Beneficial role of rapamycin in experimental autoimmune myositis

Nicolas Prevel, Yves Allenbach, David Klatzmann, Benoit Salomon, Olivier Benveniste, Nicolas Prevel, Yves Allenbach, David Klatzmann, Benoit Salomon, Olivier Benveniste

Abstract

Introduction: We developed an experimental autoimmune myositis (EAM) mouse model of polymyositis where we outlined the role of regulatory T (Treg) cells. Rapamycin, this immunosuppressant drug used to prevent rejection in organ transplantation, is known to spare Treg. Our aim was to test the efficacy of rapamycin in vivo in this EAM model and to investigate the effects of the drug on different immune cell sub-populations.

Methods: EAM is induced by 3 injections of myosin emulsified in CFA. Mice received rapamycin during 25 days starting one day before myosin immunization (preventive treatment), or during 10 days following the last myosin immunization (curative treatment).

Results: Under preventive or curative treatment, an increase of muscle strength was observed with a parallel decrease of muscle inflammation, both being well correlated (R(2) = -0.645, p<0.0001). Rapamycin induced a general decrease in muscle of CD4 and CD8 T cells in lymphoid tissues, but spared B cells. Among T cells, the frequency of Treg was increased in rapamycin treated mice in draining lymph nodes (16.9 ± 2.2% vs. 9.3 ± 1.4%, p<0.001), which were mostly activated regulatory T cells (CD62L(low)CD44(high): 58.1 ± 5.78% vs. 33.1 ± 7%, treated vs. untreated, p<0.001). In rapamycin treated mice, inhibition of proliferation (Ki-67(+)) is more important in effector T cells compared to Tregs cells (p<0.05). Furthermore, during preventive treatment, rapamycin increased the levels of KLF2 transcript in CD44(low) CD62L(high) naive T cell and in CD62L(low) CD44(high) activated T cell.

Conclusions: Rapamycin showed efficacy both as curative and preventive treatment in our murine model of experimental myositis, in which it induced an increase of muscle strength with a parallel decrease in muscle inflammation. Rapamycin administration was also associated with a decrease in the frequency of effector T cells, an increase in Tregs, and, when administered as preventive treatment, an upregulation of KFL2 in naive and activated T cells.

Trial registration: ClinicalTrials.gov NCT00525889.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Preventive administration of rapamycin permits…
Figure 1. Preventive administration of rapamycin permits to decrease severity of EAM.
A: strength (time to fall) improvement in rapamycin-treated mice. B: gastrocnemius, quadriceps, and triceps muscle inflammatory infiltrates evaluated by histological grading after hematoxylin-eosin staining as illustrated in C. C: the first two upper images represent a gastrocnemius section of an EAM mouse untreated with a histological score of 4 (HE, ×20). The middle picture also shows an invaded/tunnelized fiber (arrow, HE, ×40). The third image panel represents a gastrocnemius section of a EAM mouse treated with 3 mg/kg/day of rapamycin, displaying a histological score of 1 (HE, ×20). D: correlation between muscle strength (time to fall) and histological grade of inflammation.
Figure 2. Rapamycin treatment induced a T…
Figure 2. Rapamycin treatment induced a T cells lymphopenia sparing B cells.
A: absolute lymphocyte number in draining lymph nodes, spleen, and non-draining lymph nodes is decreased in rapamycin-treated mice compare to controls in a dose-dependent fashion. B: quantification of the different sub-populations of lymphocytes: B lymphocytes (B, CD19+B220+), T (CD3+), T CD4+ (CD3+CD4+) and T CD8+ (CD3+CD8+) cells. * p<0.05, *** p<0.001. White histogram bars: control mice, black histogram bars: rapamycin treated mice (3 mg/kg/day). C: percent of T or B lymphocytes in draining lymph nodes. D: Representative dot plot of flow cytometry analysis of draining lymph nodes for percentage of pre-activated CD4+ T cell (CD3+CD4+FoxP3−CD69+) and percentage of pre-activated CD4+ T cell (CD3+CD4+FoxP3−CD69+) in controls and rapamycin (3 mg/kg/day) treated mice.
Figure 3. Effect of rapamycin on Treg…
Figure 3. Effect of rapamycin on Treg cells.
Representative dot plot of flow cytometry analysis of draining lymph nodes for percent of Treg (CD4+CD25+FoxP3+) in CD4+ (A) and in CD4+CD44high (B) in controls and rapamycin (3 mg/kg/day) treated mice. Percent of Treg (CD4+CD25+FoxP3+) in CD4+ (A, right) and in CD4+CD44high (B, right) in controls and rapamycin (3 mg/kg/day) treated mice. C: Histogram representative of the difference in activation of Treg (CD62Llow cell) and naive Treg (CD62Lhigh) in controls and rapamycin (3 mg/kg/day) treated mice. D: Suppressive test. Horizontal lines indicate means. Suppressive activity of sorted CD4+CD25+ T cells (Treg) from controls (white bars) and rapamycin (3 mg/kg/day, black bars) mice on the proliferation of autologous CD4+CD25− T cells (responders) stimulated with irradiated (15 Gy) splenocytes. Proliferation of responder cells (Teff) was measured by 3H-Tymidine incorporation (counts per minute, cpm). Results are indicated as percentage inhibition (±SD) compared to a max (ratio 1∶0). Different Teff∶Treg ratios were tested. E: Percent of Ki-67positive cells (i.e. proliferative cells) in activated effector (aT) and activated regulatory (aTreg) T cells in controls and rapamycin (3 mg/kg/day) treated mice.
Figure 4. Rapamycin treatment did not alter…
Figure 4. Rapamycin treatment did not alter muscle infiltrates composition.
A: Gastrocnemius muscle fiber membranes are stained with an anti-laminin antibody (green). Nuclei were counterstained with DAPI (blue). Cells of muscular inflammatory infiltrates are stained with anti-CD4 (yellow) and with anti-FoxP3 (red) antibodies. B: Percent of Tregs (CD4+FoxP3+) in muscle inflammatory infiltrates.
Figure 5. Change in KLF2 pathway induced…
Figure 5. Change in KLF2 pathway induced by rapamycin treatment.
A: Histogram plot of the shift of CCR7 expression in controls and rapamycin-treated (3 mg/kg/day) mice showing naive T cell subset (nT:CD62LhighCD44low). B: Shift of CCR7 expression in controls and rapamycin-treated (3 mg/kg/day) mice in naive T cells (nT:CD62LhighCD44low) an activated T cells (eT:CD62LlowCD44high). C: Q-RT-PCR of KLF2 in naive T cells (nT:CD62highCD44low), activated T cells (aT: CD62low CD44high), naive regulatory T cells (nTreg:CD62high), activated Treg cells (aTreg: CD62low) in controls and rapamycin treated (3 mg/kg/day) mice.
Figure 6. Beneficial effect of rapamycin (3…
Figure 6. Beneficial effect of rapamycin (3 mg/kg/day) in curative treatment of EAM.
Rapamycin (or water for control animals) was given orally for 10 days at a dose of 3 mg/kg. A: strength of mice evaluated by inverted screen test (time to fall in seconds). B: Gastrocnemius muscle inflammatory infiltrates evaluated by histological grading after haematoxylin-eosin staining. C: Percent of Treg cells (CD3+CD4+CD25+FoxP3+) in draining lymph nodes from controls and rapamycin-treated (3 mg/kg/day) mice.

References

    1. Banker BQ (1975) Dermatomyostis of childhood, ultrastructural alteratious of muscle and intramuscular blood vessels. J Neuropathol Exp Neurol 34: 46–75.
    1. Dalakas MC (2007) Autoimmune inflammatory myopathies. Handb Clin Neurol 86: 273–301.
    1. Troyanov Y, Targoff IN, Tremblay JL, Goulet JR, Raymond Y, et al. (2005) Novel classification of idiopathic inflammatory myopathies based on overlap syndrome features and autoantibodies: analysis of 100 French Canadian patients. Medicine (Baltimore) 84: 231–249.
    1. van der Meulen MF, Bronner IM, Hoogendijk JE, Burger H, van Venrooij WJ, et al. (2003) Polymyositis: an overdiagnosed entity. Neurology 61: 316–321.
    1. Dalakas MC (2012) Pathogenesis and therapies of immune-mediated myopathies. Autoimmun Rev 11: 203–206.
    1. Allenbach Y, Solly S, Gregoire S, Dubourg O, Salomon B, et al. (2009) Role of regulatory T cells in a new mouse model of experimental autoimmune myositis. Am J Pathol 174: 989–998.
    1. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330–336.
    1. Gambineri E, Torgerson TR, Ochs HD (2003) Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T-cell homeostasis. Curr Opin Rheumatol 15: 430–435.
    1. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057–1061.
    1. Darrasse-Jeze G, Klatzmann D, Charlotte F, Salomon BL, Cohen JL (2006) CD4+CD25+ regulatory/suppressor T cells prevent allogeneic fetus rejection in mice. Immunol Lett 102: 106–109.
    1. Frey O, Petrow PK, Gajda M, Siegmund K, Huehn J, et al. (2005) The role of regulatory T cells in antigen-induced arthritis: aggravation of arthritis after depletion and amelioration after transfer of CD4+CD25+ T cells. Arthritis Res Ther 7: R291–301.
    1. Hsu WT, Suen JL, Chiang BL (2006) The role of CD4CD25 T cells in autoantibody production in murine lupus. Clin Exp Immunol 145: 513–519.
    1. Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI (2006) Suppression of disease in New Zealand Black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J Immunol 177: 1451–1459.
    1. Terrada C, Fisson S, De Kozak Y, Kaddouri M, Lehoang P, et al. (2006) Regulatory T cells control uveoretinitis induced by pathogenic Th1 cells reacting to a specific retinal neoantigen. J Immunol 176: 7171–7179.
    1. Saunders RN, Metcalfe MS, Nicholson ML (2001) Rapamycin in transplantation: a review of the evidence. Kidney Int 59: 3–16.
    1. Donia M, Mangano K, Amoroso A, Mazzarino MC, Imbesi R, et al. (2009) Treatment with rapamycin ameliorates clinical and histological signs of protracted relapsing experimental allergic encephalomyelitis in Dark Agouti rats and induces expansion of peripheral CD4+CD25+Foxp3+ regulatory T cells. J Autoimmun 33: 135–140.
    1. Esposito M, Ruffini F, Bellone M, Gagliani N, Battaglia M, et al. (2010) Rapamycin inhibits relapsing experimental autoimmune encephalomyelitis by both effector and regulatory T cells modulation. J Neuroimmunol 220: 52–63.
    1. Lisi L, Navarra P, Cirocchi R, Sharp A, Stigliano E, et al. (2012) Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis. J Neuroimmunol 243: 43–51.
    1. Contet C, Rawlins JN, Deacon RM (2001) A comparison of 129S2/SvHsd and C57BL/6JOlaHsd mice on a test battery assessing sensorimotor, affective and cognitive behaviours: implications for the study of genetically modified mice. Behav Brain Res 124: 33–46.
    1. Kojima T, Tanuma N, Aikawa Y, Shin T, Sasaki A, et al. (1997) Myosin-induced autoimmune polymyositis in the rat. J Neurol Sci 151: 141–148.
    1. Chi H (2012) Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev Immunol 12: 325–338.
    1. Cejka D, Hayer S, Niederreiter B, Sieghart W, Fuereder T, et al. (2010) Mammalian target of rapamycin signaling is crucial for joint destruction in experimental arthritis and is activated in osteoclasts from patients with rheumatoid arthritis. Arthritis Rheum 62: 2294–2302.
    1. Battaglia M, Stabilini A, Draghici E, Migliavacca B, Gregori S, et al. (2006) Induction of tolerance in type 1 diabetes via both CD4+CD25+ T regulatory cells and T regulatory type 1 cells. Diabetes 55: 1571–1580.
    1. Thomson AW, Turnquist HR, Raimondi G (2009) Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol 9: 324–337.
    1. Viola A, Molon B, Contento RL (2008) Chemokines: coded messages for T-cell missions. Front Biosci 13: 6341–6353.
    1. Weinreich MA, Takada K, Skon C, Reiner SL, Jameson SC, et al. (2009) KLF2 transcription-factor deficiency in T cells results in unrestrained cytokine production and upregulation of bystander chemokine receptors. Immunity 31: 122–130.
    1. Bromley SK, Thomas SY, Luster AD (2005) Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat Immunol 6: 895–901.
    1. Debes GF, Arnold CN, Young AJ, Krautwald S, Lipp M, et al. (2005) Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat Immunol 6: 889–894.
    1. Kurobe H, Liu C, Ueno T, Saito F, Ohigashi I, et al. (2006) CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity 24: 165–177.
    1. Sebzda E, Zou Z, Lee JS, Wang T, Kahn ML (2008) Transcription factor KLF2 regulates the migration of naive T cells by restricting chemokine receptor expression patterns. Nat Immunol 9: 292–300.
    1. Sinclair LV, Finlay D, Feijoo C, Cornish GH, Gray A, et al. (2008) Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking. Nat Immunol 9: 513–521.
    1. Roncarolo MG, Gregori S, Lucarelli B, Ciceri F, Bacchetta R (2011) Clinical tolerance in allogeneic hematopoietic stem cell transplantation. Immunol Rev 241: 145–163.
    1. Kaposztas Z, Etheridge WB, Kahan BD (2008) Case report: successful treatment of posttransplant lymphoproliferative disorder and quiescence of dermatomyositis with rituximab and sirolimus. Transplant Proc 40: 1744–1746.
    1. Nadiminti U, Arbiser JL (2005) Rapamycin (sirolimus) as a steroid-sparing agent in dermatomyositis. J Am Acad Dermatol 52: 17–19.
    1. Banica L, Besliu A, Pistol G, Stavaru C, Ionescu R, et al. (2009) Quantification and molecular characterization of regulatory T cells in connective tissue diseases. Autoimmunity 42: 41–49.

Source: PubMed

3
구독하다