Elsevier

Experimental Eye Research

Volume 105, December 2012, Pages 43-52
Experimental Eye Research

Everolimus improves experimental autoimmune uveoretinitis

https://doi.org/10.1016/j.exer.2012.09.003Get rights and content

Abstract

The efficacy and action mechanism of everolimus in the treatment of experimental autoimmune uveoretinitis (EAU) was analyzed. Disease was induced in B10.RIII mice by immunization with human interphotoreceptor-retinoid-binding protein peptide 161-180 (hIRBPp161-180). Everolimus was administered by oral gavage (5 mg/kg/d) beginning either two days before or 14 days after immunization. Everolimus significantly reduced the histopathological uveitis score compared to sham-treated mice. To examine the effect on the antigen-specific immune response, proliferation ([3H]-thymidine test) and delayed-type hypersensitivity (DTH) response were measured. Furthermore, content of T-helper-1, -2, and -17 cytokines were analyzed intraocularly (Bead Array) and in cell culture supernatants from splenocytes (sandwich ELISA). To study the effect on the humoral immune response the presence of antigen-specific serum antibodies was tested (indirect ELISA). The DTH, the humoral immune response, the proliferation of splenocytes and the intraocular Th1, Th2, Th17 cytokine content and in vitro production of Th1 and Th17 cytokines were impaired after everolimus treatment. The study of CD4+CD25+FoxP3+ regulatory T cells (Treg) in peripheral blood, draining lymph nodes, and spleen by flow cytometry showed an increased number of splenic Treg in mice of the everolimus therapy group. Furthermore the Treg of these mice had a higher suppressive capacity than cells from sham-treated mice. These results indicate that the immunosuppressive effect of everolimus on EAU was associated with the suppression of pathogenic effector responses and induction of regulatory T cells.

Highlights

► Experimental autoimmune uveoretinitis (EAU) was induced in B10.RIII mice. ► Mice were prophylactic- and therapeutically treated with everolimus. ► Incidence and severity were reduced under prophylactic and therapeutic treatment. ► The uveitogenic immune response was impaired in both treatment groups. ► The number and suppressive capacity of splenic Treg increased under therapy.

Introduction

Experimental autoimmune uveoretinitis (EAU) is a T cell-mediated autoimmune disease (Atalla et al., 1990) which serves as a model for noninfectious posterior uveitis in humans (Forrester et al., 1990). EAU can be induced in rats and mice by injecting a retinal antigen such as retinal S-antigen or interphotoreceptor retinoid-binding protein (IRBP) (Caspi et al., 1990, 1988; Chan et al., 1990; Mochizuki et al., 1985; Xu et al., 2000). Here, the ocular inflammation peaks 14 days after immunization (Abbas et al., 2007; Caspi et al., 1986; Jiang et al., 1999) and is characterized by a massive leukocyte infiltration (Caspi et al., 1988). The infiltrate consists mainly of CD4+ T cells, which mediate disease initiation. With disease exacerbation, mainly activated macrophages and granulocytes invade the ocular tissue (Caspi et al., 1993). Further histological features of EAU are vasculitis, retinal granuloma formation, folding and detachment of the retina, and destruction of the photoreceptors (Caspi et al., 1988).

The antigens responsible for the development of human uveitis have not yet been identified. Nevertheless, as patients with posterior uveitis often show a strong cellular immune response to the retinal antigens that are used for EAU induction in experimental animal models, the observations from rodent EAU models can possibly be applied to human disease (Chan et al., 1990).

Patients with endogenous, noninfectious uveitis are currently first treated with topical or systemic corticosteroids. If the disease is refractory to this treatment, additional immunosuppressive agents are used (e.g. antimetabolites, T-cell inhibitors, and alkylating agents) (Jabs et al., 2000). The mTor inhibitor rapamycin has demonstrated efficacy in preventing intraocular inflammation from developing in a rat EAU model (Roberge et al., 1993). Inhibition of mTor blocks the growth factor-driven signal transduction in T- and B-cells, resulting in cell cycle arrest in G1 phase (Schuler et al., 1997; Schuurman et al., 1997). In addition to suppressing effector T cells, rapamycin induces the de novo generation of Treg (Battaglia et al., 2005; Valmori et al., 2006). This could be an advantage for the treatment of autoimmune diseases with known malfunction or dysregulation of Treg (Haxhinasto et al., 2008; Kang et al., 2008; Sauer et al., 2008; Zeiser et al., 2008).

Everolimus (mTor inhibitor), a derivate of rapamycin, has a higher bioavailability, reaches the steady-state level faster without a loading dose, and has a shorter half-life than rapamycin (Schuler et al., 1997). Its immunosuppressive efficacy has also been demonstrated in transplant studies, showing prolonged allograft survival in primates after lung transplantation (Hausen et al., 2000a, 2000b; Schuurman et al., 2000; Schuurman et al., 1998) and in clinical studies for heart transplantation (Ross et al. 2010). Therefore, we hypothesized that everolimus may also be suitable for treating T-cell-mediated uveitis.

In the present study we analyzed the efficacy of everolimus for the prevention and treatment of EAU in mice. Furthermore, the inhibitory capacity of everolimus therapy on uveitogenic effector cells and the induction of CD4+CD25+FoxP3+ T cells were tested.

Section snippets

Mice

B10.RIII mice 10–12 weeks of age were purchased from Jackson Laboratory (Maine, USA). Mice were kept in accordance with the ARVO statement for the Use of Animals in Ophthalmic and Vision Research and in concordance with the guidelines of the German animal care and use committee.

Reagents

The hIRBPp161-180 (SGIPYIISYLHPGNTILHVD) was purchased from EMC Microcollection (Tuebingen, Germany); Bordetella pertussis toxin and complete Freund's adjuvant were obtained from Sigma–Aldrich (Steinheim, Germany).

Everolimus ameliorates EAU in mice

To induce EAU, B10.RIII mice were immunized with hIRBPp160-180. The first clinical signs could be observed in fundoscopic examinations 9–10 days after immunization (data not shown). Ocular inflammation with different severity was detected in histopathological sections of mice eyes 21 days after model induction. In comparison to naïve mice, typical signs of the posterior uveitis were infiltration of leukocytes in retina (Fig. 1B) and vitreous, vasculitis, granuloma formation, retinal detachment,

Discussion

To our knowledge, neither the efficacy of everolimus nor its effect on regulatory T cells has been previously evaluated for the hIRBPp161-180-induced EAU model in mice.

The present data show that everolimus can effectively prevent and treat ocular inflammation in a mouse model of EAU. Animals that received everolimus prophylaxis or treatment showed a notable decrease in histological incidence and severity of intraocular inflammation as compared to sham treatment.

Splenocytes harvested from

Conclusion

The ability of everolimus to prevent and treat the intraocular inflammation occurring in EAU by downregulating proinflammatory Th1 and Th17 effector responses, combined with its ability to induce Treg, and therefore to improve the peripheral tolerance mechanisms suggests that everolimus may be a useful therapeutic option for noninfectious intraocular inflammation.

Author disclosure information

M Hennig: None; D Bauer: None; S Wasmuth: None; M Busch: None; K Walscheid: None; S Thanos: None; A Heiligenhaus: Novartis.

Commercial interest

This study was supported by research grants from DFG Ba2248/1-2. Novartis supported this work by providing everolimus for the experimental study. Novartis did not participate in the study design, the collection, statistical analysis, or interpretation of data or in the preparation, review, or approval of the present manuscript.

Acknowledgments

We thank Dipl.-Ing. Klaus Lennartz (University of Duisburg-Essen) for cell sorting, and PD Dr. Karin Loser (University of Muenster) for possibility of using the FACSCalibur and radioactive lab.

References (51)

  • R. Zeiser et al.

    Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells

    Blood

    (2008)
  • R.K. Agarwal et al.

    Rodent models of experimental autoimmune uveitis

    Methods in Molecular Medicine

    (2004)
  • R.K. Agarwal et al.

    Pregnancy ameliorates induction and expression of experimental autoimmune uveitis

    The Journal of Immunology

    (1999)
  • A. Amadi-Obi et al.

    T(H)17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1

    Nature Medicine

    (2007)
  • L. Atalla et al.

    Inhibition of autoimmune uveitis by anti-CD4 antibody

    Investigative Ophthalmology & Visual Science

    (1990)
  • M. Battaglia et al.

    Rapamycin and interleukin-10 treatment induces T regulatory type 1 cells that mediate antigen-specific transplantation tolerance

    Diabetes

    (2006)
  • M. Battaglia et al.

    Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients

    The Journal of Immunology

    (2006)
  • F. Behrens et al.

    Imbalance in distribution of functional autologous regulatory T cells in rheumatoid arthritis

    Annals of the Rheumatic Diseases

    (2007)
  • R.R. Caspi et al.

    The mouse as a model of experimental autoimmune uveoretinitis (EAU)

    Current Eye Research

    (1990)
  • R.R. Caspi et al.

    A new model of autoimmune disease. Experimental autoimmune uveoretinitis induced in mice with two different retinal antigens

    The Journal of Immunology

    (1988)
  • R.R. Caspi et al.

    T cell lines mediating experimental autoimmune uveoretinitis (EAU) in the rat

    The Journal of Immunology

    (1986)
  • S.K. Chauhan et al.

    Autoimmunity in dry eye is due to resistance of Th17 to Treg suppression

    The Journal of Immunology

    (2009)
  • L. Chen et al.

    Decreased frequency and diminished function of CD4+CD25 high regulatory T cells are associated with active uveitis in patients with Vogt-Koyanagi-Harada syndrome

    Investigative Ophthalmology & Visual Science

    (2008)
  • J.J. Coenen et al.

    Rapamycin, not cyclosporine, permits thymic generation and peripheral preservation of CD4+ CD25+ FoxP3+ T cells

    Bone Marrow Transplantation

    (2007)
  • J.V. Forrester et al.

    Comparison of clinical and experimental uveitis

    Current Eye Research

    (1990)
  • Cited by (0)

    1

    Equal contribution.

    View full text