Elsevier

Molecular and Cellular Neuroscience

Volume 68, September 2015, Pages 284-292
Molecular and Cellular Neuroscience

Statin-modified dendritic cells regulate humoral immunity in experimental autoimmune myasthenia gravis

https://doi.org/10.1016/j.mcn.2015.08.010Get rights and content

Highlights

  • AT-BMDCs regulate humoral immune response in EAMG.

  • AT-BMDCs reduce the quantity and the relative affinity of antibodies.

  • AT-BMDCs suppress germinal center response.

  • AT-BMDCs decrease follicular helper T cells and IL-21.

  • AT-BMDCs increase regulatory B cells.

Abstract

We previously demonstrated that atorvastatin induced immature dendritic cells (DCs) derived from spleen in vitro. Administration of these tolerogenic DCs led to amelioration of experimental autoimmune myasthenia gravis (EAMG). The protective effect was mainly mediated by inhibited cellular immune response, including up-regulated regulatory T cells and shifted Th1/Th17 to Th2 cytokines. The present study employed atorvastatin-modified bone marrow-derived DCs (AT-BMDCs) to explore the effect of tolerogenic DCs on humoral immune response of EAMG and further elucidate the underlying mechanisms. Our data showed that AT-BMDCs reduced the quantity and the relative affinity of pathogenic antibodies, suppressed germinal center response, decreased follicular helper T cells and IL-21, and increased regulatory B cells. These results suggest that AT-BMDCs ameliorate EAMG by regulating humoral immune response, thus providing new insights into therapeutic approaches of myasthenia gravis and other autoimmune diseases.

Introduction

Myasthenia gravis (MG) is a classical organ specific autoimmune disease, which is antibody-mediated, T cell-dependent and complement-involved. It impairs the postsynaptic nicotinic acetylcholine receptor (AchR) at the neuromuscular junction (NMJ), leading to neuromuscular transmission failure, thereby inducing muscle weakness and fatigability (Berrih-Aknin et al., 2014). Experimental autoimmune myasthenia gravis (EAMG), induced by immunization of AchR purified from Torpedo californica (Lennon et al., 1975) or synthetic peptide corresponding to region 97–116 of the rat AChRα subunit (R97–116 peptide) (Baggi et al., 2004), serves as a well-established animal model used to explore the immunopathological mechanisms and therapeutic strategies of human MG.

Current immunosuppressive agents for MG treatment, such as corticosteroids and cytotoxic reagents, are more pan-immunosuppressive than specific cell targeted, thus causing numerous side effects. Pathogenic autoantibodies (mostly antibodies against AchR) are detected in a majority of MG patients and damage the neuromuscular transmission via many mechanisms (Meriggioli and Sanders, 2009), indicating that antibody-secreting B cells are essential for the development of MG. Accumulating evidence demonstrates that B cell targeted therapy via elimination of B cells with drugs such as rituximab (anti-CD20 mAb) can relieve symptoms of MG patients (Iorio et al., 2014), which supports the idea that autoreactive B cells may be a prominent candidate for targeted therapies in MG. Moreover, a distinct T cell subset, named follicular helper T (Tfh) cells and characterized by expression of transcription factor B-cell lymphoma 6 (Bcl6), surface molecules including CXC chemokine receptor 5 (CXCR5), inducible T-cell costimulator (ICOS) and programmed death protein-1 (PD-1), and cytokine IL-21, is indicated to supply direct assistance for antibody production of B cells (Crotty, 2011). The contributing role of Tfh cells has been confirmed in many antibody-mediated autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and autoimmune thyroid diseases (Ma et al., 2012, Simpson et al., 2010, Zhu et al., 2012). Recent studies have also reported an aberrant expansion of Tfh population in MG patients (Luo et al., 2013, Zhang et al., 2014a). Taken together, these findings suggest the pathogenic role of humoral immune response mediated by B cells and Tfh cells in MG.

Recently, the discovery of regulatory B cells (Breg) has challenged the conventional roles of B cells as antigen-presenting cells and antibody-secreting cells (Mauri and Ehrenstein, 2008). Adoptive transfer of Breg attenuates severity of some autoimmune diseases in animal models, such as experimental autoimmune encephalomyelitis (EAE) (Matsushita et al., 2010), type 1 diabetes (T1DM) (Hussain and Delovitch, 2007) and collagen-induced arthritis (CIA) (Yang et al., 2012). Sun et al. have reported that IL-10 producing Breg (B10 cells) decrease in MG patients and those who respond well to rituximab treatment exhibit rapid repopulation of B10 cells (Sun et al., 2014). However, data are still scarce about the exact role of Breg in MG.

Previously, our study reported that atorvastatin induced immature dendritic cells (DCs) derived from spleen in vitro and administration of these tolerogenic DCs ameliorated EAMG by inhibiting proliferation of lymphocytes, up-regulating regulatory T cells (Treg), shifting Th1/Th17 to Th2 response and decreasing levels of pathogenic antibodies (Li et al., 2013). The previous study focused on the effect of tolerogenic DCs on cellular immunity, however, the exact mechanisms by which tolerogenic DC treatment triggered reduction of antibodies deserve further investigations. Furthermore, given that bone marrow generates more abundant and immature DCs with higher trafficking property than spleen (Emmanouilidis et al., 2006, Fields et al., 1998), and most studies employing DCs prefer bone marrow-derived dendritic cells (BMDCs) to spleen-derived dendritic cells (SpDCs), we used BMDCs in this study instead of SpDCs in the previous study. Collectively, the aim of the present study was to explore the effect of atorvastatin-modified BMDCs (AT-BMDCs) on humoral immune response in EAMG and we attempted to explore the underlying mechanisms.

In this study, we found that atorvastatin successfully induced immature BMDCs in vitro, of which CD80, CD86 and MHC II molecules were inhibited. AT-BMDC treatment led to amelioration of ongoing EAMG. AT-BMDCs suppressed the quantity and the relative affinity of anti-R97-116 IgG as well as germinal center (GC) response. In addition, Tfh cells and IL-21 were decreased by AT-BMDC treatment. Interestingly, the amount of CD19+ B cells was slightly influenced, while Breg were increased by AT-BMDCs. Our data suggest that AT-BMDCs could ameliorate EAMG by regulating humoral immune response and might provide a potential approach and new evidence for MG therapies.

Section snippets

Atorvastatin induces immature BMDCs in vitro

To determine whether atorvastatin could induce immature BMDCs, an experiment in vitro was conducted. BMDCs were cultured with or without atorvastatin and then analyzed by flow cytometry (FACS). In the presence of atorvastatin, CD80 (89.77 ± 1.98%), CD86 (82 ± 5.31%) and MHC II (90.23 ± 1.27%) expressed on AT-BMDCs were inhibited compared with those on 0-BMDCs cultured in the absence of atorvastatin (CD80, 50.93 ± 3.01%, CD86, 51 ± 4.02%, MHC II, 63.20 ± 9.04%; t = 18.693, p < 0.001; t = 8.042, p < 0.01 and t = 

Discussion

DCs play a key role in inducing immunity as the most efficient antigen-presenting cells, however, the fact that DCs also induce tolerance has been well established and accepted, which at least partially depends on their maturation state (Manicassamy and Pulendran, 2011). Generally, mature DCs tend to induce immunity, whereas immature DCs induce tolerance. Previously, we found that atorvastatin could induce immature SpDCs in vitro and AT-SpDCs successfully ameliorated EAMG by up-regulated Treg

Animals and reagents

Six to eight-week-old female Lewis rats weighing 155–180 g were purchased from Vital River Corporation (Beijing, China) and housed under pathogen-free conditions at the local facilities with a 12/12-h light/dark cycle. Rats were fed with standard rat chow and water ad libitum. All the experimental protocols were in accordance with the Guidelines of Animal Experiments from the institutional committee. Minimum number of rats necessary was used to achieve the scientific objectives and animal

Conflict of interest statement

All authors have declared that there are no conflicts of interest with regard to this work.

Acknowledgments

We thank Beijing Garlin Pharmaceutical Co., Ltd. for assistance in providing atorvastatin. The project was funded by the National Natural Science Foundation of China (30771989, 81171128 and 81471222) and Taishan Scholars Construction Engineering of Shandong Province (ts20130914).

References (60)

  • C. Mauri et al.

    The ‘short’ history of regulatory B cells

    Trends Immunol.

    (2008)
  • A. Meraouna et al.

    The chemokine CXCL13 is a key molecule in autoimmune myasthenia gravis

    Blood

    (2006)
  • M.N. Meriggioli et al.

    Autoimmune myasthenia gravis: emerging clinical and biological heterogeneity

    Lancet Neurol.

    (2009)
  • A. Morva et al.

    Maturation and function of human dendritic cells are regulated by B lymphocytes

    Blood

    (2012)
  • L. Qian et al.

    Regulatory dendritic cells program B cells to differentiate into CD19hiFcgammaIIbhi regulatory B cells through IFN-beta and CD40L

    Blood

    (2012)
  • N. Schmitt et al.

    Human dendritic cells induce the differentiation of interleukin-21-producing T follicular helper-like cells through interleukin-12

    Immunity

    (2009)
  • J. Shaw et al.

    Plasmacytoid dendritic cells regulate B-cell growth and differentiation via CD70

    Blood

    (2010)
  • A. Vogelzang et al.

    A fundamental role for interleukin-21 in the generation of T follicular helper cells

    Immunity

    (2008)
  • Y. Wei et al.

    Infusion of dendritic cells carrying donor lymphocytes treated with 8-methoxypsoralen and ultraviolet a light induces CD19 + IL-10 + regulatory B cells and promotes skin allograft survival

    Transplant. Proc.

    (2014)
  • X. Xie et al.

    ATRA alters humoral responses associated with amelioration of EAMG symptoms by balancing Tfh/Tfr helper cell profiles

    Clin. Immunol.

    (2013)
  • N. Xin et al.

    RNA interference targeting Bcl-6 ameliorates experimental autoimmune myasthenia gravis in mice

    Mol. Cell. Neurosci.

    (2014)
  • H. Xu et al.

    Therapeutic potential of atorvastatin-modified dendritic cells in experimental autoimmune neuritis by decreased Th1/Th17 cytokines and up-regulated T regulatory cells and NKR-P1(+) cells

    J. Neuroimmunol.

    (2014)
  • M. Yang et al.

    IL-10-producing regulatory B10 cells ameliorate collagen-induced arthritis via suppressing Th17 cell generation

    Am. J. Pathol.

    (2012)
  • M. Zhang et al.

    Thymic TFH cells involved in the pathogenesis of myasthenia gravis with thymoma

    Exp. Neurol.

    (2014)
  • F. Baggi et al.

    Breakdown of tolerance to a self-peptide of acetylcholine receptor alpha-subunit induces experimental myasthenia gravis in rats

    J. Immunol.

    (2004)
  • N.A. Carter et al.

    Mice lacking endogenous IL-10-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Th1/Th17 but a decrease in regulatory T cells

    J. Immunol.

    (2011)
  • S. Crotty

    Follicular helper CD4 T cells (TFH)

    Annu. Rev. Immunol.

    (2011)
  • O. Dienz et al.

    The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by CD4 + T cells

    J. Exp. Med.

    (2009)
  • B. Dubois et al.

    Critical role of IL-12 in dendritic cell-induced differentiation of naive B lymphocytes

    J. Immunol.

    (1998)
  • N. Emmanouilidis et al.

    Immunosuppressive and trafficking properties of donor splenic and bone marrow dendritic cells

    Transplantation

    (2006)
  • Cited by (23)

    • Statins for Graves' orbitopathy (STAGO): a phase 2, open-label, adaptive, single centre, randomised clinical trial

      2021, The Lancet Diabetes and Endocrinology
      Citation Excerpt :

      By affecting these pathways, statins might influence innate immunity and regulate inflammatory cell balance. In addition, in-vitro studies and experimental models of autoimmune diseases showed that statins have direct immunoregulatory effects by inducing tolerogenic dendritic cells, a specialised subset of dendritic cells promoting immune tolerance and counteracting autoimmune responses.26 Statins might also counteract orbital changes through additional mechanisms that influence tissue remodelling because they inhibit adipogenesis in preadipocytes and fibroblasts by reducing expression of adipogenic genes.27

    • Prophylactic administration of fingolimod (FTY720) ameliorated experimental autoimmune myasthenia gravis by reducing the number of dendritic cells, follicular T helper cells and antibody-secreting cells

      2021, International Immunopharmacology
      Citation Excerpt :

      The experimental autoimmune myasthenia gravis (EAMG) is a standard animal model of MG induced by AChR antigen or synthetic peptide corresponding to 97–116 region of the rat AChR-α subunit (R97–116 peptide) [2]. A humoral response from follicular helper T (Tfh) cells is involved in the development of MG and EAMG [3], along with germinal center (GC) B cells and dendritic cells (DCs) [4,5]. Patients with generalized MG show increased levels of circulating Tfh cells, which in turn, might activate the autoreactive B cells and contribute to MG [6].

    • Statins and autoimmunity: State-of-the-art

      2020, Pharmacology and Therapeutics
      Citation Excerpt :

      Numerous clinical trials have reported statin-mediated reductions in C-reactive protein (CRP), as well as inhibiting the production of MMP1 and MMP3, which are involved in connective tissue degradation (Crouse, et al., 2007; de Jong et al., 2018; Ridker et al., 2009). Statins also inhibit DC maturation and the antigen presenting process in a dose-dependent manner (H. Li et al., 2015; Mira & Mañes, 2009; Tricarico et al., 2015; Yilmaz et al., 2004). This is mediated by inhibiting the production of IFN-γ and MHC transactivator class II expression on antigen presenting cells (APCs) (Khattri & Zandman-Goddard, 2013; Ntolkeras et al., 2019).

    • Regulatory T cells: Possible mediators for the anti-inflammatory action of statins

      2019, Pharmacological Research
      Citation Excerpt :

      Statin administration leads to in vivo consumption of GGPP and FPP which both have a central role in posttranslational prenylation of several important cell signaling proteins associated with inflammation [46]. Statins have been demonstrated to modulate the immune system (Fig. 3) is by interfering with the maturation and function of dendritic cells (DCs) [47–49]. Statins inhibit the expression of MHC-II, induced by cytokine and co-stimulatory molecules of antigen presenting cells (APC), and prevent antigen presentation to CD4 T cells.

    • Toll-like receptor 9 antagonist suppresses humoral immunity in experimental autoimmune myasthenia gravis

      2018, Molecular Immunology
      Citation Excerpt :

      Assistance of TLR9 strongly promoted GC response (Hou et al., 2011), TLR9 ligation stimulated T cell-dependent germinal centre IgG antibody response, and this stimulation required B cell MyD88 (Hou et al., 2011). In our research, we found that the GC reaction in EAMG rats could be inhibited by statin-modified DCs or a caspase-1 inhibitor (Li et al., 2015; Wang et al., 2015). This is in accordance with the results of our study that H154-treated EAMG rats exhibited attenuated GC B cells and antibody production.

    View all citing articles on Scopus
    View full text