Cancer Letters

Cancer Letters

Volume 446, 1 April 2019, Pages 38-48
Cancer Letters

Original Articles
Multiple myeloma cell-derived IL-32γ increases the immunosuppressive function of macrophages by promoting indoleamine 2,3-dioxygenase (IDO) expression

https://doi.org/10.1016/j.canlet.2019.01.012Get rights and content

Highlights

  • IL-32 was universally expressed in BM biopsies from MM patients.

  • IL-32 expression was positively correlated with MM clinical stage.

  • IL-32γ in MM cells induced IDO production in MΦs through PR3.

  • IDO-positive IL-32γ-educated MΦs inhibited proliferation and effector function of CD4+ T cells.

  • IL-32γ-induced IDO expression depended on STAT3 and NF-κB pathways.

Abstract

The interaction of multiple myeloma (MM) cells with macrophages (MΦs) contributes to the pathophysiology of MM. We previously showed that IL-32 is overexpressed in MM patients. The present study was designed to explore the clinical significance of IL-32 in MM and to further elucidate the mechanisms underlying the IL-32-mediated immune function of MΦs. Our results showed that high IL-32 expression in MM patients was associated with more advanced clinical stage. RNA-sequencing revealed that IL-32γ significantly induced the production of the immunosuppressive molecule indoleamine 2,3-dioxygenase (IDO) in MΦs, and this effect was verified by qRT-PCR, western blotting, and immunofluorescence. Furthermore, MM cells with IL-32-knockdown showed a reduced ability to promote IDO expression. As a binding protein for IL-32, proteinase 3 (PR3) was universally expressed on the surfaces of MΦs, and knockdown of PR3 or inhibition of the STAT3 and NF-κB pathways hindered the IL-32γ-mediated stimulation of IDO expression. Finally, IDO-positive IL-32γ-educated MΦs inhibited CD4+ T cell proliferation and IL-2, IFN-γ, and TNF-α production. Taken together, our results indicate that IL-32γ derived from MM cells promotes the immunosuppressive function of MΦs and is a potential target for MM treatment.

Introduction

Multiple myeloma (MM) is an incurable B cell tumor characterized by the accumulation of malignant plasma cells in the bone marrow (BM) [1]. The interaction of malignant plasma cells with the BM microenvironment contributes to their growth, proliferation, invasion, metastasis, and chemoresistance [2]. The MM–BM microenvironment is composed of cells, extracellular matrix, and soluble factors, among which macrophages (MΦs) are an abundant and important component [3,4]. Several studies have shown that the degree of MΦ infiltration can serve as a prognostic marker in newly diagnosed patients with MM [5,6]. Along with promoting angiogenesis through vasculogenic mimicry, MM-associated MΦs (mMΦs) protect MM cells from spontaneous and chemotherapy-induced apoptosis through both contact-mediated and non-contact-mediated mechanisms [7,8]. mMΦs therefore represent a potential target for myeloma treatment [9,10], and it is essential to explore the mechanisms underlying MΦ infiltration and the polarization of normal MΦs to mMΦs. Research has shown that MM cells recruit tumor-supportive MΦs and promote M2-like MΦ polarization through the CXCR4/CXCL12 axis [11]. Our previous study also showed that the chemokines CCL2, CCL3, and CCL14 promoted MΦ infiltration in the MM–BM microenvironment and increased MΦ proliferation [12]. While these studies indicated that cytokines from MM cells affect MΦs, few studies have focused on the MM cell-mediated immunosuppressive function of MΦs involving T cells.

Interleukin 32 (IL-32), a proinflammatory cytokine, was identified as natural killer cell transcript 4 (NK4) in activated natural killer cells and T cells [13]. There are more than nine alternatively spliced isoforms of IL-32, each with distinct functions. Among the variants, IL-32β is the most abundant isoform, and IL-32γ appears to be the most potent isoform [[14], [15], [16]]. The definitive receptor of IL-32 has not been identified, although proteinase 3 (PR3) has been revealed to be a specific IL-32-binding protein [17]. Along with being involved in numerous inflammatory and infectious diseases [18,19], IL-32 plays a crucial role in the development of both hematologic malignancies and solid tumors [20,21]. Some reports have shown that IL-32 promotes the progression of various malignancies, such as gastric cancer, lung cancer, and cutaneous T-cell lymphoma [[22], [23], [24]]. However, conflicting reports have shown a tumor suppressive role of IL-32 in colon cancer and melanoma [25,26]. We previously reported that IL-32 is overexpressed in the BM and peripheral blood of MM patients. Moreover, IL-32 induces the production of IL-6 in bone marrow stromal cells (BMSCs) [27]. Recently, another report showed that IL-32 promotes osteoclast differentiation in MM [28]. In this study, we further investigated the association between IL-32 expression and clinical features of newly diagnosed MM patients and explored the effects and underlying mechanisms of IL-32 and MΦs in the MM–BM microenvironment.

Section snippets

Cell culture

Human MM cell lines RPMI 8226, OPM2, CAG, ARK, MM.1S, ARP-1, H929, and LP-1 were generously provided by Dr. Qing Yi (Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA). All MM cell lines were cultured in RPMI-1640 medium containing 1% l-glutamine and 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Waltham, MA, USA). To prepare conditioned media (CM), MM cells were seeded at 5 × 105 cells/ml for 24 h. Culture supernatants were collected and

High IL-32 expression in MM correlated with clinical stage

We studied IL-32 expression in BM biopsies from newly diagnosed MM patients by immunohistochemical staining using anti-IgG as an isotype control (Sup.Fig. 1A). IL-32 was detectable in all patients, and approximately 73.9% (17/23) of the samples showed high expression (Fig. 1A). The expression levels of IL-32 and clinicopathological details of the patients are shown in Table 1. IL-32 expression was significantly and positively associated with the International Staging System (ISS) stage

Discussion

We report herein that high IL-32 expression in MM patients was associated with more advanced clinical stage. IL-32γ in MM cells induced IDO production in MΦs through PR3, which was dependent on the activation of the STAT3 and NF-κB pathways. IDO-positive IL-32γ-educated MΦs suppressed the proliferation of CD4+ T cells and their IL-2, IFN-γ, and TNF-α production in response to polyclonal stimulation.

IL-32 is a proinflammatory cytokine involved in various inflammatory diseases and cancers [18].

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgements

We thank all study participants for their contributions to this project and appreciate professor Qing Yi for guiding the experiments. We thank Elsevier (https://wileyeditingservices.com/en/) for English language editing.

References (44)

  • M. Moschetta et al.

    Targeting the bone marrow microenvironment

    Immunol. Rev.

    (2015)
  • G. Shay et al.

    Dissecting the multiple myeloma-bone microenvironment reveals new therapeutic opportunities

    J. Mol. Med.

    (2016)
  • S. Panchabhai et al.

    Tumor-associated macrophages and extracellular matrix metalloproteinase inducer in prognosis of multiple myeloma

    Leukemia

    (2016)
  • Y. Zheng et al.

    PSGL-1/selectin and ICAM-1/CD18 interactions are involved in macrophage-induced drug resistance in myeloma

    Leukemia

    (2013)
  • J. Chen et al.

    BAFF is involved in macrophage-induced bortezomib resistance in myeloma

    Cell Death Dis.

    (2017)
  • Q. Wang et al.

    Therapeutic effects of CSF1R-blocking antibodies in multiple myeloma

    Leukemia

    (2018)
  • K. Beider et al.

    Multiple myeloma cells recruit tumor-supportive macrophages through the CXCR4/CXCL12 axis and promote their polarization toward the M2 phenotype

    Oncotarget

    (2014)
  • Y. Li et al.

    Chemokines CCL2, 3, 14 stimulate macrophage bone marrow homing, proliferation, and polarization in multiple myeloma

    Oncotarget

    (2015)
  • C.A. Dahl et al.

    Identification of a novel gene expressed in activated natural killer cells and T cells

    J. Immunol.

    (1992)
  • C. Goda et al.

    Involvement of IL-32 in activation-induced cell death in T cells

    Int. Immunol.

    (2006)
  • J.D. Choi et al.

    Identification of the most active interleukin-32 isoform

    Immunology

    (2009)
  • D. Novick et al.

    Proteinase 3 is an IL-32 binding protein

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • Cited by (36)

    • IL-32 and its paradoxical role in neoplasia

      2023, Critical Reviews in Oncology/Hematology
    • Diverse and divergent functions of IL-32β and IL-32γ isoforms in the regulation of malignant pleural mesothelioma cell growth and the production of VEGF-A and CXCL8

      2023, Cellular Immunology
      Citation Excerpt :

      Interleukin (IL)-32, primarily designated as natural killer cell transcript 4 (NK4), is a novel molecule prominently expressed by immune cell lineages including macrophages, monocytes, NK cells and CD4+ and CD8+ T cells in response to stimulation with various biological mediators such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-2, IL-12 and IL-18 [1–6]. Moreover, a variety of nonimmune sells such as fibroblasts, epithelial cells, endothelial cells, hepatocytes, mesenchymal stroma cells also express IL-32 [7–12]. Among these cell types, endothelial cells and differentiated colon epithelial cells are constitutively expressing IL-32.

    • Metabolic regulation of the bone marrow microenvironment in leukemia

      2021, Blood Reviews
      Citation Excerpt :

      IDO exerts its inhibitory effect on T cells via tryptophan deprivation (through its metabolism), as well as by inducing the expansion of Treg cells [108]. In macrophages, IL-23 induces the upregulation of IDO expression via the STAT3 and NF-κB pathways [109]. AML cells and leukemic DCs induce T cell tolerance by directly converting CD4+CD25− T cells into CD4+CD25+ Treg cells and inhibiting T cell proliferation through an IDO-dependent mechanism [108,110].

    View all citing articles on Scopus
    1

    These authors have contributed equally to this work.

    View full text