Skip to main content

REVIEW article

Front. Immunol., 08 January 2021
Sec. Autoimmune and Autoinflammatory Disorders
This article is part of the Research Topic Uveitis: Immunity, Genes and Microbes View all 23 articles

Immunological Insights in Equine Recurrent Uveitis

  • Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany

Horses worldwide suffer from equine recurrent uveitis (ERU), an organ-specific, immune-mediated disease with painful, remitting-relapsing inflammatory attacks alternating with periods of quiescence, which ultimately leads to blindness. In course of disease, both eyes can eventually be affected and since blind horses pose a threat to themselves and their surroundings, these animals have to be killed. Therefore, this disease is highly relevant for veterinary medicine. Additionally, ERU shows strong clinical and pathological resemblance to autoimmune uveitis in man. The exact cause for the onset of ERU is unclear to date. T cells are believed to be the main effector cells in this disease, as they overcome the blood retinal barrier to invade the eye, an organ physiologically devoid of peripheral immune cells. These cells cause severe intraocular inflammation, especially in their primary target, the retina. With every inflammatory episode, retinal degeneration increases until eyesight is completely lost. In ERU, T cells show an activated phenotype, with enhanced deformability and migration ability, which is reflected in the composition of their proteome and downstream interaction pathways even in quiescent stage of disease. Besides the dysregulation of adaptive immune cells, emerging evidence suggests that cells of the innate immune system may also directly contribute to ERU pathogenesis. As investigations in both the target organ and the periphery have rapidly evolved in recent years, giving new insights on pathogenesis-associated processes on cellular and molecular level, this review summarizes latest developments in ERU research.

Introduction

Insights in the pathogenesis of equine recurrent uveitis (ERU) have grown in recent years, especially on cellular and molecular level in periphery and in the eye itself. ERU is an organ-specific, immune-mediated disease, which is predominantly driven by CD4+ T cells (13). These are somehow activated in periphery and then manage to overcome the blood retinal barrier (BRB), entering the eye and causing intraocular inflammation (4). Since ERU has a remitting-relapsing character (2), increasing numbers of immune cells invade and accumulate in the eye with every inflammatory attack, gradually destroying their main target, the retina. These acute phases alternate with periods of quiescence and increase in severity as the disease progresses (5). Factors associating to ERU onset are still discussed and an exact cause is elusive to date. A genome-wide association study suggested a correlation of genetic variants influencing the expression of IL-17A and IL-17F with the development of ERU (6). This study also substantiated previous reports linking ERU to MHC class I haplotype ELA-A9 in warmblood horses (7). Direct evidence for a genetic component in ERU of warmblood horses, however, is still lacking. Disease prevalence among the horse population is high, ranging from 2%–25% in the US and 8%–10% in Europe (5, 8). ERU often affects both eyes and eventually leads to blindness if left untreated (9). These two factors, high prevalence and loss of sight, explain its importance for veterinary medicine, since blind horses have to be killed due to the threat they pose to their surroundings and themselves, which has a great personal but also economic impact on horse owners. Moreover, ERU has great potential to serve as a valuable spontaneous model for autoimmune uveitis in man, due to strong clinical and pathological similarities (3, 4, 10, 11).

In the past, several studies could show that the dysregulated immune response in ERU is directed against retinal autoantigens such as interphotoreceptor retinoid-binding protein (IRBP) and cellular retinaldehyde-binding protein (CRALBP) (4, 12), which, along with other factors, suggests presence of autoimmune reactions in ERU. These autoantigens not only remain stably expressed in advanced stages of ERU, when retinal architecture is drastically destructed (13), IRBP can also induce experimental uveitis in the horse itself, which shows close similarity to the spontaneous disease on clinical, cellular and molecular level (4, 14). Moreover, studies on ERU horses firstly provided CRALBP as novel autoantigen (12), which was subsequently proven to also have high prevalence in human patients with autoimmune uveitis (15). As shown in previous studies, the recognition of these autoantigens and the subsequent spreading of epitope recognition to a new determinant may be the cause for the relapsing character of ERU (12, 16). This process known as epitope spreading describes the reaction of immune cells against the initial epitope of an ERU autoantigen, which then redirects against a different, but structurally similar epitope, re-initializing an inflammatory response after the initial inflammation has subsided (17, 18).

In addition to the retinal-autoantigen specific T cells driving ERU from periphery, further studies suggested that the retina itself promotes the intraocular inflammatory processes through active abundance downscaling of proteins which protect the maintenance of the blood-retina-barrier (19) and through secretion of the pro inflammatory cytokine interferon gamma (IFNγ) by retinal Mueller glial cells (20). Since then, research has advanced on the target organ of ERU as well as the cellular and molecular components in periphery. This review summarizes the latest immunological insights in ERU pathogenesis.

Periphery

Adaptive Immune System

Although the concept of immune privilege is increasingly discussed since its initial description (21, 22), the eye is considered to be an immune privileged organ (23, 24), and is therefore usually devoid of immune cells form peripheral blood stream. In ERU, however, we find major cell infiltrates in the eye (2, 3, 2527), which mainly consist of CD4+ lymphocytes (2, 27). To date, the exact mechanisms enabling these autoreactive cells to overcome the BRB in ERU are still elusive. Experimental autoimmune uveitis (EAU) is a valuable tool to investigate these mechanisms, not only in common rodent models (2831), but to a certain extent also in the horse (4). Immunization of healthy horses with the retinal autoantigen IRBP mimics the spontaneous disease through the formation of auto-aggressive, IRBP specific T cells, which leave the peripheral blood stream to invade the eye directly prior to a uveitic attack (4). These cells show distinct protein changes before immunization, during the inflammatory phase and in quiescent episodes (14), which points to a highly dynamic functional phenotype depending on the stage of disease. During quiescence of this IRBP induced equine EAU, protein abundance changes in immune cells show many similarities to those observed in freshly obtained cells from horses in quiescent stage of ERU (14), underlining the close resemblance of the spontaneous and the induced disease in the horse. Although this type of induced horse model may allow more in depth investigations on ERU pathomechanisms, which is especially interesting for assessment of protein and cellular dynamics throughout the different stages of ERU, it is less suitable for large-scale experiments comparable to those performed on the several existing rodent models for autoimmune uveitis.

Analysis of primary T cells from horses with spontaneously occurring ERU revealed distinct proteome changes, mainly of proteins with a role in cell adhesion, cell migration and regulation of cell shape (3234). In transmigration experiments, CD4+ T cells from ERU horses showed increased migration rates, which was linked to increased expression of the protein formin-like 1 (FMNL1) in the membranes of these cells (33). As shown in a new FMNL1 knock-out mouse model, FMNL1 deficiency impedes extravasation and trafficking of T cells to sites of inflammation (35), further undermining the importance of this protein and its functional effects in ERU pathogenesis. Furthermore, lymphocytes from ERU horses displayed significantly higher cell motility, cell speed and directness toward selected chemoattractants, especially IFNγ, and toward the ERU autoantigen CRALBP, in live-cell imaging experiments (36), which was associated to lower abundance of the protein septin 7 in these cells (32, 36). Previous studies on septin 7-depleted murine CD4+ T cells (clone D10.G4, as well as primary T cells from DO11.10 TCR transgenic mice), showed less rigidity and aberrant cell morphology in these depleted cells, resulting in the ability to migrate through narrow pores (37), which was also hypothesized for ERU (32), but never directly proven. Identification of interactors of septin 7, most importantly dedicator of cytokinesis 8 (DOCK8), which was decreased in ERU (34), pointed to impaired immunity and increased risk for recurrent inflammation, as suggested in studies on human cells from patients with DOCK8 deficiency (38). Furthermore, increased expression of DOCK8 interacting protein integrin-linked kinase (ILK) was functionally associated to a decreased apoptosis rate in ERU cells (34). Combined results from these in vitro experiments - increased life span, increased migratory reactivity toward chemokines and autoantigens and enhanced migration ability of ERU lymphocytes - underlines the dysregulated nature of these cells in ERU. Although the transfer of these interpretations from in vitro experiments to in vivo mechanisms needs to be assessed with care, these insights further support the role of CD4+ T cells as key players in ERU pathogenesis.

Recent studies showed that, although percentage of CD3+, CD8+, and CD4+ lymphocytes does not differ between healthy horses and those with ERU, only the disease-driving CD4+ T cells have an activated phenotype in ERU horses (39). These cells show significantly increased expression levels of IFNγ and decreased expression of IL‐10, indicating Th1 immune response (39). This supports previous findings of increased levels of IFNγ as well as cytokine Interferon gamma-induced protein 10 (IP-10) in serum of ERU horses (40). A further known immune response in autoimmune uveitis in man as well as EAU in rodents is mediated through Th17 cells (41). In ERU, the exact role of Th17 cells has not been established to date, however, detection of cytokines IL‐6, IL‐17, and IL‐23 via crossreactive anti-human antibodies in histological sections of the iris and the ciliary body of ERU horses provide first indications pointing toward a contribution of these cells to ERU pathogenesis (42). Similar conclusions could be drawn from M. tuberculosis (strain H37Ra) induced experimental uveitis in horses, where high levels of IL-17 were detected in aqueous humor and vitreous via Enzyme-linked Immunosorbent Assay (ELISA) (43).

Contrary to the Th1 and/or Th17 immune response inducing and maintaining inflammation in the uveitic eye, remission of each inflammatory bout in autoimmune uveitis and EAU can be associated to T regulatory cells (Treg) (4446). During an acute inflammatory uveitic attack, these cells show decreased abundance in peripheral blood as opposed to phases of quiescence, suggesting a crucial role in the remitting-relapsing character of autoimmune uveitis (47, 48). These differences, however, could not be detected in peripheral blood of ERU horses compared to healthy controls (39). To date, no direct evidence for Tregs in the equine eye could be found. Nevertheless, the role of Treg in periodic remission of ERU is highly probable and thus merits further investigations.

Although an autoreactive Th1 response against retinal antigens is widely supported as the key process in ERU pathogenesis, one pivotal question remains: how can peripheral T cells be primed against tissue specific self-antigens which are “hidden” in an immune privileged organ? Emerging investigations on autoimmune uveitis suggest that commensal microbiota, if dysregulated (dysbiosis), may show sequence similarities with self-antigens (49, 50). This in turn may induce T cell cross-activation and priming against retina-specific antigens, triggering autoimmune reactions (49, 50). Other studies suggest, that a healthy gut microbiome is needed for prevention of autoimmunity through Treg homeostasis and that dysbiosis leads to changed or insufficiently effective Treg populations (51, 52). In ERU, the involvement of commensal microbiota has not been proven to date, however it is highly likely to play a possible role.

Equine T cell activation can be markedly reduced by co-incubation with adipose‐derived equine mesenchymal stem cells (MSC) in vitro, resulting in downscaling of CD25 as well as intracellular IFNγ, IL-10, and FoxP3 (39). This feature of MSC might be useful for the development of new therapeutic approaches in ERU (53), as suggested by in equine immune-mediated keratitis, where subconjunctival injection of autologous MSCs improved clinical signs of disease (54). Although clinical application of this therapeutic strategy needs more in-depth validation, it might have the potential of broadening the currently applied methods of medical and surgical therapy, which classically comprise the systemical and topical application of immunosuppressive and anti-inflammatory medication, placement of a suprachoroidal cyclosporine sustainded-release device or pars plana vitrectomy (9, 55, 56).

Innate Immune System

The role of innate immune cells in autoimmune uveitis has been intensely investigated in the eyes of rodent EAU models (5760) and infiltration of these cells in eyes of ERU horses, although to a lesser extent, has also been observed (3). The involvement of monocytes, which might promote pro-inflammatory behavior, as well as destruction of the targeted retinal cells in ERU could be shown by expression of CD68 on infiltrating peripheral immune cells in the diseased retina (61). Monocytes are major mediators of tissue damage in EAU (62, 63) and are potent antigen-presenting cells (64), which makes their role in ERU especially interesting, particularly in regard to their interaction with specific retinal cells (65). Further indication of participation of innate immune cells in ERU was proven by identification of decreased talin 1 abundance, a key integrin regulator in leukocytes, and its interacting proteins in low-density-neutrophils which were obtained as byproduct of lymphocyte isolation (25, 66). Targeted investigations of pure granulocyte fractions of healthy and ERU horses revealed significant proteome changes in diseased state, which associated to MHC-I-mediated antigen presentation, RAF/MAP kinase cascade, and neutrophil degranulation (67). Interestingly, increased neutrophil degranulation was also described in other T-cell driven autoimmune diseases, such as multiple sclerosis, where it is linked to a generally pre-activated state of these cells (68). Moreover, neutrophil degranulation relates to deviant equine granulocyte proteome after in vitro stimulation (69), underlining a latent state of activation of granulocytes in ERU, even in quiescent stage of disease (67). The fact that neutrophils from ERU horses more readily perform NETosis supports these findings (70). The exact role of innate immune cells and their timing in and impact on ERU pathogenesis, however, is still not completely understood to date and needs more in-depth investigation.

Implication of a role of the complement system was shown by the identification of several complement factors and split factors and their increased expression in ERU sera and eyes (19, 61, 71). Namely complement factor B, as well as C3 derived split products C3d and iC3b showed highly increased abundance, indicating strong activation of the complement system not only in the peripheral blood stream but also locally in the eye itself (61, 71). Although the exact source of these complement components in ERU eyes is still unclear, they might be macrophage-derived, as suggested by studies on human retinas from patients with age-related macular degeneration (AMD) and in vivo retina analyses of rodent models for AMD (72). Other studies on C57BL/6 mice showed that complement factor B is constitutively expressed by RPE cells and this expression is positively regulated by inflammatory cytokines in the human RPE cell line ARPE19 (73), a process which might also apply to the increased occurrence in ERU eyes. Deviant regulation of the complement system is involved in the pathogenesis of various ocular diseases, including autoimmune uveitis (74, 75) and the severity of experimental autoimmune uveitis drastically decreases in complement-receptor deficient rodent models (7678). Moreover, EAU can be effectively suppressed in C57BL/6 mice when activation of complement via the alternative pathway is blocked through complement receptor CRIg-Fc. Since complement factor B is exclusive to the alternative pathway, increased intraocular levels in EAU suggest a role of this pathway in disease pathogenesis (79). Despite the high abundance of complement factor B in uveitic horse eyes, indicating a possible contribution of the alternative pathway to ERU pathogenesis, this correlation has not yet been assessed.

Serum Proteins

In the last decade, proteomic studies on sera of ERU diseased horses revealed substantial differences compared to sera of healthy horses (80). Due to breakdown of the BRB in course of multiple uveitic attacks, several of these proteins can also be detected inside the eye (3, 71, 81, 82).

Among these differentially expressed proteins, kininogen was identified with decreased abundance in sera of ERU horses (71). Contrary to this, kininogen showed increased levels in vitreous and retina in ERU, whereas it was not detectable in healthy eyes (71). Kininogen promotes angiogenesis and neovascularization (83), a process that plays a significant role in the pathogenesis of autoimmune uveitis (71). It is also a part of the kallikrein-kinin system, which promotes integrity loss of barrier systems, such as the blood-brain-barrier in a mouse model of multiple sclerosis (84). Increased vitreal and retinal kininogen levels combined with a decrease in serum of ERU horses therefore points to disruption of the BRB in course of disease. IgM levels were also increased in ERU sera compared to healthy horses, suggesting a recent immune response pattern prior to blood sampling (71).

Decreased levels of pigment epithelium derived factor (PEDF) could be shown in sera of ERU horses (82), which was also observed in the retina and the vitreous of these animals in earlier studies (19, 20). PEDF was also decreased in plasma and retina from rats with endotoxin-induced uveitis, describing PEDF as a negative acute-phase protein (85). Furthermore, PEDF plays an important role in the viability of rat and mouse retinal cells (86, 87), the protection of human retinal pigment epithelium cells (ARPE-19 cell line) against oxidative stress (88) and the protection of tight junction proteins in rat eyes (85). Therefore, the decrease of PEDF in periphery as well as the target organ suggests increased permeability of the blood retinal-barrier.

Target Organ

Retinal Pigment Epithelium

Increased BRB permeability in ERU is supported by studies on the outer BRB, which physiologically maintains ocular immune privilege through disconnection of the inner eye from peripheral blood-derived immune cells (24). The outer BRB is formed by retinal pigment epithelium (RPE) cells, which play a crucial role in the mainly avascular retina of the horse, since they maintain homeostasis of retinal cells (89). This is also reflected in the RPE surface proteome, which strongly associates to transport processes (90). Furthermore, horses with ERU display a distinctly changed RPE membrane protein repertoire in ERU (91). A significant decrease of peripherin 2, a protein that is associated with membrane fusion processes, in RPE cells of ERU horses is thought to provoke disruption of the cell-to-cell junctions which are physiolocically maintained through these cells (92), which in turn points to increased permeability of the outer BRB. As also hypothesized for PEDF (19, 20, 82), this might promote integrity loss of the BRB. Although loss of barrier function of the BRB and leukocyte infiltration in course of ERU is evident (4, 27, 91, 92), the exact chronological order of these events is still not completely clear. Integrity loss of the BRB may occur either as an initial process which subsequently facilitates migration of peripheral immune cells into the eye, or as a secondary effect in response to leukocyte recruitment. The latter could be shown in an IRBP-induced B10.RIII mouse EAU model, where Evans blue dye, which was injected shortly before killing of the mice, could be observed leaking into the retinal parenchyma only after intravascular sticking and extravasation of blood-derived leukocytes from retinal venules took place (93). Further studies on murine EAU models implied that BRB breakdown might be a result of several consecutive steps, actively triggered by intravascular adherence of primed lymphocytes, followed by changed expression of adhesion molecules and chemokine receptors in the vascular epithelium and reduced shear stress in retinal veins prior to infiltration (9397). Interestingly, adoptive transfer of EAU in lewis rats could show that antigen specificity for retinal antigens is not a prerequisite for migration of activated T cells through the BRB, however, for the induction of actual intraocular inflammation, it is indispensable (98).

Retina and Vitreous

After infiltrating the eye, peripheral immune cells accumulate in the iris, ciliary body and retina of horses with ERU (4, 27). Although the vast majority of the infiltrating cells were identified as CD4+ T cells (2, 27), pure granulocyte infiltrates can be observed in few spontaneous ERU and IRBP induced cases (3, 4). This is in stark contrast to the composition of the cellular infiltrate in IRBP-induced murine EAU models, where a more heterogeneous cell infiltrate can be observed, containing around 30% CD4+ T cells and a high percentage of macrophages (99, 100). This points to different molecular pathways and cellular processes in EAU and ERU, underlining the great variability of disease pathogenesis between species, which needs to be kept in mind while interpreting associated data. In the base of the ciliary body and the iris of ERU horses, the infiltrated cells organize into lymphoid follicle nodules, which predominantly comprise CD3+ T cells (26, 27). In the retina, we mostly see scattered areas of T cell infiltration, especially near the ora ciliaris retinae and the optic disc, but occasional lymphoid follicle formation is also observed (27). Interestingly, the majority of infiltrated CD4+ lymphocytes in the ERU retina also show surface expression of CD166, a molecule associated with activation and transmigration of T cells into inflamed tissues (26). Presence of phosphorylated signal transducer and activator of transcription (STAT) proteins 1 and 5 in these cells underline the role of activated Th1 cells in ERU but also indirectly point toward inhibition of the Th17 pathway (26). Detection of IL‐6, IL‐17, and IL‐23 in cell infiltrates of the ciliary body and iris, on the other hand, indirectly suggests that a Th17 mediated immune reaction might also present in ERU eyes (42). In rodent models, EAU can develop either as a Th1 or a Th17 response depending on the model and the antigen used for induction, showing the importance of both T cell subpopulations in disease pathogenesis (41, 101104). In ERU, on the other hand, the important role of Th1 lymphocytes as effector cells is well known (2, 3, 36), the exact impact of Th17 cells on disease pathogenesis, however, merits further investigations.

Through membrane proteome analyses of equine retinae, numerous differentially expressed proteins were identified in ERU, several of which were associated to retinal Mueller glia cell derived proteins (105). Retinal Mueller glia cells, the main macroglial cells of the retina, are indispensable for the maintenance of retinal function and integrity (106). Apart from their proinflammatory role in ERU (20) and in murine EAU (65), these cells display a gliotic phenotype, which presents with morphological changes and impaired functionality in diseased retinae (12, 20, 105, 107). One of the important functions of these cells is the regulation of potassium and water homeostasis in the retina (108, 109), which shows severe misbalance in ERU (105, 107). This is mirrored in changed abundance and distribution pattern of potassium channels Kir4.1 and Kir2.1 as well as water channels AQP4, AQP5, and AQP11 which is hypothesized to contribute to impaired Mueller cell function and intracellular fluid regulation and, subsequently, retinal edema (105, 107, 110), a severe complication of ERU (27). Furthermore, in ERU, dysfunctional RMG are connected to decreased secretion of Wnt signaling inhibitors DKK3 and SFRP2, which point to a regulatory role of Wnt signaling in ERU (111), similar to other autoimmune diseases (112, 113). These insights on RMG support the role of these cells as prime responders to autoimmune triggers in ERU, promoting inflammatory processes and increasing severity of disease pathogenesis.

Additionally, severe extracellular matrix (ECM) remodeling in ERU was suggested through the identification of the proteins fibronectin and osteopontin (OPN) (105, 114). Fibronectin is an important constituent of the vertebrate ECM, mediates cell-ECM interactions (115) and also plays a role in adhesion and migration of cultured rat Mueller cells (116). Since fibronectin expression in Mueller cell endfeet might contribute to the attachment of the retina to the vitreous body, the changed distribution of fibronectin in ERU retinae points to impaired Mueller cell adhesion (114). OPN is a multifunctional protein with pro-inflammatory as well as neuroprotective properties (117, 118). In EAU mice, an increased abundance of OPN correlates with severity of inflammation (119), whereas Mueller glia-derived OPN promotes photoreceptor survival in the Pde6brd1 mouse model of retinal degeneration (120). OPN also shows neuroprotective properties in porcine Mueller glia cells in vitro (121). Decreased expression of OPN in vitreous and retina might therefore point to reduced neuroprotection in ERU retinae (114). ERU-associated lack of neuroprotection in the retina was underlined by the identification of decreased levels and decreased activity of tissue inhibitor of metalloproteinases (TIMP)-2 in retina and vitreous (122). TIMP2 influences the activity of metalloproteinases (MMP) (123), which modulate cell-cell and cell-ECM interactions (124). It also has neuroprotective properties (125) and can inhibit migration of cells over physiological barriers (126). Hence, decrease in TIMP2 activity might promote the inflammatory responses in ERU. It has been shown that migrating immune cells, especially Th1 cells, increase their MMP2 and MMP9 expression to overcome blood-tissue barriers (127) and that inhibition of these MMPs ameliorates experimental autoimmune uveitis in rodent models (128, 129). Interestingly, infiltrating cells in ERU retinae stained highly positive for MMP9 (122).

Apart from high levels of IgG in eyes of ERU horses (3), ERU vitreous also contains various immunoglobulins of the subclass M (IgM), which show a broad reaction pattern against retinal proteins (81), whereas healthy eyes contain no IgM (or IgG). Since IgM is an activator of the complement system and complement has been proven to be highly present in the inner eye of ERU horses (61, 111), the presence of IgM in the ERU vitreous further implies involvement of innate immune system components in this T cell driven autoimmune disease. Further screening of retinal protein with vitreous from ERU horses provided potential retinal autoantigens which are targeted by IgM autoantibodies (81). One of these IgM targets was identified as neurofilament medium (NF-M) (81, 130). Interestingly, IgG response to NF-M was merely detectable, supporting the presence of a thymus-independent immune reaction with prolonged persistence of IgM response toward NF-M and lack of IgM/IgG switch (130). In the retina, on the other hand, NF-M showed decreased expression (130). The combination of active downscaling in damaged RMG and release of NF-M fragments into the adjacent vitreous might cause this combination of high vitreal occurrence and low retinal levels of NF-M (130). A further novel membrane-bound autoantigen, synaptotagmin-1, was identified with decreased expression in ERU (131), which might be a result of active downscaling of this protein in the retina through impaired neurotransmitter release in ERU. Since Synaptotagmin-1 is also expressed in the pineal gland, and pinealitis is known to concurrently develop in ERU horses (132), synaptotagmin-1 might play a role in pineal immunopathology in ERU (131). Whether these IgM-targeted retinal antigens qualify as autoantigen targets for ERU, however, remains to be proven in further studies.

Conclusions

Overall, these novel immunological insights in ERU pathogenesis point to a complex interplay of several dysregulated mechanisms, which, on the one hand, can be linked to changes in cellular and humoral components of the immune system, such as a deviant functional phenotype of T cells with increased migratory ability and a decreased apoptosis rate, latently activated granulocytes and involvement of the alternative pathway of the complement system. On the other hand, this dysregulation points to a pivotal pro-inflammatory role of retinal cells with critically impaired function in the target organ itself. Although these findings shed more light on disease mechanisms, the interaction of these dysfunctional molecular mechanisms driving ERU as well as their exact individual role and timing in ERU pathogenesis needs further assessment. Establishment of a Mueller glia cell line (133) and characterization of cultured RPE cells (90) provide valuable tools for more in depth functional examinations of ERU pathology. Additionally, the possibility of commensal microbiota involvement in ERU onset needs to be addressed.

Author Contributions

CD conceived the outline of the manuscript. CD and RD wrote the manuscript. All authors contributed to the article and approved the submitted version.

Funding

Work from research group Deeg mentioned in this review was supported by grants from the Münchener Universitätsgesellschaft 31697 (to RD) and the Deutsche Forschungsgemeinschaft DFG DE 719/4-3 (to CD).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Romeike A, Brugmann M, Drommer W. Immunohistochemical studies in equine recurrent uveitis (ERU). Vet Pathol (1998) 35(6):515–26. doi: 10.1177/030098589803500606

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Gilger BC, Malok E, Cutter KV, Stewart T, Horohov DW, Allen JB. Characterization of T-lymphocytes in the anterior uvea of eyes with chronic equine recurrent uveitis. Vet Immunol Immunopathol (1999) 71(1):17–28. doi: 10.1016/S0165-2427(99)00082-3

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Deeg CA, Kaspers B, Gerhards H, Thurau SR, Wollanke B, Wildner G. Immune responses to retinal autoantigens and peptides in equine recurrent uveitis. Invest Ophthalmol Vis Sci (2001) 42(2):393–8.

PubMed Abstract | Google Scholar

4. Deeg CA, Thurau SR, Gerhards H, Ehrenhofer M, Wildner G, Kaspers B. Uveitis in horses induced by interphotoreceptor retinoid-binding protein is similar to the spontaneous disease. Eur J Immunol (2002) 32(9):2598–606. doi: 10.1002/1521-4141(200209)32:9<2598::AID-IMMU2598>3.0.CO;2-#

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Gerding JC, Gilger BC. Prognosis and impact of equine recurrent uveitis. Equine Vet J (2016) 48(3):290–8. doi: 10.1111/evj.12451

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Kulbrock M, Lehner S, Metzger J, Ohnesorge B, Distl O. A genome-wide association study identifies risk loci to equine recurrent uveitis in German warmblood horses. PloS One (2013) 8(8):e71619. doi: 10.1371/journal.pone.0071619

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Deeg CA, Marti E, Gaillard C, Kaspers B. Equine recurrent uveitis is strongly associated with the MHC class I haplotype ELA-A9. Equine Vet J (2004) 36(1):73–5. doi: 10.2746/0425164044864651

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Spiess BM. Equine recurrent uveitis: the European viewpoint. Equine Vet J Suppl (2010) 37:50–6. doi: 10.1111/j.2042-3306.2010.tb05635.x

CrossRef Full Text | Google Scholar

9. McMullen RJ Jr., Fischer BM. Medical and Surgical Management of Equine Recurrent Uveitis. Vet Clin North Am Equine Pract (2017) 33(3):465–81. doi: 10.1016/j.cveq.2017.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Deeg CA, Hauck SM, Amann B, Pompetzki D, Altmann F, Raith A, et al. Equine recurrent uveitis–a spontaneous horse model of uveitis. Ophthalmic Res (2008) 40(3-4):151–3. doi: 10.1159/000119867

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Neutzner RV, Jager M, Friedburg C, Deeg CA, Lorenz B. [Blind spot enlargement syndrome in acute zonal occult outer retinopathy with detection of autoantibodies against the retinal antigens CRALBP and S-Ag]. Ophthalmologe (2011) 108(11):1045–9. doi: 10.1007/s00347-011-2406-x

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Deeg CA, Pompetzki D, Raith AJ, Hauck SM, Amann B, Suppmann S, et al. Identification and functional validation of novel autoantigens in equine uveitis. Mol Cell Proteomics (2006) 5(8):1462–70. doi: 10.1074/mcp.M500352-MCP200 doi: M500352-MCP200 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Deeg CA, Hauck SM, Amann B, Kremmer E, Stangassinger M, Ueffing M. Major retinal autoantigens remain stably expressed during all stages of spontaneous uveitis. Mol Immunol (2007) 44(13):3291–6. doi: 10.1016/j.molimm.2007.02.027 doi: S0161-5890(07)00096-X [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hauck SM, Lepper MF, Hertl M, Sekundo W, Deeg CA. Proteome Dynamics in Biobanked Horse Peripheral Blood Derived Lymphocytes (PBL) with Induced Autoimmune Uveitis. Proteomics (2017) 17(19):1–5. doi: 10.1002/pmic.201700013

CrossRef Full Text | Google Scholar

15. Deeg CA, Raith AJ, Amann B, Crabb JW, Thurau SR, Hauck SM, et al. CRALBP is a highly prevalent autoantigen for human autoimmune uveitis. Clin Dev Immunol (2007) 2007:39245. doi: 10.1155/2007/39245

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Deeg CA, Amann B, Raith AJ, Kaspers B. Inter- and intramolecular epitope spreading in equine recurrent uveitis. Invest Ophthalmol Vis Sci (2006) 47(2):652–6. doi: 10.1167/iovs.05-0789 doi: 47/2/652 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Vanderlugt CJ, Miller SD. Epitope spreading. Curr Opin Immunol (1996) 8(6):831–6. doi: 10.1016/s0952-7915(96)80012-4

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Ujiie H, Yoshimoto N, Natsuga K, Muramatsu K, Iwata H, Nishie W, et al. Immune Reaction to Type XVII Collagen Induces Intramolecular and Intermolecular Epitope Spreading in Experimental Bullous Pemphigoid Models. Front Immunol (2019) 10:1410. doi: 10.3389/fimmu.2019.01410

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Deeg CA, Altmann F, Hauck SM, Schoeffmann S, Amann B, Stangassinger M, et al. Down-regulation of pigment epithelium-derived factor in uveitic lesion associates with focal vascular endothelial growth factor expression and breakdown of the blood-retinal barrier. Proteomics (2007) 7(9):1540–8. doi: 10.1002/pmic.200600795

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Hauck SM, Schoeffmann S, Amann B, Stangassinger M, Gerhards H, Ueffing M, et al. Retinal Mueller glial cells trigger the hallmark inflammatory process in autoimmune uveitis. J Proteome Res (2007) 6(6):2121–31. doi: 10.1021/pr060668y

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Forrester JV, Xu H. Good news-bad news: the Yin and Yang of immune privilege in the eye. Front Immunol (2012) 3:338. doi: 10.3389/fimmu.2012.00338

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Forrester JV, McMenamin PG, Dando SJ. CNS infection and immune privilege. Nat Rev Neurosci (2018) 19(11):655–71. doi: 10.1038/s41583-018-0070-8

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Streilein JW, Takeuchi M, Taylor AW. Immune privilege, T-cell tolerance, and tissue-restricted autoimmunity. Hum Immunol (1997) 52(2):138–43. doi: 10.1016/S0198-8859(96)00288-1 doi: S0198-8859(96)00288-1 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Taylor AW, Ng TF. Negative regulators that mediate ocular immune privilege. J Leukoc Biol (2018) 103(6)1179–87. doi: 10.1002/JLB.3MIR0817-337R

CrossRef Full Text | Google Scholar

25. Degroote RL, Hauck SM, Treutlein G, Amann B, Frohlich KJ, Kremmer E, et al. Expression Changes and Novel Interaction Partners of Talin 1 in Effector Cells of Autoimmune Uveitis. J Proteome Res (2013) 12(12):5812–9. doi: 10.1021/pr400837f

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Kleinwort KJ, Amann B, Hauck SM, Feederle R, Sekundo W, Deeg CA. Immunological Characterization of Intraocular Lymphoid Follicles in a Spontaneous Recurrent Uveitis Model. Invest Ophthalmol Vis Sci (2016) 57(10):4504–11. doi: 10.1167/iovs.16-19787

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Deeg CA, Ehrenhofer M, Thurau SR, Reese S, Wildner G, Kaspers B. Immunopathology of recurrent uveitis in spontaneously diseased horses. Exp Eye Res (2002) 75(2):127–33. doi: 10.1006/exer.2002.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Caspi RR, Roberge FG, McAllister CG, el-Saied M, Kuwabara T, Gery I, et al. T cell lines mediating experimental autoimmune uveoretinitis (EAU) in the rat. J Immunol (1986) 136(3):928–33.

PubMed Abstract | Google Scholar

29. Palestine AG, Mc Allister C, Carter C, Keenan AM, Vistica B, Gery I, et al. Lymphocyte migration in the adoptive transfer of EAU. Invest Ophthalmol Vis Sci (1986) 27(4):611–5.

PubMed Abstract | Google Scholar

30. Caspi RR. Th1 and Th2 responses in pathogenesis and regulation of experimental autoimmune uveoretinitis. Int Rev Immunol (2002) 21(2-3):197–208. doi: 10.1080/08830180212063

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Caspi RR, Chan CC, Fujino Y, Najafian F, Grover S, Hansen CT, et al. Recruitment of antigen-nonspecific cells plays a pivotal role in the pathogenesis of a T cell-mediated organ-specific autoimmune disease, experimental autoimmune uveoretinitis. J Neuroimmunol (1993) 47(2):177–88. doi: 10.1016/0165-5728(93)90028-w

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Degroote RL, Hauck SM, Amann B, Hirmer S, Ueffing M, Deeg CA. Unraveling the equine lymphocyte proteome: differential septin 7 expression associates with immune cells in equine recurrent uveitis. PloS One (2014) 9(3):e91684. doi: 10.1371/journal.pone.0091684

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Degroote RL, Uhl PB, Amann B, Krackhardt AM, Ueffing M, Hauck SM, et al. Formin like 1 expression is increased on CD4+ T lymphocytes in spontaneous autoimmune uveitis. J Proteomics (2017) 154:102–8. doi: 10.1016/j.jprot.2016.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Schauer M, Kleinwort KJH, Degroote RL, Wiedemann C, Kremmer E, Hauck SM, et al. Interaction of septin 7 and DOCK8 in equine lymphocytes reveals novel insights into signaling pathways associated with autoimmunity. Sci Rep (2018) 8(1):12332. doi: 10.1038/s41598-018-30753-7

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Thompson SB, Sandor AM, Lui V, Chung JW, Waldman MM, Long RA, et al. Formin-like 1 mediates effector T cell trafficking to inflammatory sites to enable T cell-mediated autoimmunity. Elife (2020) 9:1–27. doi: 10.7554/eLife.58046

CrossRef Full Text | Google Scholar

36. Wiedemann C, Amann B, Degroote RL, Witte T, Deeg CA. Aberrant Migratory Behavior of Immune Cells in Recurrent Autoimmune Uveitis in Horses. Front Cell Dev Biol (2020) 8:101. doi: 10.3389/fcell.2020.00101

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Tooley AJ, Gilden J, Jacobelli J, Beemiller P, Trimble WS, Kinoshita M, et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol (2009) 11(1):17–26. doi: 10.1038/ncb1808 doi: ncb1808 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Aydin SE, Kilic SS, Aytekin C, Kumar A, Porras O, Kainulainen L, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options - a review of 136 patients. J Clin Immunol (2015) 35(2):189–98. doi: 10.1007/s10875-014-0126-0

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Saldinger LK, Nelson SG, Bellone RR, Lassaline M, Mack M, Walker NJ, et al. Horses with equine recurrent uveitis have an activated CD4+ T-cell phenotype that can be modulated by mesenchymal stem cells in vitro. Vet Ophthalmol (2020) 23(1):160–70. doi: 10.1111/vop.12704

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Curto E, Messenger KM, Salmon JH, Gilger BC. Cytokine and chemokine profiles of aqueous humor and serum in horses with uveitis measured using multiplex bead immunoassay analysis. Vet Immunol Immunopathol (2016) 182:43–51. doi: 10.1016/j.vetimm.2016.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Luger D, Silver PB, Tang J, Cua D, Chen Z, Iwakura Y, et al. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category. J Exp Med (2008) 205(4):799–810. doi: 10.1084/jem.20071258 doi: jem.20071258 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Regan DP, Aarnio MC, Davis WS, Carmichael KP, Vandenplas ML, Lauderdale JD, et al. Characterization of cytokines associated with Th17 cells in the eyes of horses with recurrent uveitis. Vet Ophthalmol (2012) 15(3):145–52. doi: 10.1111/j.1463-5224.2011.00951.x

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Simeonova GP, Krastev SZ, Simeonov RS. Immunological and pathological investigations in equine experimental uveitis. Vet Res Commun (2016) 40(3-4):107–15. doi: 10.1007/s11259-016-9659-4

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Sharma A, Rudra D. Emerging Functions of Regulatory T Cells in Tissue Homeostasis. Front Immunol (2018) 9:883. doi: 10.3389/fimmu.2018.00883

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Silver PB, Horai R, Chen J, Jittayasothorn Y, Chan CC, Villasmil R, et al. Retina-specific T regulatory cells bring about resolution and maintain remission of autoimmune uveitis. J Immunol (2015) 194(7):3011–9. doi: 10.4049/jimmunol.1402650

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Gilbert RM, Zhang X, Sampson RD, Ehrenstein MR, Nguyen DX, Chaudhry M, et al. Clinical Remission of Sight-Threatening Non-Infectious Uveitis Is Characterized by an Upregulation of Peripheral T-Regulatory Cell Polarized Towards T-bet and TIGIT. Front Immunol (2018) 9:907. doi: 10.3389/fimmu.2018.00907

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ruggieri S, Frassanito MA, Dammacco R, Guerriero S. Treg lymphocytes in autoimmune uveitis. Ocul Immunol Inflammation (2012) 20(4):255–61. doi: 10.3109/09273948.2012.681830

CrossRef Full Text | Google Scholar

48. Yeh S, Li Z, Forooghian F, Hwang FS, Cunningham MA, Pantanelli S, et al. CD4+Foxp3+ T-regulatory cells in noninfectious uveitis. Arch Ophthalmol (2009) 127(4):407–13. doi: 10.1001/archophthalmol.2009.32

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Horai R, Zarate-Blades CR, Dillenburg-Pilla P, Chen J, Kielczewski JL, Silver PB, et al. Microbiota-Dependent Activation of an Autoreactive T Cell Receptor Provokes Autoimmunity in an Immunologically Privileged Site. Immunity (2015) 43(2):343–53. doi: 10.1016/j.immuni.2015.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Horai R, Caspi RR. Microbiome and Autoimmune Uveitis. Front Immunol (2019) 10:232. doi: 10.3389/fimmu.2019.00232

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Ye Z, Wu C, Zhang N, Du L, Cao Q, Huang X, et al. Altered gut microbiome composition in patients with Vogt-Koyanagi-Harada disease. Gut Microbes (2020) 11(3):539–55. doi: 10.1080/19490976.2019.1700754

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Shimizu J, Kubota T, Takada E, Takai K, Fujiwara N, Arimitsu N, et al. Bifidobacteria Abundance-Featured Gut Microbiota Compositional Change in Patients with Behcet’s Disease. PloS One (2016) 11(4):e0153746. doi: 10.1371/journal.pone.0153746

PubMed Abstract | CrossRef Full Text | Google Scholar

53. MacDonald ES, Barrett JG. The Potential of Mesenchymal Stem Cells to Treat Systemic Inflammation in Horses. Front Vet Sci (2019) 6:507. doi: 10.3389/fvets.2019.00507

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Davis AB, Schnabel LV, Gilger BC. Subconjunctival bone marrow-derived mesenchymal stem cell therapy as a novel treatment alternative for equine immune-mediated keratitis: A case series. Vet Ophthalmol (2019) 22(5):674–82. doi: 10.1111/vop.12641

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Werry H, Gerhards H. [The surgical therapy of equine recurrent uveitis]. Tierarztl Prax (1992) 20(2):178–86.

PubMed Abstract | Google Scholar

56. Gilger BC, Salmon JH, Wilkie DA, Cruysberg LP, Kim J, Hayat M, et al. A novel bioerodible deep scleral lamellar cyclosporine implant for uveitis. Invest Ophthalmol Vis Sci (2006) 47(6):2596–605. doi: 10.1167/iovs.05-1540 doi: 47/6/2596 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Pepple KL, Wilson L, Van Gelder RN. Comparison of Aqueous and Vitreous Lymphocyte Populations From Two Rat Models of Experimental Uveitis. Invest Ophthalmol Vis Sci (2018) 59(6):2504–11. doi: 10.1167/iovs.18-24192

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Kerr EC, Raveney BJ, Copland DA, Dick AD, Nicholson LB. Analysis of retinal cellular infiltrate in experimental autoimmune uveoretinitis reveals multiple regulatory cell populations. J Autoimmun (2008) 31(4):354–61. doi: 10.1016/j.jaut.2008.08.006

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Jones LS, Rizzo LV, Agarwal RK, Tarrant TK, Chan CC, Wiggert B, et al. IFN-gamma-deficient mice develop experimental autoimmune uveitis in the context of a deviant effector response. J Immunol (1997) 158(12):5997–6005.

PubMed Abstract | Google Scholar

60. Kim SJ, Zhang M, Vistica BP, Chan CC, Shen DF, Wawrousek EF, et al. Induction of ocular inflammation by T-helper lymphocytes type 2. Invest Ophthalmol Vis Sci (2002) 43(3):758–65.

PubMed Abstract | Google Scholar

61. Zipplies JK, Kirschfink M, Amann B, Hauck SM, Stangassinger M, Deeg CA. Complement factor B expression profile in a spontaneous uveitis model. Immunobiology (2010) 215(12):949–55. doi: 10.1016/j.imbio.2010.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Robertson MJ, Erwig LP, Liversidge J, Forrester JV, Rees AJ, Dick AD. Retinal microenvironment controls resident and infiltrating macrophage function during uveoretinitis. Invest Ophthalmol Vis Sci (2002) 43(7):2250–7.

PubMed Abstract | Google Scholar

63. Khera TK, Copland DA, Boldison J, Lait PJ, Szymkowski DE, Dick AD, et al. Tumour necrosis factor-mediated macrophage activation in the target organ is critical for clinical manifestation of uveitis. Clin Exp Immunol (2012) 168(2):165–77. doi: 10.1111/j.1365-2249.2012.04567.x

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Chinnery HR, McMenamin PG, Dando SJ. Macrophage physiology in the eye. Pflugers Arch (2017) 469(3-4):501–15. doi: 10.1007/s00424-017-1947-5

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Okunuki Y, Mukai R, Nakao T, Tabor SJ, Butovsky O, Dana R, et al. Retinal microglia initiate neuroinflammation in ocular autoimmunity. Proc Natl Acad Sci U.S.A. (2019) 116(20):9989–98. doi: 10.1073/pnas.1820387116

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Degroote RL, Hauck SM, Kremmer E, Amann B, Ueffing M, Deeg CA. Altered expression of talin 1 in peripheral immune cells points to a significant role of the innate immune system in spontaneous autoimmune uveitis. J Proteomics (2012) 75(14):4536–44. doi: 10.1016/j.jprot.2012.01.023

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Weigand M, Hauck SM, Deeg CA, Degroote RL. Deviant proteome profile of equine granulocytes associates to latent activation status in organ specific autoimmune disease. J Proteomics (2020) 230:1–9. doi: 10.1016/j.jprot.2020.103989

CrossRef Full Text | Google Scholar

68. Naegele M, Tillack K, Reinhardt S, Schippling S, Martin R, Sospedra M. Neutrophils in multiple sclerosis are characterized by a primed phenotype. J Neuroimmunol (2012) 242(1-2):60–71. doi: 10.1016/j.jneuroim.2011.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Degroote RL, Weigand M, Hauck SM, Deeg CA. IL8 and PMA Trigger the Regulation of Different Biological Processes in Granulocyte Activation. Front Immunol (2019) 10:3064. doi: 10.3389/fimmu.2019.03064

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Fingerhut L, Ohnesorge B, von Borstel M, Schumski A, Strutzberg-Minder K, Morgelin M, et al. Neutrophil Extracellular Traps in the Pathogenesis of Equine Recurrent Uveitis (ERU). Cells (2019) 8(12):1–22. doi: 10.3390/cells8121528

CrossRef Full Text | Google Scholar

71. Zipplies JK, Hauck SM, Schoeffmann S, Amann B, van der Meijden CH, Stangassinger M, et al. Kininogen in autoimmune uveitis: decrease in peripheral blood stream versus increase in target tissue. Invest Ophthalmol Vis Sci (2010) 51(1):375–82. doi: 10.1167/iovs.09-4094

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Natoli R, Fernando N, Jiao H, Racic T, Madigan M, Barnett NL, et al. Retinal Macrophages Synthesize C3 and Activate Complement in AMD and in Models of Focal Retinal Degeneration. Invest Ophthalmol Vis Sci (2017) 58(7):2977–90. doi: 10.1167/iovs.17-21672

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Chen M, Muckersie E, Robertson M, Forrester JV, Xu H. Up-regulation of complement factor B in retinal pigment epithelial cells is accompanied by complement activation in the aged retina. Exp Eye Res (2008) 87(6):543–50. doi: 10.1016/j.exer.2008.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Mohlin C, Sandholm K, Ekdahl KN, Nilsson B. The link between morphology and complement in ocular disease. Mol Immunol (2017) 89:84–99. doi: 10.1016/j.molimm.2017.05.028

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Bora NS, Jha P, Bora PS. The role of complement in ocular pathology. Semin Immunopathol (2008) 30(2):85–95. doi: 10.1007/s00281-008-0110-y

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Zhang L, Bell BA, Yu M, Chan CC, Peachey NS, Fung J, et al. Complement anaphylatoxin receptors C3aR and C5aR are required in the pathogenesis of experimental autoimmune uveitis. J Leukoc Biol (2016) 99(3):447–54. doi: 10.1189/jlb.3A0415-157R

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Jha P, Sohn JH, Xu Q, Wang Y, Kaplan HJ, Bora PS, et al. Suppression of complement regulatory proteins (CRPs) exacerbates experimental autoimmune anterior uveitis (EAAU). J Immunol (2006) 176(12):7221–31. doi: 10.4049/jimmunol.176.12.7221

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Jha P, Sohn JH, Xu Q, Nishihori H, Wang Y, Nishihori S, et al. The complement system plays a critical role in the development of experimental autoimmune anterior uveitis. Invest Ophthalmol Vis Sci (2006) 47(3):1030–8. doi: 10.1167/iovs.05-1062

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Chen M, Muckersie E, Luo C, Forrester JV, Xu H. Inhibition of the alternative pathway of complement activation reduces inflammation in experimental autoimmune uveoretinitis. Eur J Immunol (2010) 40(10):2870–81. doi: 10.1002/eji.201040323

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Chiaradia E, Miller I. In slow pace towards the proteome of equine body fluids. J Proteomics (2020) 225:103880. doi: 10.1016/j.jprot.2020.103880

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Zipplies JK, Hauck SM, Eberhardt C, Hirmer S, Amann B, Stangassinger M, et al. Miscellaneous vitreous-derived IgM antibodies target numerous retinal proteins in equine recurrent uveitis. Vet Ophthalmol (2012) 15 Suppl;2:57–64. doi: 10.1111/j.1463-5224.2012.01010.x

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Zipplies JK, Hauck SM, Schoeffmann S, Amann B, Stangassinger M, Ueffing M, et al. Serum PEDF levels are decreased in a spontaneous animal model for human autoimmune uveitis. J Proteome Res (2009) 8(2):992–8. doi: 10.1021/pr800694y 10.1021/pr800694y [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Guo YL, Colman RW. Two faces of high-molecular-weight kininogen (HK) in angiogenesis: bradykinin turns it on and cleaved HK (HKa) turns it off. J Thromb Haemost (2005) 3(4):670–6. doi: 10.1111/j.1538-7836.2005.01218.x

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Gobel K, Asaridou CM, Merker M, Eichler S, Herrmann AM, Geuss E, et al. Plasma kallikrein modulates immune cell trafficking during neuroinflammation via PAR2 and bradykinin release. Proc Natl Acad Sci USA (2019) 116(1):271–6. doi: 10.1073/pnas.1810020116

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Zhang SX, Wang JJ, Gao G, Shao C, Mott R, Ma JX. Pigment epithelium-derived factor (PEDF) is an endogenous antiinflammatory factor. FASEB J (2006) 20(2):323–5. doi: 10.1096/fj.05-4313fje doi: 05-4313fje [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Comitato A, Subramanian P, Turchiano G, Montanari M, Becerra SP, Marigo V. Pigment epithelium-derived factor hinders photoreceptor cell death by reducing intracellular calcium in the degenerating retina. Cell Death Dis (2018) 9(5):560. doi: 10.1038/s41419-018-0613-y

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Eichler W, Savkovic-Cvijic H, Burger S, Beck M, Schmidt M, Wiedemann P, et al. Muller Cell-Derived PEDF Mediates Neuroprotection via STAT3 Activation. Cell Physiol Biochem (2017) 44(4):1411–24. doi: 10.1159/000485537

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Wang X, Liu X, Ren Y, Liu Y, Han S, Zhao J, et al. PEDF protects human retinal pigment epithelial cells against oxidative stress via upregulation of UCP2 expression. Mol Med Rep (2019) 19(1):59–74. doi: 10.3892/mmr.2018.9645

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Strauss O. The retinal pigment epithelium in visual function. Physiol Rev (2005) 85(3):845–81. doi: 10.1152/physrev.00021.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Szober CM, Hauck SM, Euler KN, Frohlich KJ, Alge-Priglinger C, Ueffing M, et al. Profound re-organization of cell surface proteome in equine retinal pigment epithelial cells in response to in vitro culturing. Int J Mol Sci (2012) 13(11):14053–72. doi: 10.3390/ijms131114053

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Uhl PB, Szober CM, Amann B, Alge-Priglinger C, Ueffing M, Hauck SM, et al. In situ cell surface proteomics reveals differentially expressed membrane proteins in retinal pigment epithelial cells during autoimmune uveitis. J Proteomics (2014) 109:50–62. doi: 10.1016/j.jprot.2014.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Uhl PB, Amann B, Hauck SM, Deeg CA. Novel localization of peripherin 2, the photoreceptor-specific retinal degeneration slow protein, in retinal pigment epithelium. Int J Mol Sci (2015) 16(2):2678–92. doi: 10.3390/ijms16022678

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Xu H, Forrester JV, Liversidge J, Crane IJ. Leukocyte trafficking in experimental autoimmune uveitis: breakdown of blood-retinal barrier and upregulation of cellular adhesion molecules. Invest Ophthalmol Vis Sci (2003) 44(1):226–34. doi: 10.1167/iovs.01-1202

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Xu H, Manivannan A, Liversidge J, Sharp PF, Forrester JV, Crane IJ. Involvement of CD44 in leukocyte trafficking at the blood-retinal barrier. J Leukoc Biol (2002) 72:1133–41, 72(6).

PubMed Abstract | Google Scholar

95. Xu H, Manivannan A, Goatman KA, Jiang HR, Liversidge J, Sharp PF, et al. Reduction in shear stress, activation of the endothelium, and leukocyte priming are all required for leukocyte passage across the blood–retina barrier. J Leukoc Biol (2004) 75(2):224–32. doi: 10.1189/jlb.1002479

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Xu H, Manivannan A, Jiang HR, Liversidge J, Sharp PF, Forrester JV, et al. Recruitment of IFN-gamma-producing (Th1-like) cells into the inflamed retina in vivo is preferentially regulated by P-selectin glycoprotein ligand 1:P/E-selectin interactions. J Immunol (2004) 172(5):3215–24. doi: 10.4049/jimmunol.172.5.3215

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Crane IJ, Xu H, Wallace C, Manivannan A, Mack M, Liversidge J, et al. Involvement of CCR5 in the passage of Th1-type cells across the blood-retina barrier in experimental autoimmune uveitis. J Leukoc Biol (2006) 79(3):435–43. doi: 10.1189/jlb.0305130

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Thurau SR, Mempel TR, Flugel A, Diedrichs-Mohring M, Krombach F, Kawakami N, et al. The fate of autoreactive, GFP+ T cells in rat models of uveitis analyzed by intravital fluorescence microscopy and FACS. Int Immunol (2004) 16(11):1573–82. doi: 10.1093/intimm/dxh158

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Zhao J, Chen M, Xu H. Experimental autoimmune uveoretinitis (EAU)-related tissue damage and angiogenesis is reduced in CCL2(-)/(-)CX(3)CR1gfp/gfp mice. Invest Ophthalmol Vis Sci (2014) 55(11):7572–82. doi: 10.1167/iovs.14-15495

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Chen M, Zhao J, Ali IHA, Marry S, Augustine J, Bhuckory M, et al. Cytokine Signaling Protein 3 Deficiency in Myeloid Cells Promotes Retinal Degeneration and Angiogenesis through Arginase-1 Up-Regulation in Experimental Autoimmune Uveoretinitis. Am J Pathol (2018) 188(4):1007–20. doi: 10.1016/j.ajpath.2017.12.021

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, Nussenblatt RB, et al. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med (2007) 13(6):711–8. doi: 10.1038/nm1585 doi: nm1585 [pii].

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Bing SJ, Shemesh I, Chong WP, Horai R, Jittayasothorn Y, Silver PB, et al. AS101 ameliorates experimental autoimmune uveitis by regulating Th1 and Th17 responses and inducing Treg cells. J Autoimmun (2019) 100:52–61. doi: 10.1016/j.jaut.2019.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Horai R, Caspi RR. Cytokines in autoimmune uveitis. J Interferon Cytokine Res (2011) 31(10):733–44. doi: 10.1089/jir.2011.0042

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Weinstein JE, Pepple KL. Cytokines in uveitis. Curr Opin Ophthalmol (2018) 29(3):267–74. doi: 10.1097/ICU.0000000000000466

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Hauck SM, Dietter J, Kramer RL, Hofmaier F, Zipplies JK, Amann B, et al. Deciphering membrane-associated molecular processes in target tissue of autoimmune uveitis by label-free quantitative mass spectrometry. Mol Cell Proteomics (2010) 9(10):2292–305. doi: 10.1074/mcp.M110.001073

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Newman E, Reichenbach A. The Muller cell: a functional element of the retina. Trends Neurosci (1996) 19(8):307–12. doi: 10.1016/0166-2236(96)10040-0

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Eberhardt C, Amann B, Feuchtinger A, Hauck SM, Deeg CA. Differential expression of inwardly rectifying K+ channels and aquaporins 4 and 5 in autoimmune uveitis indicates misbalance in Muller glial cell-dependent ion and water homeostasis. Glia (2011) 59(5):697–707. doi: 10.1002/glia.21139

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Bringmann A, Pannicke T, Grosche J, Francke M, Wiedemann P, Skatchkov SN, et al. Muller cells in the healthy and diseased retina. Prog Retin Eye Res (2006) 25(4):397–424. doi: 10.1016/j.preteyeres.2006.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, et al. Cellular signaling and factors involved in Muller cell gliosis: neuroprotective and detrimental effects. Prog Retin Eye Res (2009) 28(6):423–51. doi: 10.1016/j.preteyeres.2009.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Deeg CA, Amann B, Lutz K, Hirmer S, Lutterberg K, Kremmer E, et al. Aquaporin 11, a regulator of water efflux at retinal Muller glial cell surface decreases concomitant with immune-mediated gliosis. J Neuroinflammation (2016) 13(1):89. doi: 10.1186/s12974-016-0554-2

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Hauck SM, Hofmaier F, Dietter J, Swadzba ME, Blindert M, Amann B, et al. Label-free LC-MSMS analysis of vitreous from autoimmune uveitis reveals a significant decrease in secreted Wnt signalling inhibitors DKK3 and SFRP2. J Proteomics (2012) 75(14):4545–54. doi: 10.1016/j.jprot.2012.04.052

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Shi J, Chi S, Xue J, Yang J, Li F, Liu X. Emerging Role and Therapeutic Implication of Wnt Signaling Pathways in Autoimmune Diseases. J Immunol Res (2016) 2016:9392132. doi: 10.1155/2016/9392132

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Cici D, Corrado A, Rotondo C, Cantatore FP. Wnt Signaling and Biological Therapy in Rheumatoid Arthritis and Spondyloarthritis. Int J Mol Sci (2019) 20(22):1–15. doi: 10.3390/ijms20225552

CrossRef Full Text | Google Scholar

114. Deeg CA, Eberhardt C, Hofmaier F, Amann B, Hauck SM. Osteopontin and fibronectin levels are decreased in vitreous of autoimmune uveitis and retinal expression of both proteins indicates ECM re-modeling. PloS One (2011) 6(12):e27674. doi: 10.1371/journal.pone.0027674

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Pankov R, Yamada KM. Fibronectin at a glance. J Cell Sci (2002) 115(Pt 20):3861–3. doi: 10.1242/jcs.00059

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Wang XL, Yu T, Zhang JS, Yan QC, Luo YH. Fibronectin and focal adhesion kinase small interfering RNA modulate rat retinal Muller cells adhesion and migration. Cell Mol Neurobiol (2009) 29(4):549–56. doi: 10.1007/s10571-009-9346-x

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Braitch M, Constantinescu CS. The role of osteopontin in experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS). Inflammation Allergy Drug Targets (2010) 9(4):249–56. doi: 10.2174/187152810793358778

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Hikita ST, Vistica BP, Jones HR, Keswani JR, Watson MM, Ericson VR, et al. Osteopontin is proinflammatory in experimental autoimmune uveitis. Invest Ophthalmol Vis Sci (2006) 47(10):4435–43. doi: 10.1167/iovs.06-0064

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Kitamura M, Iwabuchi K, Kitaichi N, Kon S, Kitamei H, Namba K, et al. Osteopontin aggravates experimental autoimmune uveoretinitis in mice. J Immunol (2007) 178(10):6567–72. doi: 10.4049/jimmunol.178.10.6567

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Del Rio P, Irmler M, Arango-Gonzalez B, Favor J, Bobe C, Bartsch U, et al. GDNF-induced osteopontin from Muller glial cells promotes photoreceptor survival in the Pde6brd1 mouse model of retinal degeneration. Glia (2011) 59(5):821–32. doi: 10.1002/glia.21155

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Ruzafa N, Pereiro X, Lepper MF, Hauck SM, Vecino E. A Proteomics Approach to Identify Candidate Proteins Secreted by Muller Glia that Protect Ganglion Cells in the Retina. Proteomics (2018) 18(11):e1700321. doi: 10.1002/pmic.201700321

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Hofmaier F, Hauck SM, Amann B, Degroote RL, Deeg CA. Changes in matrix metalloproteinase network in a spontaneous autoimmune uveitis model. Invest Ophthalmol Vis Sci (2011) 52(5):2314–20. doi: 10.1167/iovs.10-6475

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Brew K, Dinakarpandian D, Nagase H. Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim Biophys Acta (2000) 1477(1-2):267–83. doi: 10.1016/s0167-4838(99)00279-4

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Murphy G, Nagase H. Progress in matrix metalloproteinase research. Mol Aspects Med (2008) 29(5):290–308. doi: 10.1016/j.mam.2008.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Magnoni S, Baker A, Thomson S, Jordan G, George SJ, McColl BW, et al. Neuroprotective effect of adenoviral-mediated gene transfer of TIMP-1 and -2 in ischemic brain injury. Gene Ther (2007) 14(7):621–5. doi: 10.1038/sj.gt.3302894

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Woods CC, Sundar K, Tessler C, Lebsack TW, Grainger L, Nielsen A, et al. Tissue inhibitor of metalloproteinase-2 inhibits T-cell infiltration and preserves pancreatic beta-cell function in an in vitro type 1 diabetes mellitus model. J Autoimmun (2006) 27(1):28–37. doi: 10.1016/j.jaut.2006.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Abraham M, Shapiro S, Karni A, Weiner HL, Miller A. Gelatinases (MMP-2 and MMP-9) are preferentially expressed by Th1 vs. Th2 cells. Cells J Neuroimmunol (2005) 163(1-2):157–64. doi: 10.1016/j.jneuroim.2005.02.001

CrossRef Full Text | Google Scholar

128. Wu B, Crampton SP, Hughes CC. Wnt signaling induces matrix metalloproteinase expression and regulates T cell transmigration. Immunity (2007) 26(2):227–39. doi: 10.1016/j.immuni.2006.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

129. El-Shabrawi Y, Walch A, Hermann J, Egger G, Foster CS. Inhibition of MMP-dependent chemotaxis and amelioration of experimental autoimmune uveitis with a selective metalloproteinase-2 and -9 inhibitor. J Neuroimmunol (2004) 155(1-2):13–20. doi: 10.1016/j.jneuroim.2004.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Swadzba ME, Hirmer S, Amann B, Hauck SM, Deeg CA. Vitreal IgM autoantibodies target neurofilament medium in a spontaneous model of autoimmune uveitis. Invest Ophthalmol Vis Sci (2012) 53(1):294–300. doi: 10.1167/iovs.11-8734

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Swadzba ME, Hauck SM, Naim HY, Amann B, Deeg CA. Retinal glycoprotein enrichment by concanavalin a enabled identification of novel membrane autoantigen synaptotagmin-1 in equine recurrent uveitis. PloS One (2012) 7(12):e50929. doi: 10.1371/journal.pone.0050929

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Kalsow CM, Dubielzig RR, Dwyer AE. Immunopathology of pineal glands from horses with uveitis. Invest Ophthalmol Vis Sci (1999) 40(7):1611–5.

PubMed Abstract | Google Scholar

133. Eberhardt C, Amann B, Stangassinger M, Hauck SM, Deeg CA. Isolation, characterization and establishment of an equine retinal glial cell line: a prerequisite to investigate the physiological function of Muller cells in the retina. J Anim Physiol Anim Nutr (Berl) (2012) 96(2):260–9. doi: 10.1111/j.1439-0396.2011.01147.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: retina, Mueller glia, vitreous, serum, lymphocyte, granulocyte, complement system, cytokines

Citation: Degroote RL and Deeg CA (2021) Immunological Insights in Equine Recurrent Uveitis. Front. Immunol. 11:609855. doi: 10.3389/fimmu.2020.609855

Received: 24 September 2020; Accepted: 30 November 2020;
Published: 08 January 2021.

Edited by:

Peizeng Yang, First Affiliated Hospital of Chongqing Medical University, China

Reviewed by:

Heping Xu, Queen’s University Belfast, United Kingdom
John Vincent Forrester, University of Western Australia, Australia

Copyright © 2021 Degroote and Deeg. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Roxane L. Degroote, r.degroote@lmu.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.