Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

24
Immunologic ReseawGh Immunol Res 1996; 15:323-346 V.K. Singh a K. Nagaraju b a Department of Immunology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, India; b Connective Tissue Disease Section, Arthritis and Rheumatism Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md., USA Experimental Autoimmune Uveitis: Molecular Mimicry and Oral Tolerance ~ l * i * * * ~ 1 7 6 1 7 6 ~ = * * * ~ 1 7 6 1 7 6 1 7 6 1 7 6 Key Words Uveitis, experimental autoimmune Molecular mimicry Oral tolerance Autoantigens . ~ ~ Q.~ o.~176 ~ 1 7 6 1 7 6 ~ D*** ~ 1 7 6 ~ ~ ~ 1 7 6 Abstract Intraocular inflammatory disease or uveitis, which affects the uveal tract and the retina of the eyes in human, is the major cause of visual impairment. Experimental autoimmune uvei- tis (EAU) is a T-cell-mediated autoimmune disease directed against retinal proteins and has been studied in several mam- malian species including subhuman primates as a model for human posterior uveitis. Autoimmune responses provoked by molecular mimicry occur when the nonself and host determi- nants are similar enough to cross-react yet different enough to break immunological tolerance, and is one of the proposed mechanisms for induction of autoimmune diseases. Thera- peutic immunomodulatory strategies have been used to in- duce antigen-specific peripheral immune tolerance in animal models of T-cell-mediated autoimmune diseases by oral ad- ministration of autoantigens. Oral tolerance leads to unique mechanisms of tissue and disease-specific immunosuppres- sion, which would circumvent the immunotherapeutic prob- lem of multiple target tissue autoreactivity. Several groups have investigated the effects of delivering autoantigens across gastric mucosal surfaces. This review briefly discusses molecu- lar mimicry and the mechanism of induction of oral tolerance with respect to immunopathogenesis of T-cell-mediated au- toimmune diseases in general and EAU in particular. KARGER E-Mail karger@karger,ch Fax+41 61 306 12 34 http://www.karger.cb 1996 S. Karger AG, Basel Dr. V.K. Singh Building 6, Room 310, MSC 2740 LRCMB, NE1, NIH Bethesda, MD 20892 (USA)

Transcript of Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Page 1: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Immuno log ic R e s e a w G h Immunol Res 1996; 15:323-346

V.K. Singh a K. Nagaraju b

a Department of Immunology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, India;

b Connective Tissue Disease Section, Arthritis and Rheumatism Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md., USA

Experimental Autoimmune Uveitis: Molecular Mimicry and Oral Tolerance

~ l * i * * * ~ 1 7 6 1 7 6 ~ = * * * ~ 1 7 6 1 7 6 1 7 6 1 7 6

Key Words Uveitis, experimental

autoimmune Molecular mimicry Oral tolerance Autoantigens

. ~ ~ Q . ~ o . ~ 1 7 6 ~ 1 7 6 1 7 6 ~ D * * * ~ 1 7 6 ~ ~ ~ 1 7 6

Abstract Intraocular inflammatory disease or uveitis, which affects the uveal tract and the retina of the eyes in human, is the major cause of visual impairment. Experimental autoimmune uvei- tis (EAU) is a T-cell-mediated autoimmune disease directed against retinal proteins and has been studied in several mam- malian species including subhuman primates as a model for human posterior uveitis. Autoimmune responses provoked by molecular mimicry occur when the nonself and host determi- nants are similar enough to cross-react yet different enough to break immunological tolerance, and is one of the proposed mechanisms for induction of autoimmune diseases. Thera- peutic immunomodulatory strategies have been used to in- duce antigen-specific peripheral immune tolerance in animal models of T-cell-mediated autoimmune diseases by oral ad- ministration of autoantigens. Oral tolerance leads to unique mechanisms of tissue and disease-specific immunosuppres- sion, which would circumvent the immunotherapeutic prob- lem of multiple target tissue autoreactivity. Several groups have investigated the effects of delivering autoantigens across gastric mucosal surfaces. This review briefly discusses molecu- lar mimicry and the mechanism of induction of oral tolerance with respect to immunopathogenesis of T-cell-mediated au- toimmune diseases in general and EAU in particular.

KARGER E-Mail karger@karger,ch Fax+41 61 306 12 34 http://www.karger.cb

�9 1996 S. Karger AG, Basel Dr. V.K. Singh Building 6, Room 310, MSC 2740 LRCMB, NE1, NIH Bethesda, MD 20892 (USA)

Page 2: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Introduction

The ocular disease developing in suscepti- ble strains of animals immunized with S anti- gen or other retinal or uveal antigens has been termed experimental autoimmune uveitis (or uveoretinitis; EAU) [1]. The S antigen prepa- rations used in most studies were obtained from bovine or guinea pig retinas and both were found to be similarly potent. The capaci- ty of S antigen to induce EAU depends on the inclusion of adjuvants in the immunization; no disease is induced in animals injected with S antigen without proper adjuvant [2]. Com- plete Freund's adjuvant (CFA), which is the adjuvant used by most investigators for in- duction of EAU, is a mixture of mineral oil with mycobacteria. However, later studies have shown that the induction of EAU is fur- ther enhanced by a simultaneous injection of the animals with Bordetella pertussis bacteria or a purified preparation of a single compo- nent of these bacteria, called pertussigen or pertussis toxin [3]. The clinical as well as his- topathological features of EAU in animals closely resemble certain uveitic conditions in humans and are considered a model for these diseases [ 1, 4, 5]. The possible role of S anti- gen in the etiopathogenesis of human uveitis is further supported by the fact that many patients with intermediate or posterior uveitis have immune responses to the human and bovine sequences [6-10].

Molecular mimicry is a process in which the presence of epitopes (either linear or con- formational) is shared between a foreign and a host protein. When the shared determinants are identical, the host shows tolerance to the foreign epitope by recognizing it as self. Ho- mologous but nonidentical determinants dif- fering in one or more amino acids may be for- eign enough to elicit an immune response. The immune response initiated against these foreign epitopes may, however, react with the

closely homologous 'self" host protein. Hu- moral and cell-mediated immune responses generated against this antigen might cross- react with a self epitope, thereby causing cel- lular injury leading to disease. Once the for- eign agent initiates these processes, it need not to be present during the autoimmune destruc- tion that follows [11, 12]. Molecular mimicry has been suggested as one of the mechanisms for induction of autoimmune diseases.

South American Indians have been re- ported to have ingested the leaves from poi- son ivy plants in order to suppress a systemic delayed hypersensitivity response that is in- duced following contact with this plant [13]. Almost 50 years ago it was first noticed that experimental drug allergies could be sup- pressed by prior feeding of the sensitizing agent [14]. In the recent past it was demon- strated that the oral administration of specific antigen could attenuate disease activity in an- imal models of various autoimmune diseases such as multiple sclerosis, uveitis, rheumatoid arthritis, thyroiditis, myasthenia gravis, and insulin-dependent diabetes mellitus (IDDM) [13, 15-17]. Initial human trials in multiple sclerosis, rheumatoid arthritis, and uveitis have shown promising results [18-20]. The exact cellular and molecular mechanisms in- volved in the generation of antigen-specific unresponsiveness in adults are not well under- stood. Therefore, it is important to under- stand the anatomy and physiology of the gut- associated lymphoid tissue, and the immuno- pathogenesis of T-cell-mediated autoimmune diseases.

Generally, autoimmune diseases charac- teristically exhibit intercalating exacerbations and remissions or complete recovery indicat- ing that disease-associated symptoms may re- solve through the activity of the immune sys- tem. This suggests that autoimmune diseases may be corrected by using natural immuno- logical agents that participate in the selection

324 Immunol Res 1996;15:323-346 Singh/Nagaraju

Page 3: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

o f the l y m p h o c y t e r epe r to i r e u n d e r phys i -

o logical cond i t i ons . One o f the mos t at-

t r ac t ive a reas o f ach iev ing i m m u n e res tora -

t ion th rough b io logica l m e a n s is by i n d u c i n g

i m m u n o l o g i c a l to le rance wi th au toan t i gens

t h rough gut. I t is a d v a n t a g e o u s because this

m o d e o f t h e r a p y arres ts i m m u n e d a m a g e by

a u t o r e a c t i v e cells w i thou t ove r i m m u n o s u p -

p ress ion a n d c o n c o m i t a n t s ide effects l ike in-

fec t ion a n d t u m o r i g e n i c i t y o b s e r v e d wi th im-

m u n o s u p p r e s s i v e drugs.

Uveitopathogenic Autoantigens in Retina

S antigen, also known as arrestin, is a 48-kDa pro- tein and is the most potent retinal antigen which ini- tiates autoimmune inflammatory reactions in the eye. It is specifically localized in visual tissues and is remarkably conserved through the phylogenetic devel- opment of vision organs, lmmunohistochemical and electron-microscopic studies showed that S antigen localizes mainly in the rod outer segments, on both sides of the disc membranes in the photoreceptor cell layer of the retina [21, 22]. The unique relationship between S antigen and the vision system is supported by the presence of this antigen in the pineal gland [23]. Pineal gland has a photoreceptor function in lower ver- tebrates and retains some anatomical features of the retina even in mammals. The mammalian pineal gland is believed to have lost its photoreceptor function as it evolved from a median eye into a secretory organ. In the course of evolution, major photoreceptor proteins like rhodopsin and ct-transducin might have been elim- inated or reduced to negligible amounts. In contrast, other photoreceptor cell proteins such as interphotore- ceptor retinoid binding protein (IRBP) and rhodopsin kinase were localized to the pineal gland [4]. It is of interest to note that in accord with its content of S anti- gen and IRBP, the pineal gland is often affected in the inflammatory processes which develop in animals im- munized with S antigen or IRBP and this condition is termed experimental autoimmune pinealitis (EAP) [24].

S antigen is one of the major proteins of the retina and pineal gland and is estimated to comprise approxi- mately 0.5 and 0.2% of the total soluble protein of the retina and pineal gland, respectively. S antigen has been purified to homogeneity and reported to have an

inhibitory role in the activated phototransduction cas- cade [25-27]. Although the exact mechanism of the inhibition is not known, it is postulated that S antigen binds to photoexcited phosphorylated rhodopsin. The inactivation of photolyzed rhodopsin requires phos- phorylation of the receptor and binding ofarrestin. By binding to phosphorylated photolyzed rhodopsin, ar- restin inhibits G protein (G1) activation and blocks premature dephosphorylation, thereby preventing the reentry of photolyzed rhodopsin in the phototransduc- tion pathway [27].

Cleavage of the S antigen molecule by enzymatic (trypsin, chymotrypsin, Staphylococcus aureus V8 pro- tease) or chemical (cyanogen bromide) agents has been used to obtain information about the peptide compo- nents involved in the immunogenic and uveitogenic activities of this molecule [28]. The complete amino acid sequence of human, bovine, rat and mouse retinal S antigen as well as rat pineal gland S antigen has been determined [29-37]. Comparison of the amino acid sequences indicates a high degree of sequence homolo- gy among these species [37]. The number of amino acid residues in S antigen are 403 (rat as well as mouse), 404 (bovine) or 405 (human). Based on amino acid sequence data, molecular weights of bovine S anti- gen and human S antigen are determined to be 45,275 Da and 45,045 Da, respectively [28, 37]. Analysis of nucleotide sequences of S antigen cDNA also showed a high degree of sequence homology among the mouse, rat, bovine and human. S antigen genes are highly homologous and have 16 exons interrupted by 15 introns spanning approximately 50 kbp in length [37]. The introns are significantly larger than the exons and the average size of the introns is approximately 3 kbp in length. The genes comprise 97% introns and 3% exons. Each splice junction was in good agreement with the GT/AG rule. Comparison of the amino acid sequences of rat pineal gland and retinal S antigen indicated a significant degree of homology. Analysis of nucleotide sequences of the above two cDNAs isolated from retinal and pineal gland libraries showed high sequence similarity [29, 36]. These results suggest that the S antigen in the retina and pineal gland has a high degree of similarity at the transcript as well as polypep- tide level. The S antigen gene is assigned to chromo- some 2q24-37 in the human and to the centromeric portion of chromosome 1 near the IDH-1 locus in the mouse [38, 39]. Secondary structure prediction and circular dichroic spectroscopy show that S antigen has predominantly a ~sheet conformation [28].

Although S antigen was previously believed to be a highly specialized single protein, it is now apparent

Experimental Autoimmune Uveitis Immunol Res 1996; 15:323-346 325

Page 4: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

that S antigens are a family of multiple similar proteins [37]. Several arrestins have been sequenced from dif- ferent organs of various species and few of them are human, bovine, rat, mouse retinal arrestin (S antigen), rat pineal S antigen, 13-adrenergic arrestins from hu- man (13-1 and 13-2), bovine (13-1 and 13-2), rat (13-1 and 13-2), human thyroid, lateral eyes of Limulus polyphe- mus (horseshoe crab), compound eyes of Drosophila rnelanogaster I and II, Drosophila miranda, locust antennae (Locusta migratoria), corn earworm anten- nae (Heliothis virescens) and human cone arrestin, cone arrestin of Xenopus laevis [40-44]. All of the above arrestin sequences have shown significant se- quence homology among them [37, 45]. Furthermore, additional arrestins have been identified by immuno- detection [46]. The existence of different subtypes of arrestins as well as the possibility of modifying the pro- teins via binding of certain ligands or via phosphoryla- tion may allow arrestin to play different roles in cellu- lar signalling.

Another major retinal antigen capable of inducing EAU and EAP is IRBP. It is a 140-kDa evolutionarily conserved glycoprotein with a 4-fold partially homolo- gous repeat structure with 30-40% amino acid se- quence homology between repeats. It has been isolated and well characterized. IRBP is thought to be involved in the transport of vitamin A derivatives between the retinal pigment epithelium and the photoreceptors. It has been sequenced from cDNA and gene [47, 48] and found to be equally potent as a uveitopathogenic mole- cule as S antigen, producing EAU at very low doses. EAU induced by IRBP resembled that induced by S antigen also in its basic clinical and histological ocular changes, and by the involvement of the pineal gland [4]. Animal experiment studies have shown that there are several uveitopathogenic epitopes in bovine IRBP, and peptide 1169-1191 has been shown to be immu- nodominant [49, 50]. Recently, Silver et al. [51] have reported an additional uveitopathogenic epitope ( 161- 180) in human IRBP for B 10.RIII mice. Furthermore, rhodopsin or opsin, A antigen, transducin and cGMP phosphodiesterase have also been reported to induce EAU [41.

EAU as a Model for Uveit is

S antigen is a highly uveitopathogenic pro- tein which induces EAU and EAP in suscepti- ble animal strains [1-3, 26]. EAU has been reproducibly induced in a variety of animals,

including primates [5], rabbits [52], guinea pigs [26], rats [53], and recently in mice [54, 55]. The clinical and histopathological ap- pearance of uveitis occurring in these models closely resembles certain uveitic conditions in man [1, 4, 5, 28]. Usually EAU has been found to be associated with EAP [24]. These auto immune inf lammatory condit ions have been studied extensively as an animal model for human uveitis [ I, 5].

The clinical symptoms observed in rats with EAU are hyperemia of the conjunctiva, pericorneal and iris vasodilation and accumu- lation of inflammatory exudate and cells in the anterior chamber and vitreous. Within 2 - 4 days after onset of the disease, the ocular inflammation reaches its peak such as pro- truding eyes due to severe periocular inflam- mation, corneal edema, intense cell infiltra- tion in the anterior chamber and vitreous, and posterior synechiae of the iris. The clinical changes disappear rapidly in the rat and the eyes become clinically quiet. The histopatho- logical changes in rats with EAU following immunizat ion with various doses of S antigen or IRBP demonstrate a high degree of confbr- mity. Initial histological changes are exudate formation and accumulation of inf lammatory cells in the photoreceptor cell layer. The retina is most often detached with accumulation of serum underneath. Other components of the eye rapidly become involved as well, with inflammatory cells infiltrating the vitreous, iris, ciliary body and anterior and posterior chambers. The choroid becomes affected 2-4 days after the disease onset. The infiltrate consists mainly of a mixture of polymorpho- nuclear and mononuclear leukocytes (lym- phocytes and histiocytes). The inf lammatory cells disappear from the anterior chamber fol- lowed by cells from the posterior chamber. During this inflammatory process the photo- receptor cell layer is partially or completely lost [4].

326 |mmunol Res 1996;l 5:323-346 Singh/Nagaraju

Page 5: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Changes in primates with EAU are of par- ticular interest because of their possible rela- tionship to those in certain human uveitic conditions. The histological changes consisted of vasculitis and chorioretinitis with remark- able involvement of plasma cells and poly- morphonuclear cells, periphlebitis, focal loss of photoreceptor cells as well as areas of sub- retinal inflammation and occasional bulging of the retina into the vitreous cavity. Some of these changes resemble those observed in hu- man disease such as birdshot retinochoroi- dopathy [5].

Histopathology of pineal glands of rats with EAU shows focal cell infiltration, mainly at the peripheral or subcapsular areas of the gland. In severe cases, however, infiltrating cells are found in central areas as well. Unlike the massive infiltration with polymorphonu- clear leukocytes in the eyes, infiltration in the pineal glands of the same rats consists virtual- ly only of mononuclear leukocytes [4].

Although humoral responses are shown to play a role in EAU [56], the cell-mediated responses play the dominant role in the patho- genesis of EAU; since the disease does not develop in nude rats, the disease develops fol- lowing the adoptive transfer of lymph node cells from animals previously immunized with S antigen, the majority of cells participat- ing in the inflammatory response can be iden- tified as T cells, and the disease can be inhibit- ed by cyclosporin A [53, 57, 58]. Homozygous nude rats failed to develop EAU on challenge with S antigen, while heterozygous rats re- sponded very well. In addition, EAU could be induced in the nonresponder nude rats by the adoptive transfer of in vitro stimulated lym- phocytes from responder rats [53].

The knowledge of S antigen amino acid sequences allowed investigators to identify the uveitopathogenic sites by the use of small synthetic peptides. Many investigators have focused on the identification of epitopes re-

sponsible for the immunogenicity and immu- nopathogenicity of S antigen. Three sites of bovine or human S antigen were identified as being immunopathogenic in Lewis rats: pep- tide 303-320 (peptide M) and 286-297 (pep- tide N) which are nondominant, and 343-362 (peptide G) which has immunodominant characteristics [59-62]. Later, de Smet et al. [63] mapped the immunogenic and immuno- pathogenic determinants of human S antigen in the Lewis rat by using overlapping synthet- ic peptides. They identified most pathogenic sequences as 180-200, 340-360 and 350-370 peptides. Ten peptide sequences induced visi- ble inflammation in the eye. A total of 23 pep- tides gave an in vitro proliferative response following immunization in animals. These studies have shown that there are several uveitopathogenic determinants within S anti- gen, sugesting the same may be true in uveitis patients. The existence of multiple determi- nants would place considerable restrictions on the feasibility of immunotherapy in patients. Immunotherapy directed against a single de- terminant is unlikely to be effective in hu- mans. Only approaches like oral tolerance that can induce nonspecific suppression, even if limited to a specific inflammatory site, are likely to be effective.

Molecular Mimicry and EAU

The phenomenon by which the host re- sponds to exogenous antigens that cross-react with self by sharing of linear or conformation- al epitopes common to microbial antigens and host structures is known as molecular mimi- cry and it may provide a mechanism for trig- gering autoimmune diseases and tolerance [11, 64]. Earlier investigations have focused on cross-reaction between B cell epitopes and the subsequent generation of autoantibodies. However, mimicry at the B cell level cannot

Experimental Autoimmune Uveitis Irnrnunol Res 1996;15:323-346 327

Page 6: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

provide the pathogenic basis for T-cell-me- diated organ-specific autoimmune diseases where CD4+ T cells either produce inflamma- tory cytokines or provide help to B ceils. Cross-reaction between self and nonselfT cell peptides has also been documented in various systems [ 12].

The question of how autoimmune diseases such as endogenous posterior uveitis are ini- tiated remains to be fully answered. Circum- stantial evidence suggests a link between au- toimmune diseases and triggering events such as a recent viral or bacterial infection. This has led to the hypothesis that circulating au- toreactive T cells, present in all healthy indi- viduals, recognize epitopes on processed for- eign antigen, which possesses extensive amino acid sequence homology with host autoanti- gens.

According to the above hypothesis of mo- lecular mimicry, invading microorganisms are rapidly attacked by innate immune mech- anisms involving acute inflammatory cells such as neutrophils and monocytes. The par- tially degraded foreign antigens are then taken up by dendritic cells and transported to lymph nodes where the immunodominant antigenic peptides are presented to T cells, most of which induce a full range of immunological sequelae including cytotoxic T cell responses, B cell responses and memory T and B cell responses. Due to their sequence homology with autoantigenic peptides, certain foreign peptides may also activate autoreactive T cells. These activated T cells then enter the circulation, but do not necessarily home to their target tissue unless the endothelium has also been activated by cytokines such as inter- leukin-1 (IL-1) and interferon-,/which induce specific adhesion molecules on their surface. This may occur if the response to the original attack by microorganisms is sufficiently se- vere and sustained to the point where a threshold concentration of cytokines and/or

endotoxin is released into the circulation [65].

Several examples of possible molecular mimicry between retinal antigens and pro- teins from microorganisms exist. For in- stance, studies have demonstrated that the onchocerca worm can induce a keratitis asso- ciated with anticomeal autoantibodies when injected into the subconjunctival space. The onchocerca parasite is responsible for en- demic river blindness, but usually does so by involving the posterior segment. Recently, an onchocercal antigen (Ov39) has been found to cross-react with a 44-kDa antigen located in retinal pigment epithelium, the neural retina and the optic nerve. The retinal antigen has not yet been fully characterized, but it does not demonstrate identity with S antigen. However, when Lewis rats were immunized with Ov39, they developed a low grade anteri- or and posterior uveitis with some similarities to EAU and experimental autoimmune ante- rior uveitis, but without evidence of autoim- munity to retinal S antigen [65]. It is probable that the autoimmune response is to the 44- kDa antigen. Ov39-induced uveitis, therefore, represents the appropriate example of com- mon linkage between immune responses to foreign and self-antigens in the eye.

The knowledge of the uveitopathogenic sites of S antigen enabled us to look for similar sequences in foreign antigens. We compared amino acid sequences of various uveito- pathogenic peptides of S antigens with a vari- ety of proteins in the National Biomedical Research Foundation data base for sequence homology. We found 4- to 6-amino acid se- quence homology between one of the uveito- pathogenic peptides of S antigen (peptide M) and various proteins, such as DNA poly- merase of hepatitis B virus, gag-pol polypro- tein ofAKV murine leukemia virus, Moloney murine leukemia virus, Moloney murine sar- coma virus, gag-pol polyprotein of baboon

328 immunol Res 1996;15:323-346 Singh/Nagaraju

Page 7: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Table 1. Sequence homology between human retinal S antigen fragment (peptide M) and other proteins

No. Source ofpeptide Amino acid sequence Ref. No.

1 Retinal S antigen (peptide M) 2 Yeast histone H3 3 E. coli elongation factor 4 E. coli hypothetical protein 5 Potato inhibitor IIa 6 Hepatitis B virus protein 7 Baboon endogenous virus 8 Moloney murine leukemia virus 9 Moloney murine sarcoma virus

10 AKV routine leukemia virus

D T N L A S S T I I K E l 59 D T N L A A I H A K R V 1 68-70 A R H L A A S I A F K E 68 L A N L A S S T Q L C K ~ 67 D T N I A S Y K S U C E 66 L T N L L S S N L S W L l 66 P T N L A K V R T I T Q I 66 P T N L A K V K G I T Q ~ 66 P T N L A K V K G I T Q 1 66 P T N L A K V K G I T Q ~ 66

Underlinings indicate identical amino acid residues with peptide M. Peptides have been found to consistently induce EAU in Lewis rats.

endogenous virus, Escherichia coli hypotheti- cal protein E-116, yeast histone H3 and pota- to proteinase inhibitor IIa (table 1) [12]. These amino acid sequences were used to syn- thesize oligopeptides of 12-20 amino acids by conventional solid phase chemistries on resin using an automated peptide synthesizer and purified by high-performance liquid chroma- tography [61]. Lewis rats were immunized intradermally with various peptides emulsi- fied in CFA followed by intravenous injection with B. pertussis. Interestingly, our results showed induction of EAU with peptides de- rived from DNA polymerase of hepatitis B virus, gag-pol polyprotein ofAKV murine leu- kemia virus, gag-pol polyprotein of baboon endogenous virus [66], E. coli hypothetical protein E-116 [67] and yeast histone H3 [68]. In addition, native yeast histone purified from Saccharomyces cerevisiae was found to be capable of inducing EAU in Lewis rats [68]. Yeast histone H3 peptide also induced EAU in monkeys [69]. Furthermore, rats de- veloping EAU as a result of immunization with microbial peptides showed an associated

EAP characterized by a lymphocytic infiltra- tion of the subcapsular area of the pineal glands [68]. The histology of the diseased reti- na and pineal glands from animals immu- nized with peptide M or various microbi- al peptides was indistinguishable [66-68]. Lymph node cells from rats immunized with either peptide M or one of the peptides men- tioned above showed a significant degree of cross-reaction, i.e. lymph node cells from ani- mals immunized with peptide M showed sig- nificant in vitro T-cell-proliferative response in the presence of one of the above microbial peptides or vice versa [70]. Lymph node cells from animals immunized either with peptide M or yeast histone H3 peptide and stimulated in vitro with either peptide M or yeast histone H3 peptide induced EAU when injected in- traperitoneally in naive syngeneic rats. The histopathotogy showed severe inflammation in the uveal tract and complete loss of photo- receptor cells which was indistinguishable from EAU induced with peptide M or native S antigen [70].

Experimental Autoimmune Uveitis lmmunol Res 1996;15:323-346 329

Page 8: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Table 2. Some important examples of molecular mimicry

No. Diseases Homologous antigens Ref. No.

1 EAU 2 EAE 3 AS 4 Myocarditis 5 Rheumatoid arthritis 6 Ovarian autoimmunity 7 Celiac disease 8 IDDM

S antigen and various microbial proteins 66-70 MBP and HBVP 73 HLA-B27 and proteins from Klebsiella, Yersinia, Salmonella, Shigella 75, 76 Heart tissue myosin and coxsackievirus capsid protein 77, 78 Human heat shock protein and mycobacterial heat shock protein 79, 80 Ovarian and nonovarian peptides 82 Wheat gluten EIB protein of adenovirus 12 83 Islet antigens and CVB protein 84, 85

HLA = H u m a n leukocyte antigen.

The likelihood of any particular autoim- mune disease and an infectious disease hav- ing a common antigenic ontogeny depends on many factors such as the degree of epitope homology between the antigens, the immuno- dominance of the common antigenic se- quence, the degree of MHC restriction and the avidity of class II binding, the dose of anti- genic peptide released for presentation, and the number of potentially primed autoreac- tive T cell clones. The chances are increased after infection with superantigens. Superanti- gens are proteins that activate the T cell recep- tor (TCR) not via the conventional MHC- peptide mechanism, but by complexing the MHC class II to the TCR directly via the TCR 13 chain outside the peptide binding site. Superantigens have been implicated as causa- tive agents in a variety of autoimmune dis- eases [71]. Infection with certain superantig- ens may lead to the activation of up to 10% of the T cell population, which significantly en- hances the chances that one or more of these clones may be responsive to presentation of a self peptide. In particular, it has recently been demonstrated that the superantigen Staphylo- coccus M peptide contains sequences homolo- gous to certain peptides of retinal S antigen that can induce EAU. In addition, these pep-

tides preferentially associate with certain V13 T cell receptor chains which appear to be used at greater frequency in patients with posterior uveitis [72].

Molecular Mimicry in Other Autoimmune Diseases

There are various examples of molecular mimicry in different systems (table 2) and some of them are discussed below. Fujinami and Oldstone [73] have reported 6-amino acid sequence homology between hepatitis B virus polymerase (HBVP) and the encephalitogenic site of myelin basic protein (MBP) which causes experimental allergic encephalomyeli- tis (EAE), the animal model for multiple scle- rosis. Rabbits injected with synthetic peptide showed antibody responses that cross-reacted with MBP. The peripheral blood mononu- clear cells proliferated significantly when in- cubated with either MBP or HBVP. In addi- tion, these rabbits showed central nervous system lesions reminiscent of EAE induced by the immunization with MBP.

Ankylosing spondylitis (AS) is a chronic inflammatory rheumatic disorder particularly affecting the axial skeleton. A link between AS

330 Immunol Res 1996;15:323-346 Singh/Nagaraju

Page 9: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

and the major histocompatibility antigen HLA-B27 has been well established. Over 95% of patients with AS are HLA-B27-posi- tive. There are seven subtypes of HLA-B27 ranging from B'2701 to B'2707. It has been suggested that B'2705 is the most common HLA-B27 subtype in the Caucasian popula- tion, whilst it is proposed that B'2703 is the most common in black populations, where AS is rare. The observation has led to the sugges- tion that B'2703 is not associated with AS. The cause of AS remains controversial, al- though an environmental agent in the form of the gram-negative bacterium, Klebsiella pneu- moniae, present in the gut, has been suggested as a possible etiological agent to which anti- bodies have been shown. Schwimmbeck et al. [74] identified an amino acid homology, QTDRED found in residues 72-77 ofB'2705 and residues 188-193 of K. pneumoniae ni- trogenase enzyme. Others have shown homol- ogy between HLA-B27 and Yersinia pseudo- tuberculosis adhesin, outer surface of protein of Yersinia enterocolitica and Salmonella ty- phimztrium and Shigella flexneri [75]. The validity of this homology, as a cross-reactive antigen, has, however, been criticized, in both biochemical and immunochemical terms. Re- cently, Fielder et al. [76] have shown novel homology between two sequences of K. pneu- moniae pulD secretion protein (DRDE) with HLA-B27 (DRED) and pulA (pullulanase) en- zyme (Gly-X-Pro) with type I, III and IV col- lagen, respectively. Elevated levels of anti- body in AS patients were detected against 16 mer synthetic peptides of HLA-B27 and pulD, pulA and type I and IV collagen.

Clinical studies show that a higher propor- tion of patients with chronic myocarditis or dilated cardiomyopathies contain serum IgM antibodies to coxsackievirus group B (CVB) than patients with heart disease of other infec- tious etiologies or immunological causes. Mo- lecular mimicry has been proposed to be a

mechanism that may explain some cases of CVB-induced chronic myocarditis in humans and in some strains of mice that model the human disease [77]. The hypothesis of shared epitope between CVB capsid proteins in and on normal cells is supported by the fact that a neutralizing monoclonal antibody (mAb) against CVB4 was shown to bind to viral cap- sid polypeptide VP 1 and to heart tissue of sev- eral animals. This mAb was directed against an epitope in heart tissue-specific myosin and not skeletal myosin. A neutralizing mAb against CVB3 bound to epitopes on normal mouse myocytes, HeLa cells and HEp2 cells. A nonneutralizing mAb that was generated against murine heart myocytes, and perhaps against the highly myocarditic CVB3 receptor was found to recognize CVB3. This mAb can partially protect BALB/c mice against CVB3- induced murine myocarditis and can also rec- ognize an epitope on the adhesin nucleotide translocator protein of murine myocytes. None of these mAbs to CVB4 and CVB3 was reported to induce cytopathic changes in cells in vitro or pathological alterations in heart tis- sues of mice inoculated with a mAb [78].

In patients with rheumatoid arthritis, pep- tides from a mycobacteria165-kDa heat shock protein have been shown to cross-react with peptides from proteoglyca_q link proteins and human heat shock protein 60 [79, 80]. More recently, viral peptides were demonstrated to activate MBP-specific T cell clones isolated from multiple sclerosis patients [81]. Garza and Tung [82] have demonstrated that close to 50% of nonovarian peptides that share some critical residues of the ovarian peptides can cause ovarian autoimmunity.

Celiac disease is characterized by small intestinal mucosal injury and malabsorption. The disease is activated by dietary exposure to wheat gluten. Patients with celiac diseases show higher titer of antibodies against A-glia- din and the E1B protein of adenovirus sub-

Experimental Autoimmune Uveitis Immunol Res 1996:15:323-346 331

Page 10: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

type 12, but not normal controls, suggesting that a cross-reactive immune response against the wheat protein A-gliadin initially induced by adenovirus 12 intestinal infection plays a role in the pathogenesis of celiac disease [83]. Recently, similarity between one peptide of the CVB and islet autoantigen, carboxypepti- dase H, autoantibodies to which have been claimed to be characteristic of IDDM, has been reported [84]. This is in addition to another homology between islet autoantigen, glutamic acid decarboxylase and the CVB which was reported earlier [85].

Oral Tolerance in Autoimmune Diseases

Gttt-Associated Lymphoid Tissue and Handling of Ingested Antigens by the Gut The lymphoid system of mammals is com-

posed of primary lymphoid organs, the bone marrow and the thymus, and secondary lym- phoid organs such as diffuse lymphoid tissue, lymphoid follicles, Peyer's patches, tonsil, lymph nodes and spleen. The bone marrow and the thymus are responsible for the matu- ration of stem cells into B and T lymphocytes, respectively. The secondary lymphoid organs are the sites where the antigen-dependent and antigen-independent interactions of mature lymphocytes take place leading to an effective immune response. The secondary lymphoid organs are conveniently situated near the vast mucosal surface, lining the digestive, respira- tory and urinogenital system. Gut-associated lymphoid tissue (GALT) consists of aggre- gated lymphoid follicles in the small intes- tines, the appendix vermiformis, colonic patches and solitary lymphoid nodules [86, 87]. GALT has two functions: to protect the host from pathogens entering through the gut, and preventing the host immune system from mounting an immune response to ingested

antigens. The wall of the intestine consists of seven layers which from the inner towards the outer surface are: epithelium, lamina propria, lamina muscularis, mucosae, submucosa, the layer of circular muscles and the layer of lon- gitudinal muscles. Of these the first three form the intestinal villi. The submucosa con- tains diffuse lymphoid tissue with dense areas of solitary lymphoid follicles especially in the ileum. This portion of the small intestine has clusters of lymphoid follicles called Peyer's patches. Each patch is seen on the inner sur- face as a group of bare dome-shaped struc- tures in the microvilli. The epithelium of the dome overlying each follicle is comprised of specialized cells with many microfolds called M cells. These M cells take up particulate antigens by endocytosis and process and present antigens either directly to T cells or alternatively transfer them to antigen-present- ing cells (APCs) such as dendritic cells or Lan- gerhans cells. In Peyer's patches the B cells are committed to immunoglobulin classA and subsequently populate the lamina propria [88]. The origin of the lymphocytes present in the lamina propria and in the intraepithelial portion of the intestine is not known. Charac- terization of lymphoid populations in these regions shows that these are predominantly T cells with et/13 TCR and are of the CD8 phe- noptype, CD45 RO antigen suggesting they are MHC class I-restricted memory T cells. These ceils are specific for a limited number of antigens [89]. A small proportion of intra- epithelial lymphocytes (IEL) have V/~5 TCR.

Traditionally, GALT is considered as a secondary lymphoid tissue. The following ob- servations made during the past few years sug- gest that GALT may have functions similar to the thymus. (1)Murine IEL can develop via an extrathymic pathway [90-96] giving rise to two distinct subsets of T cells, one with a/J3 TCR repertoire suggesting thymus selection and phenotypically with CD8 et/13 heterodi-

332 Immunol Res 1996:15:323-346 Singh/Nagaraju

Page 11: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

mer, and another population appears to have developed extrathymically with a phenotype of CD8 a/a homodimer [90, 97]. (2)Yrans- genic mouse models expressing transgene for a T/8 TCR specific for MHC class I molecule suggest the presence of anergic autoreactive T/ 8 IEL in the absence of these clones in the peripheral circulation [98]. Similar results were obtained using mice transgenic for an a/ 13 TCR specific for male H-Y antigen [97]. (3) The presence of RAG 1 and RAG2 mRNA in the adult and fetal intestines. (4) Studies on athymic radiation chimeras showed genera- tion of CD4+, CD8 a/J3+, CD4+/CD8 a/a++ mucosal T cell subsets in the gut upon recon- stitution with T-cell-depleted bone marrow or fetal liver [99].This evidence indicates that GALT may have some functions similar to those of primary lymphoid organs, such as thymus. It may be speculated that GALT might be involved in inducing clonal anergy and clonal deletion in a similar manner like the thymus.

Diet consists of a complex mixture of sub- stances which are potentially antigenic. The mucosal surface of the gut is covered for the most part by a monolayer of epithelial cells. A minority population of epithelial cells is highly specialized (the M cells) for presenting anti- gens to the cells of the immune system, where- as a major population of diverse epithelial and glandular cells selectively exports polymeric immunoglobulins to mucosal surfaces. The M cells are important because of their unique position over the organized lymphoid tissues that serve as inductive sites for mucosal im- mune responses and their ability to transport antigens with great efficiency. The immuno- logical consequences of antigen uptake and transport are determined in part by the nature and capacity of the intracellular pathways through which antigens are directed. Macro- molecules cross intestinal epithelial cells in three ways: shuttle through absorptive cells on

specific receptors and molecules that bind to a receptor pass or they can pass through M cells [100] or antigen fragments cross epithelial cells for presentation by MHC molecules at the basolateral surface [I01]. Enterocytes present peptides originated in the cytosol through MHC class I to CD8+ intraepithelial lymphocytes [102]. These CD8+ cells prolifer- ate and secrete various cytokines which in- fluence the immune response in a variety of ways. MHC class II antigens are present on the basolateral surface as well as in intracellular vesicles and on apical membranes of intestinal cells [ 103]. These pickup peptides are taken up by adsorptive or fluid phase endocytosis into common endosomes and present to CD4+ lymphocytes in the lamina propria. Since CD4+ lymphocytes are not generally found within epithelial cells, it is not known how fre- quently MHC class II-presented peptides in- teract with CD4+ lymphocytes in the lamina propria [104]. These epithelial MHC mole- cules might be involved in the development of thymus-independent IEL (TCR a/13 or T/8), suggesting that MHC class II+ enterocytes and lamina propria lymphocytes cooperate to modulate the lymphocyte repertoire in the gut. Primed T and B cells migrate through lymph to the peripheral blood circulation and extrav- agate mainly in the intestinal lamina propria. Intestinal B cells differentiate into IgA-secret- ing plasma cells in the lamina propria under the influence of macrophages, APCs and CD4+ T ceils. Most CD8+ T cells migrate into the villus epithelium, perhaps to mediate oral tolerance to food antigens.

Factors Determining the Induction and Maintenance of Oral Tolerance Various factors such as nature of the anti-

gen, concentration of the antigen, genetic makeup and the age of the animal determine the induction and maintenance of oral toler- ance.

Experimental Autoimrnune Uveitis lmmunol Res 1996;15:323-346 333

Page 12: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Live organisms such as viruses and bacte- ria induce active immune response rather than tolerance by the oral route, whereas the same organisms if killed or inactivated induce oral tolerance [ 105]. How the gut immune sys- tem can distinguish between immunogenic and tolerogenic stimuli is currently not known. It might be due to different modes of antigen processing and presentation involved in handling live and killed antigens by the gut immune system.

Testing a given antigen systemically over a wide range of concentrations reveals that low antigen doses often induce tolerance, interme- diate doses induce immunity and high doses induce tolerance. Unlike systemic responses, oral tolerance can be induced using a wide range of antigen doses from few micrograms to milligrams [106, 107], these differences might be due to tolerogenic epitope density in a particular antigen. Friedman and Weiner [108] showed that induction of anergy or ac- tive suppression following oral tolerance is determined by antigen dosage, single high dose of antigen-induced anergy and multiple low doses of antigen-induced tolerance by ac- tive suppression through transforming growth factor-J3 (TGF-[3) and IL-4. It is difficult to induce tolerance in animals previously primed via cross-reacting intestinal flora, therefore, multiple feeds of relatively large doses of antigen are required to induce oral tolerance in such cases [ 107]. How exactly the antigen doses elicit anergy or active suppres- sion through cytokines is not known. It is speculated that it might be due to differential requirements for number of epitopes, co- stimulatory signals and cytokines.

Genetic influence on the ease of tolerance induction is exemplified by the mouse strains BALB/B and BALB/c. The former is relative- ly much more difficult to tolerize than the congenic BALB/c strain [109-111]. Since these mice differ only at the H-2 locus, it is

reasonable to assume the MHC genes at least partly might have role in regulating oral toler- ance to protein antigens. The immunological basis of this MHC-linked effect remains to be investigated. It is easier to induce systemic immune tolerance in animals with an imma- ture lymphoid system than in mature ani- mals. The mice fed ovalbumin (OVA) during early postnatal life, i.e. first day or second, do not develop the tolerance of systemic immu- nity found in adults fed the same dose of anti- gen. In fact, OVA-fed infant mice develop a systemic immune response when challenged parenterally as adults. This reflects the inabil- ity of the immature immune system to re- spond appropriately to gut-derived tolerogen. The adult pattern of susceptibility to toler- ance develops within the first week of postna- tal life [ 112, 113]. Maturation into adulthood from 8 to 24 weeks of age significantly in- fluences the induction of oral tolerance in dif- ferent strains of mice. Animals from strains which are susceptible to the induction of oral tolerance to OVA at 8 weeks of age became refractory at 2 weeks of age [I 14].

Factors such as cytokine milieu, adjuvants, nutritional status of the animal [ 115], intesti- nal flora [106, 116], permeability of the in- gested antigen [117], chemical modification of the antigen by methylation and acetoacety- lation are also known to influence the toler- ance induction.

Mechanisms and Modulation of Oral Tolerance

Antigens such as foreign S antigen, type II collagen or MBP pass from the lumen of the gut across multifold cells (M cells) or APCs lying under Peyer's patches. These cells then activate a local population of T cells which secrete TGF-13 and IL-4 [ 118]. Following acti- vation, few of these cells wander out through

334 immunol Res 1996;15:323-346 Singh/Nagaraju

Page 13: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

the lymphatics and blood stream and then through tissues until they again find their recall antigens. In a patient with uveitis, rheu- matoid arthritis or multiple sclerosis these cells recognize via cross-reaction of the ex- posed selfS antigen, collagen II or MBP in the inflamed eye, joint or brain. The specialized T cells are then stimulated by their recall anti- gen to secrete TGF-13 and IL-4. These inhibi- tory cytokines suppress the activity of neigh- boring disease inducing Thl cells. The latter cells presumably recognize one or more au- toantigens, the nature of which is unknown. Any organ-specific antigen could lure the sup- pressive T cells to the target organ which need not be a known autoantigen (S antigen, type II collagen or MBP).

Apart from the secretion of inhibitory cy- tokines and nonspecific suppression, there are basically three nonmutually exclusive hypoth- eses that have been proposed to explain the induction of adult unresponsiveness to anti- gen, any or all of which may potentially oper- ate in models of oral tolerance. These are anergy, deletion, and antigen-specific sup- pression, and indeed the stability and revers- ibility of peripheral tolerance in a given situa- tion may depend on which of these mecha- nisms is operating [119].

Under certain conditions the contact of antigen with T cells in vitro would render them anergic [120, 121]. This was suggested to be caused by the lack ofa costimulatory sig- nal required for T cell activation. Specific T cells from tolerant animals are shown to be unresponsive even to stimulation directly through their antigen receptor via VI3-specific antibodies [122]. Such data are further sup- ported by analysis in which antigen-specific responses cannot be elicited in cells from tolerant animals in culture despite the pres- ence of cells expressing VI3 chains that have been associated with a response to that anti- gen [123]. In some models, anergy is associat-

ed with a downregulation of cell surface pro- teins critical to T cell activation such as TCR and/or accessory cell surface molecules like CD8 [124]. Melamed and Friedman [125] showed that a single feeding of 20 mg OVA by gastric intubation induced a state of anergy in OVA-specific T cells. These ceils are charac- terized by lack of proliferative responses to OVA, absence of IL-2 and IL-2R expression and this nonresponsive state was reversed by preculture of tolerized cells in IL-2-containing medium. Subsequently, it was shown that in- duction of anergy depends on antigen dosage and frequency of feeding [126]. Since anergy has been suggested to be a postthymic-periph- eral mechanism for tolerance, it could be val- id as a mechanism for oral tolerance to dietary antigens [ 127].

Peripheral clonal deletion is an intriguing possibility in oral tolerance. The existence of peripheral clonal deletion from studies show- ing the selective absence of VI3 6+ cells in the periphery of adult thymectomized, Mls+ mice in comparison to adult thymectomized Mls- mice has been suggested [128]. Peripheral downregulation of TCR and CD8 molecules can be a novel mechanism for the establish- ment of peripheral tolerance [124, 129, 130]. Whitacre et al. [131] showed the specific ab- sence o f t cells expressing the VI3 8.2 TCR in SJL mice fed MBP. Since the immune re- sponse in these mice is dominated by V[3 8.2+ T cells, these results support the possibility that clonal deletion of antigen-specific T cells may occur during oral tolerance. But these studies do not exclude downregulation of TCR and accessory molecules like CD8 on T cells [124]. The observations showing that the oral tolerance may be specifically abrogated by treating with cyclophosphamide, 2'-deoxy- guanosine or by transfer of TCR ~//5 IEL sug- gest that clonal deletion is a less likely possible mechanism in oral tolerance [ 111, 132-134]. A recent study by Chen et al. [135] shows that

Experimental Autoimmune Uveitis Immunol Res 1996;15:323-346 335

Page 14: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

orally administered antigen can induce toler- ance not only by active suppression and clonal anergy but by extrathymic deletion of antigen- reactive Thl and Th2 cells.

Generally the term suppression is used to describe a situation where potentially reactive T cells are present but held in check by other T cells. For several years the idea that sup- pressor cells were able to control immune responses and could be responsible for periph- eral tolerance received widespread credence [ 136, 137]. In models of transplantation toler- ance, it was shown that adoptive transfer of ceils from tolerant animals into naive animals prevented the rejection of a subsequent graft [138]. In all cases of suppression it is neces- sary to make a distinction between specific and nonspecific suppression, and in the case of specific suppression for idiotype and anti- gen-specific suppression.

Antigen-specific suppressors can be de- fined as cells which suppress effector re- sponses through recognition of the same anti- gen as that recognized by the effector popula- tion. Therefore, they may not be the same cells as those recognized by the effector popu- lation. They may not recognize exactly the same epitope, but they must be able to recog- nize a component of the stimulating antigen so that they can interfere with the overall response to the antigen. Richman et al. [ 139] showed that oral tolerance could be trans- ferred through splenic T cells to naive, syn- geneic irradiated recipients. It was also shown that orally tolerized recipients suppressed re- sponses of normal transferred spleen cells [140]. The suppression was abrogated by us- ing cyclophosphamide, antisuppressor cell an- tiserum [111,127, 140-144]. Antigen-specific suppressor cells have been described in in vitro systems [ 145-147]. One of the models of suppression suggests involvement of three dif- ferent types of cells acting in a cascade to pro- duce suppression [148]; the first level of the

cascade involves T cells which are antigen- specific CD4+ suppressor inducers. It is also possible that anti-idiotypic suppression may be operating in oral tolerance. Orally adminis- tered antigens may selectively stimulate T cell clones which recognize pathogenic T cells, thereby controlling autoaggressive clones.

Natural suppressor cells which suppress an immune response in an antigen and MHC- independent manner were described in graft versus host disease [149, 150]; the induction of such cells in oral tolerance could not be ruled out. Lider et al. [151] showed that CD8+ cells isolated from orally tolerized animals could adoptively transfer the protection. These MBP-specific CD8+ T cells mediated suppression by releasing TGF-I3 and adminis- tration of anti-TGF-[3 abrogated the protec- tion [152]. Yoshino et al. [153] showed that oral collagen II suppressed arthritis, and the effect was dose dependent occurring at low doses of collagen II, demonstrating the biolog- ical relevance of bystander suppression asso- ciated with oral tolerance.

It was shown that secretion of anti-inflam- matory cytokines as well as protection from disease can be enhanced by feeding the bacte- rial adjuvant lipopolysaccharide together with the antigen [118, 154, 155]. Rizzo et al. [155] showed that three feedings of 0.2 mg IRBP every other day before immunization did not protect against EAU, whereas a similar regi- men of five doses was protective. However, supplementation of the three-feeding regimen with IL-2 resulted in disease suppression sim- ilar to the five-feeding regimen. They also showed that this effect was due to increased production of anti-inflammatory cytokines like TGF-13, IL-4 and IL-10 induced by IL-2 administration and the tolerance induced in the five-feeding regimen was due to anergy. Using knockout models of IL-4 and IL-10, they demonstrated that these cytokines have a role in the induction of low dose tolerance,

336 Immunol Res 1996;15:323-346 Singh/Nagaraju

Page 15: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

Table 3. Oral tolerance in various autoimmune diseases

Features EAE EAU AA IDDM

Animal model Lewis rat Lewis rat Rat NOD mice Disease induction MBP in CFA S antigen in CFA Mycobacterium Spontaneous

tuberculosis Oral tolerance induction MBP S antigen Collagen type II Porcine

insulin Inflammatory infiltrate Decreased Decreased Decreased Decreased Severity/incidence Decreased Decreased Decreased Decreased Lymphoproliferation/DTH Decreased Decreased Decreased Decreased Adoptive transfer of tolerance Yes Yes Yes Yes Ref. No. 13, 126, 151 16, 17, 176, 177, 179 159, 173 15, 160

AA = Adjuvant arthritis; NOD mice = nonobese diabetic mice; DTH = delayed-type hypersensitivity.

because these mice were successfully tolerized with a high dose feeding regimen [156].

Relevance of Oral Tolerance in T-Cell-Mediated Autoimmune Diseases

Autoimmunity develops into a disease when the components of the immune system begin to damage the individual's own tissues. T lymphocytes are strongly implicated in the pathogenesis of certain autoimmune diseases like IDDM, uveoretinitis, multiple sclerosis and arthritis (table 3). In experimental models of these diseases it was shown that the patho- genic lymphocytes were able to transfer au- toimmune disease [ 157-160]. Both cytotoxic and helper T lymphocytes have been shown to cause tissue damage. The cytotoxic T cells presumably attack the target tissue directly; the helper T cells act by providing help to cytotoxic T cells but also by releasing cyto- toxic lymphokines and by contributing to a developing inflammatory response. A number of studies on effector T cells mediating organ- specific autoimmune diseases in spontaneous or induced models have documented that the

key effector cells are CD4+ T cells which mediate autoimmune disease as helper T cells for autoantibody forming B cells, as amplifi- ers for cytotoxic or as mediators of cell- mediated immune responses akin to delayed- type hypersensitivity [161-163]. These find- ings suggest the existence of two distinct pop- ulations ofCD4+ T cells in terms of maintain- ing self-tolerance and induction of autoim- mune disease, one mediating autoimmune disease and the other inhibiting it. This fur- ther leads to the hypothesis that one aspect of self-tolerance may be maintained by the cellu- lar interaction between these two CD4+ popu- lations. The autoimmune inhibiting CD4+ T cells are dominant in the normal physiological state. The experimental evidence for this hy- pothesis was provided by studies using athymic nude mice reconstituted with partic- ular T cell subpopulations CD4+ CD51o au- toimmune-inducing clones, CD4+ CD5hi suppressor of disease-inducing clone.

Our understanding of the T-cell-mediated autoimmune diseases is derived to a large extent from the study of animal models of dis- ease like EAE, EAU, IDDM and adjuvant arthritis. Considerable effort has been di-

Experimental Autoimmune Uveitis lmmunol Res 1996;15:323-346 337

Page 16: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

rected towards defining the autoreactive T cell repertoire in many human diseases in- cluding multiple sclerosis [ 164, 165], autoim- mune thyroiditis [166], myasthenia gravis [167], IDDM [168] and chronic active hepati- tis [ 169]. However, interpretation of the data obtained has not been conclusive. Some au- thors provide evidence for a restricted T cell response [ 165-169], while others find signifi- cant diversity both with respect to autoanti- genic determinants and in the range of TCR genes [167, 170]. A solution to this contradic- tion was first shown in a rodent model of EAE that relates to the MBP-specific repertoire at different time points. These data show that the expressed autoimmune repertoire is not fixed but evolves during the course of disease. The initial responses are restricted to a spe- cific self-protein in the target organ, these responses subsequently spread to additional epitopes of the same autoantigen (intramo- lecular spread) or to different autoantigen in the target organ (intermolecular spread). Thus multiple autoantigens become targets of au- toimmune responses [171, 172]. This indi- cates that therapeutic approaches for human diseases based on deleting and rendering an- ergic specific antigen-reactive clones using immunodominant peptides or anti-idiotypic strategies may not work. Generally oral toler- ance leads to unique mechanisms of tissue- directed suppression that occur following oral administration of proteins which would cir- cumvent the immunotherapeutic problem of multiple target tissue autoreactivity. Oral tol- erance is tissue- and disease-specific, i.e. oral- ly administered MBP does not affect adjuvant arthritis [173] or autoimmune uveitis [17]. Likewise, orally administered collagen or S antigen suppress these diseases, respectively, and do not affect EAE. Therefore, the induc- tion of oral tolerance may be a suitable thera- peutic intervention in T-cell-mediated auto- immune diseases.

Oral Tolerance in EAU

As discussed above, EAU is a T-cell-me- diated autoimmune disease which can be in- duced by many well-characterized photore- ceptor cell proteins including S antigen, IRBP [28] or their pathogenic peptides or sensitized cells [58-63]. It was shown that EAU can be prevented by oral administration of native bovine S antigen or its peptides prior to im- munization [16, 17, 174, 175]. The exact mechanism by which the protection is con- ferred is not well known. Vrabec et al. [174] suggested that adoptive transfer of protection from EAU or EAE using splenocytes from orally tolerized animals should not be possible if anergy was the protective mechanism. In addition, it has been suggested that oral toler- ance in EAU operates by at least two distinct mechanisms that depend on the feeding dose; low doses induce suppression, whereas high doses induce unresponsiveness [176]. Avail- able experimental evidence tilts more towards suppression as a mechanism of oral tolerance in EAU for the following reasons. (1)Oral administration of antigen induces suppressor T cells and splenic CD8+ T cells protect the animals from subsequent active induction of EAU. Furthermore, these cells also inhib- it antigen-specific in vitro prolifcrative re- sponses [17]. (2)The antigen-specific in vitro suppression was blocked by anti-CD8 anti- bodies demonstrating that suppression was CD8+ T-cell-mediated. Recently, Vistica et al. [177] suggested that CD8 T cells are not essential for the induction of low-dose oral tolerance. Additional studies showed that tol- erance to retinal antigen could be induced via the upper respiratory tract with microgram doses of antigen preventing subsequent in- duction of EAU [I 78]. The authors also spec- ulate that the inhalational tolerance may be better than oral tolerance due to small amounts of antigen used to induce the toler-

338 Immunol Res 1996;15:323-346 Singh/Nagaraju

Page 17: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

ance. Recently, we have shown that feeding of E. coli expressing retinal S antigen to Lewis rats prior to uveitopathogenic challenge with S antigen resulted in significant suppression of EAU and of the cellular responses to S anti- gen [179].

Conclusion and Future Direction

Despite the fact that several examples of sequence homology between retinal S antigen and microbial peptides and the induction of EAU by the latter group of peptides have been clearly demonstrated, the specific role for mi- crobial proteins in the pathogenesis of uveitis is obscure. Molecular mimicry has been one of the hypotheses, and no convincing evi- dence has been brought forward.

The evidence for molecular mimicry in au- toimmune disease, however, remains mostly circumstantial at the present time. An alterna- tive theory is that autoimmune diseases are, in fact, primarily the result of infection and that of reactive autoantigens, which are nor- mally sequestered from autoreactive T cells, occurs because of bystander tissue damage releasing partially degraded antigens to be presented by local APCs to primed/activated T cells. This may, in part, explain the develop- ment of autoimmunity to corneal antigens in onchocerca infection.

In general, the course of autoimmune dis- ease characteristically exhibits intercalating exacerbations and remissions, or complete re- covery indicates that associated symptoms or the disease itself may resolve through the activity of the immune system, even after periods of profound dysfunction. This indi- cates that the autoimmune disease may be appropriately corrected by using natural im- munological reagents that participate in selec- tion of the lymphocyte repertoire under physi- ological conditions. Therefore, in the future

the autoimmune disease therapy should be based on utilization of natural components of the immune system, rather than immunosup- pressive drugs. This view points to the impor- tance of understanding the anatomy and physiology of the components of the immune system. The above hypothesis also suggests that one of the means of practical immune intervention in autoimmune states is through oral tolerance. In the future, research efforts should be directed to understanding the po- tential mechanisms operating in the induc- tion and maintenance of oral tolerance. The best experimental evidence for peripheral suppression is through cytokines secreted by Tht and Th2 cells. Until now oral tolerance was investigated in rodent models of autoim- mune disease; in the future it will be impor- tant to direct efforts toward understanding the phenomenon of oral tolerance through primate and human studies.

Acknowledgements

We would like to express our thanks to Dr. Timo- thy J. Schoen for helpful comments during preparation of the manuscript. Financial assistance from the In- dian Council of Medical Research, New Delhi, and Uttar Pradesh Council of Science and Technology, Lucknow, India, to V.K.S. is gratefully acknowledged. The laboratory infrastructure was provided by a JICA grant-in-aid to the SGPGt projects.

Experimental Autoimmune Uveitis Immunol Res 1996;15:323-346 339

Page 18: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

o l o ~ 1 7 6 1 7 6 1 7 6 1 4 9 e � 9 , � 9 �9 , ~ 1 4 9 1 4 9 �9 ~ , � 9 ~ 1 7 6 ~ ~ 1 7 6 � 9 1 4 9 o � 9 1 4 9 1 4 9 1 4 9 � 9 1 7 6 1 7 6 1 7 6 ~ 1 4 9 1 4 9 1 4 9 � 9 1 4 9 �9 �9 � 9 1 4 9 1 7 6 1 7 6 ~ 1 7 4 1 4 9 1 4 9 �9 �9 � 9 1 7 6 ~ 1 4 9 , o � 9 ~ 1 7 6 1 7 6 1 7 6 1 7 6 1 7 6 1 4 9 o , �9 ~ 1 7 6 1 7 6 ~ 1 7 6 1 4 9 o � 9 � 9 1 7 6 � 9 1 7 6 �9 �9 ~ , � 9 ~ 1 7 4 1 7 6 ~ 1 7 6 . ~ 1 7 6 . o , | 1 4 9

References

1 Faure JP: Autoimmunity and the retina. Curr Top Eye Res 1980;2: 215-302.

2 Wacker WB, Lipton NM: Experi- mental allergic uveitis. I. Production in the guinea-pigs and rabbit by im- munization with retina in adjuvant. J Immuno11968;101:151-156.

3 de Kozak Y, Sakai J, Thillaye B, Faure JP: S-antigen induced experi- mental autoimmune uveoretinitis in rats. Curr Eye Res 1981; 1:327-336.

4 Gery I, Mochizuki M, Nussenblatt RB: Retinal specific antigens and immunopathogenic processes they provoke; in Osborne NN, Chader GJ (eds): Progress in Retinal Re- search. New York, Pergamon Press, 1986, vol 5, pp 75-109.

5 Nussenblatt RB, Kuwabara T, de Monasterio FM, Wacker WB: S-an- tigen uveitis in primates. A new model for human disease. Arch Ophthalmo11981;99:1090-1092.

6 Nussenblatt RB, Gery I, Ballintine EJ, Wacker WB: Cellular immune responsiveness of uveitis patients to retinal S-antigen. Am J Ophthalmol 1980;89:173-179.

7 Hirose S, Don�9 LA, Shinohara T, Palestine AG, Nussenblatt RB, Gery I: Lymphocyte responses to an 18- mer peptide derived from the retinal S-antigen in uveitis patients. Jpn J Ophthalmo11990;34:298-305.

8 Nityanand S, Singh VK, Shinohara T, Paul AK, Singh V, Agarwal PK, Agarwal SS: Cellular immune re- sponse of uveitis patients to pcptide M, a retinal S-antigen fragment. J Clin Immuno11993;13:352-358.

9 Rajasingh J, Singh VK, Singh V, Sharma K, Agarwal SS: Cellular im- mune response to retinal S-antigen and interphotoreceptor retinoid binding protein fragments in uveitis patients. Indian J Med Res 1996; 103:222-226.

10 de Smet MD, Yamamoto JH, Mo- chizuki M, Gery I, Singh VK, Shino- hara T, Wiggert B, Chader GJ, Nus- senblatt RB: Cellular immune re- sponse of uveitis patients to retinal antigens and their fragments. Am J Ophthalmo11990;110:135-142.

11 Oldstone MBA: Molecular mimicry and autoimmune disease. Cell 50; 1987:819-820.

12 Shinohara T, Singh VK, Yamaki K, Abe T, Tsuda M, Suzuki S: S-anti- gen: Molecular mimicry may play a role in the autoimmune uveitis; in Osborne NN, Chader GJ (eds): Pro- gress in Clinical and Biological Re- search, Molecular Biology of the Retina: Basic and Clinically Rele- vant Studies. New York, Wiley Liss, 1991, vo1362, pp 163-190.

13 Weiner HL, Friedman A, Miller A, Khoury S J, A1-Sabbah A, Santos L, Sayegh M, Nussenblatt RB, Trent- ham DE, Hailer D: Oral tolerance: Immunologic mechanism and treat- ment of animal and human organ- specific autoimmune diseases by oral administration of nut�9 Annu Rev Immunol 1994;12:809- 837.

14 Chase MW: Inhibition of experi- mental drug allergy by prior feeding of the sensitizing agent. Proc Soc Exp Biol Med 1946;61:257-259.

15 Zhangs ZJ, Davidson L, Eisenbarth G, Weiner HL: Suppression of dia- betes in nonobese mice by oral ad- ministration of porcine insulin. Proc Natl Acad Sci USA 1991;88:10252- 10256.

16 Singh VK, Kalra HK, Yamaki K, Shinohara T: Suppression of experi- mental autoimmune uveitis in rats by the oral administration of the uveitopathogenic S-antigen frag- ment or a cross-reactive homologous peptide. Cell Immunol 1992;139: 81-90.

17 Nussenblatt RB, Caspi RR, Mahdi R, Chan CC, Roberge F, Lider O, Weiner HL: Inhibition of S-antigen induced experimental autoimmune uveoretinitis by oral induction of tolerance with S-antigen. J Immunol 1990;144:1689-1695.

18 Weiner HL, Mackin GA, Matsui M, Orav EJ, Khoury S J, Dawson DM, Haffler DA: Double-blind pilot trial of oral tolerization with myelin anti- gens in multiple sclerosis. Science 1993;259:1321-1324.

19 Trentham DE, Dynesius-Trentham RA, Orav E J, Combitchi D, Lorenzo C, Sewell KL, Haffler DA, Weiner HL: Effect of oral administration of collagen on rheumatoid arthritis. Science 1993;261:1727-1730.

20 Nussenblatt RB, Whitcup SM, de Smct MD, Caspi RR, Kozhich AT, Weiner HL, Vistica B, Gery I: In- traocular inflammatory disease (uvcitis) and the use of oral toler- ance: A status report. Ann NY Acad Sci 1996;778:325-337.

21 Wacker WB: Retinal autoimmunity and the path�9 of uveitis; in Helmsen R J, Suran A, Gery I, Nus- senblatt RB (eds): Proceeding 'Im- munology of the Eye; Workshop II'. Washington, Information Retrieval, 1981, pp 11-32.

22 Yajima S, Seki F, Takano S, Usui M: Localization of S-antigen by en- zyme-labeled antibody method and electron microscopy. Jpn J Ophthal- mo11983:27:526-534.

23 Kalsow CM, Wacker WB: Pineal reactivity of anti-retina sera. Invest Ophthalmol Vis Sci 1977;16:181- 184.

24 Kalsow CM, Wacker WB: Pineal gland involvement in retina-in- duced experimental allergic uveitis. Invest Ophthalmol Vis Sci 1978;17: 774-783.

25 Dorey C, Cozette J, Faure JP: A sim- ple and rapid method for isolation of retinal S-antigen. Ophthalmic Res 1982;14:249-255.

26 Waeker WB, Don�9 LA, Kalsow CM, Yankeelov JA Jr, Organisciak DT: Experimental allergic uveitis. Isolation, characterization, and lo- calization of a soluble uveitopatho- genie antigen from bovine retina. J Immunol 1977;119:1949-1958.

27 Kuhn H, Hall SW, Wilden U: Light induced binding of 48 kDa protein to photoreeeptor membranes is highly enhanced by phosphorylation of rhodopsin. FEBS Lett 1984;176: 473-478.

340 Immunol Res 1996;15:323-346 Singh/Nagaraju

Page 19: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

28 Shinohara T, Donoso LA, Tsuda M, Yamaki K, Singh VK: S-antigen: Structure, function and autoim- mune uveitis (EAU); in Osborne NN, Chader GJ (eds): Progress in Retinal Research. New York, Perga- mon Press, 1988, vol 8, pp 51-66.

29 Abe T, Shinohara T: S-antigen from the rat retina and pineal gland have identical sequences. Exp Eye Res 1990;5 t: l 11-112.

30 Shinohara T, Dietzschold B, Craft CM, Wistow G, Early J J, Donoso LA, Horwitz J, Tao R: Primary and secondary, structure of bovine reti- nal S-antigen (45 kDa protein). Proc Natl Acad Sci USA 1987;84:6975- 6979.

31 Tsuda M, Syed M, Bugra K, Wheal- an JP, McGinnis JF, Shinohara T: Structural analysis of mouse S-anti- gen gene. Gene 1988;73:11-20.

32 Tsuda M. Kikuchi T, Yamaki K, Shinohara T: The mouse S-antigen. Comparison with human and Dro- sophila. Eur J Biochem 1991;200: 95-101.

33 Yamaki K, Tsuda M, Shinohara T: The sequence of human retinal S- antigen reveals similarities with ct- transducin. FEBS Lett 1988;234: 39-43.

34 Yamaki K, Tsuda M. Kikuchi T, Chan KH, Huang KP, Shinohara T: The structural organization of the human S-antigen gent. J Biol Chem 1990:265:20757-20762.

35 Yamaki K, Takahashi Y, Sakuragi S, Matsubara M: Molecular cloning of S-antigen cDNA from bovine ret- ina. Biochem Biophys Res Commun 1987:142:904-910.

36 Abe T, Yamaki K, Tsuda M, Singh VK, Suzuki S, Mckinnon R, Klein DC, Donoso LA, Shinohara T: Rat pineal S-antigen: sequence analysis reveals presence of ct-transducin ho- mologous sequence. FEBS Lett 1989:247:307-311.

37 Shinohara T, Kukuchi T, Tsuda M, Yamaki K: A family of retinal S- antigens (arrestins) and their genes; comparative analysis of human, mouse, rat, bovine and drosophila. Comp Biochem Physiol 1992;103B: 505-509.

38 Ngo JT, Klisak I, Sparkes RS, Mo- handas T, Yamaki K, Shinohara T, Bateman JB: Assignment of the S- antigen gene (SAG) to chromosome 2q24-37. Genomics 1990;7:84-87.

39 Danciger M, Kozak CA, Tsuda M, Shinohara T, Farber DM: The gene for retinal S-antigen (48 kDa pro- tein) maps to the centromeric por- tion of mouse chromosome 1 near IDH-1. Genomics 1989;5:378-381.

40 Craft CM, Whitmore DH: The ar- restin superfamily: Cone arrestins are a fourth family. FEBS Lett 1995; 362:247-255.

41 Smith DP, Shieh BH, Zucker CS: Isolation and structure of an arrestin gene from Drosophila. Proc Natl Acad Sci USA 1990;87:1003-1007.

42 Smith WC, Greenberg RM, Caiman BG, Hendrix MM, Hutchinson L, Donoso LA, Battle BA: Isolation and expression of an arrestin cDNA from the horseshoe crab lateral eye. J Neurochem 1995;64:1-13.

43 Lohse M J, Benovic JL, Codina J. Caron MG, Lefkowitz RJ: 13-Arres- tin: A protein that regulates i3-adren- ergic receptor function. Science 1990;248:1547-1550.

44 Hyde DR. Mecklenburg KL, Pol- lock JA, Vihtelic TS, Benzer S: Twenty Drosophila visual system eDNA clones: One is a homolog of human arrestin. Proc Nail Acad Sci USA 1990;87:1008-1012.

45 Raming K, Freitag J, Krieger J, Breer H: Arrestin-subtypes in insect antennae. Ceil Signal 1993;5:69-80.

46 Sakumma H, Inana G, Murakami A, Higashide T, McLaren M J: lm- munolocalization of X-arrestin in human cone photoreceptors. FEBS Lett 1996;382:105-110.

47 Redmond TM, Wiggert B, Robey FA, Chader G J: lnterspecies conser- vation of structure of interphotore- ceptor retinoid binding protein. Similarities and differences as ad- judged by peptide mapping and N- terminal sequencing, giochem J 1986;240:19-26.

48 Borst DE, Redmond TM, Elser JE, Gonda MA, Wiggert B, Chader G J, Nickerson JM: lnterphotoreceptor retinoid binding protein. Gene char- acterization, protein repeat struc- ture, and its evolution. J Biol Chem 1989;264:11 I5-t 123.

49 Sanui H, Redmond TM, Kotake S, Wiggert B, Hu LH, Margalit H, Ber- zofsky JA, Chader GJ, Gery I: Iden- tification of an immunodominant and highly immunopathogenic de- terminant in the retinal interphoto- receptor retinoid binding protein (IRBP). J Exp Med 1989;169:1947- 1960.

50 lnoue H, Takeuchi M, Tanaka T, Usui M, Taguchi O: Analysis of the uveitopathogenic determinant in re- peat structure of retinal interphoto- receptor retinoid-binding protein (IRBP). Clin Exp Immunol 1994;97: 219-225.

51 Silver PB, Rizzo LV, Chan CC, Donoso LA, Wiggert B, Caspi RR: Identification of a major pathogenic epitope in human IRBP molecule recognized by mice of the H-2V hap- lotype. Invest Ophthalmol Vis Sci 1995;36:946-954.

52 Wacker WB, Rao NA, Marak GE Jr: Immunopathogenic responses of rabbits to 'S" antigen. I. Phlogistic characteristics associated with anti- gen source and sensitizing dose. Ophthalmic Rcs 1981;13:302-311.

53 Salinas-Carmona MC, Nussenblatt RB, Gery I: Experimental autoim- mune uveitis in the athymic nude rats. Eur J Immunol 1982;12:481- 484.

54 Caspi RR, Roberge FG, Chan CC, Wiggert B, Chader GJ. Rozenszajn LA, Lando Z, Nussenblatt RB: A new model for autoimmune disease. Experimental autoimmune uveoret- initis induced in mice with two dif- ferent retinal antigens. J lmmunol 1988;140:1490-1495.

55 lwase K. Fuji Y, Nakashima I, Kato N, Fujino Y, Kawashima H, Mochi- zuki M: A new method for induction of experimental autoimmune uveo- retinitis (EAU) in mice. Curr Eye Res 1990;9:207-216.

56 De Kozak Y, Mirshahi M, Boucheix C, Faure JP: Inhibition of experi- mental autoimmune uveoretinitis in rats by S-antigen-specific monoclon- al antibodies. Eur J Immunol 1985; 15:1107-t 111.

57 Nussenblatt RB, Rodrigues MM, Wacker WB, Cevario SJ, Salinas- Carmona MC, Gery I: Cyclosporin A: Inhibition of experimental au- toimmune uveitis in Lewis rats. J Clin Invest 1981;67:1228-1231.

Experimental Autoimmune Uveitis lmmunol Res 1996;15:323-346 341

Page 20: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

58 Mochizuki M, Kuwabara T, McA1- lister C, Nussenblatt RB, Gery I: Adoptive transfer of experi- mental autoimmune uveoretinitis in rats: Immunopathogenic mecha- nisms and histological features. In- vest Ophthalmol Vis Sci 1985;26: 1-9.

59 Donoso LA, Merryman CF, Shino- hara T, Sery TH, Smith A: Experi- mental autoimmune uveitis follow- ing immunization with small syn- thetic peptide. Arch Ophthalmol 1987;105:838-840.

60 Singh VK, Yamaki K, Donoso LA, Shinohara T: S-antigen: Experimen- tal autoimmune uveitis induced in guinea pigs with two synthetic pep- tides. Curt Eye Res 1988;7:87-92.

61 Singh VK, Nussenblatt RB, Donoso LA, Yamaki K, Chan CC, Shinoha- ra T: Identification of a uveito- pathogenic and lymphocyte prolifer- ation site in bovine S-antigen. Cell Immuno11988;115:413-419.

62 Gregerson DS, Merryman CF, Obritsch WE, Donoso LA: Identifi- cation of a potent new pathogenic site in human retinal S-antigen which induces experimental au- toimmune uveoretinitis in LEW rats. Cell Immunol 1990;128:209- 219.

63 de Smet MD, Bitar G, Roberge FG, Gery I, Nussenblatt RB: Human S- antigen: Presence of multiple immu- nogenic and immunopathogenic sites in the Lewis rat. J Autoimmun 1993;6:587-599.

64 Barnett LA, Fujinami RS: Molecu- lar mimicry: A mechanism for au- toimmune injury. FASEB J 1992;6: 840-844.

65 Forrester JV, Lumsden L, Liver- sidge J, Kuppner M, Mesri M: Im- munoregulation of uveoretinal in- flammation; in Osborne NN, Chad- er GJ (eds): Progress in Retinal and Eye Research. New York, Pergamon Press, 1995, vol 14, pp 393-412.

66 Singh VK, Kalra HK, Yamaki K, Donoso LA, Abe T, Shinohara T: Molecular mimicry between the uveitopathogenic site of retinal S- antigen and viral peptides: Induc- tion of experimental autoimmune uveitis in Lewis rats. J lmmunol 1990;144:1282-1287.

67 Singh VK. Yamaki K, AbeT, Shino- hara T: Molecular mimicry between uveitopathogenic site of retinal S- antigen and Escherichia coli protein: Induction of experimental autoim- mune uveitis and lymphocyte cross- reaction. Cell lmmunol 1989;122: 262-273.

68 Singh VK, Yamaki K, Donoso LA, Shinohara T: Molecular mimicry: Yeast histone H3 induced experi- mental autoimmune uveitis. J Im- muno11989;142:1512-1517.

69 Singh VK, Usukura J, Shinohara T: Molecular mimicry: Uveitis in- duced in Macaca fasicularis by mi- crobial protein having sequence ho- mology with retinal S-antigen. Jpn J Ophthalmo11992;36:108-116.

70 Singh VK, Yamaki K, Donoso LA, Shinohara T: Sequence homology between yeast histone H3 and uvei- topathogenic site of S-antigen: Lym- phocyte cross-reaction and adoptive transfer of the disease. Cell Immu- nol 1989; 119:211-22 I.

71 Acha-Orbea H: Bacterial and viral superantigens: Roles in autoimmu- nity. Ann Rheum Dis 1993;52:$6- 16.

72 Friedman SM, Tumang JR, Crow MK: Microbial superantigens as etiopathogenic agents in autoimmu- nity. Rheum Dis Clin North Am 1993; 19:207-222.

73 Fujinami RS, Oldstone MBA: Ami- no acid homology between the en- cephalitogenic site of myelin basic protein and virus: Mechanism for autoimmunity. Science 1985;230: 1043-1045.

74 Schwimmbeck PL, Yu DTY, Old- stone MBA: Autoantibodies to HLA-B27 in the sera of HLA-B27 patients with ankylosing spondylitis and Reiter's syndrome. J Exp Med 1987;166:173-181.

75 Lahesmaa R, Skurnik M, Toivanen P: Molecular mimicry: Any role in the pathogenesis of spondyloar- thropathies. Immunol Res 1993; 12: 193-208.

76 Fielder M, Pirt S J, Tarpey I, Wilson C, Cunningham P, Ettelaie C, Bin- der A, Bansal S, Ebringer A: Molec- ular mimicry and ankylosing spon- dylitis: Possible role of a novel se- quence in pullulanase of Klebsiella pneumoniae. FEBS Lett 1995;369: 243-248.

77 Beisel KW, Srinivasappa J, Prabha- kar BS: Identification of a putative shared epitope between coxsackie virus B4 and a-cardiac myosin heavy chain. Clin Exp lmmunol 1991;86:49-55.

78 Gauntt C J, Arizpe HM, Higdon AL, Wood H J, Bowers DF, Rozek MM, Crawley R: Molecular mimicry, anti-coxsackievirus B3 neutralizing monoclonal antibodies, and myo- carditis. J Immunol 1995;154: 2983-2995.

79 Danieli MG, Markovits D, Gabrielli A, Corvetta A, Giorgi PL, van der Zee R, van Embden JDA, Danieli G, Cohen IR: Juvenile rheumatoid arthritis patients manifest immune reactivity to the mycobacterial 65 kDa heat shock protein, to its 180- 188 peptide and to a partially ho- mologous peptide of the proteogly- can link protein. Clin Immunol Im- munopathol 1992;64:121-128.

80 Quayle AJ, Wilson KB, Li SG, Kjeldsen-Kragh J, Oftung F, Shin- nick T, Sioud M, Forre O, Capra JD, Natvig JB: Peptide recognition, T- cell receptor usage and HLA restric- tion elements of human heat shock protein (hsp) 60 and mycobacterial 65 kDa hsp-restrictive T-cell clones from rheumatoid synovial fluid. Eur J Immunol 1992;22:1315-1322.

81 Wucherpfennig KW, Strominger JL: Molecular mimicry in T cell-me- diated autoimmunity: Viral pep- tides activate human T cell clones specific for myelin basic protein. Cell 1995;80:695-705.

82 Garza KM, Tung KSK: Frequency of molecular mimicry among T-cell peptides as the basis of autoimmune disease and autoantibody induction. J lmmuno11995;155:5444-5448.

83 Kagnoff MF, Paterson YJ, Kumar P J, Kasarda DD, Carbone FR, Uns- worth D J, Austin RK: Evidence for the role of a human intestinal adeno- virus in the pathogenesis of coeliac disease. Gut 1987;28:995-1001.

84 Baum H, Brusik V, Choudhuri K, Cunningham P, Vergani D, Peak- man M: MHC molecular mimicry in diabetes. Nature Med 1995; 1:388.

85 Jones DB, Armstrong NW: Cox- sackie virus and diabetes revisited. Nature Med 1995;1:284.

342 Immunol Res 1996:15:323-346 Singh/Nagaraju

Page 21: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

86 Uchida J: Electron microscopic study of microfold ceils (M cells) in normal and inflamed human appen- dix. Gastroenterol Jpn 1988;23: 251-262.

87 Owen RL, Paizza AJ, Ermak TH: Ultrastructural and cytoarchitectur- al features of lymphoreticular or- gans in colon and rectum of adult BALB/c mice. Am J Anat 1991;190: 10-18.

88 Craig SW, Cebra JJ: Peyer's patches: An enriched source of precursors for IgA-producing immunocytes in rab- bit. J Exp Med 1971;134:188-200.

89 Blumberg RS, Yockey CE, Gross GG, Ebert EC, Balk SP: Human in- testinal intraepithelial lymphocytes are derived from a limited number of T cell cIones that utilize multiple V!3 T-celt receptor genes. J Immunol 1993;150:5144-5153.

90 Guy-Grand D, Cerf-Bensussan N, Malissen B, Malassis-Seris M, Briot- tet C, Vassalli P: Two gut intraepi- thelial CD8 § lymphocyte popula- tions with different T cell receptors: A role for the gut epithelium in T cell differentiation. J Exp Med 1991; 173:471-481.

91 Rocha B, Vassalli P, Guy-Grand D: The extrathymic T cell development pathway. Immunol Today 1992;13: 449-454.

92 Mayrhofer G: Thymus dependcnt and thymus independent subpopu- lations ot" intestinal intraepithelial lymphocytes: A granular subpopula- tion of probable bone marrow origin and relationship to mucosal mast cells. Blood 1980;55:532-535.

93 Klein JR: Ontogeny of the Thy-l-, Lyt-2* murine intestinal intraepi- thelial lymphocyte. Characteriza- tion of a unique population of thy- mus independent cytotoxic effector cells in the intestinal mucosa. J Exp Med 1986; 164:309-314.

94 Mosley RL, Styre D, Klein JR: Dif- ferentiation and functional matura- tion of bone marrow derived intesti- nal epithelial T cells expressing membrane T cell receptor in athy- mic radiation chimeras. J Immunol 1990;145:1369-1375.

95 De Geus B, Van den Enden M, Coolen C, Nagelkerken L, Van der Heijden P, Rozing J: Phenotype of intraepithelial lymphocytes in eu- thymic and athymic mice: Impli- cations for differentiation of cells bearing a CD3-associated "y/8 T cell receptor. Eur J Immunol 1990; 20:291-298.

96 Bandeira A, ltohara S, Bonneville M, Burlen-Defranouex O, Mota- Santos T, Coutinho A, Tonegawa S: Extrathymic origin of intestinal in- traepithelial lymphocytes bearing T cell antigen receptor y/8. Proc Natl Acad Sci USA 1991;88:43-47.

97 Rocha B, yon Boehmer H, Guy- Grand D: Selection of intraepithe- lial lymphocytes with CD8 ala co- receptors by self antigen in the mu- rine gut. Proc Natl Acad Sci USA 1992;89:5336-5340.

98 Barrett TA, Delvy ML, Kennedy DM, Lefrancois L, Marls LA, Dent AL, Hedrick SM, Bluestone JA: Mechanism of self-tolerance of 3'/8 T cells in epithelial tissues. J Exp Med 1992;175:65-70.

99 Poussier P, Julius M: Maturation of T cells in the intestinal epithe- lium. lmmunol Today 1993;14: 140-141.

100 Bockman DE, Cooper MD: Pino- cytosis by epithelium associated with lymphoid follicles in the bur- sa of Fabricius, appendix and Peyer's patches. An electron mi- croscopic study. Am J Anat 1973; 136:455-478.

101 Hill SM, Milla PJ, Bottazzo GF, Mirakian R: Autoimmune enter- opathy and colitis: Is there a gener- alised autoimmune gut disorder? Gut 1991;32:36-42.

102 Godefroy O, Huet C, Blair LAC, Sahuquillo-Merino C, Louvard D: Differentiation of a clone isolated from HT29 cell line: polarized dis- tribution of histocompatibility an- tigens (HLA) and of transferrin re- ceptors. BiN Cell 1988;63:41-55.

103 Mayrhofer G, Spargo LDJ: Distri- bution of class II major histocom- patibility antigens in enterocytes of the rat jejunum and their asso- ciation with organelles of the endo- cytic pathway. Immunology 1990; 70:11-19.

104 Brandtzaeg P, Sollid LM, Thrane PS, Kvale D, Bjerke K, Scott H, Kett K, Rognum TO: Lymphoepi- thelial interactions in the mucosal immune system. Gut 1988;29: 1116-1130.

105 Rubin D, Weiner HL, Fields BN, Greene MI: Immunologic toler- ance after oral administration of reovirus: Requirement for two vi- ral gene products for tolerance in- duction. J Immunol 1981;127: 1697-1701.

106 Mowat AM, Thomas M J, Macken- zie S, Parrott DMV: Divergent ef- fects of bacterial lipopolysaccha- ride on immunity to orally admin- istered protein and particulate an- tigens in mice. Immunology 1986; 58:677-683.

107 Lamont AG, Mowat AM, Parrott DMV: Priming of systemic and lo- cal delayed type hypersensitivity responses by feeding low doses of ovalbumin to mice. Immunology 1989;66:595-599.

108 Friedman A, Weiner HL: Induction of anergy or active suppression fol- lowing oral tolerance is determined by antigen dosage. Proc Natl Acad Sci USA 1994;91:6688-6692.

109 Lamont AG, Mowat AM, Brown- ing M J, Parrott DMV: Genetic control of oral tolerance to ovalbu- rain in mice. Immunology 1988; 63:737-739.

110 Peng HJ, Turner MW, Strobel S: Failure to induce tolerance for de- layed hypersensitivity to protein antigens in neonatal mice can par- tially be corrected by spleen cell transfer. Pediatr Res 1989;26: 486-490.

111 Mowat AM, Lamont AG, Strobel S, Mackenzie S: The role of antigen processing and suppressor T-cells in immune responses to dietary proteins in mice. Adv Exp Med Biol 1987;216A:709-720.

112 Hanson DG: Ontogeny of orally induced tolerance to soluble pro- tein in mice. I. Priming and toler- ance in newborns. J lmmunol 1981;127:1518-1524.

113 Strobel S, Ferguson A: Immune re- sponses to fed protein antigens in mice. 3. Systemic tolerance or priming is related to age at which antigen is encountered. Pediatr Res 1984;18:588-594.

Experimental Autoimmune Uveitis Immunol Res 1996;15:323-346 343

Page 22: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

114 Faria AMC, Garcia G, Rios MJC, Michalaros CL, Vaz NM: Decrease in susceptibility to oral tolerance induction and occurrence of oral immunization to ovalbumin in 20-38-week-old mice. The effect of interval between oral exposures and the rate of antigen intake in the oral immunization. Immunol- ogy 1993;78:147-151.

115 Lamont AG, Gordon M, Ferguson A: Oral tolerance in protein de- prived mice. I. Profound antibody tolerance but impaired DTH toler- ance after antigen feeding. Immu- nology 1987:61:333-337.

116 Kitamura K, Kiyono H, Fujihashi K, Eldridge JH, Green DR, MC Ghee JR: Contrasuppressor cells that break oral tolerance are anti- gen-specific T-cells distinct from T-helper (L3T4*), T-suppressor (Lyt-2*) and B cells. J lmmunol 1987; 139:3251-3259.

117 Walker WA: Role of the mucosal barrier in antigen handling by gut; in Broshoff J, Challacomer SJ (eds): Food Allergy and Intoler- ance. Eastbourne, Saunders, 1987, pp 209-222.

118 Khoury S J, Hancock WW, Weiner HL: Oral tolerance to myelin basic protein and natural recovery from experimental autoimmune encephalomyelitis are associated with downregulation of inflamma- tory cytokines and differential upregulation of transforming growth factor 13, interleukin-4, and prostaglandin E expression in the brain. J Exp Med 1992;176:1355- 1364.

119 Arnold B, Schonrich G, Haem- merling G J: Induction of tolerance in mature peripheral T cells. Exp Nephrol 1993; 1:72-77.

120 Jenkins MK, Schwartz RH: Anti- gen presentation by chemically modified splenocytes induces anti- gen specific T cell unresponsive- ness in vitro and in vivo, J Exp Med 1987;165:302-319.

121 Schwartz RH: A cell culture model for T lymphocyte clonal anergy. Science 1990;248:1349-1356.

122 Burkly LC, Lo D, Kanagawa O, Brinster RL, Flavel RA: T cell tol- erance by clonal anergy in trans- genic mice with nonlymphoid ex- pression of MHC class-lI I-E. Na- ture 1989;342:564-566.

I23 Rammensee HG, Kroschewski R, Frangoulis B: Clonal anergy in ma- ture V136 + T lymphocytes on im- munizing Mls I a mice with Mls I a expressing cells. Nature 1989;339: 541-544.

124 Schonrich G, Kalinke U, Morn- burg F, Malissen M, Schmitt-Ver- hulst AM, Malissen B, Hammer- ling G J, Arnold B: Downregula- tion ofT cell receptors on self-reac- tive T-cells as a novel mechanism for extrathymic tolerance induc- tion. Cell 1991;65:293-304.

125 Melamed D, Friedman A: Direct evidence for anergy in T lympho- cytes tolerized by oral administra- tion ofovalbumin. Eur J Immunol 1993;23:935-942.

126 Friedman A, Weiner HL: Induc- tion of anergy and/or active sup- pression in oral tolerance is deter- mined by frequency of feeding and antigen dosage. J lmmunol 1993; 150:4A.

127 Mowat AM: The regulation of im- mune responses to dietary protein antigens. Immunol Today 1987;8: 93-98.

128 Jones LA, Chin LT, Longo DL, Kruisbeck AM: Peripheral clones elimination of functional T cells. Science 1990;250:1726-1729.

129 Kisielow P, Bluthmann H, Staerz UD, Steinmetz M, yon Boehmer H: Tolerance in T-cell receptor transgenic mice involves deletion ofnonmature CD4§ § thymocytes. Nature 1988;333:742-746.

130 Sha WC, Nelson CA, Newberry RD, Krantz DM, Russel JH, Loh DY: Positive and negative selec- tion of an antigen receptor on T cells in transgenic mice. Nature 1988;336:73-76.

131 Whitacre CC, Gienapp IE, Zhang X, Herber-Katz E: Oral tolerance in experimental autoimmune en- cephalomyelitis (EAE): A research for the MBP specific T cell recep- tor. FASEB J 1990;4:A1856.

132 Fujihashi K, Taguchi T, Aicher WK, McGhee JR, BlueStone JA, Eldridge JH, Kiyono H: Immuno- regulatory functions of murine in- traepithelial lymphocytes: ~,/5 T cell receptor positive (TCR*) cells abrogate oral tolerance, while o,/13 TCR* T cells provide B ceil help. J Exp Med 1992;175:695-707.

133 Mowat AM, Strobel S, Drummond HE, Ferguson A: Immunological response to fed protein antigens in mice. I. Reversal of oral tolerance to ovalbumin by cyclophospha- mide. Immunology 1982;45:105- 113.

134 Mowat AM: Depletion of suppres- sor T cells by 2'deoxyguanosine abrogates tolerance in mice fed ov- albumin and permits the induction of intestinal delayed type-hyper- sensitivity. Immunology 1986;58: 179-184.

135 Chen Y, lnobe J, Marks R, Gon- nella P, Kuchroo VK, Weiner HL: Peripheral deletion of antigen reactive T cells in oral tolerance. Nature 1995;376:177-180.

136 Gershon RK: T cell control of anti- body production. Contemp Top Immunobiol 1974;3:1-40.

137 Gershon RK. Liebhaber S, Ryu S: T-cell regulation of T-cell re- sponses to antigen. Immunology 1974;26:909-923.

138 Hutchison IV: Specific Immuno- suppression; in Morris PJ (ed): Kidney Transplantation: Princi- ples and Practice. Orlando. Grune & Stratton, 1984, pp 301-318.

139 Richman LK, Chiller JM, Brown WR, Hanson DG. Vaz NM: Enter- ically induced immunological tol- erance. I. Induction of suppressor T lymphocytes by intragastric ad- ministration of soluble proteins. J Immunol 1978; 121:2429-2433.

140 Hanson DG, Vaz NM, Maia LCS. Lynch JM: Inhibition of specific immune responses by feeding pro- tein antigens. III. Evidence against maintenance of tolerance to oval- bumin by orally induced anti- bodies. J lmmunol 1979;123; 2337-2343.

344 Immunol Res 1996:15:323-346 Singh/Nagaraju

Page 23: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

I41 Green DR, Gold J, St Martin S, Gershon R, Gershon RK: Mi- croenvironmental immunoregula- tion: Possible role of contra sup- pressor cells in maintaining im- mune responses in gut-associated lymphoid tissues. Proc Natl Acad Sci USA 1982;79:889-892.

142 Mowat AM, Lamont AG, Parrott DMV: Suppressor T cells, antigen presenting cells and the role of l-J restriction in oral tolerance to ov- albumin. Immunology 1988;64: 141-145.

143 Ferguson A, Mowat AM, Strobel S: Abrogation of tolerance to fed anti- gens and induction of cell me- diated immunity in the gut associ- ated lymphoreticular tissues. Ann NY Acad Sci 1983;409:486-497.

144 Mac Donald'IT: lmmunosuppres- sion caused by antigen feeding. I. Evidence for the activation of a feedback suppressor pathway in the spleens of antigen ted mice. Eur J Immunot 1982;t2:767-773.

145 Sykes M, Sachs DH: Mechanisms of suppression in mixed allogenic chimeras. Transplantation 1988; 46:135S.

i46 Damle NK, Engleman EG: Anti- gen-specific suppressor T lympho- cytes in man. Clin Immunol lm- munopatho11989;53:S17.

147 Hall BM, Jclbart ME, Gurtey KE, Dorsch SE: Specific unresponsive- ncss in rats with prolonged cardiac allograft survival alter treatment with cyclosporin: Mediation of specific suppression by T helper/ inducer cells. J Exp Med 1985;162: 1683-1694.

148 Asherson GL, Colizzi V, Zembala M: An overview of T-suppressor cell circuits. Ann Rev Immunol 1986;4:37-68.

149 Holda JH, Maier T, Claman HN: Murine graft-versus-host disease across minor harriers: lmmuno- suppressive aspects of natural sup- pressor cells, lmmunol Rev 1985: 88:87-105.

150 Strober S, Palathumpat V, Schwa- dron R, Hertel-Wulff B: Cloned natural suppressor cells prevent graft-vs-host disease. J Immunol 1987;138:699-703.

t51 Lider O, Santos LMB, Lee CSY, Higgins P J, Weiner HL: Suppres- sion of experimental autoimmune encephalomyelitis by oral admin- istration of myelin basic protein. II. Suppression of disease and in vitro response is mediated by anti- gen-specific CD8 § T-lymphocytes. J Immunol 1989; 142:748-752.

152 Miller A, Lider O, Roberts AB, Sporn MB, Weiner HL: Suppres- sor T cells generated by oral toleri- zation to myelin basic protein sup- press both in vitro and in vivo immune responses by the release of transforming growth factor-13 following antigen specific trigger- ing. Proc Natl Acad Sci USA 1992; 89:421-425.

153 Yoshino S, Quattrocchi E, Weiner HL: Suppression of antigen in- duced arthritis in Lewis rats by oral administration of type II col- lagen. Arthritis Rheum 1995;38: 1092- 1096.

154 Khoury SJ, Lider O, AI-Sabbagh A, Weiner HL: Suppression of ex- perimental autoimmune encepha- lomyelitis by oral administration of myelin basic protein, llI. Syner- gistic effect of lipopolysaccharide. Cell Immuno11990;131:302-310.

155 Rizzo LV, Miller-Rivero NE, Chan CC, Wiggcrt B, Nussenblatt RB, Caspi RR: Interleukin-2 treat- ment potentiates induction of oral tolerance in a murine model ofau- toimmunity. J Clin Invest 1994; 94:1668-1672.

156 Caspi RR, StiffLR, Morawetz R, Miller-Rivero NE, Chan CC, Wig- gert B, Nussenblatt RB, Morse HC III, Rizzo LV: Cytokine modula- tion of oral tolerance in a murine model of autoimmune uveitis. Ann NY Acad Sci 1996;778:315- 324.

157 Zamvil S, Nelson P, Trotter J, Mit- chell D, Knobler R, Fritz R, Stein- man L: T-cell clones specific for myelin basic protein induce chronic relapsing paralysis and de- myelination. Nature 1985:317: 355-358.

I58 Hu LH, Redmond MT, Sanui H, Kuwabara T, McAIlister CG, Wig- gert B, Chader GJ, Gery I: Rat T- cell lines specific to a nonimmuno- dominant determinant of a retinal protein (IRBP) produce uveoreti- nitis and pinealitis. Cell Immunol 1989;122:251-261.

159 Trentham DE, Dynesius RA, Da- vid JR: Passive transfer by cells of type II collagen-induced arthritis in rats. J Clin Invest 1978;62:359- 366.

160 Serreze DV, Leiter EH, Worthen SM, Shultz LD: NOD marrow stem cells adoptively transfer dia- betes to resistant (NOD x NON)FI mice. Diabetes 1988;37: 252-255.

161 Sakaguchi S, Fukuma K, Kuri- bayashi K, Masuda T: Organ-spe- cific autoimmune diseases in- duced in mice by elimination ofT- cell subset. I. Evidence tbr the ac- tive participation ofT-cells in nat- ural self tolerance; Deficit of a T cell subset as a possible cause of autoimmune disease. J Exp Med 1985;161:72-87.

162 Londei M, Bottazzo GF, Fold- mann M: Human T-cell clones from autoimmune thyroid glands: Specific recognition of autologous thyroid cells. Science 1985;228: 85-89.

163 Koike T, ltoh Y, Ishii T, Ito I, Takabayashi K, Maruyama N, Tomioka H, Yoshida S: Pre- ventive effect of monoclona[ anti- L3T4 antibody on development of diabetes in NOD mice. Diabetes 1987;36:539-541.

164 Ota K, Matsui M, Milford EL, Mackin GA, Weiner HL, Hailer DA: T-cell recognition of an im- munodominant myelin basic pro- tein epitope in multiple sclerosis. Nature 1990;346:183-187.

165 Kotzin BL, Karuturi S, Chou YK, Lafferty J, Forrester JM, Better M, Nedwin GE, Offner H, Vanden- bark AA: Preferential T-cell recep- tor 13-chain variable gene use in myelin basic protein-reactive T- cell clones from patients with mul- tiple sclerosis. Proc Natl Acad Sci USA 1991;88:9161-9165.

Experimental Autoimmune Uveitis Immunol Res 1996; 15:323-346 345

Page 24: Experimental autoimmune uveitis: Molecular mimicry and oral tolerance

166 Davies TF, Martin A. Concepcion ES, Graves P, Cohen L, Ben-Nun A: Evidence of limited variability of antigen receptors in intrathyroi- dal T-cell in autoimmune thyroid disease. N Engl J Med 1991;325: 238-244.

167 Harcourt GC, Sommer N, Roth- bard J, Willcox HNA, Newsom- Davis J: A juxt amembrane epi- tope on the human acetylcholine receptor recognized by T-cells in myasthenia gravis. J Clin Invest 1988;82:1295-1300.

168 Kontiainen S, Toomath R, Lowder J, Feldmann M: Selective activa- tion of T-cells in newly diagnosed insulin-dependent diabetic pa- tients: evidence of heterogeneity of T-cell receptor usage. Clin Exp lm- munol 1991;83:347-351.

169 Lohr H, Manns M, Kyriatsoulis A, Lohse AW, Trautwein C, Meyer- Zum Bushenfelde KH, Fleischer B: Clonal analysis of liver-infil- trating T-cells in patients with LKM-I antibody-positive autoim- mune chronic active hepatitis. Clin Exp Immunol 1991;84:297- 302.

170 Giegerich G, Pette M, Meinl E, Epplcn JT. Wekerle H, Hinkkancn A: Diversity of T-cell receptor ct and ~ chain genes expressed by hu- man T cells specific for similar my- elin basic protein pcptide/major histocompatibility complexes. Eur J lmmunol 1992;22:753-758.

171 Lehmann PV, Sercarz EE, Forsthuber T, Dayan CM, Gam- mon G: Determinant spreading and the dynamics of the autoim- mune T cell repertoire. Immunol Today 1993; 14:203-208.

172 Mc Carton RM, Fallis RJ, McFar- lin DE: Alterations in T-cell anti- gen specificity and class II restric- tion during the course of chronic relapsing experimental allergic en- cephalomyelitis. J Neuroimmunol 1990;29:73-79.

173 Zhang ZJ, Lee CSY, Lider O, Weiner HL: Suppression of adju- vant arthritis in Lewis rats by oral administration of type II collagen. J lmmuno11990;145:2489-2493.

174 Vrabec TR, Gregerson DS, Dua HS, Donoso LA: Inhibition of ex- perimental autoimmune uveoreti- nitis by oral administration of S- antigen and synthetic peptides. Autoimm unity 1992; 12:175-184.

175 Thurau SR, Chan CC, Suh E, Nus- senblatt RB: Induction of oral tol- erance to S-antigen induced exper- imental autoimmune uveitis by a uveitogenic 20 mer peptide. J Au- toimmun 1991;4:507-516.

176 Gregerson DS, Obritscb WF, Do- noso LA: Oral tolerance in experi- mental autoimmune uveoretinitis. Distinct mechanisms of resistance are induced by low vs high dose feeding protocols. J Immunol 1993; 151:5751-5761.

177 Vistica BP, Chanaud NP III, Felix N, Caspi RR, Rizzo LV, Nussen- blatt RB, Gery I: CD8 T-cells are not essential for the induction of low-dose oral tolerance. Clin Im- munol Immunopathol 1996;78: 196-202.

178 Dick AD, Cheng YF, Liversidge J, Forrester JV: Intranasal adminis- tration of retinal antigens sup- presses retinal antigen-induced ex- perimental autoimmune uveoret- initis. Immunology 1994;82:625- 631.

179 Singh VK, Anand R, Sharma K, Agarwal SS: Suppression of exper- iental autoimmune uveitis in Lewis rats by oral administration of recombinant Escherichia coli expressing retinal S-antigen. Cell Immunol 1996:172:158-162.

346 Immunol Res 1996; 15:323-346 Singh/Nagaraju