies with biological
activity result in thyroid dysfunction, but antibodies that sim-
ply bind do not. We have applied flow cytometry to the mea-
surements of IgG, IgA, and IgE immunoreactivity to the TSHR
in patients with Graves’ disease (GD) and thyroid eye disease
(TED) and in normal controls.
CHO cells stably expressing the extracellular domain of the
TSHR with a glycophosphatidylinositol anchor were pro-
duced and found to express approximately 4 times as many
receptors, but of similar affinity, as JP09 in TSH binding stud-
ies. Substantial increases in median fluorescence and peak
channel fluorescence were obtained by flow cytometry using
TSHR monoclonal antibodies on the glycophosphatidylinosi-
tol cells.
IgG autoantibodies were demonstrated in 55 of 65 un-
treated GD patients, 3 of 25 normal subjects, and 4 of 8 atypical
TED sera (negative for TSHR autoantibodies with biological
activity) by flow cytometry and correlated poorly with thy-
roid-stimulating antibodies. IgA antibodies were present in 1
of 12 normal, 1 of 7 treated GD with TED, and 3 of 8 atypical
TED sera. IgE binding was observed in 1 of 12 normal, 2 of 8
treated GD without TED, 1 of 6 treated GD with TED, and 0 of
8 atypical TED sera.
In conclusion, we have demonstrated autoantibodies that
bind directly to the TSHR in the majority of GD patients and
in 50% of patients with atypical TED and a small number of
normal controls lacking TSHR antibodies that affect function.
Although predominantly IgG
, TSHR autoantibodies of the
IgA and IgE isotypes are also detectable. (J Clin Endocrinol
Metab 87: 1754 –1761, 2002)
AUTOANTIBODIES to the TSH receptor (TSHR) are
found in patients with autoimmune thyroid disor-
ders, particularly Graves’ disease (GD). They are classified
according to their biological activity. Thyroid-stimulating
antibodies (TSAB) mimic the action of TSH and result in
hyperthyroidism. Thyroid-blocking antibodies (TBAB) in-
hibit TSH induced stimulation and may be the cause of
hypothyroidism in idiopathic myxedema. Both TSAB and
TBAB may also be TSH binding-inhibiting Igs (TBII), which
prevent hormone/receptor interaction (1).
Assays in current diagnostic use are not able to detect
antibodies that simply bind to the receptor without any effect
on the TSHR or TSH binding. The other major autoantigens
in autoimmune thyroid disease are Tg and thyroperoxidase
(TPO); both can be detected by direct binding, e.g. in ELISA
(2). The incidence of TPO antibodies would be grossly un-
derestimated if the only detection methods relied on a bio-
logical effect such as inhibition of enzyme activity. Direct
binding has also revealed that 10–20% of normal euthyroid
individuals, especially women, have circulating Tg and TPO
autoantibodies, but little has been reported regarding TSHR
antibodies in such a control population (3).
Previous studies investigating direct binding of autoanti-
bodies to the TSHR have concentrated on sera from known
TSAB- and/or TBII-positive GD patients. A variety of meth-
ods have been used, including ELISA, in vitro transcription/
translation, Western blotting of bacterially produced TSHR,
and flow cytometric analysis of cells expressing the full-
length receptor or the extracellular domain (ECD) anchored
via a glycophosphatidylinositol (GPI) link (3–10). Most stud-
ies report TSHR-binding autoantibodies in a proportion of
sera containing TSAB and/or TBII and demonstrate the
heterogeneity of TSHR autoantibodies, e.g. a GD patient with
monoclonal gammopathy, having antibodies that bound
to the TSHR in a Western blot, but were TBII and TSAB
negative (11).
In the case of the TSHR, antibodies without biological
effect may be of particular relevance in the extrathyroidal
manifestations of GD, thyroid eye disease (TED), and
pretibial myxedema. Pretibial myxedema is rare, but TED is
frequently associated with GD, and several lines of evidence
indicate that the TSHR may have a role in both disorders (12).
These include the demonstration of TSHR transcripts and
protein in the ocular tissues, especially in the adipose com-
partment (13–15), and the development of animal models of
TED induced by transfer of TSHR-primed T cells (16) or
TSHR genetic immunization (17). A strong argument against
the TSHR being involved in TED is the fact that not all
patients with TED have TSAB or TBII. This heterogeneous
Abbreviations: ECD, Extracellular domain; FITC, fluorescein isothio-
cyanate; GD, Graves’ disease; GPI, glycophosphatidylinositol; TBAB,
thyroid-blocking antibodies; TBII, TSH binding-inhibiting Igs; TED, thy-
roid eye disease; TPO, thyroperoxidase; TSAB, thyroid-stimulating an-
tibodies; TSHR, TSH receptor.
0013-7227/02/$15.00/0 The Journal of Clinical Endocrinology & Metabolism 87(4):1754 –1761
Printed in U.S.A. Copyright © 2002 by The Endocrine Society
1754
group of individuals with severe symptoms of TED in the
absence of TSHR autoantibodies in conventional assays,
which we have termed atypical TED, may have no evidence
of thyroid dysfunction or may have been thyrotoxic
previously.
TSAB are predominantly of the IgG1 isotype, and there is
some evidence for light chain restriction. Other isotypes have
been implicated in the extrathyroidal manifestations of GD
(18), and increases in the level of IgA secretion in tears have
also been reported in 25% of patients (19). Furthermore, in
the receptor-induced animal models of TED, mast cells were
an early indicator of orbital pathology, and elevated IgE
levels have been found in GD patients (20), both of which are
suggestive of IgE immunoreactivity to the TSHR.
The aim of the present study was to investigate antibodies
binding to the TSHR in individuals negative for TSAB and
TBII, either normal controls or patients with atypical TED,
and to determine whether TSHR autoantibodies are exclu-
sively of the IgG isotype and/or light chain restricted in these
and a panel of GD sera.
Subjects and Methods
Subjects studied
Serum samples from a total of 110 individuals were tested. This
included 65 untreated patients with GD, defined by thyrotoxicosis, a
diffuse goiter, and the presence of Tg or TPO antibodies or an appro-
priate family history or exophthalmos, and 12 patients with treated GD.
Of the latter group, 6 also had TED, i.e. exophthalmos measured by
exophthalmometry. All of these patients had TSHR autoantibodies de-
tected as TSAB or TBII. We also selected 8 atypical TED patients who
were negative for TSAB and TBII in conventional assays: 3 with no
evidence of thyroid dysfunction, 1 with newly diagnosed GD, and 4 who
were previously treated for GD. All 8 patients had exophthalmos of at
least grade II; their details are shown in Table 1. We also studied 25
biochemically euthyroid normal blood donors (16 females and 9 males)
selected at random from the local blood transfusion service. All samples
were obtained with informed consent and with the approval of the local
ethics committee in accordance with the Declaration of Helsinki.
Measurements of TSAB, TBAB, and TBII
TSAB and TBII were measured on CHO cell lines stably expressing
the human TSHR, lulu or NA-4 for TSAB (21, 22) and JP09 for TBII (23),
as previously described in detail.
Construction and characterization of GPI cell line
Construction of GPI-anchored TSHR ECD. A synthetic linker encoding a
thrombin cleavage site and an artificial GPI attachment site based on a
region of domain 3 of rat CD4 (7) was created by annealing of the
following seven oligonucleotides and cloned into the XhoI and XbaI sites
of pcDNA3.1 (Invitrogen, San Diego, CA), 5-TCGAGCTGGTGCCAA-
GAGGCTCTATCGAGGGCAGA-3,5-CGTGATGGATGTGCCTCT-
GCCCTCGATAGAGCCTCTTGGCACCAGC-3,5-GGCACATCCAT-
CACGGCCTATAAGAGTGAG-3,5-CTCCGCTGACTCCCCCTCAC-
TCTTATAGGC-3,5-GGGGAGTCAGCGGAGTTCTTCTTCCTACTC-
3,5-CTAGCTAGACGAGCACGAGCAGGAGCAGAAGGATGAGT-
AG GAAGAAGAA-3,5-ATCCTTCTGCTCCTGCTCGTGCTCGTC-
TAG-3,5-GGCTCGAGTATGTCTTCACACGGGTT-GAACTC-3, and
5-GCCGGATCCATGAGGCCGGCGGA-CTTGCTG-3.
The region coding the extracellular domain of human TSHR [amino
acids 1 (methionine) to 412 (isoleucine)] was amplified using the High
Fidelity Taq polymerase (Roche, Indianapolis, IN) and the following
oligonucleotide primers: sense, 5-GCCGGATCCATGAGGCCGGCG-
GACTTGCTG-3; antisense, 5-GGCTCGAGTATGTCTTCACACGGG-
TTGAACTC-3, and was cloned into the BamHI and XhoI sites upstream
of the GPI anchor sequence to give the plasmid pGPI-TSHR. The final
construct was verified by sequencing using an ABI 310 system (PE
Applied Biosystems, Foster City, CA). Plasmid pGPI-TSHR was trans-
fected into CHO-K1 cells using Tfx-50 (Promega Corp., Madison, WI)
according to the manufacturers instructions, and stable lines were se-
lected using G418 at a concentration of 400
g/ml. Clonal lines were
obtained and screened by FACS using TSHR monoclonal antibody 2C11
(Serotec, Oxford, UK).
The expression of TSHR ECD on the GPI cell surface was demon-
strated by flow cytometry using monoclonal antibodies A10 (1:50 dilu-
tion) (24), BA8 (1:10 dilution) (25), and 3G4 (1:50 dilution) (25), used as
culture medium, and 2
g/tube purified 2C11 (Serotec) to the human
TSHR, followed by an antimouse IgG-fluorescein isothiocyanate (Ig-
FITC) conjugate (1:32; DAKO Corp., Carpenteria, CA), as described in
detail below.
TSH binding studies were performed on 1.5 10
5
cells in 12-well
plates to estimate the numbers of receptors expressed at the surface of
the GPI cells and were compared with JP09 (and the control JP02, CHO
TABLE 1. AtypicalTED patient details: summary of flow cytometry results
No./sex/age TSAB/TBII/TPO Treatment/diagnosis IgG IgA IgE
Euthyroid TED
P1 F 53 ve ve ve TED confirmed in all 3 cases by
C.T. and/or M.R.I. scan
ve ve ve
P2 M 62 ve ve ve ve ve ve
P3 F 67 ve ve ve ve ve ve
GD with TED TSAB and TBII negative
P4 F 37 ve ve ve Previous MMI ve ve ve
P5 F 46 ve ve ve Previous PTU ve ve ve
P6 F 60 ve ve ve Previous MMI ve ve ve
P7 F 47 ve ve ve Radio-iodine ve ve ve
P8 F 32 ve ve ve Untreated ve ve ve
GD with TED TSAB positive
GD 1 F 60 ve ve ve Previous MMI ve ve ve
GD 14 F 62 ve ve ve Untreated ve ve ve
GD 15 F 49 ve ve ve Untreated ve ve ve
Normal subject
N6 F 52 ve ve N.D. ve ve N.D.
C.T., Computerized tomographic; M.R.I., magnetic resonance imaging; MMI, methimazole; PTU, propylthiouracil; N.D., not determined.
TSAB and TBII were measured on CHO cell lines stably expressing the human TSHR, lulu, or NA-4 for TSAB (21, 22) and JP09 for TBII
(23), as described previously, and TPO antibodies using a Roche Immunodiagnostics enzymun test, values greater than 32 kU/liter being positive.
Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 1755
cells expressing the neomycin resistance plasmid but no TSHR). Binding
was performed in binding buffer comprised of NaCl-free Hankssolu-
tion, 280 mmsucrose, and 0.2% BSA. Initially, saturation curves, using
increasing volumes of [
125
I]TSH (Brahms Diagnostica, Berlin, Germany)
from 50500
l/well and an equal volume of binding buffer, were
carried out. Subsequently, the optimized volume of tracer was competed
for by increasing concentrations of cold bovine TSH (Sigma, St. Louis,
MO)for2hatroom temperature. Cells were washed twice with the
binding buffer and lysed with 0.5 ml 1 nNaOH/well, and bound
radioactivity was determined in a
-counter. All measurements were
made in duplicate and performed at least three times to calculate average
EC
50
and binding capacity values from Scatchard analysis, reported as
milliunits per ml TSH.
Flow cytometry
Seventy to 90% confluent cells (GPI and JP02) were detached from
75-cm
2
culture dishes using 5 ml 5 mmEDTA and 5 mmEGTA in PBS.
The cells were washed three times in PBS containing 0.1% BSA and
adjusted to 2 10
6
cells/ml in the same buffer. Aliquots (100
l) of cells
were incubated with 2
l heat-inactivated test serum from the various
patient and control groups for1hatroom temperature. After three
washes in PBS-BSA, they were incubated on ice in the dark for 30 min
with antihuman IgA-FITC conjugate (1:32, from Sigma), antihuman IgE
(1:50, from Sigma; 1:50, from Serotec) or antihuman IgG (1:50, from
Sigma). They received an additional three washes in PBS-BSA and were
resuspended in 1 ml of the same buffer, but containing propidium iodide
FIG. 1. Characterization of the GPI cell line. A, Typical
profile of TSHR staining at the surface of GPI. The faint
trace shows the fluorescence intensity using an isotype
control monoclonal, and the bold trace shows the fluo-
rescence intensity with TSHR monoclonal 2C11. The D
value is 0.93. B, TSH binding to TSHR expressing CHO
GPI and JP09 and the control JP02 cell lines. Binding
was performed with 40,000 counts/well (JP09 and JP02)
or 80,000 counts/well (GPI). The x-axis shows the con-
centration of added cold TSH in milliunits per ml, and
the y-axis shows the total counts bound (mean of du-
plicates agreeing to within 10%).
1756 J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies
for gating out dead cells. Cells were also analyzed in the same protocol,
but omitting the first antibody (either TSHR monoclonal or patients
serum) to control for nonspecific binding of the mouse and human FITC
conjugates. In addition, 12 of the samples positive for IgG binding to GPI
were analyzed further by replacing the antihuman IgG-FITC with either
an antihuman
or an antihuman
light chain-FITC.
Flow cytometric analysis was performed on a FACS Vantage from
Becton Dickinson and Co. (Mountain View, CA), incorporating a Co-
herent Enterprise II laser emitting at 488 nm. Forward light scatter, 90°
light scatter, and fluorescence emissions were collected for 1 10
4
cells,
and the geometric mean fluorescence intensity values of GPI and JP02
were compared for all sera, including the normal samples. In addition,
the Kolmogorov-Smirnov (K-S) two-sample test, which gives the great-
est difference between the two histograms (GPI and JP02) and is quoted
as the D value, was used (26), and cut-off values were defined based on
the mean 2sd of the normal sera.
Results
Characterization of the GPI cell line
Surface expression of TSHR ECD was confirmed by flow
cytometry. Background staining with an isotype-matched
control antibody showed a median fluorescence of 4.03 and
peak channel fluorescence of 3 compared with 667 and 889
when using the 2C11 monoclonal (D 0.93). Similar results
were obtained with the other three TSHR monoclonal anti-
bodies tested, which together recognize epitopes at the ex-
treme N- and C-termini of the TSHR ECD and the native
conformation of the human TSHR. A typical experiment is
shown in Fig. 1A.
Saturation curves for [
125
I]TSH binding demonstrated that
JP09 reached saturation with 150
l (40,000 cpm) tracer, but
GPI required 300
l (80,000 cpm) to achieve this, the first
indication of the greater surface receptor expression in the
latter cells (data not shown). TSH binding curves are shown
in Fig. 1B. The affinity of the ECD TSHR expressed in the GPI
cells is similar to that of the full-length TSHR in JP09, with
EC
50
values of 4.3 (range, 26) and 3.4 (range, 35) mU/ml
TSH, respectively. Scatchard analysis of the GPI and JP09 cell
lines produced Bmax values of 76 and 19 mU/ml TSH re-
spectively, indicating approximately 4 times as many recep-
tors expressed at the surface of GPI compared with JP09.
Flow cytometry
Each patient or control serum was assayed against both the
JP02 and GPI cell lines, because of variable reactivity of
human sera with the control receptor negative cell line, e.g.
when detecting IgG binding the geometric mean ranged from
1492. The distributions of fluorescence and background
fluorescence were similar for the receptor-negative and -pos-
itive populations.
Based on the mean 2sd of the normal control serum, a
D value of more than 0.28 was considered positive, and 55
of 65 untreated GD sera displayed specific positive binding
to the GPI cell population. As shown in Fig. 2, there was a
poor correlation (r 0.22) between autoantibodies binding
directly to the TSHR and TSAB measured in a luminescent
bioassay.
Twelve of the sera positive on GPI were tested for
or
light chain restriction in their TSHR binding antibodies.
From the mean 2sd of the 12 sera and the difference in
D value between
and
, 1 sample displayed complete
restriction, and 7 of the other 11 displayed more binding
associated with
than
light chains, as summarized in
Table 2.
Positive specific IgG binding to the ECD TSHR of the GPI
cells was also observed in 3 of 25 normal and 4 of 8 of the
TSAB/TBII-negative TED sera. Representative histograms
are shown in Fig. 3.
IgA binding to the control receptor-negative cells had a
range of geometric mean from 48 112. A D value greater than
0.35 was taken as the cut-off, and positive IgA receptor au-
toantibodies were demonstrated in 1 of 12 normal sera, 1 of
7 GD with TED, and 3 of 8 of the TSAB/TBII-negative TED
sera. Representative results are shown in Fig. 4.
Circulating IgE levels are considerably lower than those
for IgG and IgA, making the production of specific antisera
more difficult. We applied two different IgE-FITC conju-
gates, both polyclonal antibodies raised in goat that pro-
duced cut-offs for the D values of 0.41 (Sigma) and 0.25
(Serotec), respectively. Positive IgE binding was demon-
strated, using both IgE-FITC conjugates, in 1 of 12 normal
sera (28-yr-old male), 2 of 6 GD without TED, 1 of 8 GD with
TED, and 0 of 8 atypical TED sera. Representative results are
shown in Fig. 5.
FIG. 2. TSHR autoantibodies in 65 patients with untreated GD.
TSAB were assessed by luminescent bioassay, and results are re-
ported as a percentage of a TSAB standard on the x-axis. Antibodies
(IgG) binding directly to the TSHR were measured by flow cytometry
on GPI cells, and results are reported as a D value on the y-axis. There
is a poor correlation (r 0.22) between the two methods. The dotted
lines depict the normal cut-off (TSAB, 40%; D ⫽⬎0.28) for TSAB
and direct TSHR binding, respectively.
TABLE 2. Lambda light chain predominance in direct binding
IgGs to the TSHR
GD sample Dvalue kappa Dvalue lambda
GD 2 0.51 0.81
GD 3 0.26 0.65
GD 4 0.44 0.60
GD 5 0.55 0.39
GD 6 0.55 0.53
GD 7 0.57 0.83
GD 8 0.55 0.46
GD 9 0.46 0.77
GD 10 0.42 0.55
GD 11 0.53 0.74
GD 12 0.69 0.67
GD 13 0.49 0.74
Based on the mean 2SD of the group as a whole, D values for
kappa 0.3 and for lambda 0.37 were considered negative. On this
basis, patient 3 displays complete lambda restriction because the
kappa value is negative.
Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 1757
Flow results for IgG, IgA, and IgE binding, showing the
presence of receptor antibodies of more than one isotype in
some individuals, are summarized in Table 1.
Discussion
We have produced a CHO cell line stably expressing the
ECD of the TSHR linked to the cell surface via a GPI anchor.
The characteristics of the GPI line are similar to those in two
earlier reports (7, 8) and indicate that the ECD has an affinity
for TSH comparable to that of the full-length TSHR and is
sufficient to detect binding of TSHR autoantibodies.
We have demonstrated IgG antibodies binding directly to
the receptor in the majority of untreated patients with GD,
in agreement with some (8, 10), but not all (9), previous
studies that have applied flow cytometric analysis. We found
a poor correlation between these direct binding antibodies
and TSAB, as reported by others (10). Our data lend further
support to the concept that TSAB comprise a small propor-
tion of TSHR autoantibodies. The results also indicate that
TSHR autoantibodies in general display a preference for the
IgG
isotype and that it is not a feature confined to TSAB (1).
The presence of TSHR autoantibodies in GD is not sur-
prising, but we have also obtained positive binding of IgG
and/or IgA antibodies to GPI cells in 50% of patients with
atypical TED and a small proportion of normal euthyroid
controls, all of them women. Binding of IgE antibodies to the
GPI cells, apart from 3 of 14 GD sera, was detected only in
one 28-yr-old normal male subject and in none of the TSAB/
TBII-negative TED patients.
The etiopathogenesis of TED remains a puzzle to endo-
crinologists, although most of the signs and symptoms can
be attributed to an increase in orbital volume as the conse-
quence of glycosaminoglycans production, edema, and fat
hypertrophy. A variety of immune cells and cytokines are
present in TED orbits, and TSHR-specific T cell lines have
been reported (27). Several additional lines of evidence favor
the TSHR as a common antigen in GD and TED, as described
above. The present study has demonstrated immunoreac-
tivity to the TSHR in a subset of patients with TED previously
assumed to be free of receptor autoimmunity. Their euthy-
roid state is explained by the neutral nature of their TSHR
autoantibodies, which lack TSAB or TBII activity. Given the
size of an Ig, it is perhaps surprising that an antibody that is
FIG. 3. IgG autoantibodies binding directly to the TSHR. Flow cytometry with fluorescence intensity shown on the x-axis and cell counts on
the y-axis. The bold trace was obtained from GPI cells, and the faint trace was obtained from JP02 receptor-negative control cells. A, Atypical
TED P4 (D 0.5); B, atypical TED P1 (D 0.2); C, normal 5 (D 0.44); D, normal 7 (D 0.19). A D value more than 0.28 is considered positive.
1758 J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies
able to bind the receptor does not inhibit TSH binding, al-
though this has been reported previously (4), as have TSAB
that lack TBII activity (11). Measurement of direct binding
TSHR antibodies could be applied to confirm the diagnosis
if TED is suspected in an individual negative for TSAB and
TBII.
There are a few reports indicating TSHR autoreactivity in
normal healthy subjects, including the demonstration of re-
ceptor-specific T cell lines (28) and peripheral blood mono-
nuclear cells (29). More recently Atger and colleagues (3)
applied an ELISA, using a solubilized TSHR preparation to
coat the plates, and found neutral antibodies in a surprisingly
high proportion of euthyroid controls. In our series of normal
subjects, TSHR antibodies were confined to three women,
aged 4652 yr, one of whom displayed both IgG and IgA
reactivity.
The idea of autoantibodies in healthy subjects is not novel,
and the other major thyroid autoantigens, Tg and TPO, are
associated with incidences of approximately 15% and 10%,
respectively; they are highest among middle-aged women
(2). It seems that the TSHR is not so different, although
studies using a larger panel of euthyroid male and female
subjects spanning all age ranges are required to determine
the prevalence of TSHR autoreactivity in the absence of
disease.
GD is very common, and our data imply that immuno-
reactivity to the target antigen may also be common. Is its
immunogenicity due to molecular mimicry of the TSHR by
structures on the surface of commensal bacteria, or does
receptor processing release/unmask an immunodominant
epitope? Immunodominant epitopes have been implicated in
a variety of autoimmune diseases, and indeed, there is a
suggestion that the development of thyroiditis requires re-
moval of immunodominant regions in a receptor-induced
animal model (30).
We were able to detect IgE immunoreactivity to the
receptor by flow cytometry in patients with GD and TED.
Interest in this arm of the immune response has been
kindled by an animal model of TED induced by passive
transfer of TSHR-primed T cells, which one of us has
reported (16). In this model, mast cell infiltration was one
of the earliest indicators of orbital pathology. Develop-
ment of TSHR autoantibodies of the IgE subclass by iso-
type switching would provide a neat explanation for the
severe TED found in some, but not all, GD patients. Re-
cently, elevated IgE levels have been reported in GD pa-
tients, although whether the antibodies were directed to
the TSHR was not investigated (20), although IgE anti-
bodies to another major thyroid autoantigen, TPO, have
been reported (31). Further studies, using a larger series of
FIG. 4. IgA autoantibodies binding directly to the TSHR. Flow cytometry with fluorescence intensity shown on the x-axis and cell counts on
the y-axis. The bold trace was obtained from GPI cells, and the faint trace was obtained from JP02 receptor-negative control cells. A, GD with
TED GD1 (D 0.38); B, atypical TED P8 (D 0.35); C, normal 6 (D 0.72); D, normal 7 (D 0.15). A D value more than 0.35 is considered
positive.
Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 1759
patients, particularly GD patients before the onset of TED,
are warranted to resolve the issue of IgE antibodies to the
receptor.
In conclusion, we report neutral IgG antibodies, predom-
inantly of the
class, in the majority of patients with un-
treated GD and in a small proportion of middle-aged eu-
thyroid female controls. We have clearly demonstrated IgG
and/or IgA antibodies recognizing receptor conformation in
a high proportion of patients with TED who are negative for
TSHR autoantibodies with biological activity measured as
TSAB or TBII.
Acknowledgments
We are grateful to Drs. Costagliola, Banga, and Prabhakar for kindly
donating antibodies to the TSHR, and to Drs. Parkes and Lazarus for
providing patientssera.
Received September 25, 2001. Accepted January 8, 2002.
Address all correspondence and requests for reprints to: Dr. M.
Ludgate, Department of Medicine, University of Wales College of Med-
icine, Heath Park, Cardiff, United Kingdom CF14 4XN. E-mail:
ludgate@cf.ac.uk.
This work was supported in part by Brahms Diagnostica GmbH
(Berlin, Germany) and grants from the Wales Office of Research and
Development and the United Kingdom Medical Research Council.
References
1. Rees Smith B, Mclachlan S, Furmaniak J 1998 Autoantibodies to the thyro-
tropin receptor. Endocr Rev 9:106120
2. Weetman AP, McGregor AM 1994 Autoimmune thyroid disease; further
developments in our understanding. Endocr Rev 15:788830
3. Atger M, Misrahi M, Young J, Jolivet A, Orgiazzi J, Schaison G, Milgrom E
1999 Autoantibodies interacting with purified native thyrotropin receptor. Eur
J Biochem 265:10221031
4. Morgenthaler NG, Hodak K, Seissler J, Steinbrenner H, Pampel I, Gupta M,
McGregor AM, Scherbaum WA, Banga JP 1999 Direct binding of thyrotropin
receptor autoantibody to in vitro translated thyrotropin receptor: a comparison
to radioreceptor assay and thyroid stimulating bioassay. Thyroid 9:466475
5. Prentice L, Sanders JF, Perez M, Kato R, Sawicka J, Oda Y, Jaskolski D,
Furmaniak J, Smith BR 1997 Thyrotropin (TSH) receptor autoantibodies do
not appear to bind to the TSH receptor produced in an in vitro transcription/
translation system. J Clin Endocrinol Metab 82:12881292
6. Bobnikova Y, Graves PN, Vlase H, Davies TF 1997 Characterization of soluble
di-sulphide bond stabilized, procaryotically expressed human thyrotropin
receptor ectodomain. Endocrinology 138:588593
7. DaCosta CR, Johnstone AP 1998 Production of the thyrotropin receptor extra-
cellular domain as a glycosylphosphatidylinositol-anchored membrane pro-
tein and its interaction with thyrotropin and autoantibodies. J Biol Chem
273:1187411880
8. Costagliola S, Khoo D, Vassart G 1998 Production of bioactive amino-terminal
domain of the thyrotropin receptor via insertion in the plasma membrane by
a glycosylphosphatidylinositol anchor. FEBS Lett 436:427433
9. Jaume JC, Kakinuma A, Chazenbalk GD, Rapoport B, McLachlan SM 1997
Thyrotropin receptor autoantibodies in serum are present at much lower levels
than thyroid peroxidase autoantibodies: analysis by flow cytometry. J Clin
Endocrinol Metab 82:500507
10. Patibandla SA, Dallas JS, Seetharamaiah GS, Tahara K, Kohn LD, Prabhakar
BS 1997 Flow cytometric analysis of antibody binding to chinese hamster ovary
cells expressing human thyrotropin receptor. J Clin Endocrinol Metab 82:
18851893
11. Tonacchera M, Costagliola S, Cetani F, Ducobu J, Stordeur P, Vassart G,
Ludgate M 1996 Patient with monoclonal gammopathy, thyrotoxicosis,
pretibial myxedema and thyroid-associated ophthalmopathy: demonstration
of direct binding of autoantibodies to the thyrotropin receptor. Eur J Endo-
crinol 134:97103
12. Perros P, Kendall-Taylor P 1995 Thyroid-associated ophthalmopathy: patho-
genesis and clinical management. Bailliere Clin Endocrinol Metab 9:115135
13. Crisp M, Lane C, Halliwell M, Wynford-Thomas D, Ludgate M 1997 Thy-
rotropin receptor transcripts in human adipose tissue. J Clin Endocrinol Metab
82:20032005
14. Bahn R, Dutton C, Natt N, Joba W, Spitzweg C, Heufelder A 1998 Thyro-
tropin receptor expression in Gravesorbital adipose/connective tissues; po-
tential autoantigen in Gravesopthalmopathy. J Clin Endocrinol Metab 83:
9981002
15. Crisp MS, Starkey KJ, Lane C, Ham J, Ludgate M 2000 Adipogenesis in
thyroid eye disease. Invest Ophthalmopathy Vis Sci 41:32493255
16. Many MC, Costagliola S, Detrait M, Denef JF, Vassart G, Ludgate M 1999
Development of an animal model of autoimmune thyroid eye disease. J Im-
munol 162:49664974
17. Costagliola S, Many MC, Denef JF, Pohlenz J, Refetoff S, Vassart G 2000
Genetic immunization of outbred mice with thyrotropin receptor cDNA pro-
vides a model of Gravesdisease. J Clin Invest 105:803811
18. Arnold K, Metcalfe A, Weetman AP 1995 Immunoglobulin A class fibroblast
antibodies in pateints with Gravesdisease and pretibial myxoedema. J Clin
Endocrinol Metab 80:34303436
19. Khalil HA, Dekeizer RJW, Bodelier VMW, Kijlstra A 1989 Secretory IgA and
lysozyme in tears of patients with Gravesophthalmopathy. Doc Ophthalmol
72:329334
20. Sato A, Takemura Y, Yamada T, Ohtsuka H, Sakai H, Miyahara Y, Aizawa
T, Terao A, Onuma S, Junen K, Kanamori A, Nakamura Y, Tejima E, Ito Y,
Kamijo K 1999 Possible role of immunoglobulin E in patients with hyper-
thyroid Gravesdisease. J Clin Endocrinol Metab 84:36023605
21. Evans C, Morgenthaler N, Lee S, Llewellyn DH, Clifton-Bligh R, John R,
Lazarus JH, Chatterjee VK, Ludgate M 1999 Development of a luminescent
bioassay for thyroid stimulating antibodies. J Clin Endocrinol Metab 84:
374377
22. Watson PF, Ajjan RA, Phipps J, Metcalfe R, Weetman AP 1998 A new
chemiluminescent assay for the rapid detection of thyroid stimulating anti-
bodies in Gravesdisease. Clin Endocrinol (Oxf) 49:577581
23. Costagliola S, Swillens S, Niccoli P, Dumont J, Vassart G, Ludgate M 1992
Binding assay for thyrotropin receptor autoantibodies using the recombinant
receptor protein. J Clin Endocrinol Metab 75:15401544
FIG. 5. IgE autoantibodies binding directly to the TSHR. Flow cy-
tometry with fluorescence intensity shown on the x-axis and cell
counts on the y-axis. The bold trace was obtained from GPI cells, and
the faint trace was obtained from JP02 receptor-negative control cells.
A, normal 15 (D 0.29); B, GD with TED GD1 (D 0.56). A D value
more than 0.41 is considered positive.
1760 J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies
24. Nicholson L, Vlase H, Graves P, Nilsson M, Molne J, Huang GC, Mor-
genthaler NG, Davies TF, McGregor AM, Banga JP 1996 Monoclonal anti-
bodies to the human TSH receptor: epitope mapping and binding to the native
recptor on the baso-lateral plasma membrane of thyroid follicular cells. J Mol
Endocrinol 16:159170
25. Costagliola S, Rodien P, Many MC, Ludgate M, Vassart G 1998 Genetic
Immunization against the human thyrotropin receptor causes thyroiditis and
allows production of monoclonal antibodies recognizing the native receptor.
J Immunol 160:14581465
26. Young IT 1977 Proof without prejudice: use of the Kolmogarov-Smirnov test
for the analysis of histograms from flow systems and other sources. J Histo-
chem Cytochem 25:935941
27. Pappa A, Calder V, Ajjan R, Fells P, Ludgate M, Weetman AP, Lightman S
1997 Analysis of extraocular muscle-infiltrating T cells in thyroid-associated
ophthalmopathy. Clin Exp Immunol 109:362369
28. Molteni M, Zulian C, Scrofani S, Della Bella S, Bonara P, Kohn LD, Scorza
R1998 High frequency of T cell lines responsive to immunodominant epitopes
of thyrotropin receptor in healthy subjects. Thyroid 8:241247
29. Tandon N, Freeman M, Weetman AP 1992 T cell responses to synthetic TSH
receptor peptides in Gravesdisease. Clin Exp Immunol 89:468473
30. Wang SH, Carayanniotis G, Zhang Y, Gupta M, McGregor AM, Banga JP
1998 Induction of thyroiditis in mice with thyrotropin receptor lacking sero-
logically dominant regions. Clin Exp Immunol 113:119125
31. Guo J, Rapoport B, McLachlan SM 1997 Thyroperoxidase autoantibodies of
IgE class in thyroid autoimmunity. Clin Exp Immunol 82:157162
FIRST WORLD CONGRESS: HORMONAL AND GENETIC BASIS OF
SEXUAL DIFFERENTIATION DISORDERS
May 17–18, 2002
Tempe, Arizona
Participants: Maria I. New (Chairman), New York, NY; Jean Wilson (Chairman), Dallas, TX; Richard
Behringer, Houston, TX; Camerino Giovanna, Pavia, Italy; Cheryl Chase, San Francisco, CA; Patricia
Donahoe, Boston, MA; Maguelone Forest, Lyon, France; James Griffin, Dallas, TX; Melvin Grumbach, San
Francisco, CA; Nathalie Josso, Montrouge, France; Ursula Kuhnle-Krahl, Munich, Germany; Robin Lovell-
Badge, London, UK; Berenice Mendonca, Sao Paolo, Brazil; Heino Meyer-Bahlburg, New York, NY; Claude
Migeon, Baltimore, MD; Dix Poppas, New York, NY; Martin Ritzen, Stockholm, Sweden; Jacques Simard,
Quebec, Canada; Garry Warne, Victoria, Australia; Amy Wisniewsky, Baltimore, MD.
The Conference lectures will include: Genetics of Sex, Anti-Mu¨llerian Hormone Mutations, Animal Models
of Intersex, Normal Sexual Differentiation, Ambiguous Genitalia in Humans, Hormones and Sexual Be-
havior, Rationale for Gender Assignment, Legal Aspects of Gender Assignment, Ethical Issues in Gender
Assignment, Role of Advocacy Groups, Prenatal Therapy in Congenital Adrenal Hyperplasia, Surgical
Reconstruction in Patients with CAH and Ambiguous Genitalia, Medical Management at Puberty, Questions
Frequently Asked by Patients and Their Families, Support Groups for CAH and Androgen Insensitivity
Syndrome, Outcome Studies in CAH, Choosing Gender in the Indian Cultural Context, Partial Androgen
Insensitivity Syndrome and Partial Gonadal Dysgenesis, Complete AIS and Congenital Micropenis, Males
with 17
-Hydroxysteroid Dehydrogenase Deficiency, 5
-Reductase Deficiency, True Hermaphrodites.
For Congress registration and scientific information, please contact: Maria New, M.D., Professor and
Chairman, Department of Pediatrics New York Presbyterian Hospital, Weill Cornell Medical College, 525
E. 68th Street, New York, NY 10021. E-mail: minew@med.cornell.edu; Phone: (212) 746-3450.
This conference was previously scheduled to occur in Gubbio, Italy, on September 12, 2001, as Update on
Androgen Disorders,but was postponed due to the September 11 disaster.
Metcalfe et al. IgG, IgA, and IgE TSHR Autoantibodies J Clin Endocrinol Metab, April 2002, 87(4):1754 1761 1761
... However, some people with TED have hypothyroidism whilst a small proportion are euthyroid [11]. Of interest, we have demonstrated the presence of TSHR autoantibodies even in euthyroid TED, although they lack biological activity when measured using conventional assays [12]. ...
... However, treatment of an unusual thyroid cancer patient who also had GD and TED, with a human monoclonal thyroid blocking antibody (TBAB) improved these disorders. The results are also compatible with my view that we do not need any additional autoantigens-but maybe we have yet to identify TED-specific TSHR autoantibodies [12]. ...
Article
Dysthyroid eye disease is a rare condition, mainly found in people with Graves’ hyperthyroidism. Autoimmune responses to thyroid/orbit shared antigens drive extensive tissue remodelling. This includes excess adipogenesis and over-production of extra-cellular matrix, which both tend to occur in the earlier ‘active’ inflammatory stages of disease. With time these give way to fibrosis, which has a profound impact on eye motility and may be life-long. Progress has been made in identifying the shared autoantigen(s) and the role of specific T cells and autoantibodies in remodelling, which have facilitated development of novel therapies. However relatively little is known of the autoimmune processes under-pinning fibrosis and currently there are no adequate medical treatments.
... The narrow sense of neutral TRAb is against the hinge region, an inert scaffold connecting the leucine-rich ectodomain that binds to TSH, to the transmembrane region of the receptor, and it does not basically affect TSH binding to the TSH-R [14][15][16]. It seems that some patients with GD have these type of autoantibodies that bind to the hinge region of TSH-R [17][18][19][20]. The neutral antibodies, in a narrow sense, do not interfere with TSH-binding to the TSH-R and, therefore, they should be negative for TBII. ...
Article
Full-text available
Context The thyrotropin (TSH) receptor (TSH-R) autoantibody activity is clinically measured by inhibition of labeled ligand (TSH or M22) binding to the TSH-R [TSH-binding inhibitory immunoglobulin (TBII)] or by stimulation [TSH-R stimulating antibody (TSAb)] or inhibition [TSH-R blocking antibody (TSBAb)] of cAMP production in isolated cells. Objective We experienced a patient with hypothyroid Graves’ disease (GD) having strong positive TBII, but with almost neutral bioactivities on the TSH-R. The aim of this study is characterization of this apparently paradoxical TBII (sera S). Design We first compared the TBII, TSAb and TSBAb activities of sera S with mixtures of stimulating (S-mAb) and blocking monoclonal Ab (B-mAb). Next, we serially measured cAMPs stimulated by various sera in the presence or absence of TSH. Results Mixtures of S-mAb and B-mAb did not reproduce the characteristics of sera S. Instead, the sera S had a unique feature which blocked the TSH-stimulated cAMP initially but disappeared the blocking activity thereafter to reach the control level. Conclusions We present here the TBIIs with neutral bioactivities found in the patient with autoimmune thyroid disease, which strongly inhibit TSH-binding to the TSH-R but exerts neither TSAb nor TSBAb activity. Differences in the methods to detect TRAb between TBII in vitro and bioassay may cause the discrepancy. Although the sera S may be an extreme example, a variety of TRAb, not only stimulating or blocking but also interfering with TSH-R binding only for a short time, may exist in the sera of GD patients.
... TSHR expression has been shown to increase during adipogenesis (5). We demonstrated that 'neutral' TSHR antibodies were capable of binding but had no effect on traditional TSHR signaling pathways (described below) (29). Indeed, TSHR signaling may be far more complex than initially thought (30). ...
Article
Full-text available
Graves’ orbitopathy (GO) is a complex and poorly understood disease in which extensive remodeling of orbital tissue is dominated by adipogenesis and hyaluronan production. The resulting proptosis is disfiguring and underpins the majority of GO signs and symptoms. While there is strong evidence for the thyrotropin receptor (TSHR) being a thyroid/orbit shared autoantigen, the insulin-like growth factor 1 receptor (IGF1R) is also likely to play a key role in the disease. The pathogenesis of GO has been investigated extensively in the last decade with further understanding of some aspects of the disease. This is mainly derived by using in vitro and ex vivo analysis of the orbital tissues. Here, we have summarized the features of GO pathogenesis involving target autoantigens and their signaling pathways.
... Although several authors have reported correlations between TSHRAb titre and the prevalence and/or severity Graves orbitopathy [125][126][127] , the fact that not all patients with Graves orbitopathy have TSHRAbs with stimulating activity on the TSHR (TSHRAbs activate the PKA-cAMP cascade) has led to two differing conclusions. The first is that TSHRAb sig nals might exist that signal to other potential cascades and, second, that there is an additional autoantigen 111,128 . ...
Article
Graves orbitopathy, also known as thyroid eye disease or thyroid-associated orbitopathy, is visually disabling, cosmetically disfiguring and has a substantial negative impact on a patient’s quality of life. There is increasing awareness of the need for early diagnosis and rapid specialist input from endocrinologists and ophthalmologists. Glucocorticoids are the mainstay of treatment; however, recurrence occurs frequently once these are withdrawn. Furthermore, in >60% of cases, normal orbital anatomy is not restored, and skilled rehabilitative surgery is required. Clinical trials have shown that considerable benefit can be derived from the addition of antiproliferative agents (such as mycophenolate or azathioprine) in preventing deterioration after steroid cessation. In addition, targeted biologic therapies have shown promise, including teprotumumab, which reduces proptosis, rituximab (anti-CD20), which reduces inflammation, and tocilizumab, which potentially benefits both of these parameters. Other strategies such as orbital radiotherapy have had their widespread role in combination therapy called into question. The pathophysiology of Graves orbitopathy has also been revised with identification of new potential therapeutic targets. In this Review we provide an up-to-date overview of the field, outline the optimal management of Graves orbitopathy and summarize the research developments in this area to highlight future research questions and direct future clinical trials. Graves orbitopathy has a negative impact on a patient’s quality of life. This Review provides an overview of the field and outlines the optimal management of Graves orbitopathy. The authors also highlight future research questions to direct future clinical trials.
... A study has shown that the TgAb and TPOAb can mediate antibody-dependent cell-mediated cytotoxicity (11). Xie et al. concluded that the humoral response was important in pathogenesis of HT (12). ...
Article
Full-text available
Background: Hashimoto's thyroiditis (HT) is a common cause of thyroid diseases in children and the role of vitamin D (VD) is controversial. Therefore, the aim of this study was to investigate the influence of VD therapy on HT in children with hypovitaminosis D. Materials and Methods: This randomized clinical trial study was conducted on 30 patients referred to Endocrine Clinic of Amirkola Children's Hospital (ACH) of Babol in Iran. The serum levels of calcium, T4, TSH, Anti thyroid peroxidase antibody (TPOAb) and Anti thyroglobulin antibody (TgAb) were checked, and ultimately the HT was diagnosed based on thyroid sonography and these findings. According to normal range of calcium>8.4 mg/dl and low level of VD, the patients were divided into deficient
... Neither TSHR nor IGF1R ticks all four boxes; IGF1R is still awaiting (b) and (c), whilst TSHR does not yet fulfil (d); although treatment with a human monoclonal thyroid-blocking antibodyimproved GD and TED in an unusual patient with these disorders in addition to thyroid cancer (ETA abstract). These results are also compatible with my view that we do not need any additional autoantigens-but may be we have yet to identify TED-specific TSHR autoantibodies [35]. ...
Article
I was privileged to be one of the co-chairs, along with Professor Tim Sullivan (Brisbane, Australia), for the Cambridge Ophthalmological Society (COS) annual international symposium, which, this year, was dedicated to thyroid eye disease (TED). Together with the organisers, Miss Rachna Murthy and Professor Keith Martin from COS, we compiled an impressive programme covering all aspects of the condition from events happening in a single orbital cell to improved surgical approaches.
... However, the fact that not all GO patients have TSAB and may even be euthyroid or hypothyroid has prompted people to seek other explanations. Work from our group [75] demonstrated 'neutral' TSHR antibodies able to bind but having no effect on known TSHR signaling. Indeed, TSHR signaling may be far more complex than initially thought (reviewed by Latif et al. [76]). ...
Article
Introduction: Thyroid eye disease or Graves’ orbitopathy (GO) is an autoimmune condition most common in people with hyperthyroid Graves’ disease. Severe GO may cause blindness and all patients have reduced quality of life and effective treatments are lacking. This has driven efforts to identify novel therapeutic options resulting in progress in our understanding of the tissue remodeling processes underpinning GO.Areas covered: We have described clinical aspects including signs and symptoms, risk factors and current treatments. We have summarized GO pathogenesis including target autoantigens, regulation of tissue remodeling and tolerance mechanisms. Finally we reflect on the possible role of the microbiome in autoimmunity. References cited were selected from pubmed using the search terms ‘Graves’ orbitopathy’ ‘thyroid eye disease.’Expert commentary: Currently the results of two clinical trials, using completely novel treatments for thyroid eye disease (TED)/GO, are awaited. The trials are the result of progress made in understanding some aspects of the pathogenesis of TED/GO (end stages of disease) mainly derived using in vitro models. However, we remain largely ignorant of the triggers that initiate and maintain the autoimmune response; studies using in vivo models are likely to shed light on the mechanisms leading to loss of tolerance.
Article
Full-text available
Emerging data and innovative technologies are re-shaping our understanding of the scope and specificity of the autoimmune response in Pemphigus vulgaris (PV), a prototypical humorally mediated autoimmune skin blistering disorder. Seminal studies identified the desmosomal proteins Desmoglein 3 and 1 (Dsg3 and Dsg1), cadherin family proteins which function to maintain cell adhesion, as the primary targets of pathogenic autoAbs. Consequently, pathogenesis in PV has primarily considered to be the result of anti-Dsg autoAbs alone. However, accumulating data suggesting that anti-Dsg autoAbs by themselves cannot adequately explain the loss of cell-cell adhesion seen in PV, nor account for the disease heterogeneity exhibited across PV patients has spurred the notion that additional autoAb specificities may contribute to disease. To investigate the role of non-Dsg autoAbs in PV, an increasing number of studies have attempted to characterize additional targets of PV autoAbs. The recent advent of protein microarray technology, which allows for the rapid, highly sensitive, and multiplexed assessment of autoAb specificity has facilitated the comprehensive classification of the scope and specificity of the autoAb response in PV. Such detailed deconstruction of the autoimmune response in PV, beyond simply tracking anti-Dsg autoAbs, has provided invaluable new insights concerning disease mechanisms and enhanced disease classification which could directly translate into superior tools for prognostics and clinical management, as well as the development of novel, disease specific treatments.
Article
Full-text available
Autoantibodies are frequently observed in healthy individuals. In a minority of these individuals, they lead to manifestation of autoimmune diseases, such as rheumatoid arthritis or Graves’ disease. Overall, more than 2.5% of the population is affected by autoantibody-driven autoimmune disease. Pathways leading to autoantibody-induced pathology greatly differ among different diseases, and autoantibodies directed against the same antigen, depending on the targeted epitope, can have diverse effects. To foster knowledge in autoantibody-induced pathology and to encourage development of urgently needed novel therapeutic strategies, we here categorized autoantibodies according to their effects. According to our algorithm, autoantibodies can be classified into the following categories: (1) mimic receptor stimulation, (2) blocking of neural transmission, (3) induction of altered signaling, triggering uncontrolled (4) microthrombosis, (5) cell lysis, (6) neutrophil activation, and (7) induction of inflammation. These mechanisms in relation to disease, as well as principles of autoantibody generation and detection, are reviewed herein.
Article
Full-text available
In this paper the Kolmogorov-Smirnov statistical test for the analysis of histograms is presented. The test is discussed for both the two-sample case (comparing fn1(X) to fn2 (X)) and the one-sample case (comparing fn1 (X) to f(X)). Presentation of the specific algorithmic steps involved is done through development of an example where the data are from an experiment discussed elsewhere in this issue. It is shown that the two histograms examined come from two different parent populations at the 99.9% confidence level.
Article
Full-text available
Twenty-eight peptides, representing the entire extracellular domain of the TSH receptor, were synthesised to investigate which parts of this autoantigen may be targets for the T cell response in Graves' disease (GD). T cells from 11 of 21 controls and 26 of 36 newly diagnosed GD patients proliferated in response to one or more peptides with a stimulation index (SI) of greater than 2.0 (chi 2 = 2.31, P greater than 0.1). The response of patients and controls to any of the individual peptides was also not statistically different. However, individual patients gave high SIs with certain peptides to which controls either gave an absent or very weak response. HLA-DR3 was not associated with any particular response to TSHR peptides. Three out of seven GD patients whose T cells were evaluated before and after treatment showed a response of this kind only early in the course of their disease. Intrathyroidal T cells from four GD patients did not give a consistent proliferative response to pools of five peptides, and depleting peripheral blood T cells of their CD8+ population did not affect the proliferative response. These results indicate that the T cell response to the TSH receptor in GD does not seem to be directed against any one particular epitope on the peptides we have tested which cover the extracellular domain.
Article
We have characterized a transfected Chinese hamster ovary cell line, JP09, which expresses high levels of the human TSH receptor (TSH-R). Based on a theoretical biological activity for TSH of 40 IU/mg, JP09 has approximately 90,000 receptors per cell, having a dissociation constant of 1.64 x 10(3) mU/L or 1.47 x 10(-9) mol/L. We have used JP09 to prepare solubilized TSH-Rs which have formed the basis of a binding assay for thyroid-binding inhibiting immunoglobulins in unfractionated sera. We have compared the JP09 assay with the TRAK assay (which is based on solubilized porcine TSH-R) and found a highly positive correlation between the two assays, r = 0.83 P < 0.0001, in 55 sera from patients with autoimmune thyroid disease. JP09 can be adapted to growth in suspension culture, permitting large scale production. The tracer in the assay is bovine [125I]TSH; surprisingly, despite the use of a hTSH-R, hTSH had no effect on the binding of the tracer up to 10(3) mU/L and only a minor effect at 10(4) mU/L.
Article
The involvement of autoantibodies in the extrathyroidal manifestations of Graves' disease has been the subject of extensive investigation, with fairly inconclusive results to date. We investigated the presence of immunoglobulin A (IgA) and IgG antibodies in patients with Graves' disease and pretibial myxedema (PTM; n = 21) as well as those with Graves' disease with thyroid-associated ophthalmopathy (TAO; n = 10), Graves' disease with no clinical evidence of extrathyroidal manifestations (n = 11), Hashimoto's thyroiditis (n = 9), type 1 diabetes mellitus (n = 10), systemic lupus erythematosus (n = 9) and normal individuals (n = 17). We looked for antibodies to both retroocular muscle and dermal fibroblasts as well as to thyroid peroxidase, thyroid microsomal antigen, thyroglobulin, and human eye muscle membranes. IgA class antibodies to microsomal antigen (30-50% of patients), thyroid peroxidase (5-20%), and human eye muscle membrane (0-26%) antigens were found in the various groups of patients with Graves...
Article
In this study we analyzed the proliferative response to the extracellular domain of thyrotropin receptor (TSHR-ECD) of T-cell lines raised from healthy subjects. We found high frequencies of cell lines reactive to TSHR-ECD, ranging from 12% to 37%. The response of the cell lines to a set of overlapping peptides of TSHR-ECD showed that the most recognized epitopes by T lymphocytes are on the C-terminal portion. In particular, the regions of residues 360-396 and 258-277 are immunodominant in T-lymphocyte reactivity. A group of cell lines specific for the peptides of TSHR-ECD lost the response to the peptides during time in culture. However, these lines were still responsive to TSHR extracellular domain. The cloning of one of these lines showed three types of T-cell clones: (1) CD4+ clones (n = 4) highly responsive to the TSHR-ECD; (2) CD4+ clones (n = 4) low responsive to TSHR-ECD; (3) CD8+ clones (n = 9) not responsive to TSHR-ECD. The first group of clones was stable during time in culture, while the second group was characterized by the loss of the specific response to TSHR-ECD after some weeks from the first analysis. The observation of a spontaneous anergy in the second group of CD4+ clones suggests that mechanisms of control of the lymphocyte response to TSHR-ECD could be activated in vitro.
Article
It is acknowledged that the TSH receptor (TSHr) on thyroid follicular cells is the autoantigen involved in the hyperthyroidism of Graves' disease. However, whether this receptor is expressed in extrathyroidal tissues, and whether it participates directly in the pathogenesis of Graves' ophthalmopathy (GO) are unclear. We sought to detect the expression of TSHr messenger ribonucleic acid (mRNA) and protein in orbital adipose/connective tissue specimens and in human orbital preadipocyte fibroblast cultures using liquid hybridization analysis and immunohistochemical methods. We demonstrated intact and variant TSHr mRNA transcripts and TSHr-like immunoreactivity in orbital adipose/connective tissue specimens from patients with GO. In addition, TSHr-like immunoreactivity was detected in early passage GO preadipocyte fibroblast cultures that were shown to include some adipose cells. In contrast, neither TSHr mRNA nor protein was detected in normal orbital adipose/connective tissue specimens or in late passage GO orbital fibroblast cultures containing no lipid-laden adipose cells. In conclusion, we showed that TSHr is expressed in the adipose/connective tissue of the diseased orbit in GO. In addition, TSHr is demonstrable in early passage GO preadipocyte orbital fibroblast cultures that contain a subpopulation of adipocytes. Subsequent passaging of these cells results in the loss of both TSHr expression and adipocyte-specific staining. These results suggest that both the expression of this receptor and the accumulation of adipose tissue in the orbit in GO may be induced in vivo by a humoral factor(s) not present in the cell culture environment.
Article
We have characterized a transfected Chinese hamster ovary cell line, JP09, which expresses high levels of the human TSH receptor (TSH-R). Based on a theoretical biological activity for TSH of 40 IU/mg, JP09 has approximately 90,000 receptors per cell, having a dissociation constant of 1.64 x 10(3) mU/L or 1.47 x 10(-9) mol/L. We have used JP09 to prepare solubilized TSH-Rs which have formed the basis of a binding assay for thyroid-binding inhibiting immunoglobulins in unfractionated sera. We have compared the JP09 assay with the TRAK assay (which is based on solubilized porcine TSH-R) and found a highly positive correlation between the two assays, r = 0.83 P < 0.0001, in 55 sera from patients with autoimmune thyroid disease. JP09 can be adapted to growth in suspension culture, permitting large scale production. The tracer in the assay is bovine [125I]TSH; surprisingly, despite the use of a hTSH-R, hTSH had no effect on the binding of the tracer up to 10(3) mU/L and only a minor effect at 10(4) mU/L.
Article
Using an enzyme linked immuno-assay (ELISA) and spectrophotometry, we determined levels of secretory IgA and lysozyme in tears of 69 patients with Graves' ophthalmopathy and 28 controls. The quantitative determination of secretory IgA and lysozyme in tears provided an impression of the functioning of the lacrimal gland in the two groups. An IgA/lysozyme ratio was calculated in both patients and controls as a parameter for the activity of the secretory IgA-producing plasma cells in the lacrimal gland. An increase in the IgA/lysozyme ratio was observed in 23 patients (33%) and one control (3%). Half of the patients who had suffered from the disease for more than 5 years showed a raised IgA/lysozyme ratio. No correlation was found between the IgA/lysozyme ratio and the NOSPECS classification. Our findings suggest that the lacrimal gland is involved in the orbital condition produced by Graves' ophthalmopathy. In most cases the involvement occurs in patients with a long history of the disease.
Article
This review considers recent developments in our understanding of the properties of TRAb, particularly measurement of the antibodies and their sites of action and synthesis. Two new assay methods have allowed considerable improvements in the sensitivity, specificity, precision, and ease of measuring TRAb. In particular: 1) receptor assays based on inhibition of receptor-purified labeled TSH binding to detergent-solubilized TSH receptors and 2) bioassays based on stimulation of cAMP release from monolayer cultures of isolated thyroid cells. Detailed studies with the two assays indicate that TSH receptor antibodies nearly always act as TSH agonists in patients with a history of Graves' hyperthyroidism. Studies in areas of dietary iodine sufficiency suggest that measurement of the antibodies at various stages in the course of treating Graves' disease can be of value in predicting the outcome of therapy. However, in areas of iodine deficiency, difficulties in the ability of patients' thyroid tissue to recover from the effects of antithyroid drugs may prevent the receptor antibodies from causing a relapse of thyrotoxicosis. Consequently, the predictive value of receptor antibody measurements would be expected to be lower in these geographical areas. Although patients with a history of Graves' hyperthyroidism nearly always have TRAb which act as TSH agonists, about 20% of patients with frank hypothyroidism due to autoimmune destruction of the thyroid have TRAb which act as TSH antagonists (blocking antibodies). There is some evidence that these blocking antibodies can cause hypothyroidism particularly in the neonate. With regard to the site of synthesis of TRAb, there is now direct evidence that they are synthesized by thyroid lymphocytes, particularly the lymphocytes in close proximity to thyroid follicular cells. This is consistent with the well established effects of antithyroid treatment (drugs, radioiodine, or surgery) on TRAb levels in addition to their effects on thyroid hormone synthesis. Recent studies using affinity labeling with 125I-labeled TSH have enabled elucidation of the structure of the TSH receptor. TSH receptors in human, porcine, and guinea pig thyroid tissue have a two-chain structure in which the TSH binding site is formed on the outside surface of the cell membrane by a water-soluble A subunit (Mr approximately 50 K). The A subunit is linked by a disulfide bridge and weak noncovalent bonds to the amphiphilic B subunit (Mr approximately 30 K). This subunit, which penetrates the lipid bilayer, probably forms the site for interaction of the receptor with the regulatory subunits of adenylate cyclase.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
The revolution in molecular techniques has allowed dissection of the autoimmune response in a way impossible to imagine 10 yr ago. There have been spectacular advances in our understanding of self-tolerance mechanisms and how these may fail, combined with a detailed comprehension of antigen presentation, functional T cell subsets, and TCR utilization in autoimmunity, albeit usually in animal models that resemble, but do not exactly duplicate, human diseases. More gradually, these findings are being translated to thyroid autoimmunity, where the major achievement of the last decade has been the molecular characterization of the three main thyroid autoantigens. This in turn has allowed epitope identification, although again the only clear data so far have come from animal models of EAT. Another advance has been the recognition that the thyrocyte is not a helpless target of autoaggression, being capable of expressing a wide array of immunologically active molecules, which may exacerbate or diminish the autoimmune response. In 1983, there was considerable excitement at the discovery of the first of these phenomena, namely MHC class II expression, but its possible role in autoantigen presentation remains to be defined. By analogy with pancreatic beta-cells, and based on our own data, we believe that class II-expressing thyrocytes have little, if any, such role and suspect that instead this may be a mechanism for inducing peripheral tolerance. Defining the contribution of thyrocytes to the intrathyroidal autoimmune response, whether from released cytokines or surface-bound molecules, will be crucial to our future understanding, as well as holding the promise that these thyroid-derived products might be therapeutic targets. Despite molecular developments in HLA analysis, there have been no really major improvements in our understanding of the immunogenetics of thyroid autoimmunity, equivalent to those made in type 1 diabetes mellitus. The available data suggest strongly that non-MHC genes play an important role in susceptibility, and novel approaches will be required to identify these. On the other hand, we know more about the importance of environmental and endogenous (most probably hormonal) factors in thyroid autoimmunity. Understanding the basic immunological changes in the postpartum period is still poor, however, as most studies to date have concentrated on epidemiology and clinical delineation. As PPTD undergoes spontaneous remission, elucidation of these mechanisms has clear implications for treatment.(ABSTRACT TRUNCATED AT 400 WORDS)