Elsevier

Journal of Autoimmunity

Volume 62, August 2015, Pages 75-80
Journal of Autoimmunity

CD4+ T cells epigenetically modified by oxidative stress cause lupus-like autoimmunity in mice

https://doi.org/10.1016/j.jaut.2015.06.004Get rights and content

Highlights

  • The current study tested whether oxidant-treated T cells can induce lupus in mice.

  • In vitro treatment of CD4+ T cells with H2O2 or ONOO decreased ERK signaling and Dnmt1 expression in these cells.

  • Oxidant treatment also upregulated genes known to be suppressed by DNA methylation in SLE patients and animal models of SLE.

  • Adoptive transfer of oxidant-treated CD4+ T cells into syngeneic mice caused anti-dsDNA antibody and glomerulonephritis.

  • Oxidative stress may contribute to SLE by reducing ERK signaling & DNA methylation, activating immune genes & autoreactivity.

Abstract

Lupus develops when genetically predisposed people encounter environmental agents such as UV light, silica, infections and cigarette smoke that cause oxidative stress, but how oxidative damage modifies the immune system to cause lupus flares is unknown. We previously showed that oxidizing agents decreased ERK pathway signaling in human T cells, decreased DNA methyltransferase 1 and caused demethylation and overexpression of genes similar to those from patients with active lupus. The current study tested whether oxidant-treated T cells can induce lupus in mice. We adoptively transferred CD4+ T cells treated in vitro with oxidants hydrogen peroxide or nitric oxide or the demethylating agent 5-azacytidine into syngeneic mice and studied the development and severity of lupus in the recipients. Disease severity was assessed by measuring anti-dsDNA antibodies, proteinuria, hematuria and by histopathology of kidney tissues. The effect of the oxidants on expression of CD40L, CD70, KirL1 and DNMT1 genes and CD40L protein in the treated CD4+ T cells was assessed by Q-RT-PCR and flow cytometry. H2O2 and ONOO decreased Dnmt1 expression in CD4+ T cells and caused the upregulation of genes known to be suppressed by DNA methylation in patients with lupus and animal models of SLE. Adoptive transfer of oxidant-treated CD4+ T cells into syngeneic recipients resulted in the induction of anti-dsDNA antibody and glomerulonephritis. The results show that oxidative stress may contribute to lupus disease by inhibiting ERK pathway signaling in T cells leading to DNA demethylation, upregulation of immune genes and autoreactivity.

Introduction

Systemic lupus erythematosus (SLE) is a chronic relapsing autoimmune disease that primarily affects women. SLE develops and flares when genetically predisposed people encounter certain environmental agents. The genes predisposing to lupus are being identified, and epidemiologic evidence indicates that environmental agents which cause oxidative stress, such as infections, sun exposure, silica and smoking, are associated with lupus onset and flares [1]. However, how environmentally induced reactive oxygen species interact with the immune system to trigger lupus flares remains unclear.

Our group reported that CD4+ T cells epigenetically altered with DNA methylation inhibitors like 5-azacytidine (5-azaC), procainamide (Pca) or hydralazine (Hyd) cause lupus-like autoimmunity in animal models [2], and that similar epigenetically modified T cells are found in lupus patients during disease flares [1]. We traced the cause of the epigenetic defect to PKCδ inactivation, which prevents upregulation of DNA methyltransferase 1 (Dnmt1) during mitosis to copy methylation patterns [3]. Others reported that serum proteins are nitrated in patients with active lupus, caused by superoxide (O2-) combining with nitric oxide (NO) to form peroxynitrite (ONOO), a highly reactive molecule that nitrates proteins and other molecules [4]. Our group subsequently found that PKCδ is similarly nitrated in T cells from patients with active lupus, and that nitrated PKCδ is unable to transmit signals that upregulate Dnmt1 to copy DNA methylation patterns in dividing T cells, causing demethylation and overexpression of genes normally suppressed by DNA methylation [5]. We extended these observations by demonstrating that treating human CD4+ T cells with the oxidizing agents H2O2 or ONOO inhibits PKCδ activation, thereby decreasing ERK pathway signaling, decreasing Dnmt1 levels and causing demethylation and overexpression of CD70 and Kir genes similar to T cells from patients with active lupus [6]. However, whether the epigenetically modified T cells are sufficient to cause lupus-like autoimmunity was unknown.

We have now tested if female murine CD4+ T cells treated with H2O2 or ONOO also overexpress methylation sensitive genes, and if the treated cells cause lupus-like autoimmunity in mice. The results demonstrate that the oxidized T cells overexpress the X-linked gene CD40L, one copy of which is silenced by DNA methylation in female human and mouse T cells [7], [8], and Kir genes, normally expressed by NK cells but silenced by DNA methylation in human and mouse T cells [9]. The results also demonstrate that adoptive transfer of the treated cells into syngeneic recipients causes anti-DNA antibodies and an immune complex glomerulonephritis, similar to T cells treated 5-azaC, Pca or Hyd. Together these studies support the contention that environmentally-induced oxidative stress may trigger lupus flares by epigenetically altering T cells through effects on T cell signaling and Dnmt1 expression.

Section snippets

Drugs and reagents

5-azaC and H2O2 were purchased from Sigma–Aldrich (St. Louis, MO), and ONOO from Calbiochem (San Diego, CA).

Mice

Female SJL mice were purchased from Jackson Laboratories, housed in filter-protected cages, and provided with standard irradiated rodent diet 5053 (Lab Diet; PMI Nutrition International) and water ad libitum. Urinary protein and hemoglobin were measured using Chemstrip 7 dipsticks (Roche, Madison, WI). The mice were maintained in a specific pathogen–free facility by the Unit for

Murine CD4+ T cells treated with oxidizing agents overexpress methylation sensitive genes

We previously reported that inhibiting the replication of human CD4+ T cell DNA methylation patterns with the Dnmt inhibitor 5-azaC causes demethylation and overexpression of genes including CD40L on the female inactive X chromosome in humans and mice [8], [12] as well as the human KIR gene family, normally expressed by NK cells but not T cells [9], [13]. Initial studies compared the effects of H2O2 and ONOO to 5-azaC on murine CD4+ T cell CD40L and Kir expression. We recently reported that

Discussion

A relatively extensive body of literature implicates CD4+ T cell DNA demethylation in lupus onset and flares. Early work demonstrated that inhibiting DNA methylation in CD4+ T cells with 5-azaC alters gene expression and converts polyclonal as well as cloned antigen-specific CD4+ T cells in autoreactive cells that respond to self class II MHC determinants without the addition of specific antigen [16], [17]. The autoreactivity was traced to overexpression of the adhesion molecule LFA-1

Acknowledgments

We thank Erin Grant for her excellent technical assistance and Ms. Stacy Fry for her excellent secretarial assistance. We also thank Dr. Gabriela Gorelik for helpful discussions and advice on oxidative stress and PKC-δ. This work was supported by PHS grants AR42525 and ES017885, a Merit grant from the Department of Veterans Affairs and the Lupus Insight Prize from the Lupus Foundation of America, the Alliance for Lupus Research and the Lupus Research Institute.

References (37)

  • J. Quddus et al.

    Treating activated CD4+ T cells with either of two distinct DNA methyltransferase inhibitors, 5-azacytidine or procainamide, is sufficient to cause a lupus-like disease in syngeneic mice

    J. Clin. Investigation

    (1993)
  • G. Gorelik et al.

    Impaired T cell protein kinase C delta activation decreases ERK pathway signaling in idiopathic and hydralazine-induced lupus

    J. Immunol.

    (2007)
  • G.J. Gorelik et al.

    Protein kinase Cdelta oxidation contributes to ERK inactivation in lupus T cells

    Arthritis Rheumatism

    (2012)
  • Y. Li et al.

    Oxidative stress, T cell DNA methylation, and lupus

    Arthritis & Rheumatology

    (2014)
  • F.M. Strickland et al.

    Diet influences expression of autoimmune-associated genes and disease severity by epigenetic mechanisms in a transgenic mouse model of lupus

    Arthritis Rheumatism

    (2013)
  • B. Richardson et al.

    Murine models of lupus induced by hypomethylated T cells (DNA hypomethylation and lupus…)

    Methods Mol. Biol.

    (2012)
  • Q. Lu et al.

    Demethylation of CD40LG on the inactive X in T cells from women with lupus

    J. Immunol.

    (2007)
  • A. Rouhi et al.

    Evidence for epigenetic maintenance of Ly49a monoallelic gene expression

    J. Immunol.

    (2006)
  • Cited by (60)

    • Approaches to autoimmune diseases using epigenetic therapy

      2023, Epigenetics in Human Disease, Third Edition
    • Regulation of the epigenetic landscape by immune cell oxidants

      2021, Free Radical Biology and Medicine
      Citation Excerpt :

      At lower concentrations H2O2 (100 μM), this change was not observed [141]. In contrast to this, two studies have shown that CD4+ T cells that were treated with 20–50 μM H2O2 showed reduced DNMT1 levels and demethylation of key genes involved in lupus disease progression [142,143]. In order to systematically investigate the effect of oxidants on methylation, our research group utilised heavy isotope mass spectrometry method in combination with cell cycle synchronisation to specifically monitor the methylation status of 15N3-deoxycytidine (15N3-dC) incorporated into newly synthesised DNA [9].

    • Epigenetics and precision medicine in skin diseases

      2021, Epigenetics in Precision Medicine
    • Autoimmune disease and epigenetics

      2021, Medical Epigenetics
    View all citing articles on Scopus
    View full text