Elsevier

Journal of Autoimmunity

Volume 37, Issue 3, November 2011, Pages 180-189
Journal of Autoimmunity

Inhibited expression of hematopoietic progenitor kinase 1 associated with loss of jumonji domain containing 3 promoter binding contributes to autoimmunity in systemic lupus erythematosus

https://doi.org/10.1016/j.jaut.2011.09.006Get rights and content

Abstract

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by T cell overactivation and B cell hyper-stimulation. Hematopoietic progenitor kinase 1 (HPK1, also called MAP4K1) negatively regulates T cell-mediated immune responses. However, the role of HPK1 and the mechanisms that regulate HPK1 expression in SLE remain poorly understood. Using chromatin immunoprecipitation (ChIP) microarray data, we identified markedly increased histone H3 lysine 27 trimethylation (H3K27me3) enrichment at the HPK1 promoter of SLE CD4+ T cells relative to controls, and confirmed this observation using ChIP and real-time PCR experiments. We further found that HPK1 mRNA and protein levels were significantly decreased in CD4+ T cells of patients with SLE, and that this decrease was not caused by exposure to standard SLE medications. Down-regulating HPK1 in healthy CD4+ T cells significantly accelerated T cell proliferation and production of IFNγ and IgG. Consistent with these findings, overexpressing HPK1 in SLE CD4+ T cells caused a significant decrease in T cell reactivity. In addition, we observed a striking decrease in jumonji domain containing 3 (JMJD3) binding, but no marked change in enhancer of zeste homolog 2 (EZH2) binding, at the HPK1 promoter region in SLE CD4+ T cells compared to healthy controls. SiRNA knock down of JMJD3 in healthy CD4+ T cells led to decreased JMJD3 binding and increased H3K27me3 enrichment at the HPK1 promoter region, thus inhibiting the expression of HPK1. Concordantly, plasmid-induced overexpression of JMJD3 in SLE CD4+ T cells led to increased JMJD3 binding, decreased H3K27me3 enrichment, and up-regulated HPK1 expression. Our results show for the first time that inhibited HPK1 expression in SLE CD4+ T cells is associated with loss of JMJD3 binding and increased H3K27me3 enrichment at the HPK1 promoter, contributing to T cell overactivation and B cell overstimulation in SLE. These findings suggest that HPK1 may serve as a novel target for effective SLE therapy.

Highlights

► HPK1 expression is significantly decreased in CD4+ T cells from SLE patients. ► Reduced JMJD3 leads to increased H3K27me3 at the HPK1 promoter in SLE CD4+ T cells. ► Increased H3K27me3 enrichment results in HPK1 down-regulation in SLE CD4+ T cells. ► Decreased HPK1 contributes to abnormal T cell reactivity in SLE.

Introduction

Autoimmune diseases are multifactorial in their pathogenesis. Contributory factors include: genetic predisposition and environmental factors [1], [2]. Autoimmune diseases are always female-predominant [3], [4], [5]. As a typical chronic autoimmune disease, systemic lupus erythematosus (SLE) is characterized by T cell overactivation and the overproduction of autoantibodies against multiple self antigens [6]. Although the molecular mechanisms that initiate these autoimmune responses in SLE remain unclear, it is widely accepted that epigenetic alterations in the promoters of certain immune-related genes play critical roles in the onset and progression of SLE [7], [8], [9], [10]. The term “epigenetics” refers to stable and heritable changes in gene expression that are not related to changes in the DNA sequence [11], [12]. The major mechanisms of epigenetic regulation include DNA methylation, histone modifications, chromatin modifications, noncoding RNA regulation, and so forth [11], [12], [13], [14]. Among histone modifications, histone H3 lysine 27 trimethylation (H3K27me3) is a hallmark of gene silencing [15], [16], [17], [18], via its binding to Pc protein, which forms part of the polycomb repressive complex 1 (PRC1). PRC1 then blocks the access of transcriptional activation factors and chromatin remodeling factors to DNA, and prevents initiation of transcription by RNA polymerase II. Furthermore, PRC1 is known to associate with histone deacetylases (HDACs, which can inhibit transcription), and PRC1 and H3K27me3 can block positively-acting imprints such as H3K4 methylation [15], [17]. It is well-known that the histone demethylase jumonji domain containing 3 (JMJD3) [19], [20], [21], [22] and the histone methyltransferase enhancer of zeste homolog 2 (EZH2) [23], [24] can both regulate H3K27me3 levels.

Hematopoietic progenitor kinase 1 (HPK1, also known as MAP4K1) is a mammalian Ste20-related serine/threonine protein kinase. It belongs to the germinal center kinase (GCK) family and can be activated by a variety of signal stimuli, such as epidermal growth factor (EGF) [25], prostaglandin E2 (PGE2) [26], transforming growth factor-β (TGF-β) [27], erythropoietin (EPO) [28], and TCR and BCR stimulation [29], [30], [31], [32]. It is also involved in various cellular events, such as MAPK [33], [34], [35], [36], NF-κB [29], [37], [38], [39] and cytokine signalings [28], [40], [41], as well as cellular proliferation and apoptosis [31], [42], [43], [44]. In addition, HPK1 negatively regulates TCR signaling and T cell-mediated immune responses [40], [43]. Shui et al. found that HPK1−/− mice T cells become hyperproliferative in response to stimulation with anti-CD3 and anti-CD28 antibodies, and that these cells can produce more proinflammatory cytokines when immunized with T cell-dependent antigens. Furthermore, T cell-dependent humoral responses in HPK1−/− immunized mice are more vigorous than in controls, and HPK1−/− mice demonstrate even more severe autoimmune phenotypes in an experimental autoimmune encephalomyelitis (EAE) model [43]. The T cell phenotypes observed in these HPK1-deficient mice are reminiscent of those observed in the T cells of patients with SLE, suggesting that HPK1 may play a causative role in SLE pathogenesis.

In then present study, chromatin immunoprecipitation (ChIP) microarray analysis revealed that H3K27me3 enrichment at the HPK1 promoter was significantly higher in CD4+ T cells of patients with SLE than in healthy controls. According to this clue, we set out to further investigate the putative roles of HPK1 in the development of autoimmunity in SLE and epigenetic regulation of its expression. To achieve this, we first confirmed that HPK1 mRNA and protein expressions were significantly decreased in SLE CD4+ T cells. Secondly, we showed that HPK1 negatively regulated CD4+ T cell activation and production of IFNγ and IgG in both healthy and SLE CD4+ T cells. We also observed elevated H3K27me3 enrichment and decreased JMJD3 binding at the HPK1 promoter in SLE CD4+ T cells, with no change in EZH2 binding. By down- and up-regulating JMJD3 expression using JMJD3-siRNA and JMJD3-plasmid, respectively, we demonstrated that changes in HPK1 expression were associated with changes in JMJD3 binding and H3K27me3 enrichment at its promoter. Together, these results provide novel insights into the mechanisms that cause SLE, and suggest a new approach for the treatment of SLE.

Section snippets

Subjects

15 patients with SLE and 15 age- and sex-matched healthy controls were enrolled in this study. Relevant patient information is listed in Table 1. All patients (n = 15, age: 28.60 ± 6.25 years) were recruited from the out-patient clinic and in-patient ward of the Department of Dermatology of the Second Xiangya Hospital, Central South University, China. All patients fulfilled at least 4 of the American College of Rheumatology (ACR) Classification Criteria for SLE [45]. Disease activity was

Increased H3K27me3 enrichment at the HPK1 promoter in SLE CD4+ T cells in the result of ChIP microarrays

We first used ChIP microarray analysis to examine H3K27me3 enrichment at various gene promoters in pooled CD4+ T cell lysates from 5 SLE patients and 5 age- and sex-matched healthy controls. Based on the microarray results, out of 20,832 distinct gene promoters screened, 552 showed a greater than two-fold difference in H3K27me3 enrichment between SLE and control CD4+ T cells. Among these, H3K27me3 enrichment at the HPK1 promoter in SLE CD4+ T cells was approximately five times higher than in

Discussion

In recent years, accumulating evidence has demonstrated that epigenetic alterations play essential roles in the pathogenesis of SLE [7], [8], [9], [10]. In particular, considerable interest has been focused on DNA demethylation, and a growing number of studies indicate that demethylation of regulatory sequences can result in the overexpression of certain immunity-related genes, including perforin (PRF1) [47], CD11a (ITGAL) [48], [49], CD70 (TNFSF7) [48], [50], and CD40 ligand (CD40L) [51], [52]

Acknowledgments

The authors gratefully acknowledge Dr. Charlie Degui Chen and Dr. Rüdiger Arnold for providing plasmids. This work was supported by the National Natural Science Foundation of China (grant 30730083 and 81101194) and the National Basic Research Program of China (973 Plan, grant 2009CB825605)

References (65)

  • H. Maciejewska-Rodrigues et al.

    Epigenetics and rheumatoid arthritis: the role of SENP1 in the regulation of MMP-1 expression

    J Autoimmun

    (2010)
  • A.H. Lund et al.

    Polycomb complexes and silencing mechanisms

    Curr Opin Cell Biol

    (2004)
  • R. Cao et al.

    The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3

    Curr Opin Genet Dev

    (2004)
  • T. Swigut et al.

    H3K27 demethylases, at long last

    Cell

    (2007)
  • F. De Santa et al.

    The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing

    Cell

    (2007)
  • S. Fujii et al.

    Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation

    J Biol Chem

    (2008)
  • S. Sawasdikosol et al.

    Hematopoietic progenitor kinase 1 (HPK1) negatively regulates prostaglandin E2-induced fos gene transcription

    Blood

    (2003)
  • G. Zhou et al.

    Hematopoietic progenitor kinase 1 is a component of transforming growth factor beta-induced c-Jun N-terminal kinase signaling cascade

    J Biol Chem

    (1999)
  • Y. Nagata et al.

    Activation of hematopoietic progenitor kinase-1 by erythropoietin

    Blood

    (1999)
  • G. Zhou et al.

    Protein phosphatase 4 is a positive regulator of hematopoietic progenitor kinase 1

    J Biol Chem

    (2004)
  • S. Sawasdikosol et al.

    Prostaglandin E2 activates HPK1 kinase activity via a PKA-dependent pathway

    J Biol Chem

    (2007)
  • Y. Ito et al.

    Interaction of hematopoietic progenitor kinase 1 and c-Abl tyrosine kinase in response to genotoxic stress

    J Biol Chem

    (2001)
  • M. Lewitzky et al.

    Mona/Gads SH3C binding to hematopoietic progenitor kinase 1 (HPK1) combines an atypical SH3 binding motif, R/KXXK, with a classical PXXP motif embedded in a polyproline type II (PPII) helix

    J Biol Chem

    (2004)
  • D. Brenner et al.

    Caspase-cleaved HPK1 induces CD95L-independent activation-induced cell death in T and B lymphocytes

    Blood

    (2007)
  • J. Schulze-Luehrmann et al.

    Hematopoietic progenitor kinase 1 supports apoptosis of T lymphocytes

    Blood

    (2002)
  • M. Zhao et al.

    Epigenetics and SLE: RFX1 downregulation causes CD11a and CD70 overexpression by altering epigenetic modifications in lupus CD4+ T cells

    J Autoimmun

    (2010)
  • Q. Lu et al.

    Effect of DNA methylation and chromatin structure on ITGAL expression

    Blood

    (2002)
  • A.H. Sawalha et al.

    Autoimmunity and Klinefelter’s syndrome: when men have two X chromosomes

    J Autoimmun

    (2009)
  • P. Invernizzi et al.

    Female predominance and X chromosome defects in autoimmune diseases

    J Autoimmun

    (2009)
  • R. Arnold et al.

    Caspase-mediated cleavage of hematopoietic progenitor kinase 1 (HPK1) converts an activator of NFkappaB into an inhibitor of NFkappaB

    J Biol Chem

    (2001)
  • D. Brenner et al.

    Concepts of activated T cell death

    Crit Rev Oncol Hematol

    (2008)
  • S. Zhao et al.

    Epigenetic perspectives in systemic lupus erythematosus: pathogenesis, biomarkers, and therapeutic potentials

    Clin Rev Allergy Immunol

    (2010)
  • Cited by (76)

    • Epigenetic basis of autoimmune disorders in humans

      2023, Epigenetics in Human Disease, Third Edition
    • The development of small-molecule inhibitors targeting HPK1

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      These signaling pathways are also related to the regulation of cell proliferation, activation and apoptosis. Therefore, abnormal expression of HPK1 and related proteins may contribute to the occurrence of autoimmune diseases and other consequences, such as systemic lupus erythematosus (SLE) [87,88]. HPK1 protein kinase has also been shown to be involved in the incidence and progression of various human malignant tumors [89–94].

    • Drug discovery for epigenetics targets

      2022, Drug Discovery Today
    • KDM6B epigenetically regulated-interleukin-6 expression in the dorsal root ganglia and spinal dorsal horn contributes to the development and maintenance of neuropathic pain following peripheral nerve injury in male rats

      2021, Brain, Behavior, and Immunity
      Citation Excerpt :

      A great number of studies have shown that KDM6B enhances proinflammatory genes expression, which is typically associated with the activation of NF-κB, STAT, and TGF-β/SMAD3 signaling (Kruidenier et al., 2012; Malinczak et al., 2020; Na et al., 2017; Przanowski et al., 2014; Salminen et al., 2014). Diverse works have reported that both pharmacological inhibition of and deficiency in KDM6B have a protective role in inflammatory diseases, such as early sepsis (Pan et al., 2018), wound healing(Na et al., 2017), neuroinflammation (Wang et al., 2020), blood spinal cord barrier disruption (Lee et al., 2016), encephalomyelitis (EAE) (Doñas et al., 2016), systemic lupus erythematosus (Zhang et al., 2011), and arthritic and inflammatory pain (Jia et al., 2018; Achuthan et al., 2016) through inhibiting the production of inflammatory cytokines in immune cells via various signaling pathways. Recent studies have revealed that abnormal expression of KDM6B in brain involves in the pathogenesis of alcohol dependence (Johnstone et al., 2021), cocaine reward memory (Zhang et al., 2018), and increased susceptibility of depression (Wang et al., 2020) by epigenetically regulation of proinflammatory signaling pathway in several preclinical rodent models.

    View all citing articles on Scopus
    View full text