key: cord-0040352-ym5z4yb5 authors: Lim, Wee-Kiak; Nussenblatt, Robert B. title: Retinal Autoimmunity date: 2012-03-21 journal: Retina DOI: 10.1016/b978-0-323-02598-0.50019-7 sha: 802833d3e1a94c220d3a9964488bd03c8ae76626 doc_id: 40352 cord_uid: ym5z4yb5 nan The eye is a unique and complex organ that has developed multiple mechanisms to protect itself against immunological attacks and inflammation in order to maintain its delicate structure, both anatomically and physiologically. It was one of the first sites of immune privilege recognized more than 100 years ago. 1 This immune privilege has served well to curb excessive inflammation and preserve function. However, this privilege may be lost in certain conditions and situations, resulting in retinal autoimmunity. For a long time, retinal autoimmunity has been perceived as pathogenic and active suppression of retinal immunity was presumed necessary to maintain the health of the eye. However, studies have shown the presence of retinal autoantibodies in normal controls, 2 and constitutive expression of proinflammatory ligands have been found in the normal retina in high concentration. 3 Moreover, animal optic nerve injury studies revealed possible beneficial roles of retinal autoimmunity in controlling collateral damage to the retinal ganglion cells. 4 Thus, retinal autoimmunity can be viewed as a "double-edged sword" with both protective and destructive effects. To understand retinal autoimmunity, we first have to understand some basic concepts in immunology and how some of these immunological components are at work to maintain the eye's immune privilege. There are two systems of immunity, innate and adaptive, coexisting to keep pathogens out. Innate immunity, phlyogenetically earlier, is nonspecific and immediate in response. It is the first immune response mounted against invading pathogens. The key components of innate immunity are a cadre of leukocytes and plasma proteins that are capable of detecting and destroying pathogens. These leukocytes include polymorphonuclear cells, monocytes, macrophages, natural killer (NK) cells, eosinophils, and basophils and the plasma proteins belong to families of the complement cascade, clotting cascade, and acute-phase reactants. In contrast, adaptive immunity, as the name implies, is antigen-specific, adaptive, transferable, and has other cardinal features like immunologic memory and tolerance. Adaptive immunity has evolved and developed novel molecules like the antibody and the T-cell receptor (TCR), which come in almost limitless varieties to recognize most molecules of biological interest, including both self and nonself. These molecules, developed in the absence of exogenous stimulation, have unique configurations that confer specificity in antigen recognition. The key cellular components of adaptive immunity are the T and B lymphocytes which possess unique receptors for recognizing billions of different antigenic epitopes. 5 Each clone of lymphocyte expresses molecularly identical receptors on the surface, hence, in order to recognize billions of different epitopes, there are billions of unique lymphocytes. Upon maturation in the thymus (T cells) and the bone marrow (B cells), cells remain quiescent in G 0 of the cell cycle until they encounter the complementary antigen of sufficient affinity to their receptors. Binding with the receptors in the presence of appropriate costimulation is the initiating event to trigger an immune response targeting elimination of the complementary molecule. If this molecule is pathogen-derived, activation will lead to the elimination of the pathogen, but if the molecule is self-derived then activation will potentially lead to autoimmunity, tissue injury, disease, and the destruction of host tissue. Activation of lymphocytes results in the emergence of clones of lymphocytes through proliferation, each with a unique receptor to recognize one epitope of the inciting antigen. Some of these clones will become memory cells, which are responsible for the enhanced immune response if the antigen is re-encountered. The rest differentiate into effector cells such as B cells, synthesizing antibodies mediating humoral immunity, and T cells secreting cytokines targeted at other cells mediating cellular immunity. Both immune and accessory cells, including those of the innate immunity system, are activated to eliminate the pathogens. In this way, adaptive immunity serves to enhance the immune protection through better-coordinated and more specific attacks using innate immunity mechanisms. Within the genome lies the ability to create antibodies and TCR for antigens that binds to self antigens with high affinity. 6 Hence, the threat of autoimmunity is inborn and regulation is crucial. The ability to distinguish between foreign and self, and the ability to regulate autoimmunity, are critical for survival. processed, transported, and finally presented to the lymphocytes. As the naive lymphocytes can only be activated in the organized lymph tissues in secondary lymphoid organs such as lymph nodes, spleen, tonsils, and Peyer's patches, it needs specialized cells to execute the immune afferent limb. These cells are professional antigen presenting cells (APCs). Dendritic cells and macrophages, both bone marrow-derived, have APC functions serving to capture antigens through phagocytosis and endocytosis, process the antigens, and present the processed antigen in conjunction with special major histocompatibility complex (MHC) molecules on their cell surface. In addition, the APCs also provide the necessary costimulation needed for lymphocyte activation by upregulating an array of surface molecules (CD80, CD86, intercellular adhesion molecule-1 (ICAM-1), lymphocyte function associated molecule-3 (LFA-3), and CD40) that function as ligands for receptors expressed by the lymphocytes. They also secrete cytokines such as interleukin (IL)-12, IL-6, IL-10, and IL-1β serving similar costimulation functions. Costimulation is required as the second signal for full activation of the lymphocytes, independent of the first signal, which is the antigen and the lymphocyte receptor engagement. 7 There are differences in antigen presentation to B and T lymphocytes. For B lymphocytes, the receptor which is a surface-bound antibody can engage a naive antigen directly while TCRs can only recognize peptide fragments presented on special surface molecules (MHC classes I and II). MHC class I, which is present in most cells, presents peptides derived from protein degradation in the cytoplasm (intracellular antigens such as viral and intracellular microbe products), while MHC class II molecules, which are present in APCs and lymphocytes, present peptides from phagocytic vesicles (extracellular antigens from the microenvironment). Upon activation, the first cells to respond are the CD4+ T cells specific for the presented peptide on the MHC class II molecules of the APCs. The CD4+ lymphocytes will proliferate and secrete an array of cytokines, including IL-2, IL-3, granulocyte-macrophage colony-stimulating factor, interferon (IFN)-gamma, and IL-4. These cytokines serve as growth and stimulation factors for the lymphocytes and APCs, hence amplifying the proliferation process. In addition, IL-2, IFN gamma and IL-4 also promote the differentiation of CD8+ T lymphocytes to mature cytotoxic T lymphoctes (CTLs) which recognize antigen-derived peptides presented on class I MHC molecules. Upon activation, these CD8+ CTLs function as effectors to lyse the targeted cells and also produce proinflammatory cytokines, especially IFN-gamma. The cytokine production by CD4+ T cells also promotes activation and differentiation of B lymphocytes. IFN-gamma and IL-2 stimulate B cells to produce complementfixing immunoglobulin G (IgG) antibodies, while IL-4, IL-5, IL-6, and IL-10 result in the production of noncomplementfixing IgG, IgE, or IgA antibodies. The final products after the central processing in the lymphoid tissues are immune effectors. These are the CD4+, CD8+ T cells and B cells that have receptors specific for the inciting antigen. They are transported, predominantly via a hematogenous route, to the site of the inciting antigens to execute their effector function, thereby completing the efferent loop of the immune response. At the target sites, sites of infection and inflammation, vessels are leaky, the vascular endothelial cells display ligands that bind to receptors on the immune cells, and chemokines secreted by the local inflammatory cells serve to attract more immune effector cells to the site. While the engagement and activation of B cells can be direct, activation of the T cells still need the APCs for antigen recognition. The activated CD4+ T cells secrete cytokines such as IFNgamma and tumor necrosis factor-alpha which in turn attract and recruit the cells of the innate immune system, such as monocytes, macrophages, and NK cells, to the site to effect the actual destruction of the antigen or pathogen through generation of cytotoxic products and phagocytosis of the pathogens. B cells secrete antibodies specific for an antigen which will result in direct killing/lysis of the target and also recruit polymorphonuclear cells via complement activation and the complement activation products respectively. In this way, the adaptive immune system serves to direct the innate immune system components to target the inciting antigen or pathogen. The immune system can be regulated at any part of the immune response loop, i.e., afferent limb, central process, and efferent limb. Regulation of the autoimmune response can be via any of the following mechanisms: central tolerance -clonal deletion and peripheral tolerance -clonal anergy, T-cell suppression, immune deviation, immunologic ignorance, and antigen sequestration. Since the genome has the capability to generate both self-and nonself-recognizing antibodies and TCRs, mechanisms must be in place to contain and prevent the activation of selfreactive T cells. During lymphocyte development, both T and B cells with receptors that recognize self molecules with high affinity are clonally deleted via apoptois (central tolerance). 8 However the process is not foolproof, as autoreactive T and B cells in the periphery exist even in normal individuals. Many tissuespecific antigens, like the eye-restricted molecules, may not be expressed in the thymus or cells may have escaped the selection process by the central tolerance mechanism in the thymus without encountering the specific self antigen. Hence, the potential for induction and expression of autoimmunity still exists, and mechanisms to contain these autoreactive immune cells are crucial to prevent autoimmunity. Evidence for peripheral tolerance can be found in all phases of the immune response, i.e., afferent limb, central process, and efferent limb. 9, 10 The afferent limb depends heavily on the functional properties of the APCs which offer many opportunities for modulation. First, the antigen can be sequestrated and prevented from contact with the APCs by physical barriers or by rendering APCs incapable of antigen capture in the microenvironment (sequestration). Second, the ability of the APC to degrade, process, and express the antigens can be inhibited. Third, the antigen-bearing APCs may be prevented from migrating to the lymphoid tissues. Lastly, the ability of the APCs to generate the costimulatory signal can be inhibited or different costimulatory signals may be produced (immune deviation). Regulation of the APC's function can be seen as a form of immunologic ignorance where reactive autoimmune cells are present but never got the opportunity to encounter its antigen in a proper immunogenic form due to a deficiency in antigen presentation. In the central processing phase of the immune response, two signals are required for full activation of the lymphocyte. The absence of costimulation in the presence of antigen-specific stimulation renders the T cells unresponsive to further antigen stimulation, even in the presence of costimulatory signals (anergy). 11 On the other hand, too much costimulation in the presence of high antigen dose may lead to profound cell activation and apoptosis. Both mechanisms will result in tolerance. It is also now known that APCs from certain immune-privileged sites such as the eye, and APCs in the gut processing orally ingested antigens, may cause immune cells to develop into regulatory cells (Th3, Tr1, CD4+ CD25+ T-regulatory cells) secreting transforming growth factor-beta (TGF-beta) and IL-10 cyokines associated with a Th2 inflammatory profile instead of the typical Th1 inflammatory cytokines one usually sees. 12 In the efferent limb, immune regulation can occur via targeting at the level of antigen-specific immune effector cells. One such mechanism would be through Fas ligand -Fas-induced apoptosis of lymphocytes. 13, 14 The immune-reactive lymphocytes express the CD95 receptor (Fas receptor). On encountering the Fas ligand, which is expressed in immuno-privileged eye tissues such as the cornea and retina, these immune cells undergo apoptosis. Other molecular mechanisms, such as tumor necrosis factoralpha production by immune and accessory cells, such as Müller cells and retinal pigment epithelial cells during antigen encounter, can also result in apoptosis, thereby deleting these immunereactive cells. Similar to the central process, effector cells need costimulatory signals to be activated and the expression of inappropriate costimulatory signals such as IL-10 or TGF-beta by APCs can lead to anergy or reprogramming of lymphocytes into immunoregulatory cells secreting immunosuppressive cytokines, thereby further suppressing autoimmunity. 15, 16 The eye has been recognized as an immune-privileged site for more than 100 years. In the 1940s Medawar demonstrated this by showing prolonged, often indefinite, survival of organs or tissue grafts in the anterior chamber of the eye. 17 It is now known that this immune privilege is a dynamic process in which immunoregulatory mechanisms combined with anatomical factors maintain the vitality of grafts in privileged sites and of privileged organs and tissues as grafts. Various tissues, including allogenic skin grafts, thyroid tissues, neuronal retinal tissue, and allogenic tumor cells, have been shown to survive in the anterior chamber of the eye for prolonged periods. 18 However, this privilege is influenced by the immunogenic strength of the antigens expressed by the cells. Tumors expressing MHC-encoded alloantigens had only a transient extension of survival compared to those that expressed weaker transplantation antigens. 19 Although most studies were done placing tissue in the anterior chamber of the eye, a number of studies demonstrated a similar immune privilege when allogenic tumor cells, foreign neuronal retinal tissue, and retinal pigment epithelium were placed in the vitreous cavity and the subretinal space. 20, 21 Certain eye tissues are known to be immune-privileged, i.e., demonstrating an altered immune response and less rejection. Ocular tissues include the cornea, retinal pigment epithelium, and probably the retina. When transplanted beneath the capsule of the kidney, a nonimmuneprivileged site, cornea demonstrated extended survival compared with other ocular tissues like the conjunctiva. 22 There are many factors contributing to ocular immune privilege. Some of these are active while the rest are passive (Table 13-1) . The blood-retinal barrier serves to keep tissue-specific antigens sequestrated. There is an absence of lymphatics in the retina, during experimental autoimmune uveitis than wild types. 32 Studies using Fas ligand-deficient mice have also revealed that ACAID is dependent on the presence of Fas ligands as these mice are unable to induce ACAID. 33 Hence, Fas-Fas ligand interaction may be more complex than just inducing apoptosis of activated immune cells, as initially presumed. Although there are active soluble factors in the aqueous humor that can inhibit T-cell activation in vitro, decrease the ability of NK cells to lyse their target, 34 and block complement activation, 35 ,36 the eye is not left defenseless against pathogens. It still retains certain immune functions to deter pathogenic invasion. Antibodies in the aqueous are capable of neutralizing viruses and cytotoxic T cells (terminally differentiated) can bind and kill their target cells as elsewhere in the body. 37 Several animal models for autoimmune posterior uveitis have been described (Table 13 .2). EAU is one of the most studied of these models since its development in 1968 by Wacker & Lipton. 38 (It is a good animal model for human ocular autoimmunity.) EAU is mostly self-limited and requires the use of adjuvant for disease induction. The EAU model has been instrumental in eliciting immune mechanisms, the identification of pathogenic epitopes of autoantigens in the eye in animals, and the evaluation of therapeutic strategies with clinical relevance. Several retinal antigens, including S-antigen (arrestin), interphotoreceptor retinoid-binding protein (IRBP), rhodopsin, recoverin, and phosduscin, have been found to have uveitogenic properties. Immunization with these antigens or their fragments can induce ocular inflammation in susceptible strains of guinea pigs, rats, mice, and in rabbits and monkeys. The disease that is induced resembles various human uveitic conditions such as ocular sarcoidosis (Fig. 13-1) , the Vogt-Koyanagi-Harada disease, sympathetic ophthalmia, and Behçet's disease ( Fig. 13-2 ). Using advanced techniques, more novel eye autoantigens have been identified, such as UACA (uveal autoantigen with coiled coil domains and ankyrin repeats) in patients with Vogt-Koyanagi-Harada disease, 39 and an unknown retinal autoantigen targeting the connecting cilium region of photoreceptors in a patient with Waldomström's macroglobulinemia, and retinal dysfunction similar to cancer-associated retinopathy. 40 Much of our understanding of retinal autoantigens is derived from EAU models using S-antigen, IRBP (peptides derived from these proteins), and, to a lesser extent, rhodopsin, recoverin, and phosducin. S-antigen, a 48-kDa protein also referred to as arrestin, is one of the most commonly used antigens used to induce EAU. 41 It was first identified in the soluble fraction of retinal extracts. As it is the first autoretina antigen to be implicated in the pathogenesis of uveitis, the sequence and its role in phototransduction have been well characterized. 42, 43 It is a highly conserved protein found in the retinal photoreceptor cells and in the pinealocyte. The main function of arrestin is to block the interaction of 256 although there is evidence that antigen may still be able to be transported to the lymph nodes. 23 Tissue drainage via the hematogenous route may alter the APCs function. 24 There is also a reduced expression of MHC class I and II molecules and an absence of bone marrow-derived cells that function within tissues as APCs in the normal retina. 25 The active factors include ligands and factors expressed on the cell surface of the ocular cells (e.g., CD95 ligands, CD59, CD55, and CD46), and soluble factors in the ocular microenvironment (e.g., TGF-beta, alpha-melanocyte-stimulating hormone, vasoactive intestinal peptide, calcitonin gene-related peptide, macrophage migration inhibitory factor, IL-1-receptor antagonist and free cortisol). All of these factors contribute to active immunoregulation in an attempt to prevent any autoimmunity. 26 ACAID is the best-studied immune-privilege phenomenon in the eye. 27 Although its name implies that it is an anterior-chamber phenomenon, there is much information to show that the same mechanisms are at work in the vitreous and subretinal space. 21 Therefore understanding ACAID could further our knowledge of retinal autoimmunity. There is a large number of bone marrowderived APCs in the iris, trabecular meshwork, and ciliary body. Studies have demonstrated that antigen captured in the anterior chamber by these APCs is processed in the spleen. 28 These APCs reach the spleen via the hematogenous route, bypassing the afferent lymphatics and the lymph nodes. The result of this is a reduction of the antigen's capacity to activate cell-mediated immune mechanisms. Functionally, these APCs are different. If transferred, these APCs induce ACAID rather than inducing conventional immune responses. The microenvironment of the eye also contains factors such as TGF-beta 2 that are able to reprogram the APCs from conventional sites, if exposed to the eye microenvironment, to secrete less IL-2, express less CD40 (a costimulatory signal for T-cell activation) and produce more mature TGF-beta. These TGF-beta 2 -treated APCs activate naive T cell to secrete IL-4 and trace amounts of IL-2, but not IFN-gamma. 29 The spleen is the critical lymphoid organ for the induction of ACAID. In the spleen, a unique population of CD4+ lymphocytes, which secrete IL-2 and TGF-beta and develop Th2 immune response upon antigen exposure, are activated. The B-cell response, occurring upon re-exposure to antigen with adjuvant, produces IgG1 isotype antibodies instead of IgG2a, IgG2b, and IgG3, consistent with a Th2 response. 30 There appears to be a concerted effort to deviate the immune response from the usual Th1 pathway to a Th2 pathway, one that is seen in allergic responses. The role of CD95 and its ligand, Fas ligand, is still unclear. It is tempting to speculate that Fas ligand acts by causing active immune cells to apoptose upon engagement, thereby resulting in the elimination of the active immune cells. 31 One would expect an upregulation of an inflammatory response if Fas ligand were absent in the eye. Contrary to the predictions, Fas ligand-deficient mice developed less severe inflammation rhodopsin with the G-protein transducin in the phototransduction cascade. Immunization of susceptible animals (such as Lewis rats but not mice) with S-antigen induces a predominantly CD4+ T-cell-mediated inflammatory response in the retina, uveal tract, and the pineal gland. Six peptide fragments of S-antigen have been identified as uveitogenic. There are six regions of S-antigen identified as targets of the T-cell epitopes. One major uveitogenic peptide is M (sequence 303 to 320). Some of the fragments are species-specific in their uveitogenicity. 44 Although the demonstration of uveitogenic properties of S-antigen has helped in the understanding of uveitis mechanisms, the relevance to human uveitis needs to be more fully explored. S-antigen has also been implicated in the pathogenesis of uveitis through molecular mimicry. It has been shown that several exogenous (baker's yeast, Escherichia coli, hepatitis B virus, streptococcal M5 protein, Moloney murine sarcoma virus, and baboon endogenous virus) and endogenous (human leukocyte antigen B-derived peptide, tropomyosin) antigens share sequence homology with peptide M of S-antigen. 45 Some have been shown to be uveitogenic in the EAU models, and lymphocytes from animals immunized with M peptide cross-reacted and proliferated when stimulated with peptides derived from some of these exogenous or endogenous antigens. Antistreptococcal monoclonal antibodies were found to recognize several uveitogenic peptides of S-antigen, providing further evidence that immunological mimicry between self and exogenous antigens from an infectious agent may be a potential mechanism in the pathogenesis of uveitis in humans. 46 Interphotoreceptor retinoid binding protein IRBP can be used to induce EAU in both mouse and rat. 47 It is a major protein (1264 amino acid residues) of the interphotoreceptor matrix, functioning as a transporter of retinoids between the retina and RPE. Similar to S-antigen, it is also found in both the eye and the pineal gland, and induction of EAU with IRBP will lead to disease in both locations. Depending on the dose of antigen used and the species of animals, a spectrum of disease ranging from hyperacute to chronic relapsing disease could be induced. The inflammation is targeted at the photoreceptor layer, producing histopathology similar to lesions seen in uveitis, such as retinal human vasculitis, granuloma, focal serous detachments, loss of photoreceptors, and formation of sub-RPE infiltrates similar to Dalen-Fuchs nodules, seen in patients with Vogt-Koyanagi-Harada disease and sympathetic ophthalmia. 48 The disease activity shows two peaks at the 5th and 10th weeks after immunization, simulating the relapsing course in human uveitis. 49 The relatively long duration of disease activity in the murine IRBP EAU model makes it a good model for evaluation of therapeutic strategies in established disease. Immunization with the particulate fraction of the retinal extract, fraction P, was shown to induce EAU. Later, rhodopsin was identified to be the uveitogenic antigen in fraction P. Rhodopsin is the visual pigment of rod photoreceptor cells, belonging to the larger family of G-protein-coupled receptors. 50 It is a membrane-bound protein comprising a polypeptide chain, the opsin, and a covalently bound chromophore, 11 cis-retinal. The function of rhodopsin is to capture photons and trigger the phototransduction cascade. The uveitis induced in susceptible animals such as guinea pigs, rabbits, rats, and monkeys is typically a retinitis with multifocal destruction of photoreceptor cells, showing dense mononuclear and polymorphonuclear cell infiltrates in the retina and anterior uvea in severe disease. 51 There are three distinct immunopathogenic sites on the molecule: sequence 230 to 250 (extracellular loop V to VI) is the most uveitogenic, comparable to S-antigen and IRBP. 52, 53 The role of rhodopsin autoantigens in human diseases is still unclear. Studies in patients with normal-pressure glaucoma have demonstrated an elevated antirhodopsin antibody titer, indicating the possibility of its role in the optic neuropathy observed in these patients. 54 Recoverin is a 23-kDa calcium-binding protein present in photoreceptor cells and functioning as a calcium sensor that regulates rhodopsin phosphorylation through inhibition of rhodopsin kinase. It is also found in bipolar cells, pineocytes, and certain tumor cells. The function in these nonphotoreceptor cells is unknown as they do not contain rhodopsin kinase. 55 Immunization of Lewis rats with high doses of recoverin produced EAU similar in severity, histology, and duration to other retinal autoantigens such as Santigen. 56 The major immunopathogenic epitope, sequence 62-81, has been characterized in Lewis rats. It has been identified as an autoantigen in paraneoplastic and degenerative diseases of the retina such as cancer-associated retinopathy. 57 High levels of antirecoverin antibodies were detected in some patients with occult cancer and retinal dysfunction. More recently, these autoantibodies have been demonstrated to induce apoptosis of photoreceptors, supporting its pathogenic role in retinal autoimmunity. 58 Phosduscin is a 33-kDa protein, a cytosolic regulator of G-proteinmediated signaling, found in the retina and also in nonretinal tissues such as liver, lung, heart, and brain. 59 Immunization with phosduscin in Lewis rats produce mild to moderate EAU characterized by late onset, low-grade severity, and predominantly involving the posterior segment. 60 Its role in human disease is unclear. After more than two decades of work on EAU models using these retinal autoantigens, it is still unclear whether there is an association between the occurrence of eye-specific autoantibodies and lymphocyte proliferation response and clinical disease, and exactly what their roles are in the pathogenesis of human uveitis. Lymphocytes reactive to retinal antigens have been demonstrated in normal individuals 4 and those with retinal disease. 61 In the normal individuals, the presence of circulating autoreactive cells suggests that immunoregulatory mechanisms must exist to prevent retinal autoimmunity resulting in disease; in the latter it is tempting to hypothesize the possible roles of these cells in the induction of retinal autoimmunity. Both the humoral and cellular responses against S-antigen and IRBP have been investigated. Tlymphocyte responses to S-antigen, IRBP, and peptides derived from patients with uveitis were investigated in a few clinical studies. An increase in T-lymphocyte responses was reported in patients with Behçet's disease during periods of active ocular inflammation. 62 In another similar study, a large proportion of the patients had lymphocytes which responded to stimulation with S-antigen, IRBP, and several uveitogenic peptides such as S-antigen peptide M but no difference was reported in the cellular response between Behçet's disease patients with and without uveitis. 63 Similar cellular responses to S-antigen, S-antigen-derived peptides (peptides M and G), and uveitogenic peptide derived from IRBP (R16) have been reported for patients with idiopathic uveitis. 64 Lymphocyte proliferation responses to 20 peptides spanning the entire sequence of S-antigen were investigated in patients with uveitis in one study. Half of the patients with idiopathic uveitis responded to at least one peptide while there was no response to the peptides in the normal controls. The peptide fragments found to elicit more lymphocyte proliferation were 61-80, 81-100, 145-160, 161-180, 221-240, and 241-260. 65 Other fragments of IRBP and S-antigen were studied in another similar study involving 82 patients with uveitis. Patients who responded to IRBP or S-antigen also responded to one or two of the fragments but to lesser magnitude. This suggests that these fragments are probably not the primary mediators of the disease. 66 Isolation of S-antigen-specific T cells from the eyes of uveitis patients, which were expanded to antigen specific Tcell lines, was reported. 67 A study evaluating oral tolerance by feeding S-antigen to uveitis patients showed a good clinical response and the patients required less immunosuppression. 68 Together, these studies provided further support for the possible pathogenic roles of retinal autoantigen in uveitis. Although both EAU in animals and uveitis in most humans are CD4+ Th1-mediated responses, there are differences in the T-cell specificities in antigen response. Patients with uveitis, due to epitope spread, can respond to more than one peptide, i.e., several epitopes are recognized in the same patient, while animals with EAU tend to respond only to the inciting antigen. There is no common pattern of lymphocyte response in clinical studies reported so far. Early clinical studies on the humoral response showed the presence of antiretinal autoantibodies, including anti-S-antigen antibodies in the sera of some uveitis patients 69, 70 (Fig. 13-3) , but there was no good correlation between presence or levels of autoantibodies and clinical disease. Moreover, anti-IRBP antibody was reported to occur in the same frequency in the sera of patients with uveitis and normal controls. The only difference has been the stronger affinity of the antibodies in patients with uveitis. 71 The relationship between humoral and cellular response to these autoantigens was studied in a group of patients with endogenous uveitis such as birdshot retinochoroidopathy, retinal vasculitis, and Behçet's disease. The antibody titers to S-antigen and IRBP were found to be decreased during active inflammation periods and the lymphocyte responses to S-antigen were found to be most significant in the period preceding a relapse of ocular inflammation, supporting the pathogenic roles of these lymphocytes in retinal autoimmunity. 72 The role of humoral and cellular responses to retinal autoantigens remains unclear and it is possible that different subsets of patients with uveitis, genetically determined, respond to different epitopes and antigens, resulting in the wide spectrum of clinical disease encountered. Retinal autoimmunity may not always be pathogenic. Recent studies on central nervous system (CNS) injury and its reparative process have revealed a possible beneficial role of autoimmunity in limiting self-injury during insults to the CNS. Axonal injury in the CNS leads not only to the degeneration of the affected axon but also the neighboring axon through self-destructive compounds released by the degenerating axon into the microenvironment. 73 Studies have revealed that autoreactive T cells directed against myelin antigens have beneficial effects on CNS myelinated axons after a mechanical crush injury 74 and it has been proposed that this neuroprotective activity exhibited by autoimmune T cells may be a physiological process which the body develops to cope with stressful conditions. 75 The observation seen in the CNS could be extrapolated to the retina. Supporting this are the findings of a recent study demonstrating vaccination with peptides derived from IRBP resulted in protection of retinal ganglion cells from glutamate-induced death or death as a consequence of optic nerve injury. 4 It appears that the immune system not only protects the body against invading pathogens but also protects it from toxic substances released by self-tissues during trauma and stress. Therefore the autoreactive cells that induce neuroprotection and those that induce autoimmune disease may share the same specificity and phenotype, indicating their potential to be protective and destructive at the same time. 76 The final outcome depends on how they are regulated. Correlation between the ability to manifest an autoimmune response with a beneficial outcome and the ability to resist the development of an autoimmune disease in animals has been reported. 77 From these recent studies, it seems clear that the ability to protect the eye from inflammation and injury does not solely depend on mechanisms conferring immune privilege but rather a precise regulation of autoimmunity. The role of infection in the pathogenesis of eye autoimmunity has long been suspected but there is no conclusive study to demonstrate it. The possible causative relationship between infections and ocular autoimmunity such as streptococcus, herpesvirus, and Behçet's disease, 78 Gram-negative bacteria, and HLA B27associated uveitis, 79 and herpesviruses and serpiginous choroiditis 80 have been reported. Infection as an exogenous trigger for inflammation in the eye is an enticing possibility as infections have been linked to the pathogenesis of many autoimmune diseases such as rheumatic fever, 81 inflammatory bowel diseases, 82 and sarcoidosis. 83 Antiretinal antibodies in the serum of patients with toxoplasmosis retinochoroiditis 84 and antiretinal and anti-RPE antibodies in murine coronavirus retinopathy and degeneration 85 were reported. These reports provided support for a role of infection in retinal autoimmunity. Even in the current EAU models in animals, bacteria-derived products like mycobacteria in complete Freund's adjuvant, lipopolysaccharides, and pertussis toxin are used to promote the development of autoimmune disease. 86 An infection could possibly play a similar role, in sensitizing the immune system to eye-restricted antigens, which are "sequestrated" or expressed in low levels below the immune activation threshold. The mechanisms involved in the pathogenesis of uveitis may be similar to that of the adjuvant in EAU, including changes in the blood-tissue barrier to allow infiltration of inflammatory cells, 87 promotion of Th1 response, 88 stimulatory effects on the APCs, 89 and enhancement of innate immune response. 90 On the other hand, there is evidence that immunologic mimicry may play a role in pathogenesis as molecular homology exists between certain retinal autoantigens and peptides derived from microbes, such as the M peptide derived from S-antigen and the streptococcal cell wall peptide, the yeast's histone peptide and certain viral agents. 46, 91, 92 Molecular mimicry has also been suspected of playing a role in conferring disease susceptibility in patients with certain HLA haplotypes, like HLA B27 for acute anterior uveitis, 93 HLA B51 for Behçet's disease, 94 and HLA-A29 for birdshot retinochoroidopathy. 95, 96 Synthetic peptides derived from HLA-B molecules such as B27 and B51 with amino acid sequence homologous with S-antigen have been reported to induce EAU in animals. 97 Lymphocytes from patients with uveitis, especially Behçet's disease, respond both to peptides derived from HLA-B molecules and peptides derived from S-antigen, demonstrating possible immunologic mimicry. 98 In addition, further studies also showed sequence homology between these peptides and rotavirus and casein protein, a protein found in milk, and cross-reaction in cellular immunity between these peptides was found in animal studies. 99 Together these studies bring forward the possibility of immunologic mimicry for exogenous factors like infection or foreign proteins breaking down tolerance and inducing autoimmunity. The guardian that prevents pathologic autoimmunity is immune tolerance. Immune tolerance, usually maintained by mechanisms described earlier, could be lost or modulated. This was demonstrated in a recent study showing that heat shock protein (HSP) 70 can modulate APC function, resulting in loss of tolerance in an animal model. 100 HSPs, especially microbial, have been implicated in the pathogenesis of Behçet's disease 101 and demonstrated to induce inflammation in the eye in animal studies. 102 Therefore, one may speculate that an infection and the subsequent release of microbial HSPs could be the initial triggering event, resulting in changes of APC function and a subsequent loss of tolerance, leading to autoimmunity. These notions remain as hypotheses for uveitis since it is a heterogeneous disease, and evidence of the initial triggering event is usually difficult to identify due to the protracted course and the delay between trigger and disease presentation. Furthermore, a disease could have multiple triggering mechanisms and different triggers may lead to the same disease, making clinical data difficult to interpret. Retinal autoimmunity may be a physiologic process rather than a pathologic one given its role in organ and tissue-specific protection from damage from injury and stress in the normal retina. That may account for the evidence of retinal autoimmunity detected in normals 2 but there is still much that is unknown about its regulation and the maintenance of the fine balance between protection and destruction given that these paradoxical roles reside in the same group of cells. With rapid technological advances in molecular immunology, new tools to unravel the roles of retina autoimmunity in both the physiological and pathological states of the eye will be available and novel targeted therapeutic strategies could be developed for the management of retina autoimmunity-induced diseases in the near future. Die Entwicklung der in fremden Grund versetzten lebenden Gewebe Cellular autoimmunity to retinal specific antigens in patients with Behçet's disease GITRL is constitutively expressed in the retina The tissue-specific self-pathogen is the protective self-antigen: the case of uveitis T cell receptor gene diversity and selection Accessory molecule and co-stimulation requirements for CD4 T cell response V(D)J recombination Central tolerance of T cells Multiple levels of peripheral tolerance Processing of self-proteins and its impact on shaping the T cell repertoire, autoimmunity and immune regulation Co-stimulation and co-inhibition: equal partners in regulation Antigen-specific suppressor factor: missing pieces in the puzzle Role of Fas-mediated cell death in the regulation of immune responses The role of FasL-induced apoptosis in immune privilege Immunosuppressive and antiinflammatory properties of interleukin 10 The immunoregulatory roles of transforming growth factor beta Immunity to homologous grafted skin. III. The date of skin homografts transplanted to the brain, to subcutaneous tissue and to the anterior chamber of the eye Deviant immune responses to allogeneic tumors injected intracamerally and subcutaneously in mice Immunogenetics basis for immunologic privilege in the anterior chamber of the eye Analysis of immune deviation elicited by antigens injected into the sub-retinal space Subretinal space and vitreous cavity as immunologically privileged sites for retinal allografts Immune privilege and the cornea Peptide-specific T cell clonal expansion in vivo following immunization in the eye, an immune-privileged site Dendritic cells in the central nervous system and eye and their associated supporting tissues Expression of major histocompatibility complex molecules in rodent retina: immunohistochemical study Ocular immunosuppressive microenvironment Anterior chamber associated immune deviation (ACAID): regulation, biological relevance, and implications for therapy Do immunologically privileged sites require a functioning spleen? TGF-beta promotes immune deviation by altering accessory signals of antigen-presenting cells Distinctive humoral immune responses following anterior chamber and intravenous administration of soluble antigen. Evidence for active suppression of IgG 2 -secreting B lymphocytes Apoptosis, tolerance, and regulatory T cells -old wine, new wineskins Fas and Fas ligand expressed on cells of the immune system, not on the target tissue, control induction of experimental autoimmune uveitis Anterior chamber inoculation of splenocytes without Fas/Fas-ligand interaction primes for a delayed-type hypersensitivity response rather than inducing anterior chamber-associated immune deviation Isolation and characterization of a unique natural killer cell inhibitory factor present in the anterior chamber of the eye Chronic low level complement activation within the eye is controlled by intraocular complement regulatory proteins Complement regulatory activity of normal human intraocular fluid is mediated by MCP, DAF, and CD59 Effect of anterior chamber-associated immune deviation (ACAID) on rat islet allograft rejection Experimental allergic uveitis. II. Serologic and hypersensitive responses of the guinea pig following immunization with homologous retina Negi identification of a novel autoantigen UACA in patients with panuveitis Waldenström's macroglobulinemiaassociated retinopathy Identification of a uveitopathogenic and lymphocyte proliferation site in bovine S-antigen An 18-mer peptide derived from the retinal S antigen induces uveitis and pinealitis in primates Characterization of a new, potent, immunopathogenic epitope in S-antigen that elicits T cells expressing V beta 8 and V alpha 2-like genes A new perspective of S-antigen from immunochemical analysis S-antigen: from gene to autoimmune uveitis Immunological mimicry between retinal S-antigen and group A streptococcal M proteins Induction of experimental autoimmune uveoretinitis and pinealitis by IRBP. Comparison to uveoretinitis induced by S-antigen and opsin A new model of autoimmune disease. Experimental autoimmune uveoretinitis induced in mice with two different retinal antigens Pathology of experimental autoimmune uveoretinitis in mice Rhodopsin and phototransduction Genetic control of antibody response to bovine rhodopsin in mice: epitope mapping of rhodopsin structure Induction of experimental autoimmune uveitis with rhodopsin synthetic peptides in Lewis rats Recoverin: a potent uveitogen for the induction of photoreceptor degeneration in Lewis rats Antirhodopsin antibodies in sera from patients with normal-pressure glaucoma Inhibition of rhodopsin phosphorylation by non-myristoylated recombinant recoverin Recoverin is highly uveitogenic in Lewis rats Role of anti-recoverin autoantibodies in cancer-associated retinopathy Autoantibody-induced apoptosis as a possible mechanism of autoimmune retinopathy Phosducin is a ubiquitous G-protein regulator Induction of experimental autoimmune uveitis by the retinal photoreceptor cell protein, phosducin Cellular immune responsiveness of uveitis patients to retinal S-antigen Prospective determination of T-cell responses to S-antigen in Behçet's disease patients and controls Cellular autoimmunity to retinal specific antigens in patients with Behçet's disease Cellular immune response to retinal S-antigen and interphotoreceptor retinoid binding protein fragments in idiopathic human uveitis Human retinal S-antigen: T cell epitope mapping in posterior uveitis patients Cellular immune responses of patients with uveitis to retinal antigens and their fragments Long-term antigen specific and non-specific T-cell lines and clones in uveitis Treatment of uveitis by oral administration of retinal antigens: results of a phase I/II randomized masked trial Anti-retinal auto-antibodies in Vogt-Koyanagi-Harada syndrome, Behçet's disease, and sympathetic ophthalmia Anti-retinal S-antigen antibodies in human sera: a comparison of reactivity in ELISA with human or bovine S-antigen Serum antibody response to human and bovine IRBP in uveitis Severe retinochoroidopathy: variations of humoral and cellular immunity to S-antigen in a longitudinal study Hydroxyl radicals generated in vivo kill neurons in the rat spinal cord: electrophysiological, histological, and neurochemical results Autoimmune T cells as potential neuroprotective therapy for spinal cord injury Protective autoimmunity is a physiological response to CNS trauma Myelin specific Th1 cells are necessary for post-traumatic protective autoimmunity Neuronal survival after CNS insult is determined by a genetically encoded autoimmune response Pathogenesis of Adamantiades-Behçet's disease Humoral immune response to Klebsiella capsular polysaccharides in HLA-B27-positive patients with acute anterior uveitis and ankylosing spondylitis Association of herpesviruses in the aqueous humor of patients with serpiginous choroiditis: a polymerase chain reaction-based study Rheumatic fever: the T cell response leading to autoimmune aggression in the heart Intestinal bacteria and ulcerative colitis Quantitative PCR of mycobacterial and propionibacterial DNA in lymph nodes of Japanese patients with sarcoidosis Human antiretinal antibodies in toxoplasma retinochoroiditis Retina and retinal pigment epithelial cell autoantibodies are produced during murine coronavirus retinopathy Immunomodulation of experimental autoimmune uveoretinitis by intravenous injection of uveitogenic peptides Acute experimental autoimmune encephalomyelitis in mice. I. Adjuvant action of Bordetella pertussis is due to vasoactive amine sensitization and increased vascular permeability of the central nervous system The requirement for pertussis to induce EAU is strain-dependent: B10.RIII, but not B10.A mice, develop EAU and Th1 responses to IRBP without pertussis treatment Pertussis toxin modulates the immune response to neuroantigens injected in incomplete Freund's adjuvant: induction of Th1 cells and experimental autoimmune encephalomyelitis in the presence of high frequencies of Th2 cells Pertussis toxin alters the innate and the adaptive immune responses in a pertussis-dependent model of autoimmunity Immunological mimicry between retinal S-antigen and group A streptococcal M proteins Molecular mimicry between a uveitopathogenic site of S-antigen and viral peptides: induction of experimental autoimmune uveitis in Lewis rats HLA-B27-associated uveitis: overview and current perspectives Human leukocyte antigen serologic and DNA typing of Behçet's disease and its primary association with B51 HLA-A29.2 subtype associated with birdshot retinochoroidopathy Molecular mimicry: a mechanism for autoimmune injury Cross-reactivity between an HLA-B27-derived peptide and a retinal autoantigen peptide: a clue to major histocompatibility complex association with autoimmune disease Induction of arthritis and uveitis in Lewis rats by antigenic mimicry of peptides from HLA-B27 and cytokeratin Autoimmune uveitis induced by molecular mimicry of peptides from rotavirus, bovine casein and retinal S-antigen Hsp70 promotes antigenpresenting cell function and converts T-cell tolerance to autoimmunity in vivo Immunology and functional genomics of Behçet's disease Heat shock proteins generate betachemokines which function as innate adjuvants enhancing adaptive immunity