Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Immune Relevant Models for Ocular Inflammatory Diseases

Immune Relevant Models for Ocular Inflammatory Diseases Abstract Ocular inflammatory diseases, such as dry eye and uveitis, are common, painful, difficult to treat, and may result in vision loss or blindness. Ocular side effects from the use of antiinflammatory drugs (such as corticosteroids or nonsteroidal antiinflammatories) to treat ocular inflammation have prompted development of more specific and safer medications to treat inflammatory and immune-mediated diseases of the eye. To assess the efficacy and safety of these new therapeutics, appropriate immune-relevant animal models of ocular inflammation are needed. Both induced and naturally-occurring models have been described, but the most valuable for translating treatments to the human eye are the animal models of spontaneous, immunologic ocular disease, such as those with dry eye or uveitis. The purpose of this review is to describe common immune-relevant models of dry eye and uveitis with an overview of the immuno-pathogenesis of each disease and reported evaluation of models from small to large animals. We will also review a selected group of naturally-occurring large animal models, equine uveitis and canine dry eye, that have promise to translate into a better understanding and treatment of clinical immune-relevant ocular disease in man. animal models, dry eye, immune-relevant, inflammatory, naturally-occurring, ocular, uveitis Introduction Blindness or low vision affects approximately 1 in 28 Americans older than 40 years of age, the underlying causes of which are commonly noninfectious immune-mediated diseases, including dry eye and uveitis.1,–3 Dry eye symptoms are experienced by 20% of adults over 45 years old, and uveitis is a leading cause of blindness in the United States.1,–6 Dry eye and uveitis are also common causes of blindness in domestic animals, and uveitis is the leading cause of blindness in horses worldwide.7,–12 There are no known cures for immune-mediated ocular diseases, and current treatment regimens are costly, require multiple daily applications, are poorly effective, and have adverse side effects. Therefore, new treatments to address these diseases are needed and for further development, there is a need for accurate and translatable immune-relevant models of ocular disease. The eye, like the brain and the uterus in pregnancy, is considered an immune privileged site.13,14 An active suppression of the immune response to endogenous and exogenous antigens occurs in the eye, as overt inflammation may compromise vision. The relative lack of antigen-presenting and MHC II-expressing cells and natural tissue barriers (i.e., the blood–ocular barrier) that physically separate ocular tissues from the systemic immune response contribute to the immune tolerance in the eye.15 With dry eye and uveitis, the normal ocular tolerance is lost (from several initiating causes) and the physical barriers become disrupted, allowing an influx of inflammatory cells. In addition, proinflammatory mediators induce T-helper cells to proliferate, activate antigen-presenting cells, expand auto-reactive B and T cell populations, and ultimately release proinflammatory and proapoptotic peptides.16,17 Current treatments for dry eye and uveitis are nonspecific and require frequent use of topical medications that may have severe ocular and systemic side effects.18,–20 Furthermore, these medications are life-long therapies and patient compliance is commonly poor, leading to treatment failures, worsening of disease, and in some cases, blindness.21 When testing effectiveness of therapeutics on models of ocular disease, there are two separate but important testing goals. The first question is whether the drug is effective in the ocular disease state that is being studied. For this goal, usually rats or mice are evaluated and dosed by a nonocular route, for example, orally, subcutaneously, or intraperitoneally. These studies help determine pathogenesis of disease-drug mechanisms; therefore, the wide array of reagents and genetically modified mice and rats are a major asset. Determination of the appropriate dose (i.e., dose ranging studies) is usually also performed in these first sets of studies. The second goal is to determine if an appropriate dose can reach the ocular target tissue and be effective in the eye using a dosing route and frequency that is clinically feasible. These studies would determine the pharmacokinetics and pharmacodynamics of a specific route of administration of a drug, typically in a normal eye, then repeated using the optimal dosing and routes in eyes of models of the disease state. For this second group of studies to be clinically valid in most instances, the animal models would have to have eyes anatomically similar to the target species and in the case of humans, use of the rabbit, dog, pig, or primate eye would be most appropriate. Finally, when selecting the appropriate animal model, the target tissue and disease state has to be paired with the most appropriate route of therapy. This determination is important for pharmacokinetic, toxicologic, and efficacy studies. Although there are many disease conditions of the human eye thought to have an immunologic pathogenesis, including allergic conjunctivitis, corneal transplant rejection, and age-related macular degeneration, as examples, the purpose of this review is to describe common immune-relevant models of dry eye and uveitis with an overview and assessment of models from small to large animals. We will also review a selected group of naturally-occurring large animal models, equine uveitis and canine dry eye, which have promise to translate into a better understanding and treatment of clinical immune-relevant ocular disease in man. Review of Commonly Used Animal Models in Inflammatory Ocular Disease Ocular Surface Disease Immune-Relevant Models Dry Eye Disease Dry eye disease (DED) is one of the most common ocular abnormalities and has multiple underlying causes. Dry eye is a disease of the tear film and ocular surface that results in symptoms of discomfort and visual disturbance with potential damage to the ocular surface.22 In one study, nearly one-half of patients claimed to have symptoms of dry eye with a negative effect on quality of life, including ocular pain, decreased activities requiring visual attention (e.g., reading, driving), and reduced productivity in the workplace.21 Dry eye develops from a deficiency of the aqueous portion of the tear fluid as a result of reduced lacrimal aqueous tear secretion or a result of increased evaporation of tears, such as the result of Meibomian gland deficiencies.23 Decreased aqueous production of the tears results in an increase of tear electrolytes (i.e., increased tear osmolality), proteins, and inflammatory mediators, resulting in damage to the surface ocular tissues, decreased visual acuity, and ocular discomfort. The relative decrease in aqueous tears on the ocular surface in patients with DED causes chronic irritation to ocular surface that disrupts the normal ocular immune tolerance.24 With breakdown of ocular surface tolerance and immune-homeostasis, autoimmunity develops through activation of NK cells and Toll-like receptors, followed by release of proinflammatory factors such as interleukin (IL)-1α, IL-1β, tumor necrosis factor α, and IL-6. These mediators amplify, activating antigen-presenting cells, which internalize autoantigens and migrate to the draining cervical lymph node where autoreactive Th1 cells, Th17 cells, or B cells (i.e., in Sjogren’s syndrome) undergo expansion. Efferent trafficking of these autoreactive T cells to the ocular surface is directed by adhesion molecules (e.g., LFA-1) and chemokine receptors. Autoreactive T-cells in ocular surface tissues potentiate the chronic autoimmune response, resulting in epithelial cell apoptosis, reduced goblet cell density, and squamous metaplasia of epithelium.17,24,25 Current treatments for DED rely on frequently applied artificial tears, punctal plugs, topical tetracycline antibiotic, and omega fatty acids, all of which provide only temporary relief of dry eye.26 Chronic DED is commonly treated with antiinflammatory medications and immunosuppressants, the latter being the mainstay of treatment in the United States.27,28 Topical cyclosporine, an immunosuppressant, used with or without corticosteroids, is effective in DED through inhibition of T-cell activation and reduction of proinflammatory cytokines.29 A recently approved topical immunosuppressive for treatment of DED, lifitegrast, is an integrin inhibitor that prevents binding of LFA-1 to ICAM-1, which is upregulated in DED. Lifitegrast thus blocks T-cell efferent recruitment to ocular tissues and reduces inflammatory cytokines.30,–32 However, both cyclosporine and lifitegrast must be administered indefinitely twice daily by the patient and are associated with burning sensation after application, leading to reduced patient compliance and hence poor treatment efficacy and success. Therefore, an effective, long-term, well-tolerated, and convenient therapy for DED is needed. There are numerous models of ocular surface disease and dry eye, but to be immune relevant, there needs to be evidence of an immuno-pathogenesis in the disease process. There are several mouse models of dry eye disease, the most common of which is a model induced by low humidity and high air flow environments, with or without the additional use of scopolamine (Table 1).33,–35 The extended environmental irritation to the surface of the eye of these mice disrupts the normal ocular immune tolerance and immunohomeostasis,24 as described previously. These mice models have been used to study the immuno-pathogenesis of dry eye and the initial evaluation of therapeutics. Another described model is the use of repeated application of topical benzalkonium chloride to the mouse or rabbit eye. This produces chronic irritation that may develop immunopathology and chronic ocular surface disease.36,37 Other induced models of DED in rodents, which may be less immunopathologic in origin, include lacrimal gland excision or injections of toxins or antigens such as botulinum toxin38 or concanavalin A.39 Genetic models, such as the MRL/lpr mouse, manifest multiple autoimmune disorders and can be helpful to study diseases such as systemic lupus erythematosus and Sjorgren’s syndrome (Table 1).40 Another example of genetic DED are neurturin-deficient mice, which may develop dry eye and serve as models for neurotrophic keratoconjunctivitis sicca, since this model lacks lacrimal innervation (Table 1).41 There are numerous other knockout and transgenic mice strains that are commonly studied that may develop DED; however, many of these models do not develop clinical signs of DED observed in large animal models, but instead develop histologic or other features characteristic of human DED.42 Table 1 Selected immune-models of dry eye disease Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Open in new tab Table 1 Selected immune-models of dry eye disease Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Open in new tab In rats, the most commonly described dry eye model is the extraorbital lacrimal gland excision model (with or without use of scopolamine).48,49 Like other models of induced dry eye, this rat lacrimal-excision model likely does not develop, substantially, an immunologic pathogenesis and therefore may not be as effective for evaluation of immunosuppressive therapies as naturally-occurring models of dry eye.33,49 All of these rodent models of dry eye are similar in that they can be used to determine proof of principal of therapeutic response to a drug, but all have similar disadvantages of having orbital and lacrimal anatomy and eye size that differs from the human eye. Rabbit or dog models are more commonly used to evaluate dry eye signs and response to therapy, because they have easily measured decreased tear production and develop ocular surface changes.42 Therefore, larger animal dry eye models are needed (see later description of canine dry eye). Rabbits are commonly used as models of ocular disease and for pharmacokinetic studies because of their relatively large eye, compared to rodents, while still being a common and economical laboratory animal. However, there are few true immune-relevant models of DED in rabbits. Most described models induce dry eye signs, but not likely an immunopathogenesis, by use of a topical irritant, such as benzalkonium chloride, or by short-term reduction in lacrimal secretion using parasympathomimetic drug, such as atropine.37,52,53 A very promising model of autoimmune dacryoadenitis in rabbits that produces a Sjögren’s-like keratoconjunctivitis is created by an intra-lacrimal or subcutaneous injection of autologous peripheral blood lymphocytes activated by purified rabbit lacrimal epithelial cells.54,55 This rabbit autoimmune dacryoadenitis model has been used to effectively evaluate immunomodulatory treatments for dry eye, including topical cyclosporine and lacrimal gland adeno-associated virus (AAV) mediated-IL-10 gene therapy.50,56 Naturally-Occurring Keratoconjunctivitis Sicca in Canines Domestic canines develop spontaneous dry eye that clinically and immunopathologically is similar to dry eye in humans (Table 1).10 Not only do dogs spontaneously develop dry eye symptoms of ocular discomfort, conjunctival hyperemia, and corneal scarring, these symptoms correlate directly with reduced aqueous tear production, a reduction readily measured using a standard Schirmer tear test strip (Figure 1). Furthermore, dogs with dry eye have a reduced tear breakup time and increased corneal staining, all abnormalities also observed in humans with DED.10 Like humans, canine dry eye is typically bilateral, develops in middle age, is more common in female dogs and in certain breeds, such as the American Cocker spaniel, Bulldog, and West Highland white terrier.57 The pathogenesis of dry eye in dogs appears similar to that of humans, where an apparent immunologic inflammation occurs with progressive lymphocytic infiltration and damage to the lacrimal gland with subsequent decreased production of the aqueous tear film.58,59 With chronicity, the ocular surface becomes progressively more dessicated and inflamed, the cornea vascularizes and scars, and ultimately the dog may lose vision.9,57 Initial proof of concept of commonly used immunosuppressive eye drops was first demonstrated to be effective in this spontaneous dog model, including topical cyclosporine, tacrolimus, and LTF-1 inhibitors.9,10,12,60,61 Figure 1 Open in new tabDownload slide Naturally-occurring dry eye in a dog. (A) Moderate dry eye disease in a dog resulting in conjunctival hyperemia, corneal vascularization, and corneal opacity. (B) Chronic dry eye disease in a dog with mucopurulent ocular discharge, hyperpigmented cornea, and conjunctival hyperemia. Figure 1 Open in new tabDownload slide Naturally-occurring dry eye in a dog. (A) Moderate dry eye disease in a dog resulting in conjunctival hyperemia, corneal vascularization, and corneal opacity. (B) Chronic dry eye disease in a dog with mucopurulent ocular discharge, hyperpigmented cornea, and conjunctival hyperemia. Uveitis Disease Models Uveitis is inflammation of the iris, ciliary body, and choroid and is associated with both infectious and noninfectious causes. Uveitis is estimated to be the third leading cause of preventable blindness worldwide.62 In the United States, the incidence of uveitis was estimated to be approximately 58 to 69 cases/100,000 people;1,6 however, another study estimated that the rate of uveitis, especially anterior uveitis, was approximately 3 times higher and it increased with increasing age of patients.4 The most common causes of uveitis in humans are human leukocyte antigen (HLA)-B27 related uveitis, acute anterior uveitis in herpes zoster disease, toxoplasmosis, sarcoidosis, and pars planitis.63 Uveitis results from several causes. The uveal tract supplies blood to the eye and is in direct contact with peripheral vasculature; therefore, diseases of the systemic circulation (e.g., septicemia, bacteremia, infection, activated lymphocytes, immune diseases, etc.) will disrupt the blood-ocular barrier.64,65 The blood-ocular barrier prevents large molecules and cells from entering the eye and thus limits the immune response to intraocular antigens. With trauma or inflammation, this barrier can be disrupted, allowing blood products and cells to enter the eye, resulting in the clinical signs typical of uveitis, such as flare, cell accumulation, and vitreous haze. Disruption of the barrier enables activation of various host immune responses, including antibody production to self-antigens that are not normally recognized by the immune system, as well as antibody production to foreign antigens inside the eye. As a result of the blood-ocular barrier, lack of lymphatics, and the presence of limited numbers of resident leukocytes, the eye is considered to have immune privilege. Naïve T cells cannot cross the normal blood-retinal barrier due to the lack of fenestration in the retinal vessels and the lack of appropriate adhesion molecules.66 Expression of chemokines in inflammation and activated T cells in the ciliary epithelium may play a role in recruitment and activation of leukocytes in diseased eyes.67 As in other autoimmune disorders, infections may trigger events, either by antigenic mimicry with a pathogen’s antigen or as a bystander effect due to the general systemic or local immune stimulation by the pathogen. Uveitogenic retinal proteins documented in experimental animals include retinal arrestin, interphotoreceptor retinoid-binding protein (IRBP), rhodopsin, recoverin, phosducin, and retinal pigment epithelium derived RPE-65.62,68,–70 Irrespective of the eliciting antigen, available experimental evidence suggests that the immunological mechanisms driving the resultant disease are similar.16 Following disruption of the blood-ocular barrier, large amounts of predominantly CD4+ T cells enter the eye and secrete proinflammatory cytokines such as IL-2 and interferon γ.71 Auto-reactive effector CD4+ T cells have been associated with the pathogenesis of inflammatory and autoimmune disorders including uveitis. Naıve CD4+ T cells differentiate into effector subsets depending on the nature of the environment in which exposure to the antigen occurs.66 Several T cell effector phenotypes have been defined, known as T helper 1 (TH1), TH2, or TH17. Early studies suggested that the interferon-γ-producing TH1 and IL-17-releasing TH17 subsets are responsible for the pathology of uveitis, with the latter being associated with development of autoimmune disease.16 Additionally, clinical uveitis frequently develops spontaneous recurrent or relapsing bouts of inflammation, likely from T cells recognizing additional autoantigens in the ocular tissue.72 Resolution of uveitis is dependent on the presence of T regulatory cells (Tregs) that are labeled as CD4+Foxp3+ cells. When Foxp3+ T cell percentages in uveitis increase to approximately 10% of the total CD4+ cells, the acute inflammation rapidly resolves. Therefore, Foxp3+ Tregs are important to induce spontaneous resolution and in maintaining remission of uveitis.73 Multiple models have been developed to evaluate the immuno-pathogenesis of uveitis and recurrent uveitis, including identification of autoantigens. Most of these models are rodent based. Other models, including those that are acute, chronic, and recurrent in nature, have been developed to evaluate therapeutics (Table 2). Large animal models, such as uveitis induced in rabbits and pigs, have been evaluated to test therapeutics in larger eyes to help translate these treatments to humans (Table 2). Table 2. Models of uveitis Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 EAU, experimental autoimmune anterior uveitis; EIU, endotoxin-induced uveitis; IL, interleukin; NZW, Zealand White. Open in new tab Table 2. Models of uveitis Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 EAU, experimental autoimmune anterior uveitis; EIU, endotoxin-induced uveitis; IL, interleukin; NZW, Zealand White. Open in new tab Rodent Models of Uveitis Endotoxin-induced uveitis A commonly used model of induced uveitis in rodents is the endotoxin-induced uveitis (EIU) model (Table 2).74,75,89 The uveitis in this model is primarily an acute anterior uveitis (i.e., iris, ciliary body) that is thought to be driven by the innate immune system (Table 2).16,76 Following intraparentoneal, subcutaneous, or hind footpad injection of endotoxin (lipopolysaccharide; 100 μg or 500 μg) in Lewis or Sprague-Dawley rats or various mouse strains (C3H),16 ocular inflammation develops within hours of injection characterized by a breakdown of the blood-aqueous barrier and the development of clinical disease. Clinical and histopathologic abnormalities peak at 24 hours and resolve by 48 to 72 hours.74,75 Experimental autoimmune uveitis (or uveoretinitis) Experimental autoimmune uveitis (EAU) is a primarily posterior uveitis (or panuveitis [i.e., inflammation of the iris, ciliary, and choroid]) that is induced by immunizing susceptible rodents with retinal antigens (e.g., S-antigen [S-ag], IRBP, recoverin, rhodopsin/opsin); while experimental melanin–protein induced uveitis, a predominantly anterior uveitis, is elicited by immunization with melanin (from RPE) or tyrosinase-related proteins 1 and 2 (Table 2).16 The predominant animal model is the Lewis rat, but other animals such as the guinea pig or mice have also been described.76,90 Injection of autoantigens into rodents, combined with bacterial adjuvants, results in EAU; EAU does not develop without the use of adjuvants. The use of complete Freund’s adjuvant (Mycobacterium cell wall product) or pertussis toxin is necessary to stimulate the innate immune response and develop inflammation73,76,78 that ultimately generates activated antigen-presenting cells capable of presenting the injected autoantigen with the coactivation factors required to activate T cells capable of recognizing the antigen. Severe EAU was induced in B6 mice by adoptive transfer of IRBP-specific T cells.79 Most of the rodent experimental models of uveitis are not recurrent. They often elicit a single, albeit chronic course of uveitis that eventually resolves. Therefore, the immunologic pathways involved in the development of these rodent models may not be the same as in naturally occurring uveitis. Spontaneous uveitis has been observed in various mouse models, including IRBP T cell receptor transgenic mice (R161H) and autoimmune regulator (AIRE)(-/-) mice.78 These mouse models have a gradual onset of chronic ocular inflammation that ultimately leads to retinal degeneration.78 Despite limitations, these rodent experimental models offer great insight into the pathogenesis and immunopathogenesis of uveitis. These models have been critical in evaluation of therapies, particularly broader immunosuppressive therapies, for treating uveitis. Rabbit Models of Uveitis Two rabbit models of uveitis have been most commonly evaluated, including the acute uveitis induced by injection of endotoxin84,91,–93 and the recurrent uveitis induced by tuberculosis antigen.82,84,94 Other uveitis models in rabbits include those following intravitreal injection of human interleukin 1 alpha,86 TNF-alpha,80 or ovalbumin in animals previously ovalbumin-immunized,85 among others (Table 2). An advantage of rabbit models of uveitis over rodent models is that the rabbit eye is more similar in size to the human eye, and therefore, more pharmacologically valid when evaluating routes of therapy. In the endotoxin-induced uveitis, after 10 to 100 ng of LPS is injected intracamerally92 or intravitreally,80,81 aqueous flare and iridal hyperemia develop within 6 hours, suggesting rapid disruption of the blood–aqueous barrier.91 The LPS induces inflammation by activating a Toll-like receptor 4-initiated signaling cascade. The inflammatory response peaks at approximately 24 hours after injection, then rapidly declines.92 Like the rat model of EIU, this endotoxin rabbit model of uveitis is not considered to have a predominantly immunopathogenesis. Experimental uveitis can be induced by unilateral intravitreal or intracameral injection of Mycobacterium tuberculosis H37Ra antigen (50 μg; 1 μg/L) in preimmunized rabbits, typically 7 to 14 days after initial subcutaneous injection.82,–84,95 To simulate chronic recurrent inflammation, eyes are re-challenged with intravitreal antigen every 14 to 21 days.96 This model has advantages similar to the endotoxin model; however, it is predominantly a T-cell lymphocyte-mediated uveitis that can be induced to be recurrent and therefore, more closely simulate endogenous human uveitis.95 Porcine Models of Uveitis The pig has been used as a large animal model of uveitis (Table 2), which, similar to the rabbit model, has an eye similar in size to the human eye; however, unlike the rabbit, it has a retinal vascular anatomy similar to humans.97 An acute model of uveitis has been used in the pig to evaluate novel therapeutics and routes of administration. In this model, similar to endotoxin uveitis in rabbits, endotoxin is injected intravitreally and the eye is monitored for up to 72 hours following injection.87 Like rodent EIU and endotoxin uveitis in rabbits, the endotoxin porcine model of uveitis is not considered to have an immunopathogenesis. Naturally-Occurring Uveitis Models Equine Recurrent Uveitis Horses spontaneously develop severe, immunologic uveitis called equine recurrent uveitis (ERU) that is frequently recurrent and chronic (Figure 2).98 ERU is the most common cause of blindness in horses.7,8,98 Spontaneous bouts of uveitis develop and blindness may occur after multiple recurrent episodes of uveitis. The immunopathology of ERU has been extensively studied and has demonstrated that T cells are the predominant mononuclear inflammatory cells infiltrating ocular tissues in horses with naturally occurring chronic uveitis, with a significant number of CD4+ cells.71,72,99 Recruitment of proinflammatory cells as well as autoreactive lymphocytes may be in part driven by the expression of the chemokine RANTES in the ciliary body.67 Figure 2 Open in new tabDownload slide Naturally-occurring uveitis in a horse. (A) Acute active uveitis in a horse, with a miotic pupil and anterior chamber opacity. (B) Chronic uveitis in a horse, from equine recurrent uveitis, with dyscoria, synechiae, and cataract. Figure 2 Open in new tabDownload slide Naturally-occurring uveitis in a horse. (A) Acute active uveitis in a horse, with a miotic pupil and anterior chamber opacity. (B) Chronic uveitis in a horse, from equine recurrent uveitis, with dyscoria, synechiae, and cataract. Study of this common, spontaneous uveitis in horses has helped understand the pathogenesis of uveitis in humans, especially the identification of autoantigens and how recurrence of uveitis develops immunologically.7,69,70,72,100,–103 Several potential autoantigens have been identified in horses that could play a role in the development of autoimmune uveitis. T cells isolated from the eyes of horses with ERU proliferate in response to two common autoantigens in rodents: retinal S-Ag and IRBP.69 In addition, several additional potential autoantigens were identified by analyzing antibodies in the sera of ERU horses that reacted with retinal proteins. These include recoverin, cellular retinaldehyde-binding protein, and malate dehydrogenase.101 While all these potential autoantigens are capable of inducing experimental uveitis in rodent models, only cellular retinaldehyde-binding protein and IRBP consistently produce uveitis in outbred horses.70,100 Additionally, studies of horses with ERU have also helped elucidate how Leptospira infections induce immunological uveitis, specifically autoimmune uveitis.104,–106 Field studies of horses in the 1950s after an outbreak of acute leptospirosis caused by L. interrogans serogroup Pomona demonstrated that one of the six horses (17%) developed intraocular inflammation during acute leptospiral disease, and all horses developed ERU 18 to 24 months after the initial infection. Subsequent studies demonstrated cross-reactivity between equine ocular tissues and Leptospira antigens,104,105 and horses with uveitis associated with Leptospira interrogans infections had high levels of IgA and IgG in their intraocular fluids that reacted to two Leptospira lipoproteins, LruA and LruB.106,107 These antibodies were also subsequently discovered in the serum of human leptospiral uveitis patients.108 Studies of spontaneous ERU have helped elucidate the immunopathogenesis of recurrent uveitis. In autoimmune disease, several autoantigens, or epitopes, participate in the immunopathogenesis; epitope spreading is accountable for disease induction, progression, and inflammatory relapses.72 Epitope spreading is defined as the diversification of epitope specificity from the initial focused, dominant, epitope-specific immune response, directed against a self or foreign protein to cryptic epitopes on that protein (intramolecular spreading) or other proteins (intermolecular spreading).72 The shifts in immunoreactivity, or epitope spreading, have been documented in ERU and are thought to be responsible for the recurring character of ERU.72 ERU, as a model of spontaneous immune-mediated uveitis, has also led to the study of promising therapeutics. For example, several sustained release ocular implants have shown much promise in the treatment of ERU.109,110 Evaluation of drug delivery to the suprachoroidal space has been shown to control ERU and prevent recurrences.111,112 Triamcinolone injections into the suprachoroidal space are currently under development for treatment of human uveitis.113 Further study of ERU and its treatment will translate well to improving the understanding and treatment of human autoimmune uveitis. Next Steps As further therapeutics are developed that more specifically target immune-mediated diseases, evaluation of these treatments in spontaneous or naturally-occurring models of ocular disease will be needed to provide proof of concept and help translate these therapies to humans. Excellent examples of developing, targeted therapies include gene therapy (especially gene addition therapy) and stem cell therapy. Our laboratory and collaborators at the Gene Therapy Center at the University of North Carolina have developed AAV delivery of immunosuppressive proteins, such as HLA-G, for suppression of ocular surface inflammation and vascularization.114 Target ocular surface diseases for AAV-HLA-G gene therapy are DED and for prevention of corneal graft rejection.114 Autologous stem cell therapy, or use of stem cell supernatant extracts, also shows much promise for immunomodulation in the eye. Effectiveness of topical ocular mesenchymal stem cell therapy was initially demonstrated in dry eye models in mice.39 Locally injected fat-derived mesenchymal stem cells near the lacrimal glands of dogs with advanced dry eye demonstrated clinical improvement and increased tear production.115 These results in a naturally-occurring model provides evidence of possible clinical translation to humans with severe dry eye. Acknowledgments and Conflicts The author thanks Jacklyn Salmon and Dr. Jorge Piedrahita for review of this manuscript. References 1 Acharya NR , Tham VM , Esterberg E , et al. Incidence and prevalence of uveitis: Results from the Pacific Ocular Inflammation Study . JAMA Ophthalmol . 2013 ; 131 ( 11 ): 1405 – 1412 . doi:10.1001/jamaophthalmol.2013.4237 . Google Scholar Crossref Search ADS PubMed WorldCat 2 Schaumberg DA , Sullivan DA , Buring JE , Dana MR . Prevalence of dry eye syndrome among US women . Am J Ophthalmol . 2003 ; 136 ( 2 ): 318 – 326 . doi:10.1016/S0002-9394(03)00218-6 . Google Scholar Crossref Search ADS PubMed WorldCat 3 Schaumberg DA , Dana R , Buring JE , Sullivan DA . Prevalence of dry eye disease among US men: Estimates from the Physicians’ Health Studies . Arch Ophthalmol . 2009 ; 127 ( 6 ): 763 – 768 . doi:10.1001/archophthalmol.2009.103 . Google Scholar Crossref Search ADS PubMed WorldCat 4 Gritz DC , Wong IG . Incidence and prevalence of uveitis in Northern California: The Northern California Epidemiology of Uveitis Study . Ophthalmology . 2004 ; 111 ( 3 ): 491 – 500 . doi:10.1016/j.ophtha.2003.06.014 . Google Scholar Crossref Search ADS PubMed WorldCat 5 Merrill PT , Kim J , Cox TA , Betor CC , McCallum RM , Jaffe GJ . Uveitis in the southeastern United States . Curr Eye Res . 1997 ; 16 ( 9 ): 865 – 874 . doi:10.1076/ceyr.16.9.865.5048 . Google Scholar Crossref Search ADS PubMed WorldCat 6 Suhler EB , Lloyd MJ , Choi D , Rosenbaum JT , Austin DF . Incidence and prevalence of uveitis in Veterans Affairs Medical Centers of the Pacific Northwest . Am J Ophthalmol . 2008 ; 146 ( 6 ). doi: 10.1016/j.ajo.2008.09.014 . WorldCat 7 Deeg CA , Hauck SM , Amann B , et al. Equine recurrent uveitis—A spontaneous horse model of uveitis . Ophthalmic Res . 2008 ; 40 ( 3–4 ): 151 – 153 . doi:10.1159/000119867 . Google Scholar Crossref Search ADS PubMed WorldCat 8 Gerding JC , Gilger BC . Prognosis and impact of equine recurrent uveitis . Equine Vet J . 2015 ; 48 ( 3 ): 290 – 298 . doi:10.1111/evj.12451 . Google Scholar Crossref Search ADS PubMed WorldCat 9 Gilger BC , Wilkie DA , Salmon JH , Peel MR . A topical aqueous calcineurin inhibitor for the treatment of naturally occurring keratoconjunctivitis sicca in dogs . Vet Ophthalmol . 2013 c; 16 ( 3 ): 192 – 197 . doi:10.1111/j.1463-5224.2012.01056.x . Google Scholar Crossref Search ADS PubMed WorldCat 10 Kaswan RL , Salisbury MA , Ward DA . Spontaneous canine keratoconjunctivitis sicca. A useful model for human keratoconjunctivitis sicca: Treatment with cyclosporine eye drops . Arch Ophthalmol . 1989 ; 107 ( 8 ): 1210 – 1216 . doi:10.1001/archopht.1989.01070020276038 . Google Scholar Crossref Search ADS PubMed WorldCat 11 Moore CP , McHugh JB , Thorne JG , Phillips TE . Effect of cyclosporine on conjunctival mucin in a canine keratoconjunctivitis sicca model . Invest Ophthalmol Vis Sci . 2001 ; 42 ( 3 ): 653 – 659 . http://www.ncbi.nlm.nih.gov/pubmed/11222523. Google Scholar PubMed WorldCat 12 Murphy CJ , Bentley E , Miller PE , et al. The pharmacologic assessment of a novel lymphocyte function-associated antigen-1 antagonist (SAR 1118) for the treatment of keratoconjunctivitis sicca in dogs . Investig Ophthalmol Vis Sci . 2011 ; 52 ( 6 ): 3174 – 3180 . doi:10.1167/iovs.09-5078 . Google Scholar Crossref Search ADS WorldCat 13 Niederkorn JY , Wang S . Immune privilege of the eye and fetus: Parallel universes? Transplantation . 2005 ; 80 ( 9 ): 1139 – 1144 . doi:10.1097/01.TP.0000173828.78382.4F . Google Scholar Crossref Search ADS PubMed WorldCat 14 Stein-Streilein J . Immune regulation and the eye . Trends Immunol . 2008 ; 29 ( 11 ): 548 – 554 . doi:10.1016/j.it.2008.08.002 . Google Scholar Crossref Search ADS PubMed WorldCat 15 Niederkorn JY . See no evil, hear no evil, do no evil: The lessons of immune privilege . Nat Immunol . 2006 ; 7 ( 4 ): 354 – 359 . doi:10.1038/ni1328 . Google Scholar Crossref Search ADS PubMed WorldCat 16 Caspi RR . Immunological disorders and autoimmunity animal models of autoimmune and immune-mediated uveitis . 2006 ; 3 ( 1 ): 3 – 9 . doi:10.1016/j.ddmod.2006.03.005 . WorldCat 17 Stern ME , Pflugfelder SC . Pathogenesis: Emphasis on dry eye and the role of the lacrimal functional unit in Sjögren’s syndrome. In: Fox R, Fox C, eds. Sjögren’s Syndrome. New York, NY: Springer; 2011 . doi:10.1007/978-1-60327-957-4_13. 18 Carnahan MC , Goldstein DA . Ocular complications of topical, peri-ocular, and systemic corticosteroids . Curr Opin Ophthalmol . 2000 ; 11 : 478 – 483 . doi:10.1097/00055735-200012000-00016 . Google Scholar Crossref Search ADS PubMed WorldCat 19 Fraunfelder FT , Sciubba JJ , Mathers WD . The role of medications in causing dry eye . J Ophthalmol . 2012 ; 2012 : 285851 . doi:10.1155/2012/285851. Google Scholar PubMed WorldCat 20 Sen HN , Vitale S , Gangaputra SS , et al. Periocular corticosteroid injections in uveitis: Effects and complications . Ophthalmology . 2014 ; 121 ( 11 ): 2275 – 2286 . doi:10.1016/j.ophtha.2014.05.021. Google Scholar Crossref Search ADS PubMed WorldCat 21 Uchino M , Schaumberg DA . Dry eye disease: Impact on quality of life and vision . Curr Ophthalmol Rep . 2013 ; 1 ( 2 ): 51 – 57 . doi:10.1007/s40135-013-0009-1. Google Scholar Crossref Search ADS PubMed WorldCat 22 DEWS . The definition and classification of dry eye disease: report of the Definition and Classification of the Dry Eye WorkShop (2007) . Ocul Surf . 2007 ; 5 ( 2 ): 75 – 92 . doi:10.1080/09273940701486803. Crossref Search ADS PubMed WorldCat 23 Lemp MA , Crews LA , Bron AJ , Foulks GN , Sullivan BD . Distribution of aqueous-deficient and evaporative dry eye in a clinic-based patient cohort . Cornea . 2012 ; 31 ( 5 ): 472 – 478 . doi:10.1097/ICO.0b013e318225415a. Google Scholar Crossref Search ADS PubMed WorldCat 24 Barabino S , Chen Y , Chauhan S , Dana R . Ocular surface immunity: Homeostatic mechanisms and their disruption in dry eye disease . Prog Retin Eye Res . 2012 ; 31 ( 3 ): 271 – 285 . doi:10.1016/j.preteyeres.2012.02.003. Google Scholar Crossref Search ADS PubMed WorldCat 25 Stern ME , Schaumburg CS , Dana R , Calonge M , Niederkorn JY , Pflugfelder SC . Autoimmunity at the ocular surface: Pathogenesis and regulation . Mucosal Immunol . 2010 ; 3 ( 5 ): 425 – 442 . doi:10.1038/mi.2010.26. Google Scholar Crossref Search ADS PubMed WorldCat 26 Gayton JL . Etiology, prevalence, and treatment of dry eye disease . Clin Ophthalmol . 2009 ; 3 ( 1 ): 405 – 412 . Google Scholar Crossref Search ADS PubMed WorldCat 27 Avunduk AM , Avunduk MC , Varnell ED , Kaufman HE . The comparison of efficacies of topical corticosteroids and nonsteroidal anti-inflammatory drops on dry eye patients: A clinical and immunocytochemical study . Am J Ophthalmol . 2003 ; 136 ( 4 ): 593 – 602 . doi:10.1016/S0002-9394(03)00326-X. Google Scholar Crossref Search ADS PubMed WorldCat 28 Sall K , Stevenson OD , Mundorf TK , Reis BL . Two multicenter, randomized studies of the efficacy and safety of cyclosporine ophthalmic emulsion in moderate to severe dry eye disease. CsA Phase 3 Study Group . Ophthalmology . 2000 ; 107 : 631 – 639 . doi:S0161642099001761 [pii]. Google Scholar Crossref Search ADS PubMed WorldCat 29 Lekhanont K , Leyngold IM , Suwan-Apichon O , Rangsin R , Chuck RS . Comparison of topical dry eye medications for the treatment of keratoconjunctivitis sicca in a botulinum toxin B-induced mouse model . Cornea . 2007 ; 26 ( 1 ): 84 – 89 . doi:10.1097/01.ico.0000240079.24583.a1. Google Scholar Crossref Search ADS PubMed WorldCat 30 Keating GM . Lifitegrast ophthalmic solution 5%: A review in dry eye disease . Drugs . 2017 ; 77 ( 2 ): 201 – 208 . doi:10.1007/s40265-016-0681-1. Google Scholar Crossref Search ADS PubMed WorldCat 31 Sheppard JD , Torkildsen GL , Lonsdale JD , et al. Semba CPOPUS-1 Study Group.Lifitegrast ophthalmic solution 5.0% for treatment of dry eye disease: Results of the OPUS-1 phase 3 study . Ophthalmology . 2014 ; 121 : 475 – 483 . doi:10.1016/j.ophtha.2013.09.015. Google Scholar Crossref Search ADS PubMed WorldCat 32 Tauber J , Karpecki P , Latkany R , et al. Semba CPOPUS-2 Investigators. Lifitegrast ophthalmic solution 5.0% versus placebo for treatment of dry eye disease results of the randomized phase III OPUS-2 study . Ophthalmology . 2015 ; 122 ( 12 ): 2423 – 2431 . doi:10.1016/j.ophtha.2015.08.001. Google Scholar Crossref Search ADS PubMed WorldCat 33 Barabino S , Chen W , Dana MR . Tear film and ocular surface tests in animal models of dry eye: Uses and limitations . Exp Eye Res . 2004 ; 79 ( 5 ): 613 – 621 . doi:10.1016/j.exer.2004.07.002. Google Scholar Crossref Search ADS PubMed WorldCat 34 Barabino S , Shen LL , Chen L , Rashid S , Rolando M , Dana MR . The controlled-environment chamber: A new mouse model of dry eye . Investig Ophthalmol Vis Sci . 2005 ; 46 ( 8 ): 2766 – 2771 . doi:10.1167/iovs.04-1326. Google Scholar Crossref Search ADS WorldCat 35 Daull P , Feraille L , Barabino S , et al. Efficacy of a new topical cationic emulsion of cyclosporine A on dry eye clinical signs in an experimental mouse model of dry eye . Exp Eye Res . 2016 ; 153 : 159 – 164 . doi:10.1016/j.exer.2016.10.016. Google Scholar Crossref Search ADS PubMed WorldCat 36 Lin Z , Liu X , Zhou T , et al. A mouse dry eye model induced by topical administration of benzalkonium chloride . Mol Vis . 2011 ; 17 : 257 – 264 . Google Scholar PubMed WorldCat 37 Xiong C , Chen D , Liu J , et al. A rabbit dry eye model induced by topical medication of a preservative benzalkonium chloride . Investig Ophthalmol Vis Sci . 2008 ; 49 ( 5 ): 1850 – 1856 . doi:10.1167/iovs.07-0720. Google Scholar Crossref Search ADS WorldCat 38 Zhu L , Shen J , Zhang C , et al. Inflammatory cytokine expression on the ocular surface in the Botulium toxin B induced murine dry eye model . Mol Vis . 2009 ; 15 : 250 – 258 . http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2633460&tool=pmcentrez&rendertype=abstract. Google Scholar PubMed WorldCat 39 Lee MJ , Ko AY , Ko JH , et al. Mesenchymal stem/stromal cells protect the ocular surface by suppressing inflammation in an experimental dry eye . Mol Ther . 2015 ; 23 ( 1 ): 139 – 146 . doi:10.1038/mt.2014.159. Google Scholar Crossref Search ADS PubMed WorldCat 40 Jabs DA , Lee B , Burek CL , Saboori AM , Prendergast RA . Cyclosporine therapy suppresses ocular and lacrimal gland disease in MRL/Mp-lpr/lpr mice . Investig Ophthalmol Vis Sci . 1996 ; 37 ( 2 ): 377 – 383 . WorldCat 41 Song XJ , Li DQ , Farley W , et al. Neurturin-deficient mice develop dry eye and keratoconjunctivitis sicca . Investig Ophthalmol Vis Sci . 2003 ; 44 ( 10 ): 4223 – 4229 . doi:10.1167/iovs.02-1319. Google Scholar Crossref Search ADS WorldCat 42 Schrader S , Mircheff AK , Geerling G . Animal models of dry eye . Dev Ophthalmol . 2008 ; 41 : 298 – 312 . doi:10.1159/000131097. Google Scholar Crossref Search ADS PubMed WorldCat 43 Guzmán M , Keitelman I , Sabbione F , Trevani AS , Giordano MN , Galletti JG . Mucosal tolerance disruption favors disease progression in an extraorbital lacrimal gland excision model of murine dry eye . Exp Eye Res . 2016 ; 151 : 19 – 22 . doi:10.1016/j.exer.2016.07.004. Google Scholar Crossref Search ADS PubMed WorldCat 44 Stevenson W , Chen Y , Lee S , et al. Extraorbital lacrimal gland excision: A reproducible model of severe aqueous tear-deficient dry eye disease . Cornea . 2014 ; 33 ( 12 ): 1336 – 1341 . doi:10.1097/ICO.0000000000000264. Google Scholar Crossref Search ADS PubMed WorldCat 45 Gao J , Morgan G , Tieu D , et al. ICAM-1 expression predisposes ocular tissues to immune-based inflammation in dry eye patients and Sjögrens syndrome-like MRL/lpr mice . Exp Eye Res . 2004 ; 78 ( 4 ): 823 – 835 . doi:10.1016/j.exer.2003.10.024. Google Scholar Crossref Search ADS PubMed WorldCat 46 Jie G , Jiang Q , Rui Z , Yifei Y . Expression of interleukin-17 in autoimmune dacryoadenitis in MRL/lpr mice . Curr Eye Res . 2010 ; 35 ( 10 ): 865 – 871 . doi:10.3109/02713683.2010.497600. Google Scholar Crossref Search ADS PubMed WorldCat 47 Ma X , Zou J , He L , Zhang Y . Dry eye management in a Sjögren’s syndrome mouse model by inhibition of p38-MAPK pathway . Diagn Pathol . 2014 ; 9 ( 1 ): 5 . doi:10.1186/1746-1596-9-5. Google Scholar Crossref Search ADS PubMed WorldCat 48 Fujihara T , Murakami T , Fujita H , Nakamura M , Nakata K . Improvement of corneal barrier function by the P2Y(2) agonist INS365 in a rat dry eye model . Invest Ophthalmol Vis Sci . 2001 ; 42 ( 1 ): 96 – 100 . http://www.ncbi.nlm.nih.gov/pubmed/11133853. Google Scholar PubMed WorldCat 49 Meng ID , Barton ST , Mecum NE , Kurose M . Corneal sensitivity following lacrimal gland excision in the rat . Investig Ophthalmol Vis Sci . 2015 ; 56 ( 5 ): 3347 – 3354 . doi:10.1167/iovs.15-16717. Google Scholar Crossref Search ADS WorldCat 50 Thomas PB , Samant DM , Selvam S , et al. Adeno-associated virus-mediated IL-10 gene transfer suppresses lacrimal gland immunopathology in a rabbit model of autoimmune dacryoadenitis . Investig Ophthalmol Vis Sci . 2010 ; 51 ( 10 ): 5137 – 5144 . doi:10.1167/iovs.10-5423. Google Scholar Crossref Search ADS WorldCat 51 Gilbard JP , Rossi SR , Gray KL , Hanninen LA , Kenyon KR . Tear film osmolarity and ocular surface disease in two rabbit models for keratoconjunctivitis sicca . Investig Ophthalmol Vis Sci . 1988 ; 29 ( 3 ): 374 – 378 . WorldCat 52 Burgalassi S , Panichi L , Chetoni P , Saettone MF , Boldrini E . Development of a simple dry eye model in the albino rabbit and evaluation of some tear substitutes . Ophthalmic Res . 1999 ; 31 ( 3 ): 229 – 235 . doi:10.1159/000055537. Google Scholar Crossref Search ADS PubMed WorldCat 53 Li C , Song Y , Luan S , et al. Research on the stability of a rabbit dry eye model induced by topical application of the preservative benzalkonium chloride . PLoS One . 2012 ; 7 ( 3 ). doi:10.1371/journal.pone.0033688. WorldCat 54 Thomas PB , Zhu Z , Selvam S , et al. Autoimmune dacryoadenitis and keratoconjunctivitis induced in rabbits by subcutaneous injection of autologous lymphocytes activated ex vivo against lacrimal antigens . J Autoimmun . 2008 ; 31 ( 2 ): 116 – 122 . doi:10.1016/j.jaut.2008.04.019. Google Scholar Crossref Search ADS PubMed WorldCat 55 Zhu Z , Stevenson D , Schechter JE , Mircheff AK , Atkinson R , Trousdale MD . Lacrimal histopathology and ocular surface disease in a rabbit model of autoimmune dacryoadenitis . Cornea . 2003 ; 22 ( 1 ): 25 – 32 . doi:10.1097/00003226-200301000-00007. Google Scholar Crossref Search ADS PubMed WorldCat 56 Thomas PB , Samant DM , Zhu Z , et al. Long-term topical cyclosporine treatment improves tear production and reduces keratoconjunctivitis in rabbits with induced autoimmune dacryoadenitis . J Ocul Pharmacol Ther . 2009 ; 25 ( 3 ): 285 – 291 . doi:10.1089/jop.2008.0138. Google Scholar Crossref Search ADS PubMed WorldCat 57 Sanchez RF , Innocent G , Mould J , Billson FM . Canine keratoconjunctivitis sicca: Disease trends in a review of 229 cases . J Small Anim Pract . 2007 ; 48 ( 4 ): 211 – 217 . doi:10.1111/j.1748-5827.2006.00185.x. Google Scholar Crossref Search ADS PubMed WorldCat 58 Izci C , Celik İ , Alkan F , Erol M , Sur E . Clinical and light microscopic studies of the conjunctival tissues of dogs with bilateral keratoconjunctivitis sicca before and after treatment with topical 2% cyclosporine . Biotech Histochem . 2015 ; 90 ( 3 ): 223 – 230 . doi:10.3109/10520295.2014.930177. Google Scholar Crossref Search ADS PubMed WorldCat 59 Kaswan RL , Martin CL , Chapman WL . Keratoconjunctivitis sicca: Histopathologic study of nictitating membrane and lacrimal glands from 28 dogs . Am J Vet Res . 1984 ; 45 ( 1 ): 112 – 118 . Google Scholar PubMed WorldCat 60 Barachetti L , Rampazzo A , Mortellaro CM , Scevola S , Gilger BC . Use of episcleral cyclosporine implants in dogs with keratoconjunctivitis sicca: Pilot study . Vet Ophthalmol . 2015 ; 18 ( 3 ): 234 – 241 . doi:10.1111/vop.12173. Google Scholar Crossref Search ADS PubMed WorldCat 61 Berdoulay A , English RV , Nadelstein B . Effect of topical 0.02% tacrolimus aqueous suspension on tear production in dogs with keratoconjunctivitis sicca . Vet Ophthalmol . 2005 ; 8 ( 4 ): 225 – 232 . doi:10.1111/j.1463-5224.2005.00390.x. Google Scholar Crossref Search ADS PubMed WorldCat 62 Siddique SS , Suelves AM , Baheti U , Foster CS . Glaucoma and uveitis . Surv Ophthalmol . 2013 ; 58 ( 1 ): 1 – 10 . doi:10.1016/j.survophthal.2012.04.006. Google Scholar Crossref Search ADS PubMed WorldCat 63 Jabs DA . Epidemiology of uveitis . Ophthalmic Epidemiol . 2008 ; 15 ( 5 ): 283 – 284 . doi:10.1080/09286580802478724. Google Scholar Crossref Search ADS PubMed WorldCat 64 Generali E , Cantarini L , Selmi C . Ocular involvement in systemic autoimmune diseases . Clin Rev Allergy Immunol . 2015 ; 49 ( 3 ): 263 – 270 . doi:10.1007/s12016-015-8518-3. Google Scholar Crossref Search ADS PubMed WorldCat 65 Levitt AE , McManus KT , McClellan AL , Davis JL , Goldhardt R , Galor A . Ocular inflammation in the setting of concomitant systemic autoimmune conditions in an older male population . Cornea . 2015 ; 34 ( 7 ): 762 – 767 . doi:10.1097/ICO.0000000000000437. Google Scholar Crossref Search ADS PubMed WorldCat 66 Caspi RR . Understanding autoimmune uveitis through animal models the Friedenwald lecture . Investig Opthalmol Vis Sci . 2011 ; 52 ( 3 ): 1873 . doi:10.1167/iovs.10-6909. Google Scholar Crossref Search ADS WorldCat 67 Gilger BC , Yang P , Salmon JH , Jaffe GJ , Allen JB . Expression of a chemokine by ciliary body epithelium in horses with naturally occurring recurrent uveitis and in cultured ciliary body epithelial cells . Am J Vet Res . 2002 ; 63 ( 7 ): 942 – 947 . doi:10.2460/ajvr.2002.63.942. Google Scholar Crossref Search ADS PubMed WorldCat 68 Deeg CA . Ocular immunology in equine recurrent uveitis . Vet Ophthalmol . 2008 ; 11 ( Suppl 1 ): 61 – 65 . doi:10.1111/j.1463-5224.2008.00625.x. Google Scholar Crossref Search ADS PubMed WorldCat 69 Deeg CA , Kaspers B , Gerhards H , Thurau SR , Wollanke B , Wildner G . Immune responses to retinal autoantigens and peptides in equine recurrent uveitis . Investig Ophthalmol Vis Sci . 2001 ; 42 ( 2 ): 393 – 398 . WorldCat 70 Deeg CA , Pompetzki D , Raith AJ , et al. Identification and functional validation of novel autoantigens in equine uveitis . Mol Cell Proteomics . 2006 b; 5 ( 8 ): 1462 – 1470 . doi:10.1074/mcp.M500352-MCP200. Google Scholar Crossref Search ADS PubMed WorldCat 71 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. Google Scholar Crossref Search ADS PubMed WorldCat 72 Deeg CA , Amann B , Raith AJ , Kaspers B . Inter- and intramolecular epitope spreading in equine recurrent uveitis . Investig Ophthalmol Vis Sci . 2006 a; 47 ( 2 ): 652 – 656 . doi:10.1167/iovs.05-0789. Google Scholar Crossref Search ADS WorldCat 73 Silver P , Horai R , Chen J , et al. Retina-specific T regulatory cells bring about resolution and maintain remission of autoimmune uveitis . J Immunol . 2015 ; 194 ( 7 ): 3011 – 3019 . doi:10.4049/jimmunol.1402650. Google Scholar Crossref Search ADS PubMed WorldCat 74 Cousins SW , Guss RB , Howes EL , Rosenbaum JT . Endotoxin-induced uveitis in the rat: Observations on altered vascular permeability, clinical findings, and histology . Exp Eye Res . 1984 ; 39 : 665 – 676 . doi:10.1016/0014-4835(84)90065-4. Google Scholar Crossref Search ADS PubMed WorldCat 75 Li Q , Peng B , Whitcup SM , Jang SU , Chan C-C . Endotoxin induced uveitis in the mouse: Susceptibility and genetic control . Exp Eye Res . 1995 ; 61 ( 5 ): 629 – 632 . doi:10.1016/S0014-4835(05)80056-9. Google Scholar Crossref Search ADS PubMed WorldCat 76 Agarwal RK , Silver PB , Caspi RR . Rodent models of experimental autoimmune uveitis . Methods Mol Biol . 2012 ; 900 : 443 – 469 . doi:10.1007/978-1-60761-720-4_22. Google Scholar Crossref Search ADS PubMed WorldCat 77 Chen J , Qian H , Horai R , Chan CC , Falick Y , Caspi RR . Comparative analysis of induced vs. spontaneous models of autoimmune uveitis targeting the interphotoreceptor retinoid binding protein . PLoS One . 2013 ; 8 ( 8 ). doi:10.1371/journal.pone.0072161. WorldCat 78 Chen J , Qian H , Horai R , Chan C-C , Caspi RR . Mouse models of experimental autoimmune uveitis: Comparative analysis of adjuvant-induced vs spontaneous models of uveitis . Curr Mol Med . 2015 ; 15 ( 6 ): 550 – 557 . http://www.ncbi.nlm.nih.gov/pubmed/26238369. Google Scholar Crossref Search ADS PubMed WorldCat 79 Shao H , Liao T , Ke Y , Shi H , Kaplan HJ , Sun D . Severe chronic experimental autoimmune uveitis (EAU) of the C57BL/6 mouse induced by adoptive transfer of IRBP1-20-specific T cells . Exp Eye Res . 2006 ; 82 ( 2 ): 323 – 331 . doi:10.1016/j.exer.2005.07.008. Google Scholar Crossref Search ADS PubMed WorldCat 80 Fleisher LN , Ferrell JB , McGahan MC . Ocular inflammatory effects of intravitreally injected tumor necrosis factor-alpha and endotoxin . Inflammation . 1990 ; 14 ( 3 ): 325 – 335 . doi:10.1007/BF00915816. Google Scholar Crossref Search ADS PubMed WorldCat 81 Goldblum D , Fausch K , Frueh BE , Theurillat R , Thormann W , Zimmerli S . Ocular penetration of caspofungin in a rabbit uveitis model . Graefe’s Arch Clin Exp Ophthalmol . 2007 ; 245 ( 6 ): 825 – 833 . doi:10.1007/s00417-006-0460-x. Google Scholar Crossref Search ADS WorldCat 82 Ang M , Ng X , Wong C , et al. Evaluation of a prednisolone acetate-loaded subconjunctival implant for the treatment of recurrent uveitis in a rabbit model . PLoS One . 2014 ; 9 ( 5 ). doi:10.1371/journal.pone.0097555. WorldCat 83 Edmond M , Yuan A , Bell BA , et al. The feasibility of spectral-domain optical coherence tomography grading of anterior chamber inflammation in a rabbit model of anterior uveitis . Investig Ophthalmol Vis Sci . 2016 ; 57 ( 9 ): OCT184 – OCT188 . doi:10.1167/iovs.15-18883. Google Scholar Crossref Search ADS WorldCat 84 Ghosn CR , Li Y , Orilla WC , et al. Treatment of experimental anterior and intermediate uveitis by a dexamethasone intravitreal implant . Invest Ophthalmol Vis Sci . 2011 ; 52 ( 6 ): 2917 – 2923 . doi:10.1167/iovs.10-5939. Google Scholar Crossref Search ADS PubMed WorldCat 85 Neumann R , Jabbur NS , Vickers F , Foster SC . Topical diclofenac sodium, dexamethasone and placebo compared in a model of immunogenic uveitis in rabbits . Ocul Immunol Inflamm . 1993 ; 1 ( 1–2 ): 87 – 98 . doi:10.3109/09273949309086543. Google Scholar Crossref Search ADS PubMed WorldCat 86 Tilden ME , Boney RS , Goldenberg MM , Rosenbaum JT . The effects of topical S(+)-ibuprofen on interleukin-1 induced ocular inflammation in a rabbit model . J Ocul Pharmacol . 1990 ; 6 ( 2 ): 131 – 135 . http://www.ncbi.nlm.nih.gov/pubmed/2394932. Google Scholar Crossref Search ADS PubMed WorldCat 87 Gilger BC , Abarca EM , Salmon JH , Patel S . Treatment of acute posterior uveitis in a porcine model by injection of triamcinolone acetonide into the suprachoroidal space using microneedles . Invest Ophthalmol Vis Sci . 2013 b; 54 ( 4 ): 2483 – 2492 . doi:10.1167/iovs.13-11747. Google Scholar Crossref Search ADS PubMed WorldCat 88 Gerding JC , Gilger BC . Prognosis and impact of equine recurrent uveitis . Equine Vet J . 2016 ; 48 ( 3 ): 290 – 298 . doi:10.1111/evj.12451. Google Scholar Crossref Search ADS PubMed WorldCat 89 Altinsoy A , Dileköz E , Kul O , et al. A cannabinoid ligand, anandamide, exacerbates endotoxin-induced uveitis in rabbits . J Ocul Pharmacol Ther . 2011 ; 27 ( 6 ): 545 – 552 . doi:10.1089/jop.2011.0049. Google Scholar Crossref Search ADS PubMed WorldCat 90 Wacker WB . Autoimmune uveitis (Choroiditis) in the guinea pig sensitized with homologous uvea and its differentiation from that following sensitization with homologous retina . Int Arch Allergy Immunol . 1972 ; 43 ( 1 ): 39 – 52 . doi:10.1159/000230819. Google Scholar Crossref Search ADS WorldCat 91 Allen JB , McGahan MC , Ferrell JB , Adler KB , Fleisher LN . Nitric oxide synthase inhibitors exert differential time-dependent effects on LPS-induced uveitis . Exp Eye Res . 1996 ; 62 : 21 – 28 . Google Scholar Crossref Search ADS PubMed WorldCat 92 Nussenblatt RB , Calogero D , Buchen SY , Leder HA , Goodkin M , Eydelman MB . Rabbit intraocular reactivity to endotoxin measured by slit-lamp biomicroscopy and laser flare photometry . Ophthalmology . 2012 ; 119 ( 7 ). doi:10.1016/j.ophtha.2012.04.004. WorldCat 93 Rafie F , Javadzadeh Y , Javadzadeh AR , et al. In vivo evaluation of novel nanoparticles containing dexamethasone for ocular drug delivery on rabbit eye . Curr Eye Res . 2010 ; 35 ( 12 ): 1081 – 1089 . doi:10.3109/02713683.2010.508867. Google Scholar Crossref Search ADS PubMed WorldCat 94 Mruthyunjaya P . Efficacy of low-release-rate fluocinolone acetonide intravitreal implants to treat experimental uveitis . Arch Ophthalmol . 2006 ; 124 ( 7 ): 1012 . doi:10.1001/archopht.124.7.1012. Google Scholar Crossref Search ADS PubMed WorldCat 95 Jaffe GJ , Yang CS , Wang XC , Cousins SW , Gallemore RP , Ashton P . Intravitreal sustained-release cyclosporine in the treatment of experimental uveitis . Ophthalmology . 1998 ; 105 ( 1 ): 46 – 56 . doi:10.1016/S0161-6420(98)91176-9. Google Scholar Crossref Search ADS PubMed WorldCat 96 Jaffe GJ , Martin D , Callanan D , Pearson PA , Levy B , Comstock T . Fluocinolone acetonide implant (retisert) for noninfectious posterior uveitis. Thirty-four-week results of a multicenter randomized clinical study . Ophthalmology . 2006 ; 113 ( 6 ): 1020 – 1027 . doi:10.1016/j.ophtha.2006.02.021. Google Scholar Crossref Search ADS PubMed WorldCat 97 Gilger BC , Abarca E , Salmon JH . Selection of appropriate animal models in ocular research: Ocular anatomy and physiology of common animal models. In: Brian C. Gilger, ed. Ocular Pharmacology and Toxicology. Springer Scientific; 2014. ISBN 978-1-62703-744-0 [2013a]. 98 Wollanke B , Brem S , Meyer P , et al. Prophylaxis of equine recurrent uveitis (ERU): First results with a leptospiral vaccine in horses . Pferdeheilkunde . 2004 ; 20 ( 5 ): 447 – 454 . Google Scholar Crossref Search ADS WorldCat 99 Kleinwort KJH , Amann B , Hauck SM , Feederle R , Sekundo W , Deeg CA . Immunological characterization of intraocular lymphoid follicles in a spontaneous recurrent uveitis model . Investig Ophthalmol Vis Sci . 2016 ; 57 ( 10 ): 4504 – 4511 . doi:10.1167/iovs.16-19787. Google Scholar Crossref Search ADS WorldCat 100 Deeg CA . A proteomic approach for studying the pathogenesis of spontaneous equine recurrent uveitis (ERU) . Vet Immunol Immunopathol . 2009 ; 128 ( 1–3 ): 132 – 136 . doi:10.1016/j.vetimm.2008.10.302. Google Scholar Crossref Search ADS PubMed WorldCat 101 Deeg CA , Raith AJ , Amann B , et al. CRALBP is a highly prevalent autoantigen for human autoimmune uveitis . Clin Dev Immunol . 2007 ; 2007 . doi:10.1155/2007/39245. WorldCat 102 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 ). doi :10.1371/journal.pone.0091684. WorldCat 103 Zipplies JK , Hauck SM , Schoeffmann S , et al. Serum PEDF levels are decreased in a spontaneous animal model for human autoimmune uveitis . J Proteome Res . 2009 ; 8 ( 2 ): 992 – 998 . doi:10.1021/pr800694y. Google Scholar Crossref Search ADS PubMed WorldCat 104 Lucchesi PMA , Parma AE . A DNA fragment of Leptospira interrogans encodes a protein which shares epitopes with equine cornea . Vet Immunol Immunopathol . 1999 ; 71 ( 3–4 ): 173 – 179 . doi:10.1016/S0165-2427(99)00084-7. Google Scholar Crossref Search ADS PubMed WorldCat 105 Lucchesi PMA , Parma AE , Arroyo GH . Serovar distribution of a DNA sequence involved in the antigenic relationship between Leptospira and equine cornea . BMC Microbiol . 2002 ; 2 : 3 . doi:10.1186/1471-2180-2-3. Google Scholar Crossref Search ADS PubMed WorldCat 106 Verma A , Artiushin S , Matsunaga J , Haake DA , Timoney JF . LruA and LruB, novel lipoproteins of pathogenic Leptospira interrogans associated with equine recurrent uveitis . Infect Immun . 2005 ; 73 ( 11 ): 7259 – 7266 . doi:10.1128/IAI.73.11.7259-7266.2005. Google Scholar Crossref Search ADS PubMed WorldCat 107 Verma A , Kumar P , Babb K , Timoney JF , Stevenson B . Cross-reactivity of antibodies against leptospiral recurrent uveitis-associated proteins A and B (LruA and LruB) with eye proteins . PLoS Negl Trop Dis . 2010 ; 4 ( 8 ). doi :10.1371/journal.pntd.0000778. WorldCat 108 Verma A , Rathinam SR , Priya CG , Muthukkaruppan VR , Stevenson B , Timoney JF . LruA and LruB antibodies in sera of humans with leptospiral uveitis . Clin Vaccine Immunol . 2008 ; 15 ( 6 ): 1019 – 1023 . doi:10.1128/CVI.00203-07. Google Scholar Crossref Search ADS PubMed WorldCat 109 Gilger BC , Malok E , Stewart T , et al. Effect of an intravitreal cyclosporine implant on experimental uveitis in horses . Vet Immunol Immunopathol . 2000 ; 76 ( 3–4 ): 239 – 255 . doi:10.1016/S0165-2427(00)00219-1. Google Scholar Crossref Search ADS PubMed WorldCat 110 Gilger BC , Wilkie DA , Davidson MG , Allen JB . Use of an intravitreal sustained-release cyclosporine delivery device for treatment of equine recurrent uveitis . Am J Vet Res . 2001 ; 62 ( 12 ): 1892 – 1896 . doi:10.2460/ajvr.2001.62.1892. Google Scholar Crossref Search ADS PubMed WorldCat 111 Gilger BC , Salmon JH , Wilkie DA , et al. A novel bioerodible deep scleral lamellar cyclosporine implant for uveitis . Invest Ophthalmol Vis Sci . 2006 ; 47 ( 6 ): 2596 – 2605 . doi:10.1167/iovs.05-1540. Google Scholar Crossref Search ADS PubMed WorldCat 112 Gilger BC , Wilkie DA , Clode AB , et al. Long-term outcome after implantation of a suprachoroidal cyclosporine drug delivery device in horses with recurrent uveitis . Vet Ophthalmol . 2010 ; 13 ( 5 ): 294 – 300 . Google Scholar Crossref Search ADS PubMed WorldCat 113 Moisseiev E , Loewenstein A , Yiu G . The suprachoroidal space: From potential space to a space with potential . Clin Ophthalmol . 2016 ; 10 : 173 – 178 . doi:10.2147/OPTH.S89784. Google Scholar Crossref Search ADS PubMed WorldCat 114 Hirsch ML , Conatser LM , Smith SM , et al. AAV vector-meditated expression of HLA-G reduces injury-induced corneal vascularization and immune cell infiltration . Sci Rep . 2017 ; 7 ( 1 ): 17840 . Google Scholar Crossref Search ADS PubMed WorldCat 115 Villatoro A , Fernández V . Use of adipose-derived mesenchymal stem cells in keratoconjunctivitis sicca in a canine model . Biomed Res Int . 2014 ; 2015 ( 2015 ): 10 . doi:10.1155/2015/527926. WorldCat © The Author(s) 2018. Published by Oxford University Press on behalf of the National Academy of Sciences. All rights reserved. For permissions, please email: journals.permissions@oup.com This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model) http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png ILAR Journal Oxford University Press

Immune Relevant Models for Ocular Inflammatory Diseases

ILAR Journal , Volume 59 (3) – Dec 31, 2018

Loading next page...
1
 
/lp/ou_press/immune-relevant-models-for-ocular-inflammatory-diseases-yxpGxVDD3u

References (123)

Publisher
Oxford University Press
Copyright
© The Author(s) 2018. Published by Oxford University Press on behalf of the National Academy of Sciences. All rights reserved. For permissions, please email: journals.permissions@oup.com
ISSN
1084-2020
eISSN
1930-6180
DOI
10.1093/ilar/ily002
Publisher site
See Article on Publisher Site

Abstract

Abstract Ocular inflammatory diseases, such as dry eye and uveitis, are common, painful, difficult to treat, and may result in vision loss or blindness. Ocular side effects from the use of antiinflammatory drugs (such as corticosteroids or nonsteroidal antiinflammatories) to treat ocular inflammation have prompted development of more specific and safer medications to treat inflammatory and immune-mediated diseases of the eye. To assess the efficacy and safety of these new therapeutics, appropriate immune-relevant animal models of ocular inflammation are needed. Both induced and naturally-occurring models have been described, but the most valuable for translating treatments to the human eye are the animal models of spontaneous, immunologic ocular disease, such as those with dry eye or uveitis. The purpose of this review is to describe common immune-relevant models of dry eye and uveitis with an overview of the immuno-pathogenesis of each disease and reported evaluation of models from small to large animals. We will also review a selected group of naturally-occurring large animal models, equine uveitis and canine dry eye, that have promise to translate into a better understanding and treatment of clinical immune-relevant ocular disease in man. animal models, dry eye, immune-relevant, inflammatory, naturally-occurring, ocular, uveitis Introduction Blindness or low vision affects approximately 1 in 28 Americans older than 40 years of age, the underlying causes of which are commonly noninfectious immune-mediated diseases, including dry eye and uveitis.1,–3 Dry eye symptoms are experienced by 20% of adults over 45 years old, and uveitis is a leading cause of blindness in the United States.1,–6 Dry eye and uveitis are also common causes of blindness in domestic animals, and uveitis is the leading cause of blindness in horses worldwide.7,–12 There are no known cures for immune-mediated ocular diseases, and current treatment regimens are costly, require multiple daily applications, are poorly effective, and have adverse side effects. Therefore, new treatments to address these diseases are needed and for further development, there is a need for accurate and translatable immune-relevant models of ocular disease. The eye, like the brain and the uterus in pregnancy, is considered an immune privileged site.13,14 An active suppression of the immune response to endogenous and exogenous antigens occurs in the eye, as overt inflammation may compromise vision. The relative lack of antigen-presenting and MHC II-expressing cells and natural tissue barriers (i.e., the blood–ocular barrier) that physically separate ocular tissues from the systemic immune response contribute to the immune tolerance in the eye.15 With dry eye and uveitis, the normal ocular tolerance is lost (from several initiating causes) and the physical barriers become disrupted, allowing an influx of inflammatory cells. In addition, proinflammatory mediators induce T-helper cells to proliferate, activate antigen-presenting cells, expand auto-reactive B and T cell populations, and ultimately release proinflammatory and proapoptotic peptides.16,17 Current treatments for dry eye and uveitis are nonspecific and require frequent use of topical medications that may have severe ocular and systemic side effects.18,–20 Furthermore, these medications are life-long therapies and patient compliance is commonly poor, leading to treatment failures, worsening of disease, and in some cases, blindness.21 When testing effectiveness of therapeutics on models of ocular disease, there are two separate but important testing goals. The first question is whether the drug is effective in the ocular disease state that is being studied. For this goal, usually rats or mice are evaluated and dosed by a nonocular route, for example, orally, subcutaneously, or intraperitoneally. These studies help determine pathogenesis of disease-drug mechanisms; therefore, the wide array of reagents and genetically modified mice and rats are a major asset. Determination of the appropriate dose (i.e., dose ranging studies) is usually also performed in these first sets of studies. The second goal is to determine if an appropriate dose can reach the ocular target tissue and be effective in the eye using a dosing route and frequency that is clinically feasible. These studies would determine the pharmacokinetics and pharmacodynamics of a specific route of administration of a drug, typically in a normal eye, then repeated using the optimal dosing and routes in eyes of models of the disease state. For this second group of studies to be clinically valid in most instances, the animal models would have to have eyes anatomically similar to the target species and in the case of humans, use of the rabbit, dog, pig, or primate eye would be most appropriate. Finally, when selecting the appropriate animal model, the target tissue and disease state has to be paired with the most appropriate route of therapy. This determination is important for pharmacokinetic, toxicologic, and efficacy studies. Although there are many disease conditions of the human eye thought to have an immunologic pathogenesis, including allergic conjunctivitis, corneal transplant rejection, and age-related macular degeneration, as examples, the purpose of this review is to describe common immune-relevant models of dry eye and uveitis with an overview and assessment of models from small to large animals. We will also review a selected group of naturally-occurring large animal models, equine uveitis and canine dry eye, which have promise to translate into a better understanding and treatment of clinical immune-relevant ocular disease in man. Review of Commonly Used Animal Models in Inflammatory Ocular Disease Ocular Surface Disease Immune-Relevant Models Dry Eye Disease Dry eye disease (DED) is one of the most common ocular abnormalities and has multiple underlying causes. Dry eye is a disease of the tear film and ocular surface that results in symptoms of discomfort and visual disturbance with potential damage to the ocular surface.22 In one study, nearly one-half of patients claimed to have symptoms of dry eye with a negative effect on quality of life, including ocular pain, decreased activities requiring visual attention (e.g., reading, driving), and reduced productivity in the workplace.21 Dry eye develops from a deficiency of the aqueous portion of the tear fluid as a result of reduced lacrimal aqueous tear secretion or a result of increased evaporation of tears, such as the result of Meibomian gland deficiencies.23 Decreased aqueous production of the tears results in an increase of tear electrolytes (i.e., increased tear osmolality), proteins, and inflammatory mediators, resulting in damage to the surface ocular tissues, decreased visual acuity, and ocular discomfort. The relative decrease in aqueous tears on the ocular surface in patients with DED causes chronic irritation to ocular surface that disrupts the normal ocular immune tolerance.24 With breakdown of ocular surface tolerance and immune-homeostasis, autoimmunity develops through activation of NK cells and Toll-like receptors, followed by release of proinflammatory factors such as interleukin (IL)-1α, IL-1β, tumor necrosis factor α, and IL-6. These mediators amplify, activating antigen-presenting cells, which internalize autoantigens and migrate to the draining cervical lymph node where autoreactive Th1 cells, Th17 cells, or B cells (i.e., in Sjogren’s syndrome) undergo expansion. Efferent trafficking of these autoreactive T cells to the ocular surface is directed by adhesion molecules (e.g., LFA-1) and chemokine receptors. Autoreactive T-cells in ocular surface tissues potentiate the chronic autoimmune response, resulting in epithelial cell apoptosis, reduced goblet cell density, and squamous metaplasia of epithelium.17,24,25 Current treatments for DED rely on frequently applied artificial tears, punctal plugs, topical tetracycline antibiotic, and omega fatty acids, all of which provide only temporary relief of dry eye.26 Chronic DED is commonly treated with antiinflammatory medications and immunosuppressants, the latter being the mainstay of treatment in the United States.27,28 Topical cyclosporine, an immunosuppressant, used with or without corticosteroids, is effective in DED through inhibition of T-cell activation and reduction of proinflammatory cytokines.29 A recently approved topical immunosuppressive for treatment of DED, lifitegrast, is an integrin inhibitor that prevents binding of LFA-1 to ICAM-1, which is upregulated in DED. Lifitegrast thus blocks T-cell efferent recruitment to ocular tissues and reduces inflammatory cytokines.30,–32 However, both cyclosporine and lifitegrast must be administered indefinitely twice daily by the patient and are associated with burning sensation after application, leading to reduced patient compliance and hence poor treatment efficacy and success. Therefore, an effective, long-term, well-tolerated, and convenient therapy for DED is needed. There are numerous models of ocular surface disease and dry eye, but to be immune relevant, there needs to be evidence of an immuno-pathogenesis in the disease process. There are several mouse models of dry eye disease, the most common of which is a model induced by low humidity and high air flow environments, with or without the additional use of scopolamine (Table 1).33,–35 The extended environmental irritation to the surface of the eye of these mice disrupts the normal ocular immune tolerance and immunohomeostasis,24 as described previously. These mice models have been used to study the immuno-pathogenesis of dry eye and the initial evaluation of therapeutics. Another described model is the use of repeated application of topical benzalkonium chloride to the mouse or rabbit eye. This produces chronic irritation that may develop immunopathology and chronic ocular surface disease.36,37 Other induced models of DED in rodents, which may be less immunopathologic in origin, include lacrimal gland excision or injections of toxins or antigens such as botulinum toxin38 or concanavalin A.39 Genetic models, such as the MRL/lpr mouse, manifest multiple autoimmune disorders and can be helpful to study diseases such as systemic lupus erythematosus and Sjorgren’s syndrome (Table 1).40 Another example of genetic DED are neurturin-deficient mice, which may develop dry eye and serve as models for neurotrophic keratoconjunctivitis sicca, since this model lacks lacrimal innervation (Table 1).41 There are numerous other knockout and transgenic mice strains that are commonly studied that may develop DED; however, many of these models do not develop clinical signs of DED observed in large animal models, but instead develop histologic or other features characteristic of human DED.42 Table 1 Selected immune-models of dry eye disease Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Open in new tab Table 1 Selected immune-models of dry eye disease Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Animal Method Advantages Disadvantages Reference Rodent Mice Environmental chambers ± scopolamine patch Reproducible, economical Small eye, anatomic differences 30 Botulinum toxin injection into lacrimal gland Reproducible, economical Above, and toxin present 29,38 Intraorbital injection of concanavalin A Above, possible orbital inflammation 39 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 36 Extraorbital lacrimal gland excision ± scopolamine Surgically induced desiccating irritation Initiates an abnormal Th1/Th17 T cell response, exogenous antigens 43,44 Neurturin-deficient (NRTN(−/−)) 41 MRL/lpr Sjogrens-like dry eye, autoimmune pathogenesis Chronic model, systemic disease 40,45,–47 Rat Extraorbital lacrimal gland excision ± scopolamine May not be immunologic 48,49 Rabbit Activated autologous lymphocytes injected into lacrimal gland Sjögren’s-like autoimmune dacryoadenitis Specialized, difficult model 50 Topical administration of benzalkonium chloride Economical Above, time to develop, may not be immunologic 37 Lacrimal gland excision May not be immunologic 51 Canine Naturally-occurring Immunologic, translatable Availability 9,–11 Open in new tab In rats, the most commonly described dry eye model is the extraorbital lacrimal gland excision model (with or without use of scopolamine).48,49 Like other models of induced dry eye, this rat lacrimal-excision model likely does not develop, substantially, an immunologic pathogenesis and therefore may not be as effective for evaluation of immunosuppressive therapies as naturally-occurring models of dry eye.33,49 All of these rodent models of dry eye are similar in that they can be used to determine proof of principal of therapeutic response to a drug, but all have similar disadvantages of having orbital and lacrimal anatomy and eye size that differs from the human eye. Rabbit or dog models are more commonly used to evaluate dry eye signs and response to therapy, because they have easily measured decreased tear production and develop ocular surface changes.42 Therefore, larger animal dry eye models are needed (see later description of canine dry eye). Rabbits are commonly used as models of ocular disease and for pharmacokinetic studies because of their relatively large eye, compared to rodents, while still being a common and economical laboratory animal. However, there are few true immune-relevant models of DED in rabbits. Most described models induce dry eye signs, but not likely an immunopathogenesis, by use of a topical irritant, such as benzalkonium chloride, or by short-term reduction in lacrimal secretion using parasympathomimetic drug, such as atropine.37,52,53 A very promising model of autoimmune dacryoadenitis in rabbits that produces a Sjögren’s-like keratoconjunctivitis is created by an intra-lacrimal or subcutaneous injection of autologous peripheral blood lymphocytes activated by purified rabbit lacrimal epithelial cells.54,55 This rabbit autoimmune dacryoadenitis model has been used to effectively evaluate immunomodulatory treatments for dry eye, including topical cyclosporine and lacrimal gland adeno-associated virus (AAV) mediated-IL-10 gene therapy.50,56 Naturally-Occurring Keratoconjunctivitis Sicca in Canines Domestic canines develop spontaneous dry eye that clinically and immunopathologically is similar to dry eye in humans (Table 1).10 Not only do dogs spontaneously develop dry eye symptoms of ocular discomfort, conjunctival hyperemia, and corneal scarring, these symptoms correlate directly with reduced aqueous tear production, a reduction readily measured using a standard Schirmer tear test strip (Figure 1). Furthermore, dogs with dry eye have a reduced tear breakup time and increased corneal staining, all abnormalities also observed in humans with DED.10 Like humans, canine dry eye is typically bilateral, develops in middle age, is more common in female dogs and in certain breeds, such as the American Cocker spaniel, Bulldog, and West Highland white terrier.57 The pathogenesis of dry eye in dogs appears similar to that of humans, where an apparent immunologic inflammation occurs with progressive lymphocytic infiltration and damage to the lacrimal gland with subsequent decreased production of the aqueous tear film.58,59 With chronicity, the ocular surface becomes progressively more dessicated and inflamed, the cornea vascularizes and scars, and ultimately the dog may lose vision.9,57 Initial proof of concept of commonly used immunosuppressive eye drops was first demonstrated to be effective in this spontaneous dog model, including topical cyclosporine, tacrolimus, and LTF-1 inhibitors.9,10,12,60,61 Figure 1 Open in new tabDownload slide Naturally-occurring dry eye in a dog. (A) Moderate dry eye disease in a dog resulting in conjunctival hyperemia, corneal vascularization, and corneal opacity. (B) Chronic dry eye disease in a dog with mucopurulent ocular discharge, hyperpigmented cornea, and conjunctival hyperemia. Figure 1 Open in new tabDownload slide Naturally-occurring dry eye in a dog. (A) Moderate dry eye disease in a dog resulting in conjunctival hyperemia, corneal vascularization, and corneal opacity. (B) Chronic dry eye disease in a dog with mucopurulent ocular discharge, hyperpigmented cornea, and conjunctival hyperemia. Uveitis Disease Models Uveitis is inflammation of the iris, ciliary body, and choroid and is associated with both infectious and noninfectious causes. Uveitis is estimated to be the third leading cause of preventable blindness worldwide.62 In the United States, the incidence of uveitis was estimated to be approximately 58 to 69 cases/100,000 people;1,6 however, another study estimated that the rate of uveitis, especially anterior uveitis, was approximately 3 times higher and it increased with increasing age of patients.4 The most common causes of uveitis in humans are human leukocyte antigen (HLA)-B27 related uveitis, acute anterior uveitis in herpes zoster disease, toxoplasmosis, sarcoidosis, and pars planitis.63 Uveitis results from several causes. The uveal tract supplies blood to the eye and is in direct contact with peripheral vasculature; therefore, diseases of the systemic circulation (e.g., septicemia, bacteremia, infection, activated lymphocytes, immune diseases, etc.) will disrupt the blood-ocular barrier.64,65 The blood-ocular barrier prevents large molecules and cells from entering the eye and thus limits the immune response to intraocular antigens. With trauma or inflammation, this barrier can be disrupted, allowing blood products and cells to enter the eye, resulting in the clinical signs typical of uveitis, such as flare, cell accumulation, and vitreous haze. Disruption of the barrier enables activation of various host immune responses, including antibody production to self-antigens that are not normally recognized by the immune system, as well as antibody production to foreign antigens inside the eye. As a result of the blood-ocular barrier, lack of lymphatics, and the presence of limited numbers of resident leukocytes, the eye is considered to have immune privilege. Naïve T cells cannot cross the normal blood-retinal barrier due to the lack of fenestration in the retinal vessels and the lack of appropriate adhesion molecules.66 Expression of chemokines in inflammation and activated T cells in the ciliary epithelium may play a role in recruitment and activation of leukocytes in diseased eyes.67 As in other autoimmune disorders, infections may trigger events, either by antigenic mimicry with a pathogen’s antigen or as a bystander effect due to the general systemic or local immune stimulation by the pathogen. Uveitogenic retinal proteins documented in experimental animals include retinal arrestin, interphotoreceptor retinoid-binding protein (IRBP), rhodopsin, recoverin, phosducin, and retinal pigment epithelium derived RPE-65.62,68,–70 Irrespective of the eliciting antigen, available experimental evidence suggests that the immunological mechanisms driving the resultant disease are similar.16 Following disruption of the blood-ocular barrier, large amounts of predominantly CD4+ T cells enter the eye and secrete proinflammatory cytokines such as IL-2 and interferon γ.71 Auto-reactive effector CD4+ T cells have been associated with the pathogenesis of inflammatory and autoimmune disorders including uveitis. Naıve CD4+ T cells differentiate into effector subsets depending on the nature of the environment in which exposure to the antigen occurs.66 Several T cell effector phenotypes have been defined, known as T helper 1 (TH1), TH2, or TH17. Early studies suggested that the interferon-γ-producing TH1 and IL-17-releasing TH17 subsets are responsible for the pathology of uveitis, with the latter being associated with development of autoimmune disease.16 Additionally, clinical uveitis frequently develops spontaneous recurrent or relapsing bouts of inflammation, likely from T cells recognizing additional autoantigens in the ocular tissue.72 Resolution of uveitis is dependent on the presence of T regulatory cells (Tregs) that are labeled as CD4+Foxp3+ cells. When Foxp3+ T cell percentages in uveitis increase to approximately 10% of the total CD4+ cells, the acute inflammation rapidly resolves. Therefore, Foxp3+ Tregs are important to induce spontaneous resolution and in maintaining remission of uveitis.73 Multiple models have been developed to evaluate the immuno-pathogenesis of uveitis and recurrent uveitis, including identification of autoantigens. Most of these models are rodent based. Other models, including those that are acute, chronic, and recurrent in nature, have been developed to evaluate therapeutics (Table 2). Large animal models, such as uveitis induced in rabbits and pigs, have been evaluated to test therapeutics in larger eyes to help translate these treatments to humans (Table 2). Table 2. Models of uveitis Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 EAU, experimental autoimmune anterior uveitis; EIU, endotoxin-induced uveitis; IL, interleukin; NZW, Zealand White. Open in new tab Table 2. Models of uveitis Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 Type of uveitis Species/Strains Agents Type of inflammation Advantages Disadvantages Reference Rodent EIU Lewis rat: Harlan Sprague Dawley Mice: (C3H and other strains) Endotoxin Anterior uveitis Rapid onset, predictable Nonimmunologic 74,75 EAU Rat Melanin from bovine RPE Tyrosinase-related proteins 1 and 3 Anterior uveitis Immunologic 76 Experimental autoimmune uveoretinitis Mice Rat Retinal arrestin (S-Ag), IRBP, recoverin, phosducin, rhodopsin/opsin Posterior segment Immunologic 73,77,–79 Spontaneous Mice RBP T cell receptor transgenic mice (R161H) Autoimmune Regulator (AIRE)(−/−) mice Adoptive transfer Retinal degeneration and persistent cellular infiltrates and lymphoid aggregation, multi-focal infiltrates and severe choroidal inflammation. 77,79 Rabbit EIU NZW Dutch belted Endotoxin Anterior and posterior segment Rapid onset, predictable Nonimmunogenic 80,81 Recurrent uveitis NZW Mycobacterium tuberculosis H37Ra antigen Ovalbumin Anterior (intracameral) or posterior (intravitreal) segment Immunologic and recurrent 82,–85 Cytokine induced uveitis NZW IL-1 TNF-alpha Acute onset (6 hours) Nonimmunologic 80,86 Porcine EIU Various Endotoxin Posterior Large eye Nonimmunogenic 87 Horse Recurrent uveitis Various Spontaneous Anterior and posterior Large eye; immunologic; recurrent Nonstandard research animal Cost 7,68,88 EAU, experimental autoimmune anterior uveitis; EIU, endotoxin-induced uveitis; IL, interleukin; NZW, Zealand White. Open in new tab Rodent Models of Uveitis Endotoxin-induced uveitis A commonly used model of induced uveitis in rodents is the endotoxin-induced uveitis (EIU) model (Table 2).74,75,89 The uveitis in this model is primarily an acute anterior uveitis (i.e., iris, ciliary body) that is thought to be driven by the innate immune system (Table 2).16,76 Following intraparentoneal, subcutaneous, or hind footpad injection of endotoxin (lipopolysaccharide; 100 μg or 500 μg) in Lewis or Sprague-Dawley rats or various mouse strains (C3H),16 ocular inflammation develops within hours of injection characterized by a breakdown of the blood-aqueous barrier and the development of clinical disease. Clinical and histopathologic abnormalities peak at 24 hours and resolve by 48 to 72 hours.74,75 Experimental autoimmune uveitis (or uveoretinitis) Experimental autoimmune uveitis (EAU) is a primarily posterior uveitis (or panuveitis [i.e., inflammation of the iris, ciliary, and choroid]) that is induced by immunizing susceptible rodents with retinal antigens (e.g., S-antigen [S-ag], IRBP, recoverin, rhodopsin/opsin); while experimental melanin–protein induced uveitis, a predominantly anterior uveitis, is elicited by immunization with melanin (from RPE) or tyrosinase-related proteins 1 and 2 (Table 2).16 The predominant animal model is the Lewis rat, but other animals such as the guinea pig or mice have also been described.76,90 Injection of autoantigens into rodents, combined with bacterial adjuvants, results in EAU; EAU does not develop without the use of adjuvants. The use of complete Freund’s adjuvant (Mycobacterium cell wall product) or pertussis toxin is necessary to stimulate the innate immune response and develop inflammation73,76,78 that ultimately generates activated antigen-presenting cells capable of presenting the injected autoantigen with the coactivation factors required to activate T cells capable of recognizing the antigen. Severe EAU was induced in B6 mice by adoptive transfer of IRBP-specific T cells.79 Most of the rodent experimental models of uveitis are not recurrent. They often elicit a single, albeit chronic course of uveitis that eventually resolves. Therefore, the immunologic pathways involved in the development of these rodent models may not be the same as in naturally occurring uveitis. Spontaneous uveitis has been observed in various mouse models, including IRBP T cell receptor transgenic mice (R161H) and autoimmune regulator (AIRE)(-/-) mice.78 These mouse models have a gradual onset of chronic ocular inflammation that ultimately leads to retinal degeneration.78 Despite limitations, these rodent experimental models offer great insight into the pathogenesis and immunopathogenesis of uveitis. These models have been critical in evaluation of therapies, particularly broader immunosuppressive therapies, for treating uveitis. Rabbit Models of Uveitis Two rabbit models of uveitis have been most commonly evaluated, including the acute uveitis induced by injection of endotoxin84,91,–93 and the recurrent uveitis induced by tuberculosis antigen.82,84,94 Other uveitis models in rabbits include those following intravitreal injection of human interleukin 1 alpha,86 TNF-alpha,80 or ovalbumin in animals previously ovalbumin-immunized,85 among others (Table 2). An advantage of rabbit models of uveitis over rodent models is that the rabbit eye is more similar in size to the human eye, and therefore, more pharmacologically valid when evaluating routes of therapy. In the endotoxin-induced uveitis, after 10 to 100 ng of LPS is injected intracamerally92 or intravitreally,80,81 aqueous flare and iridal hyperemia develop within 6 hours, suggesting rapid disruption of the blood–aqueous barrier.91 The LPS induces inflammation by activating a Toll-like receptor 4-initiated signaling cascade. The inflammatory response peaks at approximately 24 hours after injection, then rapidly declines.92 Like the rat model of EIU, this endotoxin rabbit model of uveitis is not considered to have a predominantly immunopathogenesis. Experimental uveitis can be induced by unilateral intravitreal or intracameral injection of Mycobacterium tuberculosis H37Ra antigen (50 μg; 1 μg/L) in preimmunized rabbits, typically 7 to 14 days after initial subcutaneous injection.82,–84,95 To simulate chronic recurrent inflammation, eyes are re-challenged with intravitreal antigen every 14 to 21 days.96 This model has advantages similar to the endotoxin model; however, it is predominantly a T-cell lymphocyte-mediated uveitis that can be induced to be recurrent and therefore, more closely simulate endogenous human uveitis.95 Porcine Models of Uveitis The pig has been used as a large animal model of uveitis (Table 2), which, similar to the rabbit model, has an eye similar in size to the human eye; however, unlike the rabbit, it has a retinal vascular anatomy similar to humans.97 An acute model of uveitis has been used in the pig to evaluate novel therapeutics and routes of administration. In this model, similar to endotoxin uveitis in rabbits, endotoxin is injected intravitreally and the eye is monitored for up to 72 hours following injection.87 Like rodent EIU and endotoxin uveitis in rabbits, the endotoxin porcine model of uveitis is not considered to have an immunopathogenesis. Naturally-Occurring Uveitis Models Equine Recurrent Uveitis Horses spontaneously develop severe, immunologic uveitis called equine recurrent uveitis (ERU) that is frequently recurrent and chronic (Figure 2).98 ERU is the most common cause of blindness in horses.7,8,98 Spontaneous bouts of uveitis develop and blindness may occur after multiple recurrent episodes of uveitis. The immunopathology of ERU has been extensively studied and has demonstrated that T cells are the predominant mononuclear inflammatory cells infiltrating ocular tissues in horses with naturally occurring chronic uveitis, with a significant number of CD4+ cells.71,72,99 Recruitment of proinflammatory cells as well as autoreactive lymphocytes may be in part driven by the expression of the chemokine RANTES in the ciliary body.67 Figure 2 Open in new tabDownload slide Naturally-occurring uveitis in a horse. (A) Acute active uveitis in a horse, with a miotic pupil and anterior chamber opacity. (B) Chronic uveitis in a horse, from equine recurrent uveitis, with dyscoria, synechiae, and cataract. Figure 2 Open in new tabDownload slide Naturally-occurring uveitis in a horse. (A) Acute active uveitis in a horse, with a miotic pupil and anterior chamber opacity. (B) Chronic uveitis in a horse, from equine recurrent uveitis, with dyscoria, synechiae, and cataract. Study of this common, spontaneous uveitis in horses has helped understand the pathogenesis of uveitis in humans, especially the identification of autoantigens and how recurrence of uveitis develops immunologically.7,69,70,72,100,–103 Several potential autoantigens have been identified in horses that could play a role in the development of autoimmune uveitis. T cells isolated from the eyes of horses with ERU proliferate in response to two common autoantigens in rodents: retinal S-Ag and IRBP.69 In addition, several additional potential autoantigens were identified by analyzing antibodies in the sera of ERU horses that reacted with retinal proteins. These include recoverin, cellular retinaldehyde-binding protein, and malate dehydrogenase.101 While all these potential autoantigens are capable of inducing experimental uveitis in rodent models, only cellular retinaldehyde-binding protein and IRBP consistently produce uveitis in outbred horses.70,100 Additionally, studies of horses with ERU have also helped elucidate how Leptospira infections induce immunological uveitis, specifically autoimmune uveitis.104,–106 Field studies of horses in the 1950s after an outbreak of acute leptospirosis caused by L. interrogans serogroup Pomona demonstrated that one of the six horses (17%) developed intraocular inflammation during acute leptospiral disease, and all horses developed ERU 18 to 24 months after the initial infection. Subsequent studies demonstrated cross-reactivity between equine ocular tissues and Leptospira antigens,104,105 and horses with uveitis associated with Leptospira interrogans infections had high levels of IgA and IgG in their intraocular fluids that reacted to two Leptospira lipoproteins, LruA and LruB.106,107 These antibodies were also subsequently discovered in the serum of human leptospiral uveitis patients.108 Studies of spontaneous ERU have helped elucidate the immunopathogenesis of recurrent uveitis. In autoimmune disease, several autoantigens, or epitopes, participate in the immunopathogenesis; epitope spreading is accountable for disease induction, progression, and inflammatory relapses.72 Epitope spreading is defined as the diversification of epitope specificity from the initial focused, dominant, epitope-specific immune response, directed against a self or foreign protein to cryptic epitopes on that protein (intramolecular spreading) or other proteins (intermolecular spreading).72 The shifts in immunoreactivity, or epitope spreading, have been documented in ERU and are thought to be responsible for the recurring character of ERU.72 ERU, as a model of spontaneous immune-mediated uveitis, has also led to the study of promising therapeutics. For example, several sustained release ocular implants have shown much promise in the treatment of ERU.109,110 Evaluation of drug delivery to the suprachoroidal space has been shown to control ERU and prevent recurrences.111,112 Triamcinolone injections into the suprachoroidal space are currently under development for treatment of human uveitis.113 Further study of ERU and its treatment will translate well to improving the understanding and treatment of human autoimmune uveitis. Next Steps As further therapeutics are developed that more specifically target immune-mediated diseases, evaluation of these treatments in spontaneous or naturally-occurring models of ocular disease will be needed to provide proof of concept and help translate these therapies to humans. Excellent examples of developing, targeted therapies include gene therapy (especially gene addition therapy) and stem cell therapy. Our laboratory and collaborators at the Gene Therapy Center at the University of North Carolina have developed AAV delivery of immunosuppressive proteins, such as HLA-G, for suppression of ocular surface inflammation and vascularization.114 Target ocular surface diseases for AAV-HLA-G gene therapy are DED and for prevention of corneal graft rejection.114 Autologous stem cell therapy, or use of stem cell supernatant extracts, also shows much promise for immunomodulation in the eye. Effectiveness of topical ocular mesenchymal stem cell therapy was initially demonstrated in dry eye models in mice.39 Locally injected fat-derived mesenchymal stem cells near the lacrimal glands of dogs with advanced dry eye demonstrated clinical improvement and increased tear production.115 These results in a naturally-occurring model provides evidence of possible clinical translation to humans with severe dry eye. Acknowledgments and Conflicts The author thanks Jacklyn Salmon and Dr. Jorge Piedrahita for review of this manuscript. References 1 Acharya NR , Tham VM , Esterberg E , et al. Incidence and prevalence of uveitis: Results from the Pacific Ocular Inflammation Study . JAMA Ophthalmol . 2013 ; 131 ( 11 ): 1405 – 1412 . doi:10.1001/jamaophthalmol.2013.4237 . Google Scholar Crossref Search ADS PubMed WorldCat 2 Schaumberg DA , Sullivan DA , Buring JE , Dana MR . Prevalence of dry eye syndrome among US women . Am J Ophthalmol . 2003 ; 136 ( 2 ): 318 – 326 . doi:10.1016/S0002-9394(03)00218-6 . Google Scholar Crossref Search ADS PubMed WorldCat 3 Schaumberg DA , Dana R , Buring JE , Sullivan DA . Prevalence of dry eye disease among US men: Estimates from the Physicians’ Health Studies . Arch Ophthalmol . 2009 ; 127 ( 6 ): 763 – 768 . doi:10.1001/archophthalmol.2009.103 . Google Scholar Crossref Search ADS PubMed WorldCat 4 Gritz DC , Wong IG . Incidence and prevalence of uveitis in Northern California: The Northern California Epidemiology of Uveitis Study . Ophthalmology . 2004 ; 111 ( 3 ): 491 – 500 . doi:10.1016/j.ophtha.2003.06.014 . Google Scholar Crossref Search ADS PubMed WorldCat 5 Merrill PT , Kim J , Cox TA , Betor CC , McCallum RM , Jaffe GJ . Uveitis in the southeastern United States . Curr Eye Res . 1997 ; 16 ( 9 ): 865 – 874 . doi:10.1076/ceyr.16.9.865.5048 . Google Scholar Crossref Search ADS PubMed WorldCat 6 Suhler EB , Lloyd MJ , Choi D , Rosenbaum JT , Austin DF . Incidence and prevalence of uveitis in Veterans Affairs Medical Centers of the Pacific Northwest . Am J Ophthalmol . 2008 ; 146 ( 6 ). doi: 10.1016/j.ajo.2008.09.014 . WorldCat 7 Deeg CA , Hauck SM , Amann B , et al. Equine recurrent uveitis—A spontaneous horse model of uveitis . Ophthalmic Res . 2008 ; 40 ( 3–4 ): 151 – 153 . doi:10.1159/000119867 . Google Scholar Crossref Search ADS PubMed WorldCat 8 Gerding JC , Gilger BC . Prognosis and impact of equine recurrent uveitis . Equine Vet J . 2015 ; 48 ( 3 ): 290 – 298 . doi:10.1111/evj.12451 . Google Scholar Crossref Search ADS PubMed WorldCat 9 Gilger BC , Wilkie DA , Salmon JH , Peel MR . A topical aqueous calcineurin inhibitor for the treatment of naturally occurring keratoconjunctivitis sicca in dogs . Vet Ophthalmol . 2013 c; 16 ( 3 ): 192 – 197 . doi:10.1111/j.1463-5224.2012.01056.x . Google Scholar Crossref Search ADS PubMed WorldCat 10 Kaswan RL , Salisbury MA , Ward DA . Spontaneous canine keratoconjunctivitis sicca. A useful model for human keratoconjunctivitis sicca: Treatment with cyclosporine eye drops . Arch Ophthalmol . 1989 ; 107 ( 8 ): 1210 – 1216 . doi:10.1001/archopht.1989.01070020276038 . Google Scholar Crossref Search ADS PubMed WorldCat 11 Moore CP , McHugh JB , Thorne JG , Phillips TE . Effect of cyclosporine on conjunctival mucin in a canine keratoconjunctivitis sicca model . Invest Ophthalmol Vis Sci . 2001 ; 42 ( 3 ): 653 – 659 . http://www.ncbi.nlm.nih.gov/pubmed/11222523. Google Scholar PubMed WorldCat 12 Murphy CJ , Bentley E , Miller PE , et al. The pharmacologic assessment of a novel lymphocyte function-associated antigen-1 antagonist (SAR 1118) for the treatment of keratoconjunctivitis sicca in dogs . Investig Ophthalmol Vis Sci . 2011 ; 52 ( 6 ): 3174 – 3180 . doi:10.1167/iovs.09-5078 . Google Scholar Crossref Search ADS WorldCat 13 Niederkorn JY , Wang S . Immune privilege of the eye and fetus: Parallel universes? Transplantation . 2005 ; 80 ( 9 ): 1139 – 1144 . doi:10.1097/01.TP.0000173828.78382.4F . Google Scholar Crossref Search ADS PubMed WorldCat 14 Stein-Streilein J . Immune regulation and the eye . Trends Immunol . 2008 ; 29 ( 11 ): 548 – 554 . doi:10.1016/j.it.2008.08.002 . Google Scholar Crossref Search ADS PubMed WorldCat 15 Niederkorn JY . See no evil, hear no evil, do no evil: The lessons of immune privilege . Nat Immunol . 2006 ; 7 ( 4 ): 354 – 359 . doi:10.1038/ni1328 . Google Scholar Crossref Search ADS PubMed WorldCat 16 Caspi RR . Immunological disorders and autoimmunity animal models of autoimmune and immune-mediated uveitis . 2006 ; 3 ( 1 ): 3 – 9 . doi:10.1016/j.ddmod.2006.03.005 . WorldCat 17 Stern ME , Pflugfelder SC . Pathogenesis: Emphasis on dry eye and the role of the lacrimal functional unit in Sjögren’s syndrome. In: Fox R, Fox C, eds. Sjögren’s Syndrome. New York, NY: Springer; 2011 . doi:10.1007/978-1-60327-957-4_13. 18 Carnahan MC , Goldstein DA . Ocular complications of topical, peri-ocular, and systemic corticosteroids . Curr Opin Ophthalmol . 2000 ; 11 : 478 – 483 . doi:10.1097/00055735-200012000-00016 . Google Scholar Crossref Search ADS PubMed WorldCat 19 Fraunfelder FT , Sciubba JJ , Mathers WD . The role of medications in causing dry eye . J Ophthalmol . 2012 ; 2012 : 285851 . doi:10.1155/2012/285851. Google Scholar PubMed WorldCat 20 Sen HN , Vitale S , Gangaputra SS , et al. Periocular corticosteroid injections in uveitis: Effects and complications . Ophthalmology . 2014 ; 121 ( 11 ): 2275 – 2286 . doi:10.1016/j.ophtha.2014.05.021. Google Scholar Crossref Search ADS PubMed WorldCat 21 Uchino M , Schaumberg DA . Dry eye disease: Impact on quality of life and vision . Curr Ophthalmol Rep . 2013 ; 1 ( 2 ): 51 – 57 . doi:10.1007/s40135-013-0009-1. Google Scholar Crossref Search ADS PubMed WorldCat 22 DEWS . The definition and classification of dry eye disease: report of the Definition and Classification of the Dry Eye WorkShop (2007) . Ocul Surf . 2007 ; 5 ( 2 ): 75 – 92 . doi:10.1080/09273940701486803. Crossref Search ADS PubMed WorldCat 23 Lemp MA , Crews LA , Bron AJ , Foulks GN , Sullivan BD . Distribution of aqueous-deficient and evaporative dry eye in a clinic-based patient cohort . Cornea . 2012 ; 31 ( 5 ): 472 – 478 . doi:10.1097/ICO.0b013e318225415a. Google Scholar Crossref Search ADS PubMed WorldCat 24 Barabino S , Chen Y , Chauhan S , Dana R . Ocular surface immunity: Homeostatic mechanisms and their disruption in dry eye disease . Prog Retin Eye Res . 2012 ; 31 ( 3 ): 271 – 285 . doi:10.1016/j.preteyeres.2012.02.003. Google Scholar Crossref Search ADS PubMed WorldCat 25 Stern ME , Schaumburg CS , Dana R , Calonge M , Niederkorn JY , Pflugfelder SC . Autoimmunity at the ocular surface: Pathogenesis and regulation . Mucosal Immunol . 2010 ; 3 ( 5 ): 425 – 442 . doi:10.1038/mi.2010.26. Google Scholar Crossref Search ADS PubMed WorldCat 26 Gayton JL . Etiology, prevalence, and treatment of dry eye disease . Clin Ophthalmol . 2009 ; 3 ( 1 ): 405 – 412 . Google Scholar Crossref Search ADS PubMed WorldCat 27 Avunduk AM , Avunduk MC , Varnell ED , Kaufman HE . The comparison of efficacies of topical corticosteroids and nonsteroidal anti-inflammatory drops on dry eye patients: A clinical and immunocytochemical study . Am J Ophthalmol . 2003 ; 136 ( 4 ): 593 – 602 . doi:10.1016/S0002-9394(03)00326-X. Google Scholar Crossref Search ADS PubMed WorldCat 28 Sall K , Stevenson OD , Mundorf TK , Reis BL . Two multicenter, randomized studies of the efficacy and safety of cyclosporine ophthalmic emulsion in moderate to severe dry eye disease. CsA Phase 3 Study Group . Ophthalmology . 2000 ; 107 : 631 – 639 . doi:S0161642099001761 [pii]. Google Scholar Crossref Search ADS PubMed WorldCat 29 Lekhanont K , Leyngold IM , Suwan-Apichon O , Rangsin R , Chuck RS . Comparison of topical dry eye medications for the treatment of keratoconjunctivitis sicca in a botulinum toxin B-induced mouse model . Cornea . 2007 ; 26 ( 1 ): 84 – 89 . doi:10.1097/01.ico.0000240079.24583.a1. Google Scholar Crossref Search ADS PubMed WorldCat 30 Keating GM . Lifitegrast ophthalmic solution 5%: A review in dry eye disease . Drugs . 2017 ; 77 ( 2 ): 201 – 208 . doi:10.1007/s40265-016-0681-1. Google Scholar Crossref Search ADS PubMed WorldCat 31 Sheppard JD , Torkildsen GL , Lonsdale JD , et al. Semba CPOPUS-1 Study Group.Lifitegrast ophthalmic solution 5.0% for treatment of dry eye disease: Results of the OPUS-1 phase 3 study . Ophthalmology . 2014 ; 121 : 475 – 483 . doi:10.1016/j.ophtha.2013.09.015. Google Scholar Crossref Search ADS PubMed WorldCat 32 Tauber J , Karpecki P , Latkany R , et al. Semba CPOPUS-2 Investigators. Lifitegrast ophthalmic solution 5.0% versus placebo for treatment of dry eye disease results of the randomized phase III OPUS-2 study . Ophthalmology . 2015 ; 122 ( 12 ): 2423 – 2431 . doi:10.1016/j.ophtha.2015.08.001. Google Scholar Crossref Search ADS PubMed WorldCat 33 Barabino S , Chen W , Dana MR . Tear film and ocular surface tests in animal models of dry eye: Uses and limitations . Exp Eye Res . 2004 ; 79 ( 5 ): 613 – 621 . doi:10.1016/j.exer.2004.07.002. Google Scholar Crossref Search ADS PubMed WorldCat 34 Barabino S , Shen LL , Chen L , Rashid S , Rolando M , Dana MR . The controlled-environment chamber: A new mouse model of dry eye . Investig Ophthalmol Vis Sci . 2005 ; 46 ( 8 ): 2766 – 2771 . doi:10.1167/iovs.04-1326. Google Scholar Crossref Search ADS WorldCat 35 Daull P , Feraille L , Barabino S , et al. Efficacy of a new topical cationic emulsion of cyclosporine A on dry eye clinical signs in an experimental mouse model of dry eye . Exp Eye Res . 2016 ; 153 : 159 – 164 . doi:10.1016/j.exer.2016.10.016. Google Scholar Crossref Search ADS PubMed WorldCat 36 Lin Z , Liu X , Zhou T , et al. A mouse dry eye model induced by topical administration of benzalkonium chloride . Mol Vis . 2011 ; 17 : 257 – 264 . Google Scholar PubMed WorldCat 37 Xiong C , Chen D , Liu J , et al. A rabbit dry eye model induced by topical medication of a preservative benzalkonium chloride . Investig Ophthalmol Vis Sci . 2008 ; 49 ( 5 ): 1850 – 1856 . doi:10.1167/iovs.07-0720. Google Scholar Crossref Search ADS WorldCat 38 Zhu L , Shen J , Zhang C , et al. Inflammatory cytokine expression on the ocular surface in the Botulium toxin B induced murine dry eye model . Mol Vis . 2009 ; 15 : 250 – 258 . http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2633460&tool=pmcentrez&rendertype=abstract. Google Scholar PubMed WorldCat 39 Lee MJ , Ko AY , Ko JH , et al. Mesenchymal stem/stromal cells protect the ocular surface by suppressing inflammation in an experimental dry eye . Mol Ther . 2015 ; 23 ( 1 ): 139 – 146 . doi:10.1038/mt.2014.159. Google Scholar Crossref Search ADS PubMed WorldCat 40 Jabs DA , Lee B , Burek CL , Saboori AM , Prendergast RA . Cyclosporine therapy suppresses ocular and lacrimal gland disease in MRL/Mp-lpr/lpr mice . Investig Ophthalmol Vis Sci . 1996 ; 37 ( 2 ): 377 – 383 . WorldCat 41 Song XJ , Li DQ , Farley W , et al. Neurturin-deficient mice develop dry eye and keratoconjunctivitis sicca . Investig Ophthalmol Vis Sci . 2003 ; 44 ( 10 ): 4223 – 4229 . doi:10.1167/iovs.02-1319. Google Scholar Crossref Search ADS WorldCat 42 Schrader S , Mircheff AK , Geerling G . Animal models of dry eye . Dev Ophthalmol . 2008 ; 41 : 298 – 312 . doi:10.1159/000131097. Google Scholar Crossref Search ADS PubMed WorldCat 43 Guzmán M , Keitelman I , Sabbione F , Trevani AS , Giordano MN , Galletti JG . Mucosal tolerance disruption favors disease progression in an extraorbital lacrimal gland excision model of murine dry eye . Exp Eye Res . 2016 ; 151 : 19 – 22 . doi:10.1016/j.exer.2016.07.004. Google Scholar Crossref Search ADS PubMed WorldCat 44 Stevenson W , Chen Y , Lee S , et al. Extraorbital lacrimal gland excision: A reproducible model of severe aqueous tear-deficient dry eye disease . Cornea . 2014 ; 33 ( 12 ): 1336 – 1341 . doi:10.1097/ICO.0000000000000264. Google Scholar Crossref Search ADS PubMed WorldCat 45 Gao J , Morgan G , Tieu D , et al. ICAM-1 expression predisposes ocular tissues to immune-based inflammation in dry eye patients and Sjögrens syndrome-like MRL/lpr mice . Exp Eye Res . 2004 ; 78 ( 4 ): 823 – 835 . doi:10.1016/j.exer.2003.10.024. Google Scholar Crossref Search ADS PubMed WorldCat 46 Jie G , Jiang Q , Rui Z , Yifei Y . Expression of interleukin-17 in autoimmune dacryoadenitis in MRL/lpr mice . Curr Eye Res . 2010 ; 35 ( 10 ): 865 – 871 . doi:10.3109/02713683.2010.497600. Google Scholar Crossref Search ADS PubMed WorldCat 47 Ma X , Zou J , He L , Zhang Y . Dry eye management in a Sjögren’s syndrome mouse model by inhibition of p38-MAPK pathway . Diagn Pathol . 2014 ; 9 ( 1 ): 5 . doi:10.1186/1746-1596-9-5. Google Scholar Crossref Search ADS PubMed WorldCat 48 Fujihara T , Murakami T , Fujita H , Nakamura M , Nakata K . Improvement of corneal barrier function by the P2Y(2) agonist INS365 in a rat dry eye model . Invest Ophthalmol Vis Sci . 2001 ; 42 ( 1 ): 96 – 100 . http://www.ncbi.nlm.nih.gov/pubmed/11133853. Google Scholar PubMed WorldCat 49 Meng ID , Barton ST , Mecum NE , Kurose M . Corneal sensitivity following lacrimal gland excision in the rat . Investig Ophthalmol Vis Sci . 2015 ; 56 ( 5 ): 3347 – 3354 . doi:10.1167/iovs.15-16717. Google Scholar Crossref Search ADS WorldCat 50 Thomas PB , Samant DM , Selvam S , et al. Adeno-associated virus-mediated IL-10 gene transfer suppresses lacrimal gland immunopathology in a rabbit model of autoimmune dacryoadenitis . Investig Ophthalmol Vis Sci . 2010 ; 51 ( 10 ): 5137 – 5144 . doi:10.1167/iovs.10-5423. Google Scholar Crossref Search ADS WorldCat 51 Gilbard JP , Rossi SR , Gray KL , Hanninen LA , Kenyon KR . Tear film osmolarity and ocular surface disease in two rabbit models for keratoconjunctivitis sicca . Investig Ophthalmol Vis Sci . 1988 ; 29 ( 3 ): 374 – 378 . WorldCat 52 Burgalassi S , Panichi L , Chetoni P , Saettone MF , Boldrini E . Development of a simple dry eye model in the albino rabbit and evaluation of some tear substitutes . Ophthalmic Res . 1999 ; 31 ( 3 ): 229 – 235 . doi:10.1159/000055537. Google Scholar Crossref Search ADS PubMed WorldCat 53 Li C , Song Y , Luan S , et al. Research on the stability of a rabbit dry eye model induced by topical application of the preservative benzalkonium chloride . PLoS One . 2012 ; 7 ( 3 ). doi:10.1371/journal.pone.0033688. WorldCat 54 Thomas PB , Zhu Z , Selvam S , et al. Autoimmune dacryoadenitis and keratoconjunctivitis induced in rabbits by subcutaneous injection of autologous lymphocytes activated ex vivo against lacrimal antigens . J Autoimmun . 2008 ; 31 ( 2 ): 116 – 122 . doi:10.1016/j.jaut.2008.04.019. Google Scholar Crossref Search ADS PubMed WorldCat 55 Zhu Z , Stevenson D , Schechter JE , Mircheff AK , Atkinson R , Trousdale MD . Lacrimal histopathology and ocular surface disease in a rabbit model of autoimmune dacryoadenitis . Cornea . 2003 ; 22 ( 1 ): 25 – 32 . doi:10.1097/00003226-200301000-00007. Google Scholar Crossref Search ADS PubMed WorldCat 56 Thomas PB , Samant DM , Zhu Z , et al. Long-term topical cyclosporine treatment improves tear production and reduces keratoconjunctivitis in rabbits with induced autoimmune dacryoadenitis . J Ocul Pharmacol Ther . 2009 ; 25 ( 3 ): 285 – 291 . doi:10.1089/jop.2008.0138. Google Scholar Crossref Search ADS PubMed WorldCat 57 Sanchez RF , Innocent G , Mould J , Billson FM . Canine keratoconjunctivitis sicca: Disease trends in a review of 229 cases . J Small Anim Pract . 2007 ; 48 ( 4 ): 211 – 217 . doi:10.1111/j.1748-5827.2006.00185.x. Google Scholar Crossref Search ADS PubMed WorldCat 58 Izci C , Celik İ , Alkan F , Erol M , Sur E . Clinical and light microscopic studies of the conjunctival tissues of dogs with bilateral keratoconjunctivitis sicca before and after treatment with topical 2% cyclosporine . Biotech Histochem . 2015 ; 90 ( 3 ): 223 – 230 . doi:10.3109/10520295.2014.930177. Google Scholar Crossref Search ADS PubMed WorldCat 59 Kaswan RL , Martin CL , Chapman WL . Keratoconjunctivitis sicca: Histopathologic study of nictitating membrane and lacrimal glands from 28 dogs . Am J Vet Res . 1984 ; 45 ( 1 ): 112 – 118 . Google Scholar PubMed WorldCat 60 Barachetti L , Rampazzo A , Mortellaro CM , Scevola S , Gilger BC . Use of episcleral cyclosporine implants in dogs with keratoconjunctivitis sicca: Pilot study . Vet Ophthalmol . 2015 ; 18 ( 3 ): 234 – 241 . doi:10.1111/vop.12173. Google Scholar Crossref Search ADS PubMed WorldCat 61 Berdoulay A , English RV , Nadelstein B . Effect of topical 0.02% tacrolimus aqueous suspension on tear production in dogs with keratoconjunctivitis sicca . Vet Ophthalmol . 2005 ; 8 ( 4 ): 225 – 232 . doi:10.1111/j.1463-5224.2005.00390.x. Google Scholar Crossref Search ADS PubMed WorldCat 62 Siddique SS , Suelves AM , Baheti U , Foster CS . Glaucoma and uveitis . Surv Ophthalmol . 2013 ; 58 ( 1 ): 1 – 10 . doi:10.1016/j.survophthal.2012.04.006. Google Scholar Crossref Search ADS PubMed WorldCat 63 Jabs DA . Epidemiology of uveitis . Ophthalmic Epidemiol . 2008 ; 15 ( 5 ): 283 – 284 . doi:10.1080/09286580802478724. Google Scholar Crossref Search ADS PubMed WorldCat 64 Generali E , Cantarini L , Selmi C . Ocular involvement in systemic autoimmune diseases . Clin Rev Allergy Immunol . 2015 ; 49 ( 3 ): 263 – 270 . doi:10.1007/s12016-015-8518-3. Google Scholar Crossref Search ADS PubMed WorldCat 65 Levitt AE , McManus KT , McClellan AL , Davis JL , Goldhardt R , Galor A . Ocular inflammation in the setting of concomitant systemic autoimmune conditions in an older male population . Cornea . 2015 ; 34 ( 7 ): 762 – 767 . doi:10.1097/ICO.0000000000000437. Google Scholar Crossref Search ADS PubMed WorldCat 66 Caspi RR . Understanding autoimmune uveitis through animal models the Friedenwald lecture . Investig Opthalmol Vis Sci . 2011 ; 52 ( 3 ): 1873 . doi:10.1167/iovs.10-6909. Google Scholar Crossref Search ADS WorldCat 67 Gilger BC , Yang P , Salmon JH , Jaffe GJ , Allen JB . Expression of a chemokine by ciliary body epithelium in horses with naturally occurring recurrent uveitis and in cultured ciliary body epithelial cells . Am J Vet Res . 2002 ; 63 ( 7 ): 942 – 947 . doi:10.2460/ajvr.2002.63.942. Google Scholar Crossref Search ADS PubMed WorldCat 68 Deeg CA . Ocular immunology in equine recurrent uveitis . Vet Ophthalmol . 2008 ; 11 ( Suppl 1 ): 61 – 65 . doi:10.1111/j.1463-5224.2008.00625.x. Google Scholar Crossref Search ADS PubMed WorldCat 69 Deeg CA , Kaspers B , Gerhards H , Thurau SR , Wollanke B , Wildner G . Immune responses to retinal autoantigens and peptides in equine recurrent uveitis . Investig Ophthalmol Vis Sci . 2001 ; 42 ( 2 ): 393 – 398 . WorldCat 70 Deeg CA , Pompetzki D , Raith AJ , et al. Identification and functional validation of novel autoantigens in equine uveitis . Mol Cell Proteomics . 2006 b; 5 ( 8 ): 1462 – 1470 . doi:10.1074/mcp.M500352-MCP200. Google Scholar Crossref Search ADS PubMed WorldCat 71 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. Google Scholar Crossref Search ADS PubMed WorldCat 72 Deeg CA , Amann B , Raith AJ , Kaspers B . Inter- and intramolecular epitope spreading in equine recurrent uveitis . Investig Ophthalmol Vis Sci . 2006 a; 47 ( 2 ): 652 – 656 . doi:10.1167/iovs.05-0789. Google Scholar Crossref Search ADS WorldCat 73 Silver P , Horai R , Chen J , et al. Retina-specific T regulatory cells bring about resolution and maintain remission of autoimmune uveitis . J Immunol . 2015 ; 194 ( 7 ): 3011 – 3019 . doi:10.4049/jimmunol.1402650. Google Scholar Crossref Search ADS PubMed WorldCat 74 Cousins SW , Guss RB , Howes EL , Rosenbaum JT . Endotoxin-induced uveitis in the rat: Observations on altered vascular permeability, clinical findings, and histology . Exp Eye Res . 1984 ; 39 : 665 – 676 . doi:10.1016/0014-4835(84)90065-4. Google Scholar Crossref Search ADS PubMed WorldCat 75 Li Q , Peng B , Whitcup SM , Jang SU , Chan C-C . Endotoxin induced uveitis in the mouse: Susceptibility and genetic control . Exp Eye Res . 1995 ; 61 ( 5 ): 629 – 632 . doi:10.1016/S0014-4835(05)80056-9. Google Scholar Crossref Search ADS PubMed WorldCat 76 Agarwal RK , Silver PB , Caspi RR . Rodent models of experimental autoimmune uveitis . Methods Mol Biol . 2012 ; 900 : 443 – 469 . doi:10.1007/978-1-60761-720-4_22. Google Scholar Crossref Search ADS PubMed WorldCat 77 Chen J , Qian H , Horai R , Chan CC , Falick Y , Caspi RR . Comparative analysis of induced vs. spontaneous models of autoimmune uveitis targeting the interphotoreceptor retinoid binding protein . PLoS One . 2013 ; 8 ( 8 ). doi:10.1371/journal.pone.0072161. WorldCat 78 Chen J , Qian H , Horai R , Chan C-C , Caspi RR . Mouse models of experimental autoimmune uveitis: Comparative analysis of adjuvant-induced vs spontaneous models of uveitis . Curr Mol Med . 2015 ; 15 ( 6 ): 550 – 557 . http://www.ncbi.nlm.nih.gov/pubmed/26238369. Google Scholar Crossref Search ADS PubMed WorldCat 79 Shao H , Liao T , Ke Y , Shi H , Kaplan HJ , Sun D . Severe chronic experimental autoimmune uveitis (EAU) of the C57BL/6 mouse induced by adoptive transfer of IRBP1-20-specific T cells . Exp Eye Res . 2006 ; 82 ( 2 ): 323 – 331 . doi:10.1016/j.exer.2005.07.008. Google Scholar Crossref Search ADS PubMed WorldCat 80 Fleisher LN , Ferrell JB , McGahan MC . Ocular inflammatory effects of intravitreally injected tumor necrosis factor-alpha and endotoxin . Inflammation . 1990 ; 14 ( 3 ): 325 – 335 . doi:10.1007/BF00915816. Google Scholar Crossref Search ADS PubMed WorldCat 81 Goldblum D , Fausch K , Frueh BE , Theurillat R , Thormann W , Zimmerli S . Ocular penetration of caspofungin in a rabbit uveitis model . Graefe’s Arch Clin Exp Ophthalmol . 2007 ; 245 ( 6 ): 825 – 833 . doi:10.1007/s00417-006-0460-x. Google Scholar Crossref Search ADS WorldCat 82 Ang M , Ng X , Wong C , et al. Evaluation of a prednisolone acetate-loaded subconjunctival implant for the treatment of recurrent uveitis in a rabbit model . PLoS One . 2014 ; 9 ( 5 ). doi:10.1371/journal.pone.0097555. WorldCat 83 Edmond M , Yuan A , Bell BA , et al. The feasibility of spectral-domain optical coherence tomography grading of anterior chamber inflammation in a rabbit model of anterior uveitis . Investig Ophthalmol Vis Sci . 2016 ; 57 ( 9 ): OCT184 – OCT188 . doi:10.1167/iovs.15-18883. Google Scholar Crossref Search ADS WorldCat 84 Ghosn CR , Li Y , Orilla WC , et al. Treatment of experimental anterior and intermediate uveitis by a dexamethasone intravitreal implant . Invest Ophthalmol Vis Sci . 2011 ; 52 ( 6 ): 2917 – 2923 . doi:10.1167/iovs.10-5939. Google Scholar Crossref Search ADS PubMed WorldCat 85 Neumann R , Jabbur NS , Vickers F , Foster SC . Topical diclofenac sodium, dexamethasone and placebo compared in a model of immunogenic uveitis in rabbits . Ocul Immunol Inflamm . 1993 ; 1 ( 1–2 ): 87 – 98 . doi:10.3109/09273949309086543. Google Scholar Crossref Search ADS PubMed WorldCat 86 Tilden ME , Boney RS , Goldenberg MM , Rosenbaum JT . The effects of topical S(+)-ibuprofen on interleukin-1 induced ocular inflammation in a rabbit model . J Ocul Pharmacol . 1990 ; 6 ( 2 ): 131 – 135 . http://www.ncbi.nlm.nih.gov/pubmed/2394932. Google Scholar Crossref Search ADS PubMed WorldCat 87 Gilger BC , Abarca EM , Salmon JH , Patel S . Treatment of acute posterior uveitis in a porcine model by injection of triamcinolone acetonide into the suprachoroidal space using microneedles . Invest Ophthalmol Vis Sci . 2013 b; 54 ( 4 ): 2483 – 2492 . doi:10.1167/iovs.13-11747. Google Scholar Crossref Search ADS PubMed WorldCat 88 Gerding JC , Gilger BC . Prognosis and impact of equine recurrent uveitis . Equine Vet J . 2016 ; 48 ( 3 ): 290 – 298 . doi:10.1111/evj.12451. Google Scholar Crossref Search ADS PubMed WorldCat 89 Altinsoy A , Dileköz E , Kul O , et al. A cannabinoid ligand, anandamide, exacerbates endotoxin-induced uveitis in rabbits . J Ocul Pharmacol Ther . 2011 ; 27 ( 6 ): 545 – 552 . doi:10.1089/jop.2011.0049. Google Scholar Crossref Search ADS PubMed WorldCat 90 Wacker WB . Autoimmune uveitis (Choroiditis) in the guinea pig sensitized with homologous uvea and its differentiation from that following sensitization with homologous retina . Int Arch Allergy Immunol . 1972 ; 43 ( 1 ): 39 – 52 . doi:10.1159/000230819. Google Scholar Crossref Search ADS WorldCat 91 Allen JB , McGahan MC , Ferrell JB , Adler KB , Fleisher LN . Nitric oxide synthase inhibitors exert differential time-dependent effects on LPS-induced uveitis . Exp Eye Res . 1996 ; 62 : 21 – 28 . Google Scholar Crossref Search ADS PubMed WorldCat 92 Nussenblatt RB , Calogero D , Buchen SY , Leder HA , Goodkin M , Eydelman MB . Rabbit intraocular reactivity to endotoxin measured by slit-lamp biomicroscopy and laser flare photometry . Ophthalmology . 2012 ; 119 ( 7 ). doi:10.1016/j.ophtha.2012.04.004. WorldCat 93 Rafie F , Javadzadeh Y , Javadzadeh AR , et al. In vivo evaluation of novel nanoparticles containing dexamethasone for ocular drug delivery on rabbit eye . Curr Eye Res . 2010 ; 35 ( 12 ): 1081 – 1089 . doi:10.3109/02713683.2010.508867. Google Scholar Crossref Search ADS PubMed WorldCat 94 Mruthyunjaya P . Efficacy of low-release-rate fluocinolone acetonide intravitreal implants to treat experimental uveitis . Arch Ophthalmol . 2006 ; 124 ( 7 ): 1012 . doi:10.1001/archopht.124.7.1012. Google Scholar Crossref Search ADS PubMed WorldCat 95 Jaffe GJ , Yang CS , Wang XC , Cousins SW , Gallemore RP , Ashton P . Intravitreal sustained-release cyclosporine in the treatment of experimental uveitis . Ophthalmology . 1998 ; 105 ( 1 ): 46 – 56 . doi:10.1016/S0161-6420(98)91176-9. Google Scholar Crossref Search ADS PubMed WorldCat 96 Jaffe GJ , Martin D , Callanan D , Pearson PA , Levy B , Comstock T . Fluocinolone acetonide implant (retisert) for noninfectious posterior uveitis. Thirty-four-week results of a multicenter randomized clinical study . Ophthalmology . 2006 ; 113 ( 6 ): 1020 – 1027 . doi:10.1016/j.ophtha.2006.02.021. Google Scholar Crossref Search ADS PubMed WorldCat 97 Gilger BC , Abarca E , Salmon JH . Selection of appropriate animal models in ocular research: Ocular anatomy and physiology of common animal models. In: Brian C. Gilger, ed. Ocular Pharmacology and Toxicology. Springer Scientific; 2014. ISBN 978-1-62703-744-0 [2013a]. 98 Wollanke B , Brem S , Meyer P , et al. Prophylaxis of equine recurrent uveitis (ERU): First results with a leptospiral vaccine in horses . Pferdeheilkunde . 2004 ; 20 ( 5 ): 447 – 454 . Google Scholar Crossref Search ADS WorldCat 99 Kleinwort KJH , Amann B , Hauck SM , Feederle R , Sekundo W , Deeg CA . Immunological characterization of intraocular lymphoid follicles in a spontaneous recurrent uveitis model . Investig Ophthalmol Vis Sci . 2016 ; 57 ( 10 ): 4504 – 4511 . doi:10.1167/iovs.16-19787. Google Scholar Crossref Search ADS WorldCat 100 Deeg CA . A proteomic approach for studying the pathogenesis of spontaneous equine recurrent uveitis (ERU) . Vet Immunol Immunopathol . 2009 ; 128 ( 1–3 ): 132 – 136 . doi:10.1016/j.vetimm.2008.10.302. Google Scholar Crossref Search ADS PubMed WorldCat 101 Deeg CA , Raith AJ , Amann B , et al. CRALBP is a highly prevalent autoantigen for human autoimmune uveitis . Clin Dev Immunol . 2007 ; 2007 . doi:10.1155/2007/39245. WorldCat 102 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 ). doi :10.1371/journal.pone.0091684. WorldCat 103 Zipplies JK , Hauck SM , Schoeffmann S , et al. Serum PEDF levels are decreased in a spontaneous animal model for human autoimmune uveitis . J Proteome Res . 2009 ; 8 ( 2 ): 992 – 998 . doi:10.1021/pr800694y. Google Scholar Crossref Search ADS PubMed WorldCat 104 Lucchesi PMA , Parma AE . A DNA fragment of Leptospira interrogans encodes a protein which shares epitopes with equine cornea . Vet Immunol Immunopathol . 1999 ; 71 ( 3–4 ): 173 – 179 . doi:10.1016/S0165-2427(99)00084-7. Google Scholar Crossref Search ADS PubMed WorldCat 105 Lucchesi PMA , Parma AE , Arroyo GH . Serovar distribution of a DNA sequence involved in the antigenic relationship between Leptospira and equine cornea . BMC Microbiol . 2002 ; 2 : 3 . doi:10.1186/1471-2180-2-3. Google Scholar Crossref Search ADS PubMed WorldCat 106 Verma A , Artiushin S , Matsunaga J , Haake DA , Timoney JF . LruA and LruB, novel lipoproteins of pathogenic Leptospira interrogans associated with equine recurrent uveitis . Infect Immun . 2005 ; 73 ( 11 ): 7259 – 7266 . doi:10.1128/IAI.73.11.7259-7266.2005. Google Scholar Crossref Search ADS PubMed WorldCat 107 Verma A , Kumar P , Babb K , Timoney JF , Stevenson B . Cross-reactivity of antibodies against leptospiral recurrent uveitis-associated proteins A and B (LruA and LruB) with eye proteins . PLoS Negl Trop Dis . 2010 ; 4 ( 8 ). doi :10.1371/journal.pntd.0000778. WorldCat 108 Verma A , Rathinam SR , Priya CG , Muthukkaruppan VR , Stevenson B , Timoney JF . LruA and LruB antibodies in sera of humans with leptospiral uveitis . Clin Vaccine Immunol . 2008 ; 15 ( 6 ): 1019 – 1023 . doi:10.1128/CVI.00203-07. Google Scholar Crossref Search ADS PubMed WorldCat 109 Gilger BC , Malok E , Stewart T , et al. Effect of an intravitreal cyclosporine implant on experimental uveitis in horses . Vet Immunol Immunopathol . 2000 ; 76 ( 3–4 ): 239 – 255 . doi:10.1016/S0165-2427(00)00219-1. Google Scholar Crossref Search ADS PubMed WorldCat 110 Gilger BC , Wilkie DA , Davidson MG , Allen JB . Use of an intravitreal sustained-release cyclosporine delivery device for treatment of equine recurrent uveitis . Am J Vet Res . 2001 ; 62 ( 12 ): 1892 – 1896 . doi:10.2460/ajvr.2001.62.1892. Google Scholar Crossref Search ADS PubMed WorldCat 111 Gilger BC , Salmon JH , Wilkie DA , et al. A novel bioerodible deep scleral lamellar cyclosporine implant for uveitis . Invest Ophthalmol Vis Sci . 2006 ; 47 ( 6 ): 2596 – 2605 . doi:10.1167/iovs.05-1540. Google Scholar Crossref Search ADS PubMed WorldCat 112 Gilger BC , Wilkie DA , Clode AB , et al. Long-term outcome after implantation of a suprachoroidal cyclosporine drug delivery device in horses with recurrent uveitis . Vet Ophthalmol . 2010 ; 13 ( 5 ): 294 – 300 . Google Scholar Crossref Search ADS PubMed WorldCat 113 Moisseiev E , Loewenstein A , Yiu G . The suprachoroidal space: From potential space to a space with potential . Clin Ophthalmol . 2016 ; 10 : 173 – 178 . doi:10.2147/OPTH.S89784. Google Scholar Crossref Search ADS PubMed WorldCat 114 Hirsch ML , Conatser LM , Smith SM , et al. AAV vector-meditated expression of HLA-G reduces injury-induced corneal vascularization and immune cell infiltration . Sci Rep . 2017 ; 7 ( 1 ): 17840 . Google Scholar Crossref Search ADS PubMed WorldCat 115 Villatoro A , Fernández V . Use of adipose-derived mesenchymal stem cells in keratoconjunctivitis sicca in a canine model . Biomed Res Int . 2014 ; 2015 ( 2015 ): 10 . doi:10.1155/2015/527926. WorldCat © The Author(s) 2018. Published by Oxford University Press on behalf of the National Academy of Sciences. All rights reserved. For permissions, please email: journals.permissions@oup.com This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Journal

ILAR JournalOxford University Press

Published: Dec 31, 2018

There are no references for this article.