Sei sulla pagina 1di 10

REVIEWS

AUTOIMMUNE THYROID DISEASE:


NEW MODELS OF CELL DEATH IN
AUTOIMMUNITY
Giorgio Stassi* and Ruggero De Maria‡
Autoimmunity to thyroid antigens leads to two distinct pathogenic processes with opposing
clinical outcomes: hypothyroidism in Hashimoto’s thyroiditis and hyperthyroidism in Graves’
disease. The high frequency of these diseases and easy accessibility of the thyroid gland has
allowed the identification of key pathogenic mechanisms in organ-specific autoimmune diseases.
In early investigations, antibody- and T-cell-mediated death mechanisms were proposed as
being responsible for autoimmune thyrocyte depletion. Later, studies on apoptosis have provided
new insights into autoimmune target destruction, indicating the involvement of death receptors
and cytokine-regulated apoptotic pathways in the pathogenesis of thyroid autoimmunity.

CENTRAL TOLERANCE Classically, autoimmune diseases are characterized by the pathogenesis of myasthenia gravis and rheumatic
Autoreactive T cells that react the activity of autoreactive lymphocytes, which cause fever, but might also have a role in other common
strongly with self-ligands are tissue or organ damage through the formation of anti- autoimmune conditions, such as type 1 diabetes and
eliminated during development
bodies that react against host tissues, or effector T cells, multiple sclerosis8,9. Moreover, a transient viral infec-
in the thymus by a process that
is known as negative selection. which are specific for endogenous self-peptides1. tion might initiate an autoimmune response that
Environmental and genetic factors cooperate in the could subsequently be amplified through the exposure
CRYPTIC EPITOPES induction of autoimmunity. Although several genes, of CRYPTIC EPITOPES that are released by dying cells, a
Antigenic peptides that are including certain major histocompatibility complex phenomenon known as epitope spreading10.
generated at sub-threshold
levels. When cryptic epitopes
(MHC) genotypes, are clearly associated with increased Autoimmune diseases are broadly classified as organ
become visible to the immune susceptibility to autoimmunity, monozygotic twins do specific or systemic1. Organ-specific autoimmunity
system, they might elicit an not show complete concordance2,3. So, a disease-prone involves chronic T-cell or antibody targeting of a partic-
immune response that is genetic background might not be sufficient for the ular organ. By contrast, systemic autoimmunity is often
responsible for the
clinical onset of autoimmunity. the result of the breakdown of immunological tolerance
autoimmune disease.
Responses to self-tissue antigens result from ineffec- to ubiquitous self-molecules, which generates immune-
tive control mechanisms that allow survival and expan- complex-mediated damage in several body sites11,12.
sion of autoreactive lymphocyte clones. The activation of Thyroid autoimmune diseases represent more than
autoreactive T cells that have escaped CENTRAL TOLERANCE 30% of all organ-specific autoimmunity. Hashimoto’s
*Department of Surgical mechanisms in the thymus is prevented through thyroiditis is the first described and most common
and Oncological Sciences, peripheral tolerance mechanisms that include the elim- organ-specific autoimmune disease, which affects
University of Palermo,
Palermo, Italy. ination of autoreactive lymphocytes4,5 and the about 3% of the population and represents the arche-

Laboratory of Haematology inhibitory activity of CD4+CD25+ regulatory T cells6,7. type for other T-cell-mediated degenerative diseases,
and Oncology, In some circumstances, autoimmunity can arise fol- such as type 1 diabetes and multiple sclerosis13–15.
Istituto Superiore di Sanità, lowing protective T- and B-cell responses against However, the concept of cytotoxic T lymphocyte
Rome, Italy.
Correspondence to R.DM.
foreign antigens that cross-react with self-antigens (CTL)-mediated target destruction in thyroid autoim-
email: rdemaria@tin.it expressed by normal tissues. This phenomenon, which munity has recently been challenged16–19. Progress in
DOI: 10.1038/nri750 is known as molecular mimicry, has been implicated in apoptosis research and studies of Graves’ disease, a

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | MARCH 2002 | 1 9 5


© 2002 Macmillan Magazines Ltd
REVIEWS

a Hashimoto's thyroiditis b Graves' disease

help help help


TSH-reactive
CD4 CD4 B cell
T cell T cell

B cell CD8
T cell B cell

Autoreactive Autoreactive

Plasma
cell TSI

CTL

Thyroid cell
TSHR

Necrosis/apoptosis Thyroid Apoptosis Thyroid cell


cell death survival

Hypothyroidism Hyperthyroidism

Figure 1 | Thyroid autoimmunity produces two opposite pathogenetic processes and clinical outcomes. a | During
Hashimoto’s thyroiditis, self-reactive CD4+ T lymphocytes recruit B cells and CD8+ T cells into the thyroid. Disease progression leads
to the death of thyroid cells and hypothyroidism. Both autoantibodies and thyroid-specific cytotoxic T lymphocytes (CTLs) have
been proposed to be responsible for autoimmune thyrocyte depletion. b | In Graves’ disease, activated CD4+ T cells induce B cells
to secrete thyroid-stimulating immunoglobulins (TSI) against the thyroid-stimulating hormone receptor (TSHR), resulting in
unrestrained thyroid hormone production and hyperthyroidism.

non-destructive form of thyroid autoimmunity, have histopathological feature in glands of patients with
led to a new model of autoimmune target destruction in Hashimoto’s thyroiditis. Activated CD8+ and CD4+ T
Hashimoto’s thyroiditis. cells, B cells, plasma cells and macrophages (sometimes
with the formation of ectopic GERMINAL CENTRES) consti-
Hashimoto’s thyroiditis and Graves’ disease tute the immunocyte infiltrate. Thyroid autoantibodies
Hashimoto’s thyroiditis and Graves’ disease are the most have been proposed to participate in clinical hypothy-
common forms — and the two extremes — of a wide roidism by blocking the thyroid-stimulating hormone
spectrum of mixed thyroid autoimmune conditions (TSH) receptor or by directly contributing to thyrocyte
that lead to thyrocyte death or hyperfunction15. destruction15 (FIG. 1).
Hashimoto’s thyroiditis is characterized by an In patients with Graves’ disease, the production of
inflammatory infiltrate of immunocytes that replace anti-TSH-receptor antibodies promotes thyrocyte
GERMINAL CENTRES
Secondary lymphoid follicles the parenchyma and induce thyroid enlargement, growth and unrestrained thyroid hormone secretion,
that contain reactive B cells which eventually leads to gland fibrosis. Progressive resulting in the opposite clinical outcome to
which undergo intense thyrocyte depletion results in impaired thyroid hor- Hashimoto’s thyroiditis — hyperthyroidism with
proliferation, maturation and mone production and clinical hypothyroidism, a con- increased gland vascularity, mild lymphocytic infiltra-
death after encountering their
specific antigens.
dition that involves a marked reduction of metabolic tion, OPHTHALMOPATHY and GOITRE15. The autoimmune
activity in various cells and tissues15. Hashimoto’s thy- process that leads to Graves’ disease is believed to begin
ENDOCRINE roiditis has been suggested to be a largely genetic disor- with the activation of thyroid-specific CD4+ T cells,
OPHTHALMOPATHY der of cell-mediated immunity that promotes inflam- with subsequent recruitment into the thyroid of
A complex eye disease that is
mation and leads to autoantibody formation20. After autoreactive B cells and production of anti-thyroid
characterized by lymphocyte
and chronic inflammatory-cell initiation of the autoreactive immune response by antibodies (FIG. 1). Although the pathogenetic mecha-
infiltration in orbital tissues, CD4+ T cells, thyroid cells are induced to express MHC nisms that are responsible for hyperthyroidism have
oedema and proliferation of class II molecules by interferon-γ (IFN-γ)-producing been extensively characterized, the aetiology of Graves’
connective tissue. activated lymphocytes, possibly contributing to the disease remains unclear.
GOITRE
expansion of autoreactive T-cell populations and pro-
Diffuse enlargement of the longing the inflammatory response21,22. As a result, Animal models of thyroid autoimmunity. Although a
thyroid gland. massive lymphocyte accumulation is the main spontaneous animal model of Graves’ disease is not

196 | MARCH 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

a Plasma
cell

CTL

c Death
receptor
Thyroid
cell

Suicide

Death
ligand
Complement
activation

Necrosis Apoptosis Apoptosis


Figure 2 | Three different mechanisms of thyrocyte depletion in Hashimoto’s thyroiditis have been sequentially
proposed. a | Autoantibodies deposited as immune complexes on the follicular basement membranes might activate complement
and mediate thyrocyte necrosis. b | After a T-cell receptor/peptide–MHC interaction, cytotoxic T lymphocytes (CTLs) might kill target
thyrocytes through the release of granules that contain perforin and granzyme B. c | Autocrine or paracrine interactions between
death receptors and their ligands might result in thyrocyte apoptosis. MHC, major histocompatibility complex.

available, immunization of selected strains of mice human studies, at present, the primary source of infor-
with either MHC-class-II-positive fibroblasts that mation for understanding the pathogenesis of thyroid
express the TSH receptor23,24, or with an expression autoimmunity.
vector that contains the TSH receptor complementary
DNA25, results in hyperthyroidism in female mice. Mechanism of thyrocyte destruction
Although the genetic immunization model has a lower Three different mechanisms have been sequentially pro-
frequency of hyperthyroidism compared with fibrob- posed to be responsible for autoimmune thyrocyte
last immunization, it relates more closely to the depletion (FIG. 2).
human disease owing to the presence of lymphocytic
infiltration and ophthalmopathy. However, this model Antibodies. The cytotoxic activity of antibodies against
has become available only recently and, so far, has thyrocytes, as a result of immune-complex deposition
made only a limited contribution to the understanding in follicular basement membranes and complement
of the pathogenesis of Graves’ disease. activation, was first suggested in 1977 (REF. 36; FIG. 2). In
Several experimental models of autoimmune thy- subsequent experiments, terminal complement com-
roiditis have been established on the basis of genetic plexes that produce the membrane-attack complexes
predisposition26–28, thyroid-antigen immunization29 or were found around thyroid follicles15,37. Antibodies
transfer of cells primed with thyroid antigen30,31. against thyroid peroxidase in thyroiditis have the ability
Studies of these animals have confirmed the central to fix complement and have been proposed to promote
role of CD4+ T cells in the induction32 and suppres- antibody-dependent cell-mediated cytotoxicity against
sion33 of experimental autoimmune thyroiditis (EAT), thyroid cells in vitro 38–40. These findings gave support to
whereas B-cell responses and autoantibody production the hypothesis that antibodies against the main thyroid
might not have a primary role in thyrocyte targeting, antigens, such as thyroglobulin and thyroid peroxidase,
but seem to contribute to the amplification of the were directly responsible for the autoimmune destruc-
inflammatory response34. tion of thyrocytes. Moreover, thyrocytes that were
Although experimental models of thyroiditis have attacked by complement were shown to release proin-
provided considerable insights, significant thyrocyte flammatory molecules, such as prostaglandin E2, IL-1
destruction is not observed during EAT, unless thy- and IL-6, which might cause infiltration and activation
roglobulin-reactive T cells are treated in vitro with of lymphocytes, and direct injury of thyroid cells during
anti-CD25 and interleukin-12 (IL-12) before injection. Hashimoto’s thyroiditis41. However, the presence and
However, this induces a granulomatous form of thy- titre of autoantibodies in euthyroid individuals and in
roiditis35, which is histologically distinct from Hashimoto’s thyroiditis patients are variable and
Hashimoto’s thyroiditis. So, high disease frequency scarcely correlate with the extent of thyrocyte deple-
and easy accessibility of the thyroid gland make tion15. Therefore, it is possible that the pathogenetic role

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | MARCH 2002 | 1 9 7


© 2002 Macmillan Magazines Ltd
REVIEWS

a b
T cell T cell

Ligand

Man-6-P/IGF II R Death receptor

Perforin

Granzymes

BCL2 FADD
Caspase-8
Caspase-8 BID BCL-XL BCL2
cFLIP BCL-XL

Active BID
Caspase-3 Caspase-9 caspase-8
Cer GD3

Caspase-3 Caspase-9
Mitochondria
Apoptotic
substrates
Cytochrome c
Apoptotic
APAF-1 substrates
Cytochrome c
Apoptosis
APAF-1
Apoptosis
Figure 3 | Two independent pathways mediate T-cell cytotoxicity. a | After specific recognition of peptide–MHC complexes, cytotoxic T lymphocytes release lytic
granules that contain perforin and granzyme B. Perforin polymerizes and forms pores in the target cell membrane, while mannose 6-phosphate/insulin-like growth
factor II receptor (man-6-P/IGF II R) allows granzyme B — a serine protease able to directly activate effector caspases and apoptosis — to enter the cell. Caspase
activation is amplified by granzyme-mediated cleavage and activation of BID, a pro-apoptotic member of the BCL2 family that leads to cytochrome c release from
mitochondria, allowing the formation of a high-molecular-weight complex that contains cytochrome c, APAF-1 and caspase-9, which activates downstream caspases
(mitochondrial apoptotic pathway). This process is antagonized by BCL2 and BCL-X L. b | Binding of ligands to death receptors, such as CD178 to CD95 and TRAIL
(tumour-necrosis factor (TNF)-related apoptosis-inducing ligand) to TRAIL receptors, induces the recruitment of the adaptor molecule FADD/MORT-1 and procaspase-
8 to form the death-inducing signalling complex, which results in caspase-8 activation. Active caspase-8 migrates to the cytoplasm to cleave caspase-3 and BID and
amplify the apoptotic cascade. Ceramide (Cer) accumulation participates in this process by increasing the GD3 ganglioside levels and targeting mitochondria.
Although BCL2 and BCL-X L can antagonize this signal, a more effective inhibition of the apoptotic process is mediated by cellular FLICE/caspase-8 inhibitory protein
(cFLIP), which is recruited into the death-receptor signalling complex and prevents caspase-8 activation. GD3, disialoganglioside 3.

of thyroid-specific antibodies is essentially confined to thyroiditis contain perforin46. During autoimmune


their potential ability to amplify inflammation. inflammation, thyrocytes express MHC class II mole-
cules, and are therefore potential targets of both CD4+
CTL activity. Later, attention was focused on the role of and CD8+ cytotoxic T cells47,48 (FIG. 2). T-cell lines and
T lymphocytes as effector cells responsible for thyrocyte CD8+ T-cell clones obtained from Hashimoto’s thyroidi-
destruction. The engagement of death receptors and the tis glands have shown specific cytotoxic activity against
release of cytotoxic granules mediate effector-T-cell autologous human thyroid cells49–51. However, freshly
cytotoxicity against target cells during the immune isolated autoreactive T cells that are infiltrating the thy-
response42,43. These two events are independent, but roid gland do not show any spontaneous cytotoxic
exploit similar biochemical pathways to induce target- activity against thyroid cell targets15. Therefore, it is not
cell apoptosis, which involves proteolytic cleavage of key clear whether or not these cells contribute directly to
cellular substrates by caspases or granzymes44,45 (FIG. 3). thyrocyte depletion by acting as cytotoxic effector cells.
The exocytosis of perforin-containing granules by
CTLs on cognate recognition of target cells, and the Death receptors. Death receptors are a subgroup of the
engagement of death receptors on cognate or neigh- tumour-necrosis-factor receptor (TNFR) superfamily,
bouring target cells by effector T cells, mediates CTL which is characterized by a cytoplasmic death domain
activity 42,43. Similar to normal peripheral lymphocytes, that is responsible for the transmission of the apop-
a discrete portion of intrathyroidal T cells that are iso- totic signal. The interaction of death receptors with
lated from the thyroids of patients with Hashimoto’s their ligands activates apoptotic pathways in several

198 | MARCH 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

Table 1 | Expression of apoptosis-related proteins in thyrocytes


Name Type Control GD HT References
CD95 (Fas/APO-1) Death receptors + ++ +++ 16,17,19,54,57–60,94
TRAILR1 (DR4) + ++ ++ 68, our unpublished data
TRAILR2 (DR5) + ND ND 68, our unpublished data
CD178 (FasL/CD95L) Death ligands + ++ +++ 16–19,57,59,60
TRAIL + ND +++ 67,68
FAP-1 Death inhibitors ++ ND ND 70
cFLIP + +++ + 19
Caspase-3 Pro-apoptotic + + +++ 19
molecules
Caspase-8 + + +++ 19
BAX – ND ++ 95, our unpublished data
BAK + ND ++ 95, our unpublished data
BCL2 Anti-apoptotic ++ +++ + 62,95
molecules
BCL-X L ++ +++ + 19
The table summarizes absent (–), low (+), high (++) and very high (+++) expression of pro-apoptotic and anti-apoptotic proteins in control,
Graves’ disease (GD) and Hashimoto’s thyroiditis (HT) thyroid follicular cells. cFLIP, cellular FLICE/caspase-8 inhibitory protein; FAP-1,
Fas-associated phosphatase-1; FasL, Fas ligand; ND, not determined; TRAIL, tumour-necrosis factor (TNF)-related apoptosis-inducing
ligand; TRAILR, TRAIL receptor.

physiological and pathological cell-death processes. CD95 expression in normal thyrocytes is hindered by
Triggering of death receptors transduces apoptotic sig- the low sensitivity of immunohistochemistry and by the
nals through the activation of caspases, a family of spontaneous upregulation of CD95 that is observed in
fourteen (known) proteases that induce proteolytic thyrocytes cultured ex vivo. Also, the absence of reliable
cleavage of several cellular targets52. Activation of ini- anti-CD178 antibodies in the earlier studies generated
tiator caspases, such as caspase-8 and caspase-9, trig- controversy between different groups16,58–60. A further
gers a cascade of biochemical signals that results in the setback derives from the minimal availability of normal
activation of downstream caspases and apoptotic cell thyroid specimens and the use of samples from non-
death52 (FIG. 3). BCL2 family members show pro-apop- toxic goitre or the contralateral lobe of tumour glands57
totic and anti-apoptotic activity by acting on mito- — which might have abnormal expression of CD95 and
chondria. The pro-apoptotic members, such as BAX, CD178 — as a source of control thyrocytes. Although it
BAK and BID, induce mitochondrial membrane per- is not possible to draw a conclusive evaluation from
meabilization and cytochrome c release. This promotes data in the literature, we have analysed normal thyroid
the formation of the apoptosome, a high-molecular- tissues from two laringectomy patients. We found that
weight complex that consists of apoptosis protease-acti- both CD95 and, to a lesser extent, CD178 are expressed
vating factor 1 (APAF-1) and caspase-9, which activates at low levels in normal cells (G.S. and R.D.M., unpub-
executioner caspases. By contrast, BCL2 and BCL-XL lished observations). By contrast, there is general agree-
antagonize this process by preventing the generation of ment that both CD95 (REFS 19,61) and CD178 (REFS 19,62)
apoptotic signals by mitochondria53 (FIG. 3). are substantially upregulated and simultaneously
Inappropriate CD95 (Fas/APO-1)-mediated apop- expressed by thyrocytes during Hashimoto’s thyroiditis.
tosis has been proposed to be a common pathogenetic Although the expression of CD178 in the thyroid
mechanism in organ-specific autoimmune dis- inhibits the development of experimental autoimmune
eases17,54–56. In the past few years, several reports have thyroiditis by inducing the death of infiltrating T
investigated the expression and function of CD95 and cells63,64, implying a protective role for CD178 in this
its ligand (CD178, also known as Fas ligand) in normal model, Hashimoto’s thyroiditis thyrocytes are sensitive
and pathological tissues that are frequent targets of to CD95 stimulation and probably die by autocrine or
destructive autoimmune responses, such as the thyroid, paracrine expression of CD178 (REF 19; FIG. 2; also see
pancreas and brain16,55,56. The results of these studies later discussion).
indicate a potential involvement of CD95/CD178- Other death-receptor ligands might participate in
mediated apoptosis in the pathogenesis of Hashimoto’s thyrocyte killing, including TNF-α and the TNF-related
thyroiditis, type 1 diabetes and multiple sclerosis. apoptosis-inducing ligand (TRAIL) (TABLE 1). TNF-α is
The pattern of CD95 and CD178 expression in nor- produced by immunocytes and possibly by thyrocytes
mal and Graves’ disease thyrocytes is controversial. during autoimmune thyroiditis65. The ability of TNFR1
Whereas some authors reported the absence of CD95 to promote CD95-induced thyrocyte apoptosis indicates
and presence of CD178 in control thyrocytes, others that TNF-α and CD178 might act in concert to produce
showed the opposite (reviewed in REF. 57). Analysis of thyrocyte depletion in Hashimoto’s thyroiditis66.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | MARCH 2002 | 1 9 9


© 2002 Macmillan Magazines Ltd
REVIEWS

Although there is no experimental evidence as yet, destruction in Hashimoto’s thyroiditis, as the extent of
TRAIL and its receptors (TRAILR1 and TRAILR2) thyrocyte apoptosis in vivo strictly correlates with the
might contribute to thyrocyte apoptosis in autoimmune intensity of caspase activation. In line with the apoptosis
thyroiditis in a similar way to CD95 and CD178 (REFS 67, pattern that is observed in thyroid autoimmune dis-
68 and G.S. and R.D.M., unpublished results). eases, caspase-3 and caspase-8 are upregulated and acti-
vated in Hashimoto’s thyroiditis thyrocytes, whereas
Regulation of thyrocyte survival they seem to be normal in Graves’ disease19.
It is becoming increasingly clear that the rate of thyrocyte
apoptosis dictates the clinical outcome of thyroid T cells and thyrocytes: who kills whom?
autoimmunity. The balance between the activities of It has been proposed that CD178 expression in selected
pro-apoptotic and anti-apoptotic proteins regulates thy- tissues, such as the anterior chamber of the eye, con-
rocyte survival. The best-characterized apoptosis-related tributes to the formation of immune-privileged sites
genes that are expressed in thyrocytes are death receptors through the CD95-mediated killing of potentially
and their ligands, caspases and BCL2 family members harmful lymphocytes71,72. Similarly, CD178 expression
(TABLE 1). in cancer cells might contribute to tumour immune
After complete thyroid formation, normal thyro- privilege by inducing the deletion of T lymphocytes that
cytes have a very slow turnover rate and are supposed approach the tumour73–75. These hypotheses have been
to undergo very few divisions. Accordingly, apoptosis challenged by some investigators, who believe that
has been observed only occasionally in normal thyroid CD178 expression does not promote immune privilege
in vivo. By contrast, thyrocyte apoptosis is a common or tumour escape76,77. Using CD178-transgenic mice or
finding during the active phases of Hashimoto’s thy- exogenous CD178 expression in transplanted cells, sev-
roiditis19,61,69. Analysis of thyrocytes purified from eral studies reported accelerated destruction rather than
Hashimoto’s thyroiditis glands has shown that a vari- protection of tissues that express CD178 (REFS 76,78).
able percentage of these cells undergoes spontaneous These experiments indicate that CD178-mediated
apoptosis ex vivo, indicating that a substantial number immune privilege might be restricted to very few cell
of thyrocytes are committed to cell death by some types, such as corneal epithelial cells72.
apoptogenic factors during autoimmune thyroidi- Normal thyrocytes are not sensitive to CD95 stimu-
tis19,61. The duration of the autoimmune process, lation and constitutively express low levels of CD178
which lasts several years, indicates that thyrocyte (REFS 19,54). During autoimmune thyroiditis, simultane-
depletion is slow and partially counterbalanced by the ous receptor and ligand upregulation in thyrocytes
capacity of thyroid replacement. In line with the clini- could have remarkable effects on the survival of these
cal course of the disease, thyrocyte apoptosis is not cells and neighbouring lymphocytes. In Hashimoto’s
detectable in glands from most patients with Graves’ thyroiditis, T cells that are located in proximity to thy-
disease, unless they undergo persistent treatment with roid follicles are apoptotic or seem committed to apop-
antithyroid drugs. tosis, as shown by DNA fragmentation and cytoplasmic
Normal thyrocytes constitutively express low levels of accumulation of the pro-apoptotic GD3 (disialogan-
several death receptors (TABLE 1). Thyrocyte priming by glioside 3) ganglioside18. In agreement with this, the
cytokines or the use of cyclohexamide are required for killing of infiltrating T cells by thyrocytes that are
obtaining sensitivity to death-receptor ligands19,54,66. So, ectopically or spontaneously expressing CD178 has
low production of death-receptor ligands by normal been shown in mouse models of EAT63,79. It is likely that
thyrocytes is not harmful. However, after pathological those intrathyroidal T lymphocytes that interact during
modifications of the thyroid microenvironment, thyro- Hashimoto’s thyroiditis with thyroid follicular cells that
cytes might increase their susceptibility to apoptosis and are producing large amounts of CD178 do not survive
acquire sensitivity to death-receptor stimulation. The and are gradually replaced by new infiltrating T cells18.
mechanisms responsible for the refractoriness of nor- As a consequence of CD178 expression, thyroid epithe-
mal thyrocytes to CD95 stimulation might involve high lium might constitute an immune-privileged site80.
expression of Fas-associated phosphatase-1 (FAP-1), a However, increased CD95 expression and sensitivity dur-
protein that interacts with the negative regulatory ing human autoimmune thyroiditis might result in
domain of CD95 and blocks thyrocyte apoptosis70. thyrocyte death by autocrine or paracrine CD95–CD178
The activity of apoptotic and anti-apoptotic genes interactions16,19,62. Opposing this hypothesis, ectopic
during thyroid autoimmunity is a key determinant of expression of CD178 in the mouse thyroid was not
thyrocyte fate. A correlation between thyrocyte destruc- accompanied by autocrine or paracrine CD95-mediated
tion and decreased expression of BCL2 has been pro- death63,64. In these animal models, thyrocyte survival in
posed as a pathogenetic factor that regulates the survival the presence of CD178 might result from the strong inhi-
of autoimmune thyrocytes62. Moreover, increased bition of the inflammatory response64, which is required
expression of other anti-apoptotic genes in Graves’ dis- for promoting thyrocyte sensitivity to CD95 stimula-
ease thyrocytes, such as cellular FLICE/caspase-8 tion19,66. In humans, however, both thyrocytes19 and lym-
inhibitory protein (cFLIP) and BCL-XL, could help to phocytes18 that are resident in Hashimoto’s thyroiditis
prevent thyrocyte apoptosis during autoimmune glands are susceptible to CD95-induced apoptosis ex vivo
aggression19. Expression and activation of caspases and probably die in vivo after intense thyrocyte CD178
could represent an important event leading to thyrocyte production (FIG. 4).

200 | MARCH 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

Paracrine thyroid-cell
Thyroid cell apoptosis (fratricide) Suicide
Death
ligand

Death
receptor

T-cell
apoptosis

Infiltrating
activated
T cell
T cell T cell

Fratricide

Suicide
Figure 4 | Infiltrating T cells might be killed by thyrocytes during Hashimoto’s thyroiditis. Activated T lymphocytes in
autoimmune thyroiditis are extremely sensitive to CD95 stimulation and probably die after an interaction with CD178-expressing
thyrocytes. However, because Hashimoto’s thyroiditis thyrocytes acquire sensitivity to CD95 stimulation, they might also die through
suicide or fratricide mechanisms.

The rate of apoptosis in Hashimoto’s thyroiditis which acts as an autocrine and paracrine T-cell growth
glands seems higher for lymphocytes than for thyrocytes factor and dictates the extent of the T-cell response. IL-2
both in vitro and in vivo18. However, dying T cells are also stimulates the synthesis of other T-cell-derived
continuously replaced during the inflammatory process cytokines, such as IFN-γ 87. This cytokine is a potent
and infiltrating T cells probably have an important role macrophage activator and promotes inflammation and
in the amplification and perpetuation of the autoim- effector CD8+ T-cell cytotoxicity. Moreover, IFN-γ con-
mune response through the production of cytokines. tributes to the amplification of the immune response by
inducing the expression of MHC class II molecules on
Cytokines and thyroid autoimmunity thyroid cells, an effect that is probably potentiated by
Cytokines drive thyroid autoimmune responses by thyroid-stimulating antibodies in Graves’ disease and by
influencing both immune and target cells at several high TSH levels in hypothyroidal Hashimoto’s thyroiditis
levels. T-cell-derived cytokines are the key regulators of patients21,22. On the other hand, in Graves’ disease, IL-4
the autoimmune processes. They provide essential and IL-10 might protect thyrocytes from TH1 responses
support for the cell-mediated and humoral immune by inducing T-cell ANERGY, suppression of cytotoxic
response and dictate thyrocyte fate. responses and TH1 to TH2 switching88,89. Intrathyroidal
IL-4 and IL-10 production in Graves’ disease might
Cytokine expression in thyroid autoimmunity. Two inhibit macrophage activation and block most of the
functionally distinct subsets of T-helper (TH) cells have activating effects of IFN-γ, including increased produc-
been characterized on the basis of cytokine production. tion of IL-1, nitric oxide and prostaglandins90,91. In sup-
TH1 cells secrete IFN-γ and other cytokines that are port of this model, after promising experimental evi-
associated with inflammation and cell-mediated dence in transgenic mice, IL-10 has been proposed as a
immune responses, whereas TH2 cells promote the potential therapeutic factor for preventing autoimmune
humoral immune response and inhibit TH1 responses thyrocyte destruction92. It remains to be determined
by the release of IL-4, IL-5 and IL-10 (REF. 81). In fact, whether CD4+CD25+ regulatory T cells have a role in
TH2 cytokines have been shown to inhibit target thyroid autoimmunity. These professional suppressor
destruction in organ-specific autoimmunity82,83. cells produce IL-10 (REF. 93) in vivo and might therefore
In line with their pathogeneses, analysis of cytokine influence the behaviour of both immune and target cells.
expression in autoimmune thyroid diseases has shown,
with a few exceptions84, a prevalence of TH2 cytokines in Cytokines and thyrocyte apoptosis. There is extensive evi-
Graves’ disease and TH1 cytokines in Hashimoto’s thy- dence that cytokines modify the expression of several
ANERGY
A condition of complete
roiditis19,85, particularly at the level of activated intrathy- genes that are involved in the survival or death of
unresponsiveness to antigens roidal T cells86. During the active phase of Hashimoto’s autoimmune target cells. In type 1 diabetes, the produc-
that can affect both T and B cells. thyroiditis, infiltrating T cells produce IL-2 (REF. 86), tion of IL-1β by infiltrating macrophages induces CD95

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | MARCH 2002 | 2 0 1


© 2002 Macmillan Magazines Ltd
REVIEWS

a Hashimoto's thyroiditis b Graves disease


IL-4
IFN-γ IL-10

Caspases cFLIP and


Thyroid cell and CD95 BCL-XL
upregulated upregulated

Death Death
ligand receptor

Cell death Cell survival

Figure 5 | Model of thyrocyte fate in thyroid autoimmune diseases. a | Infiltrating CD4+ T cells seem to preferentially produce
T-helper type 1 cell (TH1) cytokines in Hashimoto’s thyroiditis and TH2 cytokines in Graves’ disease. Interferon-γ (IFN-γ) primes
thyrocytes for CD95-mediated destruction by inducing massive upregulation of CD95 and pro-apoptotic caspases in Hashimoto’s
thyroiditis. b | By contrast, interleukin-4 (IL-4) and IL-10 production might protect Graves’ disease thyrocytes from CD95-mediated
apoptosis through the upregulation of cellular FLICE/caspase-8 inhibitory protein (cFLIP) and BCL-XL.

expression in pancreatic β-cells through the activation of According to this model, the predominance of TH1
inducible nitric-oxide synthase, resulting in a specific or TH2 cytokines in the autoimmune microenviron-
destruction of insulin-producing cells in the presence of ment of the thyroid dictates the outcome of both
CD178+ T lymphocytes55. Similarly, IFN-γ released by destructive and non-destructive immune-mediated
autoreactive T cells might contribute to autoimmune processes by acting at two levels. First, they orchestrate
oligodendrocyte destruction in multiple sclerosis by the immune response, and second, they directly regulate
increasing CD95 expression and susceptibility to CD178- the balance between pro- and anti-apoptotic proteins in
mediated apoptosis17,56. Although accurate analysis is Hashimoto’s thyroiditis and Graves’ disease thyrocytes.
hampered by early spontaneous CD95 upregulation in
cultured thyrocytes, both IL-1β and IFN-γ have been Future prospects
shown to increase CD95 expression in thyrocytes The mechanisms that are responsible for initiating thy-
in vitro94. However, IFN-γ is more effective than IL-1β in roid autoimmune responses remain to be determined.
promoting CD95 expression and thyrocyte apoptosis, However, the high disease frequency and easy accessi-
and is therefore the most likely candidate for priming bility of the thyroid gland have allowed substantial
CD178-mediated thyrocyte destruction in Hashimoto’s advances in the definition of the immune mechanisms
thyroiditis19,66. Altered expression of apoptosis-related that are involved in the survival or death of thyrocytes.
proteins, as a result of different cytokine expression pro- The relative contributions of individual death recep-
files, could explain the opposite fates of thyrocytes in tors in thyrocyte destruction are not known at present.
Graves’ disease and Hashimoto’s thyroiditis. The suscep- In this context, it would be of great interest to deter-
tibility or resistance of thyrocytes to CD95-induced mine how CD178 and TRAIL production by thyrocytes
apoptosis depend on the thyroid microenvironment and is regulated, and whether the death receptor–ligand sys-
are only temporary in thyrocytes analysed ex vivo 19. So, tems have a role in normal thyroid homeostasis. Weak
the role of cytokines in the determination of thyrocyte autocrine or paracrine interactions between CD178 or
survival or death has been comparatively investigated in TRAIL and their receptors might, for example, explain
Graves’ disease and Hashimoto’s thyroiditis. the absence of thyrocyte growth observed in vivo,
Increased sensitivity of Hashimoto’s thyroiditis thy- which is rapidly reversed in vitro after thyroid follicle
rocytes to apoptosis might depend on the IFN-γ-medi- disaggregation.
ated upregulation of key pro-apoptotic genes. IFN-γ In addition, the role of CD4+CD25+ regulatory
treatment increases caspase-3 and caspase-8 expression T cells in the pathogenesis of thyroid autoimmunity
and primes Hashimoto’s thyroiditis thyrocytes for remains to be determined. These cells could markedly
CD95-mediated destruction. By contrast, IL-4 and IL-10 influence both the extent of the autoimmune response
upregulate BCL-XL and cFLIP, two key anti-apoptotic and the survival of thyrocytes through the release of
proteins that can prevent CD95-induced apoptosis in anti-inflammatory and anti-apoptotic cytokines such as
Graves’ disease thyrocytes19 (FIG. 5). IL-10 and transforming growth factor-β (TGF-β).

202 | MARCH 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

Although it is not clear whether the anti-apoptotic the regulation of cell death in both effector and target
activity of TH2 cytokines is a general phenomenon or cells, explaining the success or failure of cytotoxic
restricted to limited cell types, the model of cell death immune processes, such as antiviral and antitumour
versus cell survival presented in this article is not neces- responses. In this regard, the possibility of modulating
sarily confined to organ-specific autoimmune diseases. the production of TH cytokines in selected anatomical
Information on molecules and pathways that are sites might yield new hopes for improving therapeutic
involved in destructive and non-destructive thyroid intervention in various diseases that involve excessive or
autoimmunity could contribute to the understanding of insufficient immune-mediated target destruction.

1. Sinha, A. A., Lopez, M. T. & McDevitt, H. O. Autoimmune 21. Todd, I., Pujol-Borrell, R., Hammond, L. J., Bottazzo, G. F. & 38. Khoury, E. L., Hammond, L., Bottazzo, G. F. & Doniach, D.
diseases: the failure of self tolerance. Science 248, Feldmann, M. Interferon-γ induces HLA-DR expression by Presence of the organ-specific ‘microsomal’ autoantigen on
1380–1388 (1990). thyroid epithelium. Clin. Exp. Immunol. 61, 265–273 (1985). the surface of human thyroid cells in culture: its involvement
2. Metcalfe, K. A. et al. Concordance for type 1 diabetes in 22. Hamilton, F., Black, M., Farquharson, M. A., Stewart, C. & in complement-mediated cytotoxicity. Clin. Exp. Immunol.
identical twins is affected by insulin genotype. Diabetes Care Foulis, A. K. Spatial correlation between thyroid epithelial 45, 316–328 (1981).
24, 838–842 (2001). cells expressing class II MHC molecules and interferon-γ- 39. Bogner, U., Schleusener, H. & Wall, J. R. Antibody-
3. Brix, T. H., Kyvik, K. O. & Hegedus, L. A population-based containing lymphocytes in human thyroid autoimmune dependent cell mediated cytotoxicity against human thyroid
study of chronic autoimmune hypothyroidism in Danish disease. Clin. Exp. Immunol. 83, 64–68 (1991). cells in Hashimoto’s thyroiditis but not Graves’ disease.
twins. J. Clin. Endocrinol. Metab. 85, 536–539 (2000). 23. Shimojo, N. et al. Induction of Graves-like disease in mice by J. Clin. Endocrinol. Metab. 59, 734–738 (1984).
4. Wesselborg, S., Janssen, O. & Kabelitz, D. Induction of immunization with fibroblasts transfected with the 40. Guo, J., Jaume, J. C., Rapoport, B. & McLachlan, S. M.
activation-driven death (apoptosis) in activated but not thyrotropin receptor and a class II molecule. Proc. Natl Recombinant thyroid peroxidase-specific Fab converted to
resting peripheral blood T cells. J. Immunol. 150, Acad. Sci. USA 93, 11074–11079 (1996). immunoglobulin G (IgG) molecules: evidence for thyroid cell
4338–4345 (1993). The first experimental model of Graves’ disease. damage by IgG1, but not IgG4, autoantibodies. J. Clin.
5. Klas, C., Debatin, K. M., Jonker, R. R. & Krammer, P. H. 24. Kita, M. et al. Regulation and transfer of a murine model of Endocrinol. Metab. 82, 925–931 (1997).
Activation interferes with the APO-1 pathway in mature thyrotropin receptor antibody mediated Graves’ disease. 41. Weetman, A. P., Tandon, N. & Morgan, B. P. Antithyroid
human T cells. Int. Immunol. 5, 625–630 (1993). Endocrinology 140, 1392–1398 (1999). drugs and release of inflammatory mediators by
The first demonstration that activated T cells become 25. Costagliola, S. et al. Genetic immunization of outbred mice complement-attacked thyroid cells. Lancet 340, 633–636
susceptible to CD95-induced apoptosis. with thyrotropin receptor cDNA provides a model of Graves’ (1992).
6. Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M. & disease. J. Clin. Invest. 105, 803–811 (2000). 42. Kagi, D. et al. Fas and perforin pathways as major
Toda, M. Immunologic self-tolerance maintained by Description of a new model of Graves’ disease that mechanisms of T cell-mediated cytotoxicity. Science 265,
activated T cells expressing IL-2 receptor α-chains (CD25). shows lymphocytic infiltration and ophthalmopathy. 528–530 (1994).
Breakdown of a single mechanism of self-tolerance causes 26. Dietrich, H. M., Oliveira-dos-Santos, A. J. & Wick, G. 43. Lowin, B., Hahne, M., Mattmann, C. & Tschopp, J. Cytolytic
various autoimmune diseases. J. Immunol. 155, 1151–1164 Development of spontaneous autoimmune thyroiditis in T-cell cytotoxicity is mediated through perforin and Fas lytic
(1995). Obese strain (OS) chickens. Vet. Immunol. Immunopathol. pathways. Nature 370, 650–652 (1994).
Identification of a key population of regulatory T cells. 57, 141–146 (1997). References 42 and 43 were the first to show that both
7. Suri-Payer, E., Amar, A. Z., Thornton, A. M. & Shevach, E. M. 27. Wright, J. R., Jr, Senhauser, D. A., Yates, A. J., Sharma, H. M. perforin and death-receptor pathways mediate T-cell
CD4+CD25+ T cells inhibit both the induction and effector & Thibert, P. Spontaneous thyroiditis in BB Wistar diabetic cytotoxicity.
function of autoreactive T cells and represent a unique rats. Vet. Pathol. 20, 522–530 (1983). 44. Darmon, A. J., Nicholson, D. W. & Bleackley, R. C.
lineage of immunoregulatory cells. J. Immunol. 160, Description of a spontaneous model of autoimmune Activation of the apoptotic protease CPP32 by cytotoxic
1212–1218 (1998). thyroiditis in mammals. T-cell-derived granzyme B. Nature 377, 446–468 (1995).
28. Rasooly, L., Burek, C. L. & Rose, N. R. Iodine-induced
8. Wucherpfennig, K. W. & Strominger, J. L. Molecular mimicry The first demonstration that granzyme B activates the
autoimmune thyroiditis in NOD-H-2h4 mice. Clin. Immunol.
in T cell-mediated autoimmunity: viral peptides activate apoptotic caspase cascade.
Immunopathol. 81, 287–292 (1996).
human T cell clones specific for myelin basic protein. Cell 45. Thornberry, N. A. et al. A combinatorial approach defines
29. Tomazic, V. & Rose, N. R. Autoimmune murine thyroiditis.
80, 695–705 (1995). specificities of members of the caspase family and
VIII. Role of different thyroid antigens in the induction of
9. Olson, J. K., Croxford, J. L., Calenoff, M. A., Dal Canto, M. C. granzyme B. Functional relationships established for key
experimental autoimmune thyroiditis. Immunology 30,
& Miller, S. D. A virus-induced molecular mimicry model of mediators of apoptosis. J. Biol. Chem. 272, 17907–17911
63–68 (1976).
multiple sclerosis. J. Clin. Invest. 108, 311–318 (2001). (1997).
One of the concluding papers of the initial description
10. Lehmann, P. V., Forsthuber, T., Miller, A. & Sercarz, E. E. 46. Wu, Z., Podack, E. R., McKenzie, J. M., Olsen, K. J. &
of the experimental autoimmune thyroiditis model.
Spreading of T-cell autoimmunity to cryptic determinants of Zakarija, M. Perforin expression by thyroid-infiltrating T cells
30. Romball, C. G. & Weigle, W. O. Transfer of experimental
an autoantigen. Nature 358, 155–157 (1992). in autoimmune thyroid disease. Clin. Exp. Immunol. 98,
autoimmune thyroiditis with T cell clones. J. Immunol. 138,
11. Bachmaier, K. et al. Negative regulation of lymphocyte 470–477 (1994).
1092–1098 (1987).
activation and autoimmunity by the molecular adaptor Cbl-b. 47. Pujol-Borrell, R., Hanafusa, T., Chiovato, L. & Bottazzo, G. F.
Demonstration that thyroid-specific T-cell clones can
Nature 403, 211–216 (2000). Lectin-induced expression of DR antigen on human cultured
transfer autoimmune thyroiditis.
12. Robey, E. & Urbain, J. Tolerance and immune regulation. follicular thyroid cells. Nature 304, 71–73 (1983).
31. Knight, S. C. et al. Induction of autoimmunity with dendritic
Immunol. Today 12, 175–177 (1991). cells: studies on thyroiditis in mice. Clin. Immunol. 48. Londei, M., Lamb, J. R., Bottazzo, G. F. & Feldmann, M.
13. Wucherpfennig, K. W. & Eisenbarth, G. S. Type 1 diabetes. Immunopathol. 48, 277–289 (1988). Epithelial cells expressing aberrant MHC class II
Nature Immunol. 2, 767–768 (2001). 32. Flynn, J. C., Conaway, D. H., Cobbold, S., Waldmann, H. & determinants can present antigen to cloned human T cells.
14. Steinman, L. Multiple sclerosis: a two-stage disease. Nature Kong, Y. C. Depletion of L3T4+ and Lyt-2+ cells by rat Nature 312, 639–641 (1984).
Immunol. 2, 762–764 (2001). monoclonal antibodies alters the development of adoptively References 47 and 48 were the first to show that
15. Weetman, A. P. & McGregor, A. M. Autoimmune thyroid transferred experimental autoimmune thyroiditis. Cell. thyrocytes can express MHC class II molecules and
disease: further developments in our understanding. Endocr. Immunol. 122, 377–390 (1989). act as antigen-presenting cells.
Rev. 15, 788–830 (1994). 33. Parish, N. M., Roitt, I. M. & Cooke, A. Phenotypic 49. Mackenzie, W. A. & Davies, T. F. An intrathyroidal T-cell clone
A comprehensive review on thyroid autoimmunity. characteristics of cells involved in induced suppression to specifically cytotoxic for human thyroid cells. Immunology
16. Giordano, C. et al. Potential involvement of Fas and its murine experimental autoimmune thyroiditis. Eur. J. 61, 101–103 (1987).
ligand in the pathogenesis of Hashimoto’s thyroiditis. Immunol. 18, 1463–1477 (1988). 50. MacKenzie, W. A., Schwartz, A. E., Friedman, E. W. &
Science 275, 960–963 (1997). 34. Vladutiu, A. O. Experimental autoimmune thyroiditis in mice Davies, T. F. Intrathyroidal T cell clones from patients with
17. De Maria, R. & Testi, R. Fas–FasL interactions: a common chronically treated from birth with anti-IgM antibodies. Cell. autoimmune thyroid disease. J. Clin. Endocrinol. Metab. 64,
pathogenetic mechanism in organ-specific autoimmunity. Immunol. 121, 49–59 (1989). 818–824 (1987).
Immunol. Today 19, 121–125 (1998). 35. Braley-Mullen, H. et al. Interleukin-12 promotes activation of References 49 and 50 describe T-cell clones that are
18. Stassi, G. et al. Fas/Fas ligand-driven T cell apoptosis as a effector cells that induce a severe destructive selectively cytotoxic to autologous thyroid cells.
consequence of ineffective thyroid immunoprivilege in granulomatous form of murine experimental autoimmune 51. Sugihara, S., Fujiwara, H., Niimi, H. & Shearer, G. M. Self-
Hashimoto’s thyroiditis. J. Immunol. 162, 263–267 (1999). thyroiditis. Am. J. Pathol. 152, 1347–1358 (1998). thyroid epithelial cell (TEC)-reactive CD8+ T cell lines/clones
Unexpected findings indicating that thyrocytes kill Description of a destructive model of experimental derived from autoimmune thyroiditis lesions. They recognize
infiltrating T cells in Hashimoto’s thyroiditis. autoimmune thyroiditis. self-thyroid antigens directly on TEC to exhibit T helper cell
19. Stassi, G. et al. Control of target cells survival in thyroid 36. Kalderon, A. E. & Bogaars, H. A. Immune complex deposits 1-type lymphokine production and cytotoxicity against TEC.
autoimmunity by T helper cytokines via regulation of in Graves’ disease and Hashimoto’s thyroiditis. Am. J. Med. J. Immunol. 155, 1619–1628 (1995).
apoptotic proteins. Nature Immunol. 1, 1–6 (2000). 63, 729–734 (1977). 52. Hengartner, M. O. The biochemistry of apoptosis. Nature
A demonstration that production of T-helper cytokines First demonstration of immune-complex deposits in 407, 770–776 (2000).
promotes survival or death of thyrocytes during the thyroid autoimmunity. 53. Adams, J. M. & Cory, S. Life-or-death decisions by the Bcl-2
autoimmune response. 37. Weetman, A. P., Cohen, S. B., Oleesky, D. A. & Morgan, B. P. protein family. Trends Biochem. Sci. 26, 61–66 (2001).
20. Davies, T. F., Roti, E., Braverman, L. E. & DeGroot, L. J. Terminal complement complexes and C1/C1 inhibitor 54. Arscott, P. L. et al. Fas (APO-1, CD95)-mediated apoptosis
Thyroid controversy — stimulating antibodies. J. Clin. complexes in autoimmune thyroid disease. Clin. Exp. in thyroid cells is regulated by a labile protein inhibitor.
Endocrinol. Metab. 83, 3777–3785 (1998). Immunol. 77, 25–30 (1989). Endocrinology 138, 5019–5027 (1997).

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | MARCH 2002 | 2 0 3


© 2002 Macmillan Magazines Ltd
REVIEWS

55. Stassi, G. et al. Nitric oxide primes pancreatic β cells for Fas- 71. Griffith, T. S., Brunner, T., Fletcher, S. M., Green, D. R. & 87. Lombardi, G. et al. Antigen presentation by interferon-γ-
mediated destruction in insulin-dependent diabetes mellitus. Ferguson, T. A. Fas ligand-induced apoptosis as a treated thyroid follicular cells inhibits interleukin-2 (IL-2) and
J. Exp. Med. 186, 1193–1200 (1997). mechanism of immune privilege. Science 270, 1189–1192 supports IL-4 production by B7-dependent human T cells.
Describes a new pathway that is responsible for (1995). Eur. J. Immunol. 27, 62–71 (1997).
autoimmune β-cell destruction in human diabetes. 72. Stuart, P. M. et al. CD95 ligand (FasL)-induced apoptosis is 88. Racke, M. K. et al. Cytokine-induced immune deviation as a
56. Zipp, F., Krammer, P. H. & Weller, M. Immune (dys)regulation necessary for corneal allograft survival. J. Clin. Invest. 99, therapy for inflammatory autoimmune disease. J. Exp. Med.
in multiple sclerosis: role of the CD95–CD95 ligand system. 396–402 (1997). 180, 1961–1966 (1994).
Immunol. Today 20, 550–554 (1999). 73. Bennett, M. W. et al. The Fas counterattack in vivo: apoptotic 89. Buer, J. et al. Interleukin 10 secretion and impaired effector
57. Borgerson, K. L., Bretz, J. D. & Baker, J. R. Jr. The role of depletion of tumor-infiltrating lymphocytes associated with function of major histocompatibility complex class II-
Fas-mediated apoptosis in thyroid autoimmune disease. Fas ligand expression by human esophageal carcinoma. restricted T cells anergized in vivo. J. Exp. Med. 187,
Autoimmunity 30, 251–264 (1999). J. Immunol. 160, 5669–5675 (1998). 177–183 (1998).
58. Leithauser, F. et al. Constitutive and induced expression of 74. Hahne, M. et al. Melanoma cell expression of Fas(Apo-1/ 90. Fiorentino, D. F., Zlotnik, A., Mosmann, T. R., Howard, M.
APO-1, a new member of the nerve growth factor/tumor CD95) ligand: implications for tumor immune escape. & O’Garra, A. IL-10 inhibits cytokine production by
necrosis factor receptor superfamily, in normal and Science 274, 1363–1366 (1996). activated macrophages. J. Immunol. 147, 3815–3822
neoplastic cells. Lab. Invest. 69, 415–429 (1993). 75. O’Connell, J., O’Sullivan, G. C., Collins, J. K. & Shanahan, F. (1991).
An extensive analysis of CD95 expression in human The Fas counterattack: Fas-mediated T cell killing by colon 91. Cunha, F. Q., Moncada, S. & Liew, F. Y. Interleukin-10 (IL-10)
tissues. cancer cells expressing Fas ligand. J. Exp. Med. 184, inhibits the induction of nitric oxide synthase by interferon-γ
59. Stokes, T. A. et al. Constitutive expression of FasL in 1075–1082 (1996). in murine macrophages. Biochem. Biophys. Res. Commun.
thyrocytes. Science 279, 2015A (1998). 76. Allison, J., Georgiou, H. M., Strasser, A. & Vaux, D. L. 182, 1155–1159 (1992).
60. Stassi, G. & De Maria, R. Response to ‘Thyrocytes — not Transgenic expression of CD95 ligand on islet β-cells 92. Batteux, F., Trebeden, H., Charreire, J. & Chiocchia, G.
innocent bystanders in autoimmune disease’. Nature induces a granulocytic infiltration but does not confer Curative treatment of experimental autoimmune
Immunol. 2, 183 (2001). immune privilege upon islet allografts. Proc. Natl Acad. Sci. thyroiditis by in vivo administration of plasmid DNA
61. Hammond, L. J. et al. Analysis of apoptosis in relation to USA 94, 3943–3947 (1997).
coding for interleukin-10. Eur. J. Immunol. 29, 958–963
tissue destruction associated with Hashimoto’s autoimmune 77. Restifo, N. P. Not so Fas: re-evaluating the mechanisms of
(1999).
thyroiditis. J. Pathol. 182, 138–144 (1997). immune privilege and tumor escape. Nature Med. 6,
93. Asseman, C., Mauze, S., Leach, M. W., Coffman, R. L. &
62. Mitsiades, N. et al. Fas/Fas ligand up-regulation and Bcl-2 493–595 (2000).
Powrie, F. An essential role for interleukin 10 in the function
down-regulation may be significant in the pathogenesis of 78. Kang, S. M. et al. Immune response and myoblasts that
of regulatory T cells that inhibit intestinal inflammation.
Hashimoto’s thyroiditis. J. Clin. Endocrinol. Metab. 83, express Fas ligand. Science 278, 1322–1324 (1997).
J. Exp. Med. 190, 995–1004 (1999).
2199–2203 (1998). 79. Wei, Y., Chen, K., Sharp, G. C., Yagita, H. & Braley-Mullen, H.
94. Kawakami, A. et al. Thyroid-stimulating hormone inhibits Fas
63. Batteux, F., Tourneur, L., Trebeden, H., Charreire, J. & Expression and regulation of Fas and Fas ligand on
antigen-mediated apoptosis of human thyrocytes in vitro.
Chiocchia, G. Gene therapy of experimental autoimmune thyrocytes and infiltrating cells during induction and
Endocrinology 137, 3163–3169 (1996).
thyroiditis by in vivo administration of plasmid DNA coding resolution of granulomatous experimental autoimmune
95. Koga, M. et al. Immunohistochemical analysis of Bcl-2, Bax,
for Fas ligand. J. Immunol. 162, 603–608 (1999). thyroiditis. J. Immunol. 167, 6678–6686 (2001).
64. Batteux, F., Lores, P., Bucchini, D. & Chiocchia, G. 80. Tourneur, L. et al. Transgenic expression of CD95 ligand on and Bak expression in thyroid glands from patients with
Transgenic expression of Fas ligand on thyroid follicular cells thyroid follicular cells confers immune privilege upon thyroid subacute thyroiditis. J. Clin. Endocrinol. Metab. 84,
prevents autoimmune thyroiditis. J. Immunol. 164, allografts. J. Immunol. 167, 1338–1346 (2001). 2221–2225 (1999).
1681–1688 (2000). 81. Abbas, A. K., Murphy, K. M. & Sher, A. Functional diversity
65. Aust, G. et al. Expression of tumour necrosis factor-α (TNF-α) of helper T lymphocytes. Nature 383, 787–793 (1996). Acknowledgements
mRNA and protein in pathological thyroid tissue and 82. Van der Veen, R. C. & Stohlman, S. A. Encephalitogenic TH1 G.S. and R.D.M. are supported by the Associazione Italiana per la
carcinoma cell lines. Clin. Exp. Immunol. 105, 148–154 (1996). cells are inhibited by TH2 cells with related peptide Ricerca sul Cancro.
66. Bretz, J. D., Arscott, P. L., Myc, A. & Baker, J. R. Jr. specificity: relative roles of interleukin (IL)-4 and IL-10.
Inflammatory cytokine regulation of Fas-mediated apoptosis J. Neuroimmunol. 48, 213–220 (1993).
in thyroid follicular cells. J. Biol. Chem. 274, 25433–25438 83. Gallichan, W. S., Balasa, B., Davies, J. D. & Sarvetnick, N. Online links
(1999). Pancreatic IL-4 expression results in islet-reactive TH2 cells
67. Bretz, J. D. et al. TRAIL death pathway expression and that inhibit diabetogenic lymphocytes in the nonobese DATABASES
induction in thyroid follicular cells. J. Biol. Chem. 274, diabetic mouse. J. Immunol. 163, 1696–1703 (1999). The following terms in this article are linked online to:
23627–23632 (1999). 84. Paschke, R., Schuppert, F., Taton, M. & Velu, T. LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/
Demonstration of a new potential pathway for Intrathyroidal cytokine gene expression profiles in APAF-1 | BAK | BAX | BCL2 | BCL-XL | BID | caspase-3 |
thyrocyte apoptosis. autoimmune thyroiditis. J. Endocrinol. 141, 309–315 (1994). caspase-8 | caspase-9 | CD95 | CD178 | cFLIP | cytochrome c |
68. Bretz, J. D. & Baker, J. R. Jr. Apoptosis and autoimmune 85. Heuer, M., Aust, G., Ode-Hakim, S. & Scherbaum, W. A. FAP-1 | IFN-γ | IL-1 | IL-1β | IL-2 | IL-4 | IL-5 | IL-6 | IL-10 | IL-12 |
thyroid disease: following a TRAIL to thyroid destruction? Different cytokine mRNA profiles in Graves’ disease, inducible nitric-oxide syntase | perforin | TGF-β | thyroglobulin |
Clin. Endocrinol. (Oxf.) 55, 1–11 (2001). Hashimoto’s thyroiditis, and nonautoimmune thyroid thyroid peroxidase | TNF-α | TNFR superfamily | TRAIL |
69. Kotani, T. et al. Apoptosis in thyroid tissue from patients with disorders determined by quantitative reverse transcriptase TRAILR1 | TRAILR2 | TSH receptor
Hashimoto’s thyroiditis. Autoimmunity 20, 231–236 (1995). polymerase chain reaction (RT-PCR). Thyroid 6, 97–106 OMIM: http://www.ncbi.nlm.nih.gov/Omim/
70. Myc, A., Arscott, P. L., Bretz, J. D., Thompson, N. W. & (1996). Graves’ disease | Hashimoto’s thyroiditis | multiple sclerosis |
Baker, J. R. Jr. Characterization of FAP-1 expression and 86. Roura-Mir, C. et al. Single-cell analysis of intrathyroidal myasthenia gravis | type 1 diabetes
function in thyroid follicular cells. Endocrinology 140, lymphocytes shows differential cytokine expression in
5431–5434 (1999). Hashimoto’s and Graves’ disease. Eur. J. Immunol. 27, FURTHER INFORMATION
A possible explanation for the refractoriness to CD95- 3290–3302 (1997). Encyclopedia of Life Sciences: http://www.els.net/
induced apoptosis that is observed in normal The first analysis of cytokine production at a single- autoimmune disease
thyrocytes. cell level in activated T cells that infiltrate the thyroid. Access to this interactive links box is free online.

204 | MARCH 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd

Potrebbero piacerti anche