Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

6
Contrasting Roles of IFN-c in Murine Models of Autoimmune Thyroid Diseases Yujiang Fang, 1, * Shiguang Yu, 1, * and Helen Braley-Mullen 1–3 Interferon-gamma (IFN-g), a prototypic proinflammatory cytokine produced by several different cell types, in- cluding the Th1 subset of CD4 + T cells, plays an important role in inflammation and autoimmune diseases. This review focuses on the varied and often contrasting roles of IFN-g in three murine models of autoimmune thyroid disease, experimentally induced autoimmune thyroiditis, the model of iodine-induced spontaneous autoimmune thyroiditis in NOD.H-2h4 mice and several different murine models of Graves’ disease. Introduction I nterferon-gamma (IFN-g) is a multifunctional cyto- kine that plays an important role in many autoimmune diseases, including thyroiditis. IFN-g is the prototypic Th1 cytokine produced by CD4 þ Th1 cells, and it is also produced by CD8 þ T cells and natural killer (NK) cells (1). IFN-g in- duces MHC class I and II on antigen presenting cells (APCs) and other cells, upregulates adhesion molecules as well as certain chemokines and chemokine receptors to recruit T cells to sites of inflammation, activates macrophages, and promotes IgG2A antibody production (1). Neutralization or genetic ablation of IFN-g or the IFN-g receptor (IFN-gR) can have both positive and negative effects on many autoim- mune diseases, including autoimmune thyroid diseases. In this brief review, we discuss the various and often con- trasting effects of neutralization, transgenic overexpression, or genetic ablation of IFN-g, IFN-gR, and interleukin (IL)- 12 in mouse models of autoimmune thyroid diseases. Autoimmune thyroid diseases encompass several condi- tions that have in common cellular and humoral immune responses targeted to the thyroid gland. The most common autoimmune thyroid diseases in humans are Graves’ disease (GD) and Hashimoto’s thyroiditis (2). While reviewing the role of IFN-g in autoimmune thyroid diseases, the primary focus will be on the models of experimental autoimmune thyroiditis (EAT) in mice and the iodine-induced model of spontaneous autoimmune thyroiditis (SAT) in NOD.H-2h4 mice. Although both animal models are often considered to represent animal models of Hashimoto’s thyroiditis, the effects of IFN-g ablation or neutralization on EAT and SAT are very different, suggesting basic differences in underlying mechanisms in these two animal models. Experimental Autoimmune Thyroiditis EAT is an organ-specific autoimmune disease inducible in genetically susceptible strains of mice by injection of mouse thyroglobulin (MTg) and adjuvant (active immunization) (2– 4), or by transfer of MTg-primed donor spleen cells activated with MTg in vitro (adoptive transfer model) (5,6). Thyroid lesions in both models of EAT are characterized by infiltra- tion of the thyroid by mononuclear cells, including CD4 þ and CD8 þ T cells, plasma cells, and macrophages (2–6). In- flammation tends to be long lasting (chronic), most mice produce anti-Tg antibodies, and they typically have normal serum thyroxine (T4) levels throughout the course of disease (3,5). CD4 þ T cells are the primary effector cells for both the active immunization and adoptive transfer EAT models (7,8). CD8 þ T cells reportedly have effector function in some active immunization models of EAT (3,9) but not in the adoptive transfer model studied in our laboratory (7,10). Granulomatous (G)-EAT A severe and histologically distinct granulomatous form of EAT (G-EAT) studied extensively in our laboratory is in- duced when cells from MTg-sensitized donors are activated in vitro with MTg and IL-12 or with MTg and anti-IL-2R monoclonal antibody (mAb) (7,11,12). Thyroid lesions in G-EAT, like those in EAT, are characterized by infiltration of the thyroid by CD4 þ and CD8 þ T cells, plasma cells, and macrophages. In the adoptive transfer model studied in our laboratory, inflammation in G-EAT is more severe than in EAT, and there are thyroid epithelial cell (TEC) proliferation, large numbers of histocytes, multinucleated giant cells, fibro- sis, and variable numbers of neutrophils, in addition to mono- nuclear infiltration (7,11,12). CD4 þ T cells are the primary Departments of 1 Internal Medicine and 2 Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri. 3 Department of Veterans Affairs, Harry S. Truman VA Medical Center, Columbia, Missouri. *Yujiang Fang and Shiguang Yu contributed equally to this review. THYROID Volume 17, Number 10, 2007 ª Mary Ann Liebert, Inc. DOI: 10.1089=thy.2007.0261 989 Thyroid 2007.17:989-994. Downloaded from online.liebertpub.com by Columbia Univ on 12/08/14. For personal use only.

Transcript of Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

Page 1: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

Contrasting Roles of IFN-c in Murine Modelsof Autoimmune Thyroid Diseases

Yujiang Fang,1,* Shiguang Yu,1,* and Helen Braley-Mullen1–3

Interferon-gamma (IFN-g), a prototypic proinflammatory cytokine produced by several different cell types, in-cluding the Th1 subset of CD4+ T cells, plays an important role in inflammation and autoimmune diseases. Thisreview focuses on the varied and often contrasting roles of IFN-g in three murine models of autoimmune thyroiddisease, experimentally induced autoimmune thyroiditis, the model of iodine-induced spontaneous autoimmunethyroiditis in NOD.H-2h4mice and several different murine models of Graves’ disease.

Introduction

Interferon-gamma (IFN-g) is a multifunctional cyto-kine that plays an important role in many autoimmune

diseases, including thyroiditis. IFN-g is the prototypic Th1cytokine produced by CD4þ Th1 cells, and it is also producedby CD8þ T cells and natural killer (NK) cells (1). IFN-g in-duces MHC class I and II on antigen presenting cells (APCs)and other cells, upregulates adhesion molecules as well ascertain chemokines and chemokine receptors to recruit Tcells to sites of inflammation, activates macrophages, andpromotes IgG2A antibody production (1). Neutralization orgenetic ablation of IFN-g or the IFN-g receptor (IFN-gR) canhave both positive and negative effects on many autoim-mune diseases, including autoimmune thyroid diseases. Inthis brief review, we discuss the various and often con-trasting effects of neutralization, transgenic overexpression,or genetic ablation of IFN-g, IFN-gR, and interleukin (IL)-12 in mouse models of autoimmune thyroid diseases.

Autoimmune thyroid diseases encompass several condi-tions that have in common cellular and humoral immuneresponses targeted to the thyroid gland. The most commonautoimmune thyroid diseases in humans are Graves’ disease(GD) and Hashimoto’s thyroiditis (2). While reviewing therole of IFN-g in autoimmune thyroid diseases, the primaryfocus will be on the models of experimental autoimmunethyroiditis (EAT) in mice and the iodine-induced model ofspontaneous autoimmune thyroiditis (SAT) in NOD.H-2h4mice. Although both animal models are often consideredto represent animal models of Hashimoto’s thyroiditis, theeffects of IFN-g ablation or neutralization on EAT and SATare very different, suggesting basic differences in underlyingmechanisms in these two animal models.

Experimental Autoimmune Thyroiditis

EAT is an organ-specific autoimmune disease inducible ingenetically susceptible strains of mice by injection of mousethyroglobulin (MTg) and adjuvant (active immunization) (2–4), or by transfer of MTg-primed donor spleen cells activatedwith MTg in vitro (adoptive transfer model) (5,6). Thyroidlesions in both models of EAT are characterized by infiltra-tion of the thyroid by mononuclear cells, including CD4þ

and CD8þ T cells, plasma cells, and macrophages (2–6). In-flammation tends to be long lasting (chronic), most miceproduce anti-Tg antibodies, and they typically have normalserum thyroxine (T4) levels throughout the course of disease(3,5). CD4þ T cells are the primary effector cells for both theactive immunization and adoptive transfer EAT models (7,8).CD8þ T cells reportedly have effector function in some activeimmunization models of EAT (3,9) but not in the adoptivetransfer model studied in our laboratory (7,10).

Granulomatous (G)-EAT

A severe and histologically distinct granulomatous formof EAT (G-EAT) studied extensively in our laboratory is in-duced when cells from MTg-sensitized donors are activatedin vitro with MTg and IL-12 or with MTg and anti-IL-2Rmonoclonal antibody (mAb) (7,11,12). Thyroid lesions inG-EAT, like those in EAT, are characterized by infiltration ofthe thyroid by CD4þ and CD8þ T cells, plasma cells, andmacrophages. In the adoptive transfer model studied in ourlaboratory, inflammation in G-EAT is more severe than inEAT, and there are thyroid epithelial cell (TEC) proliferation,large numbers of histocytes, multinucleated giant cells, fibro-sis, and variable numbers of neutrophils, in addition tomono-nuclear infiltration (7,11,12). CD4þ T cells are the primary

Departments of 1Internal Medicine and 2Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri.3Department of Veterans Affairs, Harry S. Truman VA Medical Center, Columbia, Missouri.*Yujiang Fang and Shiguang Yu contributed equally to this review.

THYROIDVolume 17, Number 10, 2007ª Mary Ann Liebert, Inc.DOI: 10.1089=thy.2007.0261

989

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.

Page 2: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

effector cells for G-EAT (7,12). CD8þ T cells are not requiredfor effector function in G-EAT, but function primarily topromote resolution of thyroid lesions (7,10). Thyroid in-flammation reaches maximal severity 19–21 days after celltransfer. Inflammation then spontaneously resolves if CD8þ

T cells are present, or when essentially all thyroid follicles aredestroyed, there is ongoing inflammation and developmentof fibrosis. Mice with these very severe lesions and fibrosisusually have low serum T4 (7,12–14). While reviewing therole of IFN-g in experimentally induced models of thyroiditisin mice, EAT and G-EAT will be considered together, andareas where there may be differences and=or where infor-mation is known for only one model will be mentioned.

Role of IFN-c in EAT and G-EAT

As an important Th1 cytokine (15), IFN-g plays an importantrole in immune and inflammatory responses, and it has beenshown to both promote and suppress development of EAT inmice. In some early studies, injection of IFN-g into thyroids ofCBA=J mice promoted development of EAT and productionof anti-MTg autoantibodies, and development of EAT wasinhibited by anti-IFN-g mAb (16–19). These results all suggestthat IFN-g plays an important role in thyroid destruction inEAT. However, this idea is challenged by other results show-ing that systemic administration of IFN-g suppresses EAT (20),and mice unable to respond to IFN-g (IFN-gR–=– mice) (21) orproduce IFN-g (IFN-g–=– mice) [(22) and our unpublished re-sults] are susceptible to EAT. In addition, interferon regulatoryfactor-1 (IRF-1) is an important regulator of IFN-g-mediatedimmune reactions, and IRF-1–=– NOD mice develop EAT andanti-MTg antibodies comparable to IRF-1þ=þ and IRF-1þ=–

mice (23). In the adoptive transfer model of G-EAT, neutrali-zation of IFN-g during in vitro activation of effector cells withMTg (no exogenous cytokines) promotes development ofmoresevere EAT with granulomatous histology (24), and spleno-cytes from IFN-g–=– mice activated with MTg and IL-12 trans-fer severe G-EAT to IFN-g–=– recipients (22).

The fact that IFN-gR–=–, IFN-g–=–, and IRF-1–=– mice candevelop EAT and that IFN-g–=– mice can develop severeG-EAT clearly indicates that IFN-g is not absolutely requiredfor induction of EAT. However, IFN-gR–=– mice have a milderdisease phenotype and thyroid lesions that resolve earlierthan in wild-type (WT) controls (21). In our studies, G-EATdiffers histologically in WT and IFN-g–=– mice (22), and in theabsence of IFN-g, there is earlier resolution of G-EAT and lessfibrosis (13,14). Specifically, thyroids of IFN-g–=– recipients ofIFN-g–=– effector cells have many eosinophils infiltrating thethyroids, they develop minimal fibrosis, and thyroid lesionsresolve considerably earlier than in WT mice (13,22). In con-trast, thyroids ofWT recipients ofWT effector cells havemanyneutrophils and almost no eosinophils, they develop exten-sive fibrosis, and inflammation is prolonged (13,22,25).Compared with WT mice, thyroids of IFN-g–=– recipients ofIFN-g–=– effector cells with severe G-EAT have lower mRNAexpression of Th1 cytokines and proinflammatory media-tors such as tumor necrosis factor-� (TNF-�) and induciblenitric oxide synthetase (iNOS), and higher expression of mostTh2 cytokines, particularly IL-10 (13,22). In WT mice, ex-pression of IFN-g and of other proinflammatory cytokinessuch as TNF-� and IL-17 is generally higher in thyroids withdelayed resolution and fibrosis, while Th2 cytokines such as

IL-10, IL-4, and IL-13 are generally higher in thyroids withearly resolution and less fibrosis (13,25). IFN-g can inducenecrosis or cooperate in the presence of TNF-� to inducetissue damage by apoptosis of epithelial cells (26). Increasedapoptosis of TEC and necrosis are evident in thyroids thatexpress high amounts of IFN-g and where inflammationprogresses to fibrosis (13,14), and IFN-g and other proin-flammatory cytokines can promote TEC apoptosis and moredestructive thyroiditis in mice (27,28). IFN-g also modulateschemokine production, including IFN-g-inducible protein 10(IP-10)=CXC chemokine ligand 10 (CXCL10), monokine in-duced by IFN-g (Mig)=CXCL9, and IFN-g-inducible T cell-� chemoattractant=CXCL11. These and other chemokines areimportant in promoting migration of lymphocytes to sites ofinflammation in thyroiditis and other autoimmune diseases(29–36), indicating that IFN-g plays an important role incontrolling leukocyte migration. Thus, IFN-g plays a criticalrole in regulating the inflammatory response in G-EAT,promoting chronic inflammation and fibrosis, and inhibitingresolution of thyroid lesions. The profibrotic function of IFN-g in G-EAT is in marked contrast to the antifibrotic functionof IFN-g in SAT, as discussed below, and is also distinctfrom other inflammatory processes where the primary pro-fibrotic cytokines are IL-13 and transforming growth factor-b (TGF-b) (37–39).

Transgenic Mice

Transgenic mice have also provided useful tools to inves-tigate the function of specific cytokines in EAT. Transgenicexpression of IFN-g on TEC of the EAT resistant C57Bl=6strain resulted in thyroid abnormalities, hypothyroidism,and limited CD4þ T cell infiltration (40). When the IFN-gtransgene was expressed on thyrocytes of SAT- and EAT-susceptible NOD.H-2h4 mice, there was increased expressionof MHC class II on thyrocytes, but the mice did not developspontaneous thyroiditis (41). After immunization with MTg,IFN-g-transgenic NOD.H-2h4 mice developed less severe dis-ease and reduced IgG1 responses compared toWT littermates(41). These studies are consistent with a disease-limiting roleof IFN-g in experimentally induced models of autoimmunethyroiditis, and suggest that local IFN-g activity in the thy-roid is sufficient for disease suppression (20). IL-12, a cyto-kine that promotes IFN-g production, was also expressed as atransgene on thyrocytes (42). IL-12 transgenic mice also de-veloped hypothyroidism. Although they developed onlymildthyroiditis spontaneously, immunization with MTg and ad-juvant induced more severe lymphocytic thyroiditis thanin similarly immunized WT littermates (42). The disease-promoting effect of IL-12 in transgenic mice was shown to beindependent of IFN-g (42). IL-12 also promotes developmentof very severe EAT in nontransgenic mice (12,43,44). Thepresence of IL-12 during in vitro activation of sensitizedsplenocytes promotes activation of cells that transfer verysevere G-EAT, and neutralization of IL-12 in vitro inhibitsactivation of effector cells for G-EAT (12). However, IL-12,like IFN-g, can have different effects on development of EATor G-EAT depending on when it is administered or neu-tralized. For example, cells from IL-12-deficient donors sen-sitized with MTg and adjuvant can be activated to transfersevere EAT and G-EAT, and IL-12 can inhibit EAT andG-EAT when administered in vivo (43,44). Some effects of

990 FANG ET AL.

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.

Page 3: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

IL-12 are independent of IFN-g; that is, splenocytes fromIFN-g–=– mice transfer severe G-EAT after activation withMTg and IL-12 in vitro (22). The effects of IL-12 deficiency onEAT and G-EAT are complicated by the fact that the IL-12–=–

mice used in the studies mentioned above were IL-12 p40–=–

mice, which are now known to be deficient in both IL-12and IL-23 (45). To our knowledge, IL-12 p35–=– mice have notbeen tested for their ability to develop EAT or G-EAT, andthe role of the IL-17-producing subset of CD4þ T cells [whichwould be compromised by IL-23 deficiency (46)] has not yetbeen addressed in murine models of EAT or G-EAT.

These apparently contradictory results of the effects ofIFN-g in EAT and G-EAT may reflect actual opposing effectsof IFN-g in the pathogenesis of EAT; that is, IFN-g may haveeffects that are both disease promoting and disease resolvingin EAT and G-EAT. This would be consistent with the knownopposing and often paradoxical functions of IFN-g in manymodels of inflammation and autoimmunity (47). IFN-g canupregulate expression of MHC class I and MHC class IImolecules on thyrocytes (1,18,48,49), providing targets forrecognition of TEC by CD4þ T cells and CD8þ T cells. IFN-gcan also be proapoptotic and antiproliferative, and can in-crease damage to the thyroid by inducing apoptosis ofthyrocytes or inhibit inflammation by inducing apoptosis ofinfiltrating inflammatory cells (14,26–28,50,51). The balanceof these opposing deleterious and beneficial effects of IFN-g(or IL-12 through its ability to promote IFN-g production)may direct the progression of inflammation in EAT andG-EAT, as well as in other autoimmune diseases.

Role of IFN-c in SAT

The model of SAT in NOD.H-2h4mice, like the EATmodels discussed above, has some similarities to Hashimoto’sthyroiditis in humans, with chronic infiltration of the thyroidby mononuclear cells and circulating autoantibody to thyro-globulin (52–55). When NOD.H-2h4mice are given 0.05%sodium iodide (NaI) in their drinking water, thyroid lesionsdevelop in nearly 100%of mice of both sexes in 8weeks (52–54). Previous work by our group and by others have shownthat both CD4þ and CD8þ T cells as well as B cells are re-quired for development of SAT (53,54,56–58). Both Th1 andTh2 cytokines are expressed in thyroids of NOD.H-2h4micewith SAT (53,59), and cells producing IL-12 and IFN-g aredetected in NOD.H-2h4 thyroids during the early phase ofSAT development (59). In contrast to the ability of IFN-g toboth promote and suppress EAT and G-EAT in mice, IFN-g isessential for development of typical lymphocytic (L)-SAT inNOD.H-2h4mice. IFN-g–=– NOD.H-2h4mice do not developL-SAT (60,61), and L-SAT develops only in mice with thy-rocytes able to respond to IFN-g; that is, IFN-gR–=– NOD.H-2h4mice also do not develop L-SAT (62). Thus the roleof IFN-g in SAT is very different from its role in EAT orG-EAT. Indeed, L-SAT in WT NOD.H-2h4mice is one ofrelatively few spontaneous or experimentally induced auto-immune diseases in mice that is absolutely dependent onIFN-g; that is, IFN-g–=– mice develop many other autoim-mune diseases, including scleroderma, diabetes, experi-mental allergic encephalomyelitis (EAE), collagen-inducedarthritis, experimental autoimmune uveitis, and systemiclupus erythematosus (SLE), in addition to EAT and G-EATas discussed above.

IFN-g has a dual role in SAT. It is absolutely required fordevelopment of classical L-SAT, but IFN-g suppresses devel-opment of another autoimmune lesion in NOD.H-2h4 mice,characterized by abnormal TEC hyperplasia=proliferationand fibrosis (60–62). Our laboratory developed IFN-g–=– andIFN-gR–=– NOD.H-2h4mice (60–62). Although neither straindevelops L-SAT, all IFN-g–=– and IFN-gR–=– mice given NaIin their drinking water develop abnormal proliferation andhyperplasia of thyroid epithelial cells (TEC H=P). About 60–70%of IFN-g–=– NOD.H-2h4 mice develop very severe TECH=P, in which almost the entire thyroid consists of largemasses of proliferating thyrocytes. All thyroids with severeTEC H=P also have some infiltrating cells, primarily T cells,macrophages, and some eosinophils (61). The infiltratinglymphocytes are presumably required for development ofsevere TEC H=P because IFN-g–=– NOD.H-2h4 SCID mice donot develop TEC H=P (61). Although thyroids of IFN-g–=–

NOD.H-2h4mice with TEC H=P have few infiltrating B cellsor plasma cells (both of which are prevalent in thyroids ofWT mice with L-SAT), all mice with TEC H=P produce anti-Tg autoantibodies. All NOD.H-2h4mice with severe TECH=P have low serum T4, and there is extensive deposition ofcollagen (fibrosis) surrounding the clusters of proliferatingthyrocytes (61). Splenocytes from IFN-g–=– mice with severeTEC H=P transfer severe TEC H=P with accelerated kineticsto IFN-g–=– NOD.H-2h4 SCID mice, suggesting that TEC H=Phas an autoimmune basis (61). Therefore, a single cytokine,IFN-g, is absolutely essential for development of L-SAT andto inhibit thyrocyte hyperplasia in WT NOD.H-2h4mice. Inthe absence of IFN-g, mice are resistant to L-SAT but theydevelop TEC H=P, which also has an autoimmune basis.

Adoptive transfer of WT splenocytes (as a source of IFN-g)to IFN-g–=– mice results in typical L-SAT, and TEC H=P issuppressed (60,62). In IFN-g–=– mice given WT splenocytes,both donor (WT) and recipient (IFN-g–=–) T cells are present inthe thyroid infiltrates (62), indicating that cells from IFN-g–=–

mice are able to migrate to the thyroid if IFN-g is provided.Since cells in IFN-gR–=– mice cannot respond to IFN-g,IFN-gR–=– mice were used to distinguish the effects of IFN-gon lymphocytes versus thyrocytes. Transfer of WT spleno-cytes or bone marrow as a source of IFN-g to IFN-gR–=–

NOD.H-2h4mice did not result in L-SAT or inhibit TEC H=P,although the same pool of WT splenocytes or bone marrowinduced L-SAT and inhibited TEC H=P in IFN-g–=– recipients(62). These results indicate that thyrocytes must be able torespond to IFN-g for development of L-SAT and inhibition ofTEC H=P. Unexpectedly, IFN-gR–=– splenocytes do not in-duce L-SAT in IFN-g–=– mice even though IFN-gR–=– lym-phocytes produce as much IFN-g as lymphocytes from WTdonors. Further studies indicated that recipients of IFN-gR–=–

splenocytes or bone marrow express less mRNA for IFN-g-inducible chemokines compared to recipients of WT cells(62), and upregulation of IFN-g-inducible chemokines maybe required to induce optimal migration of lymphocytes tothyroids (29–33). These results suggest that lymphocytesmust be able to respond to IFN-g to be induced to migrate tothe thyroid in sufficient numbers to result in L-SAT, andthyrocytes must express IFN-gR for SAT to develop.

Thyrocytes responding to IFN-g produced locally bythyroid-infiltrating inflammatory cells upregulate MHC classII molecules (41,48,49), and locally produced IFN-g may alsofunction to induce expression of chemokines or adhesion

IFN-c IN AUTOIMMUNE THYROID DISEASES 991

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.

Page 4: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

molecules on thyrocytes that are needed for development ofL-SAT. In support of this idea, it has been shown that iodineand IFN-g act synergistically to increase ICAM-1 expressionon NOD.H-2h4 thyrocytes (63,64).

Role of IFN-c in Thyroid Fibrosis

Another example of the opposing functions of IFN-g inautoimmune thyroid diseases in mice concerns its role in fi-brosis in G-EAT versus SAT. As mentioned above, mice withvery severe G-EAT have extensive collagen deposition (fibro-sis) in their thyroids (13,14,25), and thyroids of IFN-g–=–

NOD.H-2h4mice with severe TEC H=P also have extensivefibrosis (61). In G-EAT, thyroid fibrosis occurs as a result of anexcessive proinflammatory response, in which there is a pre-ponderance of proinflammatory cytokines (including IFN-g),overproduction of the profibrotic cytokine TGF-b, and de-creased production of IL-10 in the thyroid (13,14,25,65). Fi-brosis in G-EAT is associated with increased apoptosis ofthyrocytes, and apoptosis of inflammatory cells is decreased(13,14). Neutralization of TGF-b inhibits fibrosis and promotesresolution of G-EAT (65). Although IFN-g–=– DBA=1 mice alsodevelop severe G-EAT, they have less fibrosis and increasedapoptosis of inflammatory cells resulting in earlier resolutionof lesions in comparison to their WT littermates (13,14). Thus,IFN-g tends to be profibrotic in G-EAT since fibrosis is in-creasedwhen IFN-g is highly expressed and is decreasedwhenIFN-g is absent. In the NOD.H-2h4 model, fibrosis is inhibitedby IFN-g, since WT NOD.H-2h4 mice that produce IFN-g anddevelop L-SAT never develop thyroid fibrosis, while NOD.H-2h4mice that develop severe TEC H=P due to lack of IFN-gor IFN-gR have extensive fibrosis (60–62). In addition, whenWT splenocytes (as a source of IFN-g) are transferred to IFN-g–=– NOD.H-2h4mice, fibrosis is inhibited (61). Fibrosis in IFN-g–=– NOD.H-2h4mice with TEC H=P is due, at least in part, tooverproduction of the profibrotic cytokine TGF-bby prolifer-ating thyrocytes. Transgenic mice expressing TGF-bon TECdevelop severe thyrocyte hyperplasia and extensive fibrosis,and neutralization of TGF-b inhibits development of severeTEC H=P and fibrosis in SCID recipients of IFN-g–=– spleno-cytes (Yu et al., manuscript in preparation). Therefore, IFN-g is primarily antifibrotic in SAT, as has also been observed inseveral other experimental models (37–39).

Role of IFN-c in Mouse Models of GD

Autoimmune GD, the most common endocrine disorderin humans, is mediated by autoantibodies that bind to thethyrotropin receptor (TSHR) and stimulate thyroid hormoneproduction (66). GD in humans is considered to be primarilya Th2-type disease with production of Th2 cytokines such asIL-4 and IL-10 generally predominating over Th1 cytokinessuch as IFN-g (67). There are a number of studies concerningthe role of Th1 versus Th2 cytokines in various murinemodels of GD. Although some studies suggest a role for bothIFN-g and IL-4 in some murine models of GD (68–70),knockout of IFN-g generally does not prevent developmentof GD, whereas knockout of IL-4 does inhibit disease de-velopment (69,71). Similarly, knockout of the transcriptionfactor STAT-6, which is needed for production of IL-4, in-hibited development of GD, whereas knockout of STAT-4,which is needed for production of IFN-g, resulted in a higherincidence and greater severity of hyperthyroidism (72).

However, IFN-g was shown to play an important role in amouse model of GD induced by TSHR antigen immuniza-tion (73). It is not clear which of the various animal models ofGD most closely mimics human GD, and multiple animalmodels must be analyzed to gain an insight into the patho-genesis of human GD (66).

Summary

Studies from murine models of autoimmune thyroiditisindicate that IFN-g is critical for the development of SAT inNOD.H-2h4mice, but it is not absolutely required for de-velopment of EAT, G-EAT, or GD in mice. In EAT, G-EAT,and GD, IFN-g can have augmenting or suppressive effectson disease development depending on various factors suchas the amount, site of expression, and the presence or absenceof IFN-g at various stages of the autoimmune inflammatoryresponse. The diverse and often opposing roles of IFN-gin autoimmune thyroid diseases are not unexpected, sinceparadoxical roles of IFN-g in other inflammatory and auto-immune diseases have been recognized by others (47). UsingIFN-g as an example of only one of many cytokines involvedin regulation of autoimmune diseases suggests that it is im-portant to exercise caution in making generalizations withregard to the role played by one specific cytokine in a givenautoimmune disease.

Acknowledgments

The authors thank Gordon Sharp for his helpful insights ininterpretation of studies from our laboratory cited in thisreview. The work from our laboratory was supported by theNIH Grant DK 35527 and by a VA Merit Review Grant.

References

1. Boehm U, Klamp T, Groot M, Howard J 1997 Cellular re-sponses to interferon-g. Annu Rev Immunol 15:749–795.

2. Lira SA, Martin AP, Marinkovic T, Furtado GC 2005 Me-chanisms regulating lymphocytic infiltration of the thyroid inmurine models of thyroiditis. Crit Rev Immunol 25:251–262.

3. Charreire J 1989 Immune mechanisms in autoimmune thy-roiditis. Adv Immunol 46:263–334.

4. Stafford EA, Rose NR 2000 Newer insights into the patho-genesis of experimental autoimmune thyroiditis. Int Rev Im-munol 19:501–533.

5. Braley-Mullen H, Johnson M, Sharp GC, Kyriakos M 1985 In-duction of experimental autoimmune thyroiditis in mice within vitro activated splenic T cells. Cell Immunol 93:132–143.

6. Conaway DH, Giraldo AA, David CS, Kong YC 1990 In situanalysis of T cell subset composition in experimental auto-immune thyroiditis after adoptive transfer of activated spleencells. Cell Immunol 125:247–253.

7. Braley-Mullen H, Sharp GC 2000 Adoptive transfer murinemodel of granulomatous experimental autoimmune thyroid-itis. Int Rev Immunol 19:535–555.

8. Stull SJ, Kyriakos M, Sharp GC, Braley-Mullen H 1988 Pre-vention and reversal of experimental autoimmune thyroid-itis (EAT) in mice by administration of anti-L3T4 antibody atdifferent stages of disease development. Cell Immunol 117:188–198.

9. Kong YM, Waldmann H, Cobbold S, Giraldo AA, Fuller BE,Simon LL 1989 Pathogenic mechanisms in murine autoim-mune thyroiditis: short- and long-term effects of in vivo deple-tion of CD4þ and CD8þ T cells. Clin Exp Immunol 77:428–433.

992 FANG ET AL.

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.

Page 5: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

10. Braley-Mullen H, McMurray RW, Sharp GC, Kyriakos M1994 Regulation of the induction and resolution of granu-lomatous experimental autoimmune thyroiditis in mice byCD8þ T cells. Cell Immunol 153:492–504.

11. Braley-Mullen H, Sharp GC, Bickel JT, Kyriakos M 1991Induction of severe granulomatous experimental autoim-mune thyroiditis in mice by effector cells activated in thepresence of anti-interleukin 2 receptor antibody. J Exp Med173:899–912.

12. Braley-Mullen H, Sharp GC, Tang H, Chen K, Kyriakos M,Bickel JT 1998 Interleukin-12 promotes activation of effectorcells that induce a severe destructive granulomatous form ofmurine experimental autoimmune thyroiditis. Am J Pathol152:1347–1358.

13. Chen K, Wei Y, Sharp GC, Braley-Mullen H 2003 Mechan-isms of spontaneous resolution versus fibrosis in granulo-matous experimental autoimmune thyroiditis. J Immunol171:6236–6243.

14. Chen K, Wei Y, Sharp GC, Braley-Mullen H 2005 Balance ofproliferation and cell death between thyrocytes and myo-fibroblasts regulates thyroid fibrosis in granulomatous ex-perimental thyroiditis (G-EAT). J Leukoc Biol 77:166–172.

15. Seder RA, Paul WE 1994 Acquisition of lymphokine-producing phenotype by CD4þ T cells. Annu Rev Immunol12:635–673.

16. Tang H, Mignon-Godferoy K, Meroni PL, Garotta G, Char-reire J, Nicoletti F 1993 The effects of a monoclonal antibodyto interferon-g on experimental autoimmune thyroiditis(EAT): prevention of disease and decrease of EAT-specificT cells. Eur J Immunol 23:275–278.

17. Remy JJ, Salamero J, Michel-Bechet M, Charreire J 1987 Ex-perimental autoimmune thyroiditis induced by recombinantinterferon-g. Immunol Today 8:73.

18. Frohman M, Francfort JW, Cowing C 1991 T-dependentdestruction of thyroid isografts exposed to IFNg. J Immunol146:2227–2234.

19. Kawakami Y, Kuzuya N, Watanabe T, Uchiyama Y, Yama-shita K 1990 Induction of experimental thyroiditis in mice byrecombinant interferon gamma administration. Acta Endo-crinol 122:41–48.

20. Vladutiu AO, Sulkowsky EJ 1980 Inhibition of murine auto-immune thyroiditis by interferon. Biomedicine 33:173–174.

21. Alimi E, Huang S, Brazillet MP, Charreire J 1998 Experi-mental autoimmune thyroiditis (EAT) in mice lacking theIFN-g receptor gene. Eur J Immunol 28:201–208.

22. Tang H, Sharp GC, Peterson KP, Braley-Mullen H 1998 IFN-g-deficient mice develop severe granulomatous experimentalautoimmune thyroiditis with eosinophil infiltration in thy-roids. J Immunol 160:5105–5112.

23. Jin Z, Mori K, Fujimori K, Hoshikawa S, Tani J, Satoh J, Ito S,Satomi S, Yoshida K 2004 Experimental autoimmune thy-roiditis in nonobese diabetic mice lacking interferon regu-latory factor-1. Clin Immunol 113:187–192.

24. Stull SJ, Sharp GC, Kyriakos M, Bickel JT, Braley-Mullen H1992 Induction of granulomatous experimental autoimmunethyroiditis in mice with in vitro activated effector T cells andanti-IFN-g antibody. J Immunol 149:2219–2226.

25. Fang Y, Wei Y, DeMarco V, Chen K, Sharp GC, Braley-Mullen H 2007 Murine FLIP transgene expressed on thyroidepithelial cells promotes resolution of granulomatous experi-mental autoimmune thyroiditis. Am J Pathol 170:875–887.

26. Suk K, Chang I, Kim YH, Kim S, Kim JY, Kim H, Lee MS2001 Interferon g and tumor necrosis factor � synergism inME-180 cervical cancer cell apoptosis and necrosis: IFNg

inhibits cytoprotective NF-kB through STAT1=IRF-1 path-ways. J Biol Chem 276:13153–13159.

27. Wang SH, Bretz JD, Phelps E, Mezosi E, Arscott PL, Utsugi S,Baker JR Jr. 2002 A unique combination of inflammatory cy-tokines enhances apoptosis of thyroid follicular cells andtransforms nondestructive to destructive thyroiditis in exper-imental autoimmune thyroiditis. J Immunol 168:2470–2474.

28. Bretz JD, Arscott PL, Myc A, Baker JR Jr. 1999 Inflammatorycytokine regulation of Fas-mediated apoptosis in thyroidfollicular cells. J Biol Chem 274:25433–25438.

29. Tran EH, Prince EN, Owens, T 2000 IFN-g shapes immuneinvasion of the central nervous system via regulation of che-mokines. J Immunol 164:2759–2768.

30. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, LusterAD 2002 IFNg-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell gen-eration and trafficking. J Immunol 168:3195–3204.

31. Savinov AY, Wong FS, Chernovsky AV 2001 IFNg directshoming of diabetogenic T cells. J Immunol 167:6637–6643.

32. Rotondi M, Lazzeri E, Romagnani P, Serio M 2003 Role forinterferon-g inducible chemokines in endocrine autoimmu-nity: an expanding field. J Endocrinol Invest 26:177–180.

33. MartinAP,Coronel EC, SanoG,ChenS,VassilevaG,Canasto-Chibuque C, Sedgwick JD, Frenette PS, Lipp M, Furtado GC,Lira SA 2002 A novel model for lymphocyte infiltration of thethyroid gland generated by transgenic expression of the CCchemokine CCL21. J Immunol 173:4791–4798.

34. Kimura H, Kimura M, Rose NR, Caturegli P 2004 Earlychemokine expression induced by interferon-g in a murinemodel of Hashimoto’s thyroiditis. Exp Mol Pathol 77:161–167.

35. Chen K, Wei Y, Alter A, Sharp GC, Braley-Mullen H 2005Chemokine expression during development of fibrosis ver-sus resolution in a murine model of granulomatous experi-mental autoimmune thyroiditis. J Leukoc Biol 78:716–724.

36. Gouivestre C, Batteux F, Charreire J 2002 Chemokinesmodulate experimental autoimmune thyroiditis through at-traction of autoreactive or regulatory T cells. Eur J Immunol32:3435–3442.

37. Kim JH, Kim HY, Kim S, Chung JH, Park WS, Chung DH2005 Natural killer T (NKT) cells attenuate bleomycin-induced pulmonary fibrosis by producing interferon-gamma.Am J Pathol 167:1231–1241.

38. Oldroyd SD, Thomas GL, Gabbiani G, El Nahas AM 1999Interferon-g inhibits experimental renal fibrosis. Kidney Int56:2116–2121.

39. Kodera T, McGaha T, Phelps R, Paul WE, Bona C 2002Disrupting the IL-4 gene rescues mice homozygous for thetight-skin mutation from embryonic death and diminishesTGF-bproduction by fibroblasts. Proc Natl Acad Sci USA99:3800–3804.

40. Caturegli P, Hejazi M, Suzuki K, Dohan O, Carrasco N,Kohn LD, Rose NR 2000 Hypothyroidism in transgenic miceexpressing IFN-gamma in the thyroid. Proc Natl Acad SciUSA 97:1719–1724.

41. Barin JG, Afanasyeva M, Talor MV, Rose NR, Burek CL,Caturegli P 2003 Thyroid-specific expression of IFN-g limitsexperimental autoimmune thyroiditis by suppressing lym-phocyte activation in cervical lymph nodes. J Immunol 170:5523–5529.

42. Kimura H, Tzou SC, Rocchi R, Kimura M, Suzuki K, ParlowAF, Rose NR, Caturegli P 2005 Interleukin (IL)-12–drivenprimary hypothyroidism: the contrasting roles of two Th1cytokines (IL-12 and IFN-g). Endocrinology 146:3642–3651.

IFN-c IN AUTOIMMUNE THYROID DISEASES 993

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.

Page 6: Contrasting Roles of IFN-γ in Murine Models of Autoimmune Thyroid Diseases

43. Chen K, Wei Y, Sharp GC, Braley-Mullen H 2001 Inductionof experimental autoimmune thyroiditis in IL-12–=–mice.J Immuno1 167:1720–1727.

44. Zaccone P, Fehervari Z, Cooke A 2003 Tumour necrosisfactor-� is a fundamental cytokine in autoimmune thyroiddisease induced by thyroglobulin and lipopolyssacharide ininterleukin-12 p40 deficient C57BL=6 mice. Immunology108:50–54.

45. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, SeymourB, Lucian L, To W, Kwan S, Churakova T, Zurawski S,Wiekowski M, Lira SA, Gorman D, Kastelein RA, SedgwickJD 2003 Interleukin-23 rather than interleukin-12 is the crit-ical cytokine for autoimmune inflammation of the brain.Nature 421:744–748.

46. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH,Wang Y, Hood L, Zhu Z, Tian Q, Dong C 2005 A distinctlineage of CD4 T cells regulates tissue inflammation byproducing interleukin-17. Nat Immunol 6:1133–1141.

47. Zhang J 2007 Yin and yang interplay of IFN-g ininflammation and autoimmune disease. J Clin Invest 117:

871–873.48. Kimura H, Kimura M, Tzou S-C, Chen Y-C, Suzuki K, Rose

NR, Caturegli P 2005 Expression of class II major histocom-patibility complex molecules on thyrocytes does not causespontaneous thyroiditis but mildly increases its severity afterimmunization. Endocrinology 146:1154–1162.

49. Hamilton FM, Black M, Farquharson MA, Stewart C,Foulis AK 1991 Spatial correlation between thyroid epithe-lial cells expressing class II MHC molecules and interferon-g-containing lymphocytes in human thyroid autoimmune dis-ease. Clin Exp Immunol 83:64–68.

50. Dalton DK, Haynes I, Chu CQ, Swain SL, Wittmer S 2000Interferon-g eliminates responding CD4 T cells during myco-bacterial infection by inducing apoptosis of activated CD4T cells. J Exp Med 192:117–122.

51. Refaeli Y, VanParijs L, Alexander SL, Abbas AK 2002Interferon-g is required for activation-induced death ofT lymphocytes. J Exp Med 196:999–1005.

52. Rasooly L, Burek CL, Rose NR 1996 Iodine-induced auto-immune thyroiditis in NOD-H-2h4 mice. Clin ImmunolImmunopathol 81:287–292.

53. Braley-Mullen H, Sharp GC, Medling B, Tang H 1999Spontaneous autoimmune thyroiditis in NOD.H-2h4 mice.J Autoimmun 12:157–165.

54. Hutchings PR, Verma S, Phillips JM, Harach SZ, Howlett S,Cooke A 1999 Both CD4þ and CD8þ T cells are required foriodine accelerated thyroiditis in NOD mice. Cell Immunol192:113–121.

55. Rose NR, Bonita R, Burek CL 2002 Iodine: an environmentaltrigger of thyroiditis. Autoimmun Rev 1:97–103.

56. Verma S, Hutchings P, Guo J, McLachlan S, Rapoport B,Cooke A 2000 Role of MHC class I expression and CD8þ

T cells in the evolution of iodine-induced thyroiditis inNOD.H-2h4 mice. Eur J Immunol 30:1191–1202.

57. Yu S, Medling B, Yagita H, Braley-Mullen H 2001 Char-acteristics of inflammatory cells in spontaneous autoimmunethyroiditis in NOD.H-2h4 mice. J Autoimmun 16:37–46.

58. Braley-Mullen H, Yu S 2000 Early requirement for B cells fordevelopment of spontaneous autoimmune thyroiditis inNOD.H-2h4 mice. J Immunol 165:7262–7269.

59. Bonita RE, Rose NR, Rasooly L, Caturegli P, Burek CL 2003Kinetics of mononuclear cell infiltration and cytokine ex-pression in iodine-induced thyroiditis in the NOD-H2h4mice. Exp Mol Pathol 74:1–12.

60. Yu S, Sharp GC, Braley-Mullen H 2002 Dual roles for IFN-g,but not for IL-4, in spontaneous autoimmune thyroiditis inNOD.H-2h4 mice. J Immunol 169:3999–4007.

61. Yu S, Sharp GC, Braley-Mullen H 2006 Thyroid epithelialcell hyperplasia in IFN-g deficient NOD.H-2h4 mice. ClinImmunol 118:92–100.

62. Yu S, Sharp GC, Braley-Mullen H 2006 Thyrocytes respond-ing to IFN-g are essential for development of lymphocyticspontaneous autoimmune thyroiditis and for inhibition ofthyrocyte hyperplasia. J Immunol 176:1259–1265.

63. Sharma RB, Alegria JD, Talor MV, Rose NR, Caturegli P,Burek CL 2005 Iodine and IFN-g synergistically enhanceintercellular adhesion molecule 1 expression on NOD.H2h4mouse thyrocytes. J Immunol 174:7740–7745.

64. Bonita RE, Rose NR, Rasooly L, Caturegli P, Burek CL 2002Adhesion molecules as susceptibility factors in spontaneousautoimmune thyroiditis in the NOD.H-2h4 mouse. Exp MolPathol 73:155–163.

65. Chen K, Wei Y, Sharp GC, Braley-Mullen H 2002 Inhibitionof TGFb1 by anti-TGFb1 antibody or lisinopril reduced thy-roid fibrosis in granulomatous experimental autoimmunethyroiditis. J Immunol 169:6530–6538.

66. McLachlan SM, Nagayama Y, Rapoport B 2005 Insight intoGraves’ hyperthyroidism from animal models. EndocrineRev 26:800–832.

67. Stassi G, DeMaria R 2002 Autoimmune thyroid disease: newmodels of cell death in autoimmunity. Nat Rev Immunol 2:195–204.

68. Yan XM, Guo J, Pichurin P, Tanaka K, Jaume JC, Rapoport B,McLachlan SM 2000 Cytokines, IgG subclasses and costimu-lation in a mouse model of thyroid autoimmunity inducedby injection of fibroblasts co-expressing MHC class II andthyroid autoantigens. Clin Exp Immunol 122:170–179.

69. Nagayama Y, Saitoh O, McLachlan SM, Rapoport B, KanoH, Kumazawa Y 2004 TSH receptor-adenovirus inducedGraves’ hyperthyroidism is attenuated in both interferon-gamma and interleukin-4 knockout mice: implications forthe Th1=Th2 paradigm. Clin Exp Immunol 138:417–422.

70. Nagayama Y, Mizuguchi H, Hawakawa T, Niwa M, Mc-Lachlan SM, Rapoport B 2003 Prevention of autoantibody-mediated Graves’-like hyperthyroidism in mice with IL-4, aTh2 cytokine. J Immunol 170:3522–3527.

71. Dogan RN, Vasu C, Holterman MJ, Prabhakar BS 2003 Ab-sence of IL-4, and not suppression of the Th2 response, pre-vents development of experimental autoimmune Graves’disease. J Immunol 170:2195–2204.

72. Land KJ, Gudapati P, Kaplan MH, Seetharamaiah GS 2006Differential requirement of signal transducer and activatorof transcription-4 (Stat4) and Stat6 in a thyrotropin receptor-289-adenovirus-induced model of Graves’ hyperthyroidism.Endocrinology 147:111–119.

73. Pichurin P, Yan XM, Farilla L, Guo J, Chazenbalk GD, Ra-poport B, McLachlan SM 2001 Naked thyrotropin receptorDNA vaccination: a Th1 response in which interferon-gproduction, rather than antibody, dominates the immuneresponse in mice. Endocrinology 142:3530–3536.

Address reprint requests to:Helen Braley-Mullen

Division of ImmunologyDepartment of MedicineUniversity of MissouriColumbia, MO 65212

E-mail: [email protected]

994 FANG ET AL.

Thy

roid

200

7.17

:989

-994

.D

ownl

oade

d fr

om o

nlin

e.lie

bert

pub.

com

by

Col

umbi

a U

niv

on 1

2/08

/14.

For

per

sona

l use

onl

y.