Sunteți pe pagina 1din 10

Original Paper

HORMONE Horm Res 2009;71:350–358 Received: January 11, 2008


RESEARCH DOI: 10.1159/000223420 Accepted: August 1, 2008
Published online: June 9, 2009

Interaction of Lymphocytes and Thyrocytes


in Graves’ Disease and Nonautoimmune
Thyroid Diseases in Immunohistochemical and
Ultrastructural Investigations
Iwona Ben-Skowronek a Jadwiga Sierocinska-Sawa b Leszek Szewczyk a
Elzbieta Korobowicz b
Departments of a Paediatric Endocrinology and Neurology, and b Pathomorphology, Lublin Medical University,
Lublin, Poland

Key Words B between thyrocytes results in the thickening of the basal


Lymphocyte subsets ⴢ Graves’ disease ⴢ Nodular goiter ⴢ membrane of the thyroid follicle. No cytotoxic effect of T cy-
Simple goiter totoxic/suppressor CD8+ cells on thyrocytes was observed
in Graves’ disease, while a mild cytotoxic effect was observed
in non-autoimmune thyroid disease.
Abstract Copyright © 2009 S. Karger AG, Basel
Background: Graves’ disease is the archetype for organ-spe-
cific autoimmune disorders. It is very important for our un-
derstanding of the mechanisms responsible for progression Introduction
of autoimmunity. The aim of this study was to present inter-
actions of lymphocytes and thyrocytes in the thyroid tissue Autoimmune diseases in children occur much less fre-
in Graves’ disease and nonautoimmune thyroid diseases. quently than in adults, and most often they are not ac-
Methods: The study involved 30 children with Graves’ dis- companied by any other illnesses. The syndromes com-
ease, 30 children with nodular goiter, 30 with simple goiter prising autoimmune thyroid disease (AITD) are the three
and 30 healthy children. After thyroidectomy, T cells were intimately related illnesses: Graves’ disease with goiter,
detected in the thyroid specimens by CD3, CD4, CD8 anti- hyperthyroidism and, in many patients, associated oph-
bodies, B cells by CD79␣ antibodies and the antigen-pre- thalmopathy, Hashimoto thyroiditis with goiter and eu-
senting dendritic cells with CD1a antibodies (DakoCyto- thyroidism or hypothyroidism. The syndromes are bound
mation) and were examined in the EM 900 Zeiss Germany together by their similar thyroid pathology, similar im-
Electron Microscope. Results: The most enhanced immune mune mechanism, co-occurrence in family groups, and
reaction was observed in the thyroid from children with transition from one clinical picture to another within the
Graves’ disease. The cells of the immune system infiltrated same individual over time [1].
the thyroid follicles and interfollicular compartments; they The immune response proceeds via T-cell receptor
also formed lymph follicles. Conclusion: The immune reac- antigen recognition, followed by activation of the T cell
tion in Graves’ disease and migration of lymphocytes T and through the combined effect of antigen recognition and

© 2009 S. Karger AG, Basel Iwona Ben-Skowronek


0301–0163/09/0716–0350$26.00/0 Department of Paediatric Endocrinology and Neurology
Fax +41 61 306 12 34 Lublin Medical University
E-Mail karger@karger.ch Accessible online at: PL–20-093 Lublin DSK, ul. Chodzki 2 (Poland)
www.karger.com www.karger.com/hre Tel. +48 81 718 5440, Fax +48 81 718 4373, E-Mail skowroneki@interia.pl
Table 1. Description of patients

Diagnosis Children TSH, mU/l fT4, ng/dl TPO Ab, U/l TG Ab, U/l TRAB, U/l

Normal ranges 0.27–4.2 0.8–2.4 <12 <35 <1.1


Control group 30 0.27–4.2 – – – –
Struma colloides 30 0.21–3.9 0.8–2.3 0–14 0–36 –
Struma nodosa 30 0.2–4.1 0.8–2.1 0–16 0–40 –
Graves’ disease 30 0.007–0.001 3.3–5.1 21–6,663 25–13,351 7–462

costimulatory signals, including interleukin (IL)-1 action Numerous investigations evaluated lymphocyte sub-
leading to T cell IL-2 secretion and IL-2 receptor expres- sets in the peripheral blood [9–15], but the reactions in
sion and, subsequently, to proliferation of the T cell into the thyroid tissue are very important in a situation when
an active clone [2]. The antigen can be presented to CD4+ new experimental methods of treatment by B-lympho-
cells by conventional antigen-presenting cells, particu- cyte depletion during monoclonal anti-CD20 antibody
larly dendritic cells (DCs) [3] and also by B cells and ac- therapy are elaborated [11, 12].
tivated T cells, and less effectively by a variety of other The aim of the study was to present interactions of
cells (thyrocytes, fibroblasts) [4]. Antigen presentation to lymphocytes and thyrocytes in the thyroid tissue in
T cells leads to a variety of responses, which include pro- Graves’ disease and nonAITDs in children.
liferative or suppressive functions, development of cell
cytotoxic responses, control of immunoglobulin secre-
tion, and many more. Responding lymphocytes can be Materials and Methods
segregated into groups based on whether they are naive
or memory cells, CD4+ helper cells, or CD8+ cytotoxic/ Patients
suppressor cells. CD8+ T cell activation requires addi- The study concerned 90 children: 30 children aged 10–19
tional costimulatory pathways to the B7-dependent path- (mean 16 8 2.3) years, affected with Graves’ disease, 30 children
with nodular goiter, 30 children with simple goiter. The children
way largely used by CD4+ T cells, and ICAM-1 is par- were treated in the Department of Pediatric Endocrinology and
ticularly important [5, 6]. The CD4+ T cells are catego- Neurology in Lublin and in the Pediatric Department in Rzeszow
rized into Th1 and Th2 cells. Th1 cells produce IL-2, INF in the years 1994–2007 (table 1).
and TNF and are predominant in ‘delayed hypersensitiv- The TSH, fT4 and TT3 hormones were assayed by MEIA (Ab-
ity’ type of reactions, whereas Th2 cells produce IL-4, IL- bott Kit). The levels of TSH receptor antibodies were measured by
RIA (TRAK assay, BRAHMS Diagnostica GmbH, Berlin, Ger-
5, stimulate B cells to immunoglobulin synthesis, and are many). TPO and TG antibodies were assayed by LIA (Lumitest,
involved especially in antibody-mediated reactions. Cy- BRAHMS Diagnostica GmbH).
tokines produced by Th1 cells enhance the activity of In the patients with Graves’ disease, symptoms of thyrotoxi-
these subsets but inhibit Th2 cells and vice versa [2]. Some cosis and an increase in fT4 (mean 3.8 8 0.7 ng/dl) and TT3
CD4+ and CD8+ T cells appear to provide suppressor (mean 363 8 175.3 ng/dl) were observed, and also a decrease in
TSH (mean 0.004 8 0.003 mU/l). The levels of antibodies against
signals. Most recently, attention has been focused on al- TSH receptor (TRAB; 7–462 U/ml) and usually the levels of TPO
ternates of local secretion TGF-␤ and direct cell contact, antibodies (21–6,663 U/ml) and TG antibodies (25–13,351 U/ml)
which involves binding of CTLA-4 to the T regulatory were increased.
cell [7]. In addition to the standard T cell function de- The patients were treated with methimazole at initial doses of
scribed above, other cells participate in immune respons- 0.9–0.5 mg/kg body weight/day during 4–6 weeks, and after that
time, when in euthyreosis, they received maintenance doses of
es. Macrophages may destroy cells having immune com- approx. 0.1 mg/kg body weight/day (mainly 5 mg/day) in combi-
plexes on their surface. Other cells which do not bear the nation with a low dose of L-thyroxin (25 ␮g/day) during 18–24
CD3 marker of T cell lineage exist (K and NK cells) and months. Children with Graves’ disease in whom early relapses of
have the ability to spontaneously kill other cells (espe- hyperthyreosis were observed and led to surgery treatment (thy-
cially those expressing HLA antigens). NK cells can be roidectomy) after 18–36 months were included in the investiga-
tion.
detected by specific monoclonal antibodies such as anti- The patients with nodular goiter and simple goiter were euthy-
CD16 and are recognized phenotypically as large gran- roid or hypothyroid at the beginning of the treatment. They were
ular lymphocytes (LGLs) [8]. treated with L-thyroxin at a dose of 25–100 ␮g/day.

Lymphocytes and Thyrocytes in GD and Horm Res 2009;71:350–358 351


Non-AITD
Table 2. Subsets of lymphocytes as percentage of cells in vision fields in thyroid tissue

Group Subsets of lymphocytes


CD3+ CD4+ CD8+ CD79␣+ CD1a+
T p T helper p T suppressor/ p B p dendritic p
cytotoxic cells

Control group 1.0280.01 0.1980.28 0.6580.001 4.0780.67 0.1683.62


Simple goiter 6.3680.45 0.063 1.0282.71* 0.049 4.3885.54* 0.038 0.2880.67 0.071 0.2880.69 0.084
Nodular goiter 14.982.82* 0.032 2.185.5 0.051 985.5* 0.034 5.9811.6* 0.030 0.180.3 0.066
Graves’ disease 17.73817.9* 0.003 2.9582.51* 0.034 6.4986.93* 0.035 23.21823.6* 0.001 0.3780.3* 0.047

* p < 0.05 vs. control.

Specimens from thyroids of children who had died in acci- Ultrastructural Investigations
dents or who had been operated due to thyroglossal cysts, neck Specimens for ultrastructural investigations were obtained
injuries and during surgery of parathyroid glands were investi- during thyroidectomy or from paraffin-dewaxed preparations.
gated as controls. Small segments of the thyroid were cut into 0.5-mm3 pieces and
fixed in 4% glutaraldehyde in 0.1 M cacodylate buffer, pH 7.4 for
Immunohistochemical Investigations 24 h at 4 ° C, postfixed in 2% OsO4 in the same buffer for 1 h at
After thyroidectomy, 4-␮m thyroid slices were histologically room temperature, dehydrated in the graded series (up to 100%)
examined with hematoxylin-eosin. Immunohistochemical reac- of ethanol and embedded in 812 Epon. They were then polymer-
tions in paraffin specimens with monoclonal antibodies against ized at 60 ° C. Epon blocks were cut with ultramicrotome RMC
T-cell markers CD3, CD4, CD8 as well as against B-cells – MT-7 (RMC, Tucson, Ariz., USA). Ultrathin sections were con-
CD79- ␣ and the antigen presenting DCs – CD1a antibodies (Da- trasted with uranyl acetate and lead citrate and examined with the
koCytomation Denmark) were performed. The specimens were EM 900 Zeiss Germany Electron Microscope.
dewaxed in xylene, hydrated by rinsing with numerous alcohols The investigation was approved by the local Ethical Commit-
and distilled water. Next, antigens were revealed in citrate buffer tee of the Medical University in Lublin.
at pH 6.0. After the specimens had been washed in distilled water
and TBS buffer, endogenous peroxidase was blocked with 3%
H2O2, which was followed by a subsequent wash of the specimens
in TBS buffer. Thus prepared thyroid specimens were incubated Results
with a primary antibody and then with dextran marked with
horseradish peroxidase (DakoCytomation EnVision + Peroxidase In the thyroid specimens of the control group, lym-
kit) and with the chromogene of DAB horseradish. When the im-
munohistochemical reaction was finished, cell nuclei were stained phatic cells constituted an insignificant percentage. They
with Mayer’s hematoxylin; the specimens were dehydrated in al- were dispersed among the thyroid follicles and were often
cohol, exposed to xylene and embedded in Canada balsam. present in the proximity of blood vessels. The most prev-
The specimens were estimated in Axiostar plus microscope. alent were CD79␣+ B lymphocytes, their contents being
The lymphocytes were counted in Sony Color Camera Exwave- approximately 4.07%. T lymphocytes with a CD3+ phe-
HAD and the lymphocyte subsets were analyzed in MultiScan5
software and hardware with Show Time Plus, S-VHS frame grab- notype comprised 1.02% of the cells in the thyroid speci-
ber using an IBM Pentium computer. We determined the number mens. CD8+ suppressor/cytotoxic T lymphocytes were
of lymphocyte subpopulations in the thyroid tissue by counting more frequently observed (0.65%) than T helper cells
lymphocytes marked with CD3+, CD4+, CD8+, CD79␣+ and (0.19%). The least frequently occurring were the CD1a+
CD1a+ monoclonal antibodies in every 1,000 cells present in 10 antigen-presenting cells (0.16%; table 2). In ultrastructur-
vision fields of the microscope and by estimating their content
percentage. al investigations, we most often observed T lymphocytes
The results were expressed as means 8 SD. Comparison of the (with poor cytoplasm and the nucleus filling the cell) in
percentage of lymphocytes expressing receptors to monoclonal the basal membrane of thyroid follicles. The basal mem-
antibodies was carried out using the t test confirmed by the Mann- brane itself was thin. The thyrocytes had a follicular cell
Whitney U test. T value was considered statistically significant at nucleus. The mitochondria were concentrated in the bas-
p ! 0.05. For correlation analysis, Pearson’s and Spearman’s tests
were used. Mathematical and statistical calculations were per- al pole, and in the apical pole there were the Golgi appa-
formed with Statistica 5.0. ratus and secretion follicles containing colloid. The cell
membrane formed microvilli in the lumen of the thyroid

352 Horm Res 2009;71:350–358 Ben-Skowronek /Sierocinska-Sawa /


Szewczyk /Korobowicz
Fig. 1. Control group. Cuboidal thyrocytes with a round nucleus, Fig. 2. Colloid goiter. Cuboidal thyrocytes with a round nucleus,
numerous mitochondria in the basal pole and Golgi apparatus, mitochondria in the basal pole and small colloid vesicles and mi-
colloid vesicles and microvilli in the apical pole. Thin basal mem- crovilli in the apical pole. Thin basal membrane. Lymphocyte in
brane. !10,000. N = Nucleus; V = vesicles with colloid; RER = the interstitium. !10,000.
rough endoplasmatic reticulum; M = mitochondrion; R = ribo-
some; BM = basal membrane; Lu = lumen of the thyroid follicle.

follicles (fig. 1). In the thyroid vesicle epithelium, sporad- blood vessels. They sometimes formed inconsiderable in-
ically there appeared dying cells with apoptotic features: filtrations, but no lymphatic follicles. Among them, CD8+
a pycnotic cell nucleus, electron-dense cytoplasm and in- suppressor/cytotoxic T lymphocytes were prevalent
distinct structural membranes of the cell organelles. (9.0%), while CD4+T-helper lymphocytes constituted
Immunohistochemical investigations of the lympho- 1.02% of the observed cells. Antigen-presenting cells were
cyte subpopulation in the simple colloidal goiter indicate very scarce (table 2; fig. 3).
an insignificant contribution of lymphatic cells to the Electron microscope investigations revealed that the
goiter structure. The percentage of B CD79␣+ lympho- thyrocytes within the thyroid nodules frequently had a
cytes was merely 0.28%; they were far less numerous than different shape than that in the control group. In the api-
in the control group. The number of CD3+ lymphocytes cal region, there were fewer colloidal granules; the cell
did not exceed 6.36%. The most dominant were the CD8+ membrane projected into the lumen of the vesicles with
suppressor/cytotoxic T lymphocytes (4.38%) compared scarce microvilli. More frequently, there appeared dying,
with T-helper lymphocytes (1.02%). Antigen-presenting apoptotic cells with dark cytoplasm and a pycnotic nucle-
cells were similarly scarce (0.28%; table 2). Morphologi- us. Small lymphocytes were present in the basal mem-
cal interrelations of thyrocytes and lymphocytes in the brane of the thyroid follicles, especially in the sites where
ultrastructural investigations were similar to those in the thyroid epithelium cells were dying. Single T lympho-
control group (fig. 2). cytes (small lymphocytes with poor cytoplasm) or NK
Immunological reactions in the nodular colloidal goi- cells (big, granular lymphocytes) were observed in the lu-
ter were slightly more intense. CD79␣+ B lymphocytes men of thyroid vesicles (fig. 4).
constituted 5.9% of the cells. CD3+ T lymphocytes were The most enhanced immunological reaction was ob-
the most abundant (14.9%). They were present in the bas- served in the thyroid specimens from children with
al membrane of the thyroid follicles and in the vicinity of Graves’ disease. The cells of the immune system infil-

Lymphocytes and Thyrocytes in GD and Horm Res 2009;71:350–358 353


Non-AITD
Fig. 3. Nodular goiter. Cuboidal thyrocytes with the folded nucle- Fig. 4. Nodular goiter. Thyrocytes: one thyrocyte with extremely
ar membrane, small mitochondria in basal pole and numerous enlarged rough endoplasmatic reticulum and pycnotic nucleus
colloid follicle and microvilli in apical pole. Enlarged rough en- characteristic of thyrocyte in cytolysis stage. In the lumen of thy-
doplasmatic reticulum and spaces between thyrocytes. Lympho- roid follicle, a small T lymphocyte and LGL lymphocyte, pheno-
cyte in interstitium near the blood vessel. !10,000. Mv = Micro- typically cytotoxic lymphocyte, are seen. !10,000. Ly = Lyso-
villi. some.

trated the thyroid follicles and interfollicular compart-


ments; they also formed lymph follicles with typical pro-
liferation centers. T lymphocytes constituted 17.73% of
the cells in the specimens. 6.49% of T lymphocytes were
suppressor/cytotoxic T lymphocytes (CD8+) and 2.95%
T-helper cells (CD4+). The other T lymphocytes did not
positively react with either CD8 or CD4 antibodies. CD8+
lymphocytes were located in the basal membranes of the
thyroid follicles and among the thyrocytes. They were
also present in the concentration regions of lymph nod-
ules. CD4+ lymphocytes occurred in proliferation cen-
ters, at the peripheries of lymph nodules and in the inter-
follicular space. Lymphocytic infiltrations were domi-
nated by CD79␣+ B lymphocytes, which constituted
23.21% of the observed cells. They were visible in the bas-
al membranes of the thyroid follicles, and also infiltrated
the walls as well as the interior part of the follicles. They Fig. 5. Graves’ disease. Lymphocyte between the thyrocytes in the
constituted a remarkable percentage of lymph nodule wall of the thyroid follicle. Ultrastructure of the lymphocyte char-
cells. They were found both in the proliferation centers acteristic for T lymphocytes. !10,000. CM = Cell membrane;
RBC = red blood cell.
and at the peripheries of the nodules. CD1a+ antigen-pre-
senting cells appeared more frequently with statistical
significance than in the other examined groups, still they

354 Horm Res 2009;71:350–358 Ben-Skowronek /Sierocinska-Sawa /


Szewczyk /Korobowicz
b

Fig. 6. Graves’ disease. a Lymphocyte be-


tween the thyrocytes in the wall of the thy-
roid follicle. Ultrastructure of the lympho- c
cyte characteristic of T lymphocytes. c
!10,000. C = Cytoplasm; CG = cytoplasm
granules. Lymphocyte connected with
thyrocyte with immunological synapse
(b) and direct (c). !20,000. a

were not numerous in the investigated thyroid specimens secretion vacuoles and well-developed microvilli in the
(0.37%). In the apical region of some thyrocytes in pa- apical region. In some places, we could observe mitotic
tients with Graves’ disease, a positive reaction with CD1a cell division. Symptoms of thyroid cell death were ob-
in the form of CD1a+ granules was observed (table 2). served when they were in contact with LGLs; the cyto-
The analysis of the thyroid ultrastructure in patients plasm in the thyrocytes was filled with electron-dense
with Graves’ disease demonstrated numerous lympho- structures, the endoplasmic reticulum was enlarged and
cytes near the blood vessels (fig. 5). They were T lympho- the mitochondria and pycnotic cell nuclei were swollen.
cytes – small cells with poor cytoplasm and with a big cell In some specimens, there were B lymphocytes with a de-
nucleus filled with dark chromatin, or B lymphocytes – veloped rough endoplasmic reticulum surrounding the
plasmatic cells with characteristic layers of rough endo- cell nucleus. B lymphocytes were frequently in contact
plasmic reticulum around the nucleus. We also observed with T lymphocytes.
cells with a dark nucleus, cytoplasm with vacuoles filled Correlation between the levels of TRAB and CD19+
with dark electron-dense substance and single projec- subset B lymphocytes was observed only in Graves’ dis-
tions of the cell membrane morphologically related to ease patients (r = 0.62, p = 0.002). No correlation was ob-
LGLs of the NK type. T lymphocytes were also observed served between the levels of TPO Ab and TG Ab and lym-
beneath the basal membrane of the thyroid follicles. They phocyte subsets. In the other groups, no correlation was
were also often present among the thyrocytes and in the noticed between TRAB, TPO AB and TG AB levels and
thyroid follicle lumen (fig. 5–7). The lymphocytes formed the percentage of lymphocyte subsets in the thyroid tis-
processes binding with thyrocytes through adhesive pro- sue.
tein substances or directly; the structure of this binding
resembled a tight junction (fig. 6b). It should be empha-
sized that the basal membrane was thickened, with in- Discussion
creased electron density compared with that in the nodu-
lar or simple goiter or in the control. In some regions of In our investigations, a small number of DCs present-
the basal membrane, there were electron-dense deposits, ing CD1a, which characteristic of immature DCs, was ob-
which coincided with plasmocytes (fig. 8). The thyro- served in the thyroids without AITD, but it was signifi-
cytes did not show any traits of destruction, they were in cantly higher in the thyroids from Graves’ disease pa-
fact fairly active: they had big, light cell nuclei, numerous tients. The DCs were immature – antigen CD1a had the

Lymphocytes and Thyrocytes in GD and Horm Res 2009;71:350–358 355


Non-AITD
Fig. 7. Graves’ disease. Lymphocyte and plasmocyte in contact Fig. 8. Plasmocyte in contact with thyrocytes. Thick basal mem-
with each other in the lumen of the thyroid follicle. Lymphocyte brane of the thyroid follicle with protein deposits (asterisk) –
in the interstitium between the thyroid follicles. Many collagen probably immunological complexes. !30,000. R = Ribosome.
fibers are seen in the interstitium. Basal membrane is partially
thickened. !10,000. CF = Fibers of collagen.

structure of an ␣-chain connected with ␤-microglobu- mediated by the professional antigen-presenting cells in
lins [15, 16]. Similar observations were described by secondary lymphoid organs.
Quadbeck et al. [16], who suggested that the phenotype In nontoxic simple and nodular goiter, only lympho-
of intrathyroidal DCs in Graves’ disease supports their cytes CD4+ and CD8+ are observed in bigger numbers
role as potential costimulators of thyroid immunity [16]. in the interstitium of the thyroid, and they occasionally
Other authors observed a higher number of DC in healthy form mononuclear cell infiltrations. In ultrastructure in-
thyroid (2–3% interstitial cell population) [17]. There are vestigations, thyrocytes of different morphology than
indications that such DCs are able to proliferate [18], that in the control group were observed. Probably, it was
which means that not all of the thyroid DCs need to have the effect of somatic mutations of the thyroid [22]. Small
recently immigrated with the bloodstream. Thyroidal T lymphocytes were present mainly in the interstitium or,
DCs are often in close contact with thyrocytes, are clear- sporadically, in the follicle lumen.
ly in immature state and often show monocyte marker Our immunohistochemical investigations and other
characteristics [19]. It is thought that soluble factors pro- observations in Graves’ disease suggested that T and B
duced by TSH-stimulated thyrocytes such as GM-CSF, lymphocytes accumulating in the thyroid do not form
TGF-␤, IL-6 keep intrathyroidal DCs in their immature destructive infiltrates, but they are organized as a periph-
state [18]. The presence of positive reaction to CD1a pro- eral lymphatic tissue. Graves’ disease patients seem to
tein in the granules of the apical part of some thyrocytes have mixed Th1/Th2 profiles [23]. In the in vitro investi-
suggested that the thyrocytes probably initiated thyroid gations, Estienne et al. [24] indicated the possibility of
autoimmune reactivity [20]. The investigations in trans- forming a so-called immunological synapse of a tight
genic mice by Kimura et al. [21] suggested that expression junction between lymphocytes and thyrocytes with par-
of class II MHC molecules on epithelial thyroid cells is ticipation of adhesive proteins. This physical contact may
not required for the initiation of an autoimmune attack result in the establishment of an immunological synapse
on the thyroid. The initiation, then, seems to be mainly [25] able to stimulate intrathyroid T lymphocyte prolif-

356 Horm Res 2009;71:350–358 Ben-Skowronek /Sierocinska-Sawa /


Szewczyk /Korobowicz
eration and differentiation [24]. In our immunohisto- of granules in the apical pole, long microvilli). It is known
chemical investigations, it was the CD8+ lymphocytes that B cells are involved in the production of thyroid au-
that most frequently entered the basal membrane of thy- toantibodies [34, 39–41], and their depletion leads to im-
roid follicles. The latest investigations [26–28] suggested provement of thyroid function in Graves’ disease [11, 12].
that a crucial role in peripheral tolerance or autoreactive The migration of lymphocytes into thyroid follicles fa-
T cells is played by T regulatory subsets (Tregs) divided cilitates direct contact with the antigens of the thyrocyte
into two populations: naturally occurring and inducible apical pole – especially TPO – and production of anti-
[29, 30]. Tregs so far identified as participating in the TPO antibodies as well as formation of LGL phenotype
pathogenesis of Graves’ disease include naturally occur- NK cells, which destroy thyrocytes [1, 42–44]. The mech-
ring CD4+CD25+T cells [26], C8+CD122+T cells [27, 28] anism of migration of lymphocytes into thyroid follicles
and NK cells [31]. The comparison of immunohisto- is unknown, but is probably dependent on chemokines
chemical localization of CD4+ T cells with ultrastruc- [44] and expression of HLA molecules on the surface of
tural investigations showed that CD4+T lymphocytes thyrocytes [45].
were small cells with large nuclei and that a small amount
of cytoplasm was in contact with thyrocytes and other
lymphocytes. We observed T lymphocytes in close con- Conclusions
tact with plasmocytes, which suggested the regulation/
helper function of the cells stimulating plasmocytes to The immune reaction in Graves’ disease and migra-
production of autoantibodies. tion of lymphocytes T and B between thyrocytes results
It is known, and we observed it too, that patients with in the thickening of the basal membrane of the thyroid
Graves’ disease have an increased number of circulating follicle. Lymphocytes T and B can migrate into the thy-
B cells [32–35]. The close contact with T cells (probably roid follicle lumen.
Th2 cells) and plasmocytes was frequently observed only No cytotoxic effect of T cytotoxic/suppressor CD8+
in Graves’ disease and sporadically in the nodular goi- cells on thyrocytes was observed in Graves’ disease, while
ter. a mild cytotoxic effect was observed in non-AITD.
The other subset of T cells, T suppressor/cytotoxic
cells, was detected in immunohistochemical investiga-
tions between the basal membrane of the thyroid follicle Acknowledgements
and thyrocytes and in lymphatic infiltrations. Rifa’i et al.
This work was supported by grant 2P05E04327 from the Pol-
[30] and Endharti et al. [31] described the subsets of nat-
ish Ministry of Science and Higher Education.
urally occurring Tregs CD8+CD25+. It is possible that
when T CD8+ cells, which we observed in our study,
come into contact with thyrocytes, they play the role of
Tregs in the pathogenesis of Graves’ disease. The investi- 1 Weetmann AP, De Groot LJ: Autoimmunity
References
gations of Negrini et al. [36] characterized a subpopula- to the thyroid; in De Groot LJ, Reed Larsen
tion of CD8 T suppressor lymphocytes able to inhibit P, Hennemann G (eds): The Thyroid and
Its Diseases. Philadelphia, Saunders, 2007.
both cell proliferation and cytotoxicity, and observed www.thyroidmanager.org.
that glucocorticoid-induced TNF-like receptor is ex- 2 Von Adrian UH, Mackay CR: T cell function
pressed on such CD8 T suppressor cells and that its acti- and migration. N Engl J Med 2000; 343:
1020–1034.
vation by specific antibody inhibits generation, but not 3 Drakesmith H, Chain B, Beverly P: How can
function of these cells [36]. The papers of Nakano et al. dendritic cells cause autoimmune disease?
[37] and Nagayama [38] suggested a preventive role of Immunol Today 2000;21:215–217.
4 Yadav D, Judkowski V, Flodstrom-Tullberg
Tregs in autoimmunological reaction in the thyroid with M, Sterling L, Redmond WL, Sherman L,
AITD. Sarvetic N: B7-2 (CD86) controls the prim-
In Graves’ disease, the immunological deposits ob- ing of autoreactive CD4 T cell response
against pancreatic islets. J Immunol 2004;
served in the basal membrane of the thyroid follicles lead 173:3631–3939.
to the thickening of this membrane and probably chang- 5 Deeths MJ, Mesher MF: ICAM-1 and B7-1
es in polarization of cell membranes. The thyrocytes in provide similar but distinct costimulation
for CD8+T cells, while CD4+ T cells are
this region were columnar, with signs of increased activ- poorly costimulated by ICAM-1. Eur J Im-
ity (big nuclei, active, enlarged mitochondria, big amount munol 1999;29:45–53.

Lymphocytes and Thyrocytes in GD and Horm Res 2009;71:350–358 357


Non-AITD
6 Sepulveda H, Cerwenka A, Morgan T, Dut- accessory cells isolated from the thyroids of 34 Ben-Skowronek I, Walter-Croneck B, Szew-
ton RW: CD28, Il-2-independent costimula- Wistar and autoimmune-prone BB-DP rats. czyk L: The increase of thyroid autoantibod-
tory pathways for CD8 T lymphocyte activa- J Autoimmun 2000;15:417–424. ies and CD5+B cells in children with Graves’
tion. J Immunol 1999;163:1133–1142. 20 Lam-Tse WK, Drexhage HA: Dendritic cells disease. Horm Res 1998;50(suppl 3):64.
7 Von Boehmer H: Mechanism of suppression in thyroid autoimmune disease. Hot Thyroi- 35 Volpe R: The immunomodulatory effects of
by suppressor T cells. Nat Immunol 2005; 6: dology, 2002; www.hotthyroidology.com. anti-thyroid drugs are mediated via actions
388–344. 21 Kimura H, Kimura M, Tzou S H, Chen YC, on thyroid cells, affecting thyrocyte-immu-
8 Natarajan K, Dimasi N, Wang J, Mariuzza Suzuki K, Rose RN, Caturegli P: Expression nocyte signalling: a review. Curr Pharm Des
RA, Margulies DH: Structure and function of class II major histocompatibility complex 2001;7:451–460.
of natural killer receptors: multiple molecu- molecules on thyrocytes does not cause 36 Negrini S, Fenoglio , Balestra P, Fravega M,
lar solutions to self, non-self discrimination. spontaneous thyroiditis but mildly increases Filaci G, Indiverdi F: Endocrine regulation
Annu Rev Immunol 2002;24:853–885. its severity after immunization. Endocrinol- of suppressor lymphocytes. Role of the glu-
9 Weetman AP: Autoimmune thyroid disease: ogy 2005;146:1154–1162. cocorticoid-induced TNF-like receptor. Ann
propagation and progression. Eur J Endocri- 22 Henneman G: Multinodular goiter in the NY Acad Sci 2006;1069:377–385.
nol 2003;148:1–9. thyroid and its diseases – book chapters. 37 Nakano A, Watanabe M, Iida T, Kuroda S,
10 Cooper DS: Antithyroid drugs. N Engl J Med 2007; www.endotext.org. Matsuzuka F, Miyauchi A, Iwatani Y: Apop-
2005;352:905–917. 23 Ajjan RA, Weetman AP: Cytokines in thy- tosis-induced decrease of intrathyroidal
11 El Fassi D, Nielsen CH, Hasselbach HC, He- roid autoimmunity. Autoimmunity 2003;36: CD4+CD25+ regulatory cells in autoim-
gedus L: The rationale for B lymphocyte de- 351–359. mune thyroid disease. Thyroid 2007; 17: 25–
pletion in Graves’ disease. Monoclonal anti 24 Estienne V, Brisbarre N, Blanchin S, Du- 31.
CD20 antibody therapy as a novel treatment rand-Gorde JM, Carayon P, Ruf J: An in vitro 38 Nagayama Y: Graves’ animal models of
option. Eur J Endocrinol 2006;14:623–632. model based on cell monolayers grown on Graves’ hyperthyroidism. Thyroid 2007; 17:
12 Salvi M, Vannucchi G, Campi I, Curro N, the underside of large-pore filters in bicam- 1–8.
Dazzi D, Simonetta S, Bonara P, Rossi S, Sina eral chambers for studying thyrocyte-lym- 39 Segundo C, Rodriguez C, Aguilar M, Garcia-
C, Guastella C, Ratgla R, Becc-Peccoz P: phocyte interactions. Am J Physiol Cell Poley A, Gavilan L, Bellas C, Brieva JA: Dif-
Treatment of Graves’ disease and associated Physiol 2004;287:C1763–C1768. ferences in thyroid infiltrating B-lympho-
ophthalmopathy with the anti CD20 mono- 25 Dustin ML: Membrane domains and the im- cytes in patients with Graves’ disease:
clonal antibody rituximab: an open study. munological synapse: keeping T cells resting relationship to antibody detection. Thyroid
Eur J Endocrinol 2007;156:33–40. and ready. J Clin Invest 2002;109:155–160. 2004;14:337–344.
13 Bossowski A, Urban M, Stasiak Barmuta A: 26 McLachlan SM, Pichurin PN, Nagayama Y, 40 Segundo C, Rodriguez C, Garcia-Poley A,
Analysis of changes in the percentage of B Aliesky HA, Chen CR, Rapoport B: Toler- Aguilar M, Gavilan L, Bellas C, Brieva JA:
(CD19) and T(CD23) lymphocytes, subsets ance, regulatory T-cells and autoimmunity to Thyroid infiltrating B-lymphocytes in
CD4,CD8 and their memory (CD45RO), and the thyrotropin receptor: insight from trans- Graves’ disease are related to marginal zone
naïve (CD45RA) T cells in children with im- genic mice expressing the human TSHR A and memory B-compartments. Thyroid
mune and non-immune thyroid diseases. J subunit in the thyroid; in Abstr Annu Meet 2001;14:337–344.
Pediatr Endocriol Metab 2003;16:63–70. 77th Am Thyroid Assoc, Phoenix, 2006. 41 Metcalfe RA, Oh YS, Stround C, Arnold K,
14 Marazuela M, Garcia-Lopez Na, Figueroa- 27 Saitoh O, Nagayama Y: Regulation of Graves Weetman AP: Analysis of antibody depen-
Vega N, de la Fuente H, Alvarado-Sanchez B, hyperthyroidism with naturally occurring dent cell-mediated cytotoxicity in autoim-
Monsivais Urenda A, Sanchez-Madrid F, CD4+, CD25+ regulatory cells in a mouse mune thyroid disease. Autoimmunity 1997;
Gonzales-Amaro R: Regulatory T-cells in model. Endocrinology 2006; 14:72417–2422. 25:65–72.
human autoimmune thyroid disease. J Clin 28 Sakaguchi S: Naturally arising Foxp3-ex- 42 Arao T, Morimoto I, Kakinuma A, Ishida O,
Endocrinol Metab 2006;91:3639–3643. pressing CD4+CD25+ regulatory T cells in Teki K, Tanaka Y, Ishikawa N, Ito K, Eto S:
15 Roura-Mir C, Catalfamo M, Cheng TY, Mar- immunological tolerance to self and non- Thyrocyte proliferation by cellular adhesion
qusee E, Gurdyal SB, Jaraquemada D, Moody self. Nat Immunol 2005;6:345–352. T infiltrating lymphocytes through the in-
DB: CD1a and CD1c activate intrathyroidal 29 Beissert S, Schwartz T: Regulatory cells. In- tercellular adhesion molecule-1/lymphocyte
T cells during Graves’ disease and Hashimo- vest Dermatol 2006; 126:14–24. function associated antigen-1 pathway in
to thyroiditis. J Immunol 2005; 174: 3773– 30 Rifa’i M, Kawamoto Y, Nakshima I, Suzuki Graves’ disease. J Clin Endocrinol Metab
3780. H: Essential role of CD8+CD122+T cells in 2000;85: 382–389.
16 Quadbeck B, Ekstein AK, Tews S, Walz M, the maintenance of T cell homeostasis. J Exp 43 Hidka Y, Amino N, Iwatani Y, et al: Increase
Hoermann R, Man K, Gieseler R: Matura- Med 2004;200:1123–1134. in peripheral natural killer cell activity in pa-
tion of thyroid dendritic cells in Graves’ dis- 31 Endharti AT, Rifa’I M, Shi Z, Fukuoka Y, Na- tients with autoimmune disease. Autoim-
ease. Scand J Immunol 2003;55:612–620. kahara Y Kawamoto Y, Takeda K, Isobe K, munity 1992;11:239–246.
17 Croizet K, Rabilloud R, Kostrouch Z, et al: Suzuki H: CD8+CD122+ regulatory cells 44 Armengol MP, Cardoso-Schmidt CB, Far-
Culture of dendritic cells from a non lym- produce IL10 to suppress INF-gamma pro- nandez M, Fernandez M, Ferrer X, Pujol-
phatic organ, the thyroid gland: evidence for duction and proliferation CD8+cells. J Im- Borrel R, Juan M: Chemokines determine
TNF alpha-dependent phenotypic changes munol 2005;175:7093–7097. local lymphogenesis and a reduction of cir-
of thyroid-derived dendritic cells. Lab Invest 32 Nagayama Y, Niwa M, McLachlan SM, Rapo- culating CXCR4+T and CCR7 B and T lym-
2000;80:1215–1225. port B: Schistosoma mansoni and alpha-ga- phocytes in thyroid autoimmune diseases. J
18 Croizet K, Trouttet-Masson S, Kostrouch Z, lactosylceramide: prophylactic effect of Th1 Immunol 2003; 170:6320–6328.
et al: Signaling from epithelial to dendritic immune suppression in a mouse model of 45 Catalfamo M, Serradell L, Roura Mir C,
cells of the thyroid gland: evidence for thy- Graves’ hyperthyroidism. J Immunol 2004; Kolkowski E, Sospedra M, Vives-Pi M, Var-
rocyte-derived factors controlling the sur- 173:2167–2173. gas-Nieto F, Pujoll-Borrel R, Jaraquemada D:
vival, multiplication, and endocytic activity 33 Ben-Skowronek I, Walter-Croneck B, Szew- HLA-DM and invariant chain are expressed
of dendritic cells. Lab Invest 2001; 81: 1601– czyk L: Analysis of thyroid autoantibodies by thyroid follicular cells, enabling the ex-
1013. and lymphocyte subsets at girls with hyper- pression of compact DR molecules. Inter Im-
19 Simons PJ, Delemarre FG, Drexhage HA: A thyroidism; in Abstr 24th Annu Meet Eur munol 1999;11:269–277.
functional and phenotypic study on immune Thyroid Assoc, 1997, Munich.

358 Horm Res 2009;71:350–358 Ben-Skowronek /Sierocinska-Sawa /


Szewczyk /Korobowicz
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

S-ar putea să vă placă și