Elsevier

Clinical Immunology

Volume 128, Issue 2, August 2008, Pages 172-180
Clinical Immunology

Regulatory T cells induced by GM-CSF suppress ongoing experimental myasthenia gravis

https://doi.org/10.1016/j.clim.2008.03.509Get rights and content

Abstract

We had previously observed that treatment utilizing granulocyte–macrophage colony-stimulating factor (GM-CSF) had profound effects on the induction of experimental autoimmune myasthenia gravis (EAMG), a well-characterized antibody-mediated autoimmune disease. In this study, we show that EAMG induced by repeated immunizations with acetylcholine receptor (AChR) protein in C57BL6 mice is effectively suppressed by GM-CSF treatment administered at a stage of chronic, well-established disease. In addition, this amelioration of clinical disease is accompanied by down-modulation of both autoreactive T cell, and pathogenic autoantibody responses, a mobilization of DCs with a tolerogenic phenotype, and an expansion of regulatory T cells (Tregs) that potently suppress AChR-stimulated T cell proliferation in vitro. These observations suggest that the mobilization of antigen-specific Tregs in vivo using pharmacologic agents, like GM-CSF, can modulate ongoing anti-AChR immune responses capable of suppressing antibody-mediated autoimmunity.

Introduction

Myasthenia gravis (MG) is an antibody-mediated autoimmune disease in which pathogenic autoantibodies are directed against the skeletal muscle nicotinic acetylcholine receptor (AChR) [1]. Experimental autoimmune myasthenia gravis (EAMG) induced in C57BL/6 mice after repeated immunizations with emulsified Torpedo californica AChR (tAChR) is a useful model for the study of pathogenic mechanisms and therapeutic strategies relevant to MG in humans [2]. Although antibodies to the AChR are directly responsible for the destruction of the muscle endplate resulting in both MG and EAMG, the autoantibody response is T cell dependent, with CD4+ T cells providing help for B cells to produce anti-AChR antibodies [3], [4]. Despite the fact that the target antigen is so well defined, there is currently no specific immunosuppressive therapy or cure for MG. Nonspecific immunotherapy utilizing corticosteroids and other immunosuppressive drugs combined with symptomatic therapy with acetylcholinesterase inhibitors results in clinical improvement and substantial control of symptoms in most patients. However, there are significant potential side effects and risks associated with global nonspecific suppression of the immune response, including infections and malignancy.

Ideal specific therapies for MG would have little effect on overall immunity, while targeting the mechanisms that initiate and sustain the autoimmune response to the AChR. While these mechanisms are not completely understood, multiple lines of evidence indicate that the immune system's professional antigen-presenting cells, the dendritic cells (DCs), participate in the onset and progression of autoimmune diseases [5], [6]. Animal models show that the transfer of DCs isolated from donors with acute autoimmune disease or propagated in vitro under conditions that induce maturation, generates a strong T helper (Th)-1 response, eventually culminating in autoimmune disease [7]. Conversely, DCs have been shown to have the ability to educate T cells to tolerate self antigens, and to promote the mobilization of regulatory T cell (Treg) subsets [8], [9], [10]. It has been shown that the interaction of DCs with antigen-specific Tregs can suppress experimental autoimmunity [11]. Current evidence indicates that the immunogenic or tolerogenic function of DCs is largely determined by differentiation status which may be manipulated using growth factors such as granulocyte–macrophage colony-stimulating factor (GM-CSF) [12], and that DC functional state is important in determining Treg biology and antigen-specific control of experimental autoimmunity [13], [14]. Previous work has been published examining the potential of in vivo administration of GM-CSF in experimental autoimmune thyroiditis (EAT) [15], [16], and in the experimental model of autoimmune diabetes [14], and mobilization of specific DC subsets and Tregs was reported to critical to the observed effects. But, EAT and autoimmune diabetes are T-cell mediated diseases, and in general, the role of dendritic cells (DCs) in the biology of regulatory T cells and subsequent control of autoimmunity has been studied primarily in T cell mediated autoimmune diseases. We, however, have previously observed that GM-CSF had profound effects on the induction of experimental autoimmune myasthenia gravis (EAMG), a well-characterized antibody-mediated autoimmune disease [17].

In the current study, we examine the therapeutic potential of GM-CSF in chronic EAMG, and demonstrate that GM-CSF effectively ameliorates clinical disease in mice with ongoing, well-established disease. Furthermore, we show not only an effect of GM-CSF on particular subpopulations of DCs, T cells, and T cell proliferative response to the AChR, but also a significant down-modulation of pathogenic anti-AChR autoantibody production.

Section snippets

Mice

Eight-week old female C57BL6/J mice were purchased from the Jackson Laboratories (Bar Harbor, ME). Mice were housed in the Biologic Resources Laboratory facilities at the University of Illinois (Chicago, IL) and provided food and water ad libitum. All mice were cared for in accordance with the guidelines set forth by the University of Illinois Animal Care and Use committee.

Purification of tAChR and mouse AChR

Torpedo AChR (tAChR) was purified from the electric organs of T. californica by affinity chromatography using a conjugate

GM-CSF treatment effect in established EAMG

At the end of the disease induction (initial priming immunization and 3 boosters) stage, greater than 90% of the animals had clinically demonstrable fatigable muscle weakness. We then divided these animals into two groups (n = 20) with an equal balance of the various disease severities in each group. We treated one group of animals with GM-CSF (2 µg IP daily for 10 days) and the other group of animals with PBS. The day of treatment initiation was designated “day 0” and the mice were clinically

Discussion

In this study, we expanded upon our previous observations that treatment with GM-CSF protected against the induction of EAMG [17], by investigating the therapeutic potential of GM-CSF in the treatment of ongoing, active EAMG, a well-characterized antibody-mediated autoimmune disease. We found that treatment with GM-CSF at a stage of chronic, well-established disease, effectively induced improvement in signs of muscle weakness and potently suppressed disease progression. Understanding the

Acknowledgments

This work was supported by the NIH (National Institute of Neurologic Disorders and Stroke, K08NS058800-01, MNM; and National Institute of Allergy and Infectious Diseases, RO1 AI 058190-01, BSP); and by the Myasthenia Gravis Foundation of America — Postdoctoral Fellowship Award to JRS.

References (40)

  • M. Arpinati et al.

    Granulocyte-colony stimulating factor mobilizes T helper 2-inducing dendritic cells

    Blood

    (2000)
  • J.A. Hamilton

    GM-CSF in inflammation and autoimmunity

    Trends Immunol.

    (2002)
  • H. Groux et al.

    Role of dendritic cells in the generation of regulatory T cells

    Semin. Immunol.

    (2004)
  • A. Vincent

    Unraveling the pathogenesis of myasthenia gravis

    Nat. Rev. Immunol.

    (2002)
  • P.W. Berman et al.

    Experimental myasthenia gravis: a murine system

    J. Exp. Med.

    (1980)
  • B.W. Hughes et al.

    Pathophysiology of myasthenia gravis

    Semin. Neurol.

    (2004)
  • J.A. Hardin

    Dendritic cells: potential triggers of autoimmunity and targets for therapy

    Ann. Rheum. Dis.

    (2005)
  • B.N. Dittel et al.

    Presentation of the self antigen myelin basic protein by dendritic cells leads to experimental autoimmune encephalomyelitis

    J. Immunol.

    (1999)
  • M. Rossi et al.

    Human dendritic cells: Potent antigen-presenting cells at the crossroads of innate and adaptive immunity

    J. Immunol.

    (2005)
  • J. Banchereau et al.

    Dendritic cells and the control of immunity

    Nature

    (1998)
  • Cited by (61)

    • Development of an extended half-life GM-CSF fusion protein for Parkinson's disease

      2022, Journal of Controlled Release
      Citation Excerpt :

      Daily treatment with recombinant GM-CSF protein is generally well-tolerated with mild-to-moderate adverse events including injection site reactions, elevated white blood cells, and bone pain; the latter two primarily due to stimulation and migration of hematopoietic components from bone marrow to the peripheral circulation [13,22,37,38]. The neuroprotective efficacy of daily dosed recombinant GM-CSF treatment has been well-established not only in PD, but also in other neurodegenerative diseases such as myasthenia gravis, AD, and traumatic brain injury models of human disease [39–42]. GM-CSF treatment promotes Treg number and function, restores the phenotypic balance between Treg and Teff, and suppresses the activation of microglia, thus exerting a profound control over neurodegenerative processes [16,40,43].

    • Granulocyte-macrophage colony-stimulating factor mRNA and Neuroprotective Immunity in Parkinson's disease

      2021, Biomaterials
      Citation Excerpt :

      In support of this idea, adoptive transfer of CD4+CD25+ Treg into MPTP-intoxicated animals attenuates neuroinflammation leading to nigrostriatal protection [4]. We and others, have shown that GM-CSF protein readily increases Treg numbers and activity with parallel neuroprotective activities in PD, AD, myasthenia gravis, and traumatic brain injury (TBI) models of human disease [3,6,7,10,41,42,44]. However, allergic treatment site reactions, muscle and bone pain, and humoral responses targeted against the protein are all side effects associated with its daily clinical use [10,25,26,29].

    • IL-10 derived from CD1d<sup>hi</sup>CD5<sup>+</sup> B cells regulates experimental autoimmune myasthenia gravis

      2015, Journal of Neuroimmunology
      Citation Excerpt :

      All animal use procedures were conducted in strict accordance to the National Institutes of Health and University of Chicago institutional guidelines. AChR was purified from the electric organs of T. californica by affinity chromatography using a conjugate of neurotoxin coupled to agarose, as previously described (Sheng et al., 2006, 2008, 2011). Purified tAChR was used to induce EAMG and as antigen for in vitro studies of immune responses.

    View all citing articles on Scopus
    View full text