Cancer Letters

Cancer Letters

Volume 364, Issue 2, 10 August 2015, Pages 118-124
Cancer Letters

Original Articles
Lipid mediator lipoxin A4 inhibits tumor growth by targeting IL-10-producing regulatory B (Breg) cells

https://doi.org/10.1016/j.canlet.2015.04.030Get rights and content

Highlights

  • Lipoxin A4 (LXA4), an arachidonic acid-derived anti-inflammatory lipid mediator, has demonstrated anti-tumor effects.

  • This study reveals that LXA4 inhibits the generation of IL-10-producing Breg cells in the tumor microenvironment, and downregulates the development of Breg cell-dependent Foxp3+ Treg cells, leading to enhanced antitumor immunity.

  • LXA4 doesn’t have direct effect on the induction of Treg cells in vitro, but it can inhibit Treg cells differentiation in B cell-dependent manner.

  • LXA4 inhibits the generation of IL-10-producing Breg cells by inhibiting phosphorylation of transcriptional factor ERK and STAT3.

Abstract

Lipoxin A4 (LXA4), an arachidonic acid-derived anti-inflammatory lipid mediator, shows anti-tumor potential by regulating tumor immune microenvironments. However, the underlying molecular and cellular basis of this function remains unclear. IL-10-producing B (Breg) cells display tumor-promoting effects by negatively regulating anti-tumor immunity. Here we show that LXA4 inhibits tumor growth by suppressing the generation of Breg cells in tumor-bearing mice. The administration of LXA4 inhibited the induction of Breg cells. Breg cell deficiency, in turn, resulted in LXA4 losing its anti-tumor properties. Intriguingly, regulatory T (Treg) cells also had a role in this process. Targeting Breg cells by LXA4 decreased the number of Treg cells in draining lymph nodes and tumor tissues as well as enhanced cytotoxic T cell activities. In addition, we further demonstrated that LXA4 inhibited Breg cells through its dephosphorylating STAT3 and ERK. These findings unveil a new anti-tumor mechanism underlying LXA4 targeting Breg cells with potential clinical applications.

Introduction

B lymphocytes are well known to function as antibody-synthesizing machines to protect the host against invasive pathogens. However, mounting evidence indicates that B cells also play an essential role in various immune pathologies including malignant diseases [1], [2]. Previous studies showed that B cells inhibited the induction of T cell-dependent tumor immunity and B cells can induce CD8 T cells to become anergic [3]. On the other hand, depletion of B cells enhances anti-tumor cytotoxic T cell response, leading to effective clearance of tumor cells [4]. In clinic, there are reports that the presence of B cells is correlated with worse outcome [5], [6], regardless of other contrary findings. It has now become clear that a subset of IL-10-producing regulatory B (Breg) cells play a pivotal role in mediating immunosuppression. It has been reported that Breg cells can suppress Th1 and Th17 responses [7]. In murine models, adoptive transfer of Breg cells can ameliorate the symptoms of experimental autoimmune encephalomyelitis (EAE) [8]. In human systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), disease activities are closely related to the number and function of Breg cells [9], [10]. Initially, IL-10 production is considered as a hallmark of Breg cells; however, other cytokines like lymphotoxin and IL-35 produced by B cells also display suppressive properties [11], [12]. In addition to producing immunosuppressive cytokines, Breg cells also act directly on natural killer (NK) and dendritic cells to restrain their activities [13], [14]. Furthermore, Breg cells indirectly induce and maintain Treg cells as immune suppressors [15]. Currently, though the mechanisms by which Breg cells regulate immune responses have been widely investigated, how Breg cell numbers and their function are controlled remains poorly understood.

Lipoxin A4, an endogenous eicosanoid, is highlighted in the regulation of inflammation [16]. Lipoxin A4 is synthesized locally from arachidonic acid at inflammation sites via a transcellular biosynthesis mechanism [17]. By specifically binding to two distinct receptors: formyl peptide receptor 2 (FPR2), a surface membrane G-protein-coupled receptor, and the aryl hydrocarbon receptor (AhR), a ligand-activated nuclear transcription factor, LXA4 strongly inhibits the trafficking of leukocytes to the inflammatory site and stimulates the phagocytosis of apoptotic cells by tissue macrophages [18], [19]. Roles of LXA4 in tumorigenesis have been explored using different tumor models [20], [21], [22]. Although those results have shown anti-tumor effects of LXA4, the underlying mechanism remains unclear. In this study, we provide evidence that LXA4 exerts the anti-tumor effect through inhibiting the generation of IL-10 producing Breg cells. Our findings suggest that LXA4 functions as an endogenous immune modulator against tumors.

Section snippets

Cell lines and mice

Murine hepatocarcinoma H22 (BALB/c), melanoma B16 (C57BL/6) and colorectal carcinoma CT26 (BALB/c) cell lines were purchased from the China Center for Type Culture Collection (Wuhan, China) and cultured according to the guidelines given. Female BALB/c and C57BL/6 mice, 8- to 10-week-old, were purchased from the Center of Medical Experimental Animals of Hubei Province (Wuhan, China). Female BALB/c nude and CB-17 SCID mice, 6- to 8-week-old, were purchased from Vital River Laboratory Animal

Anti-tumor effects of LXA4 in different murine tumor models

To investigate the anti-tumor effect of LXA4, a murine H22 hepatocarcinoma tumor model was first tested. After subcutaneous injection of H22 tumor cells (2 × 105), the tumor-bearing mice were intraperitoneally administered with BML-111, an agonist of LXA4 receptor, once per two days. The result showed that the administration of BML-111 significantly inhibited H22 tumor growth (Fig. 1A). Besides the H22 tumor model, murine B16 melanoma and CT26 colon cancer tumor models were also conducted.

Discussion

Lipid metabolites are critical in the immunoregulation of both innate and adaptive immune responses. Many lipid mediators show immunosuppressive action, and potentially favor tumor immune evasion. However little is known about the process of restraining tumor immune evasion by lipid mediators. In this study, we demonstrate that arachidonic acid-derived lipid mediator LXA4 can upregulate anti-tumor immunity by targeting an important immunosuppressive B cell subset, Breg cells.

The role of B cells

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

This work was supported by National Basic Research Program of China (2014CB542100), National Science Fund for Distinguished Young Scholars of China (81225021), National Natural Science Foundation of China (81472653), and Special Fund of Health Public Welfare Profession of China (201302018).

References (43)

  • T. Matsushita et al.

    Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression

    J. Clin. Invest

    (2008)
  • P.A. Blair et al.

    Selective targeting of B cells with agonistic anti-CD40 is an efficacious strategy for the generation of induced regulatory T2-like B cells and for the suppression of lupus in MRL/lpr mice

    J. Immunol

    (2009)
  • F. Flores-Borja et al.

    CD19+ CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation

    Sci. Transl. Med

    (2013)
  • M. Ammirante et al.

    B-cell-derived lymphotoxin promotes castration-resistant prostate cancer

    Nature

    (2010)
  • P. Shen et al.

    IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases

    Nature

    (2014)
  • S. Inoue et al.

    Inhibitory effects of B cells on antitumor immunity

    Cancer Res

    (2006)
  • Y. Zhang et al.

    B lymphocyte inhibition of anti-tumor response depends on expansion of Treg but is independent of B-cell IL-10 secretion

    Cancer Immunol. Immunother

    (2013)
  • C.N. Serhan et al.

    Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators

    Nat. Rev. Immunol

    (2008)
  • G. Folco et al.

    Eicosanoid transcellular biosynthesis: from cell-cell interactions to in vivo tissue responses

    Pharmacol. Rev

    (2006)
  • A. Ariel et al.

    Aspirin-triggered lipoxin A4 and B4 analogs block extracellular signal-regulated kinase-dependent TNF-α secretion from human T cells

    J. Immunol

    (2003)
  • B. Zhang et al.

    Depletion of regulatory T cells facilitates growth of established tumors: a mechanism involving the regulation of myeloid-derived suppressor cells by lipoxin A4

    J. Immunol

    (2010)
  • Cited by (53)

    • Characteristics of tumor microenvironment and novel immunotherapeutic strategies for non-small cell lung cancer

      2022, Journal of the National Cancer Center
      Citation Excerpt :

      In patients with NSCLC, the immunoglobulin kappa C expressing plasma cells in stroma could predict the prognosis, while intratumoral plasma cells could predict outcomes of PD-L1 blockade.54,55 In addition, IL-10-producing Bregs can be targeted by lipid mediator lipoxin A4 by dephosphorylating signal transducer and activator of transcription 3 (STAT3) and ERK, thus inhibiting tumor growth.56 Overall, TIL-Bs have both antitumor and pro-tumor functions, and their functional phenotypes change dynamically under the influence of the TME.

    • Colorectal cancer: Metabolic interactions reshape the tumor microenvironment

      2022, Biochimica et Biophysica Acta - Reviews on Cancer
    • Role of immune regulatory cells in breast cancer: Foe or friend?

      2021, International Immunopharmacology
      Citation Excerpt :

      The production of these antigens can increase the frequency of infiltrated effector T cells, which is associated with the stimulation of anti-tumor responses and more remarkable survival in patients with breast cancer [29]. Besides, regulatory cells including Tregs, Bregs, TAMs, TANs, tDCs, MDSCs, MSCs, and NKregs play an essential role in tolerance to self-antigens preventing autoimmunity in a protective manner [31–33]. In patients with autoimmune diseases, the function of these cells is usually impaired, and the self-tolerance is broken [31].

    • Carcinogenesis: Failure of resolution of inflammation?

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      The lipoxin analogue BML-111 inhibited serum VEGF production in vivo in H-22 tumor bearing mice (Chen et al., 2010). In murine hepatocarcinoma, melanoma and CRC xenograft models, LXA4 suppressed tumor growth by targeting regulatory B cells (Bregs) through the inhibition of important signaling pathways (Wang et al., 2015)(). Bregs inhibit functions of CD8+ cytotoxic T cell in the tumor microenvironment.

    View all citing articles on Scopus
    1

    Z. Wang and Q. Cheng contributed equally to this work.

    View full text