Lipid metabolism as a mechanism of immunomodulation in macrophages: the role of liver X receptors

https://doi.org/10.1016/j.coph.2020.02.003Get rights and content

Macrophages are immune myeloid cells with an extreme ability to modulate their phenotype in response to insults and/or pathogens. The immunomodulatory capacity of macrophages is also patent during development as they adapt their phenotype to the host tissue environment establishing the heterogeneous populations of tissue-resident macrophages. An important mechanism of immunomodulation in macrophages occurs through the regulation of transcriptional activity. Numerous transcription factors are associated with macrophage plasticity, among them, several nuclear receptors. The nuclear receptors Liver X Receptors (LXRα and LXRβ) have also revealed as active players during macrophage adaptations in diverse scenarios. This review will address the different mechanisms by which LXRs contribute to immunomodulation in macrophages by connecting lipid metabolism and immunity through transcriptional regulation.

Introduction

The Liver X Receptors, LXRs, are crucial transcription factors in macrophages that act as important mediators in cholesterol metabolism and modulate several anti-inflammatory pathways [1]. There are two isoforms of LXRs, LXRα, encoded by the gene Nr1h3, and LXRβ, encoded by the gene Nr1h2, with highly similar sequence but diverse tissue distribution. While LXRα shows a higher expression in adipose tissue, liver and macrophages, LXRβ expression is equally found in all tissues and cell types analyzed. Their transcriptional regulation depends on activation by their endogenous ligands, such as oxidized forms of cholesterol, desmosterol and elevated concentrations of d-glucose, as well as synthetic ligands that provide promising therapeutic approaches [2]. Numerous studies investigating Lxrα−/−, Lxrβ−/−, or Lxrα/β−/− mice show mostly overlapping, but also some specific functions of both isoforms of LXR. For example, mice deficient in LXRα lose their natural resistance to a diet rich in cholesterol, showing an accumulation of hepatic cholesterol and a substantially altered conversion of cholesterol into bile acid [3]. Moreover, LXRα is specifically implicated in the developmental pathways that lead to the establishment of tissue-resident macrophages both in the liver and in the spleen [4,5]. It has been long demonstrated that LXRs play a central role in the anti-inflammatory response in macrophages and that a deficient regulation of their activity can lead to chronic inflammatory conditions [6]. Consequently, targeting LXR nuclear receptors activity in macrophages has a great immunomodulatory potential in metabolic and inflammatory disease. An important challenge to overcome in the therapeutic approaching to target LXRs is the connection between metabolism and immune regulation. Hence, it is crucial to understand LXR transcriptional activity as a central regulator of immunometabolic pathways that determine macrophage immunomodulation. Although evident links between lipid metabolism and immunity are regulated through LXR transcriptional activity, the specific mechanisms of this regulation in vivo have remained difficult to decipher. In this review, we address the central role of the LXR nuclear receptors as regulators of macrophage immune plasticity during metabolic and inflammatory diseases, as well as during development. Moreover, this review gives an overview on the diverse mechanisms by which LXR-dependent lipid metabolic pathways interconnect with immune functions in macrophages, defining novel mechanisms of immunomodulation.

Section snippets

Pleiotropic effects of liver X receptors in lipid-related pathways in macrophages

The first evidences of the role of LXRs in lipid metabolism were provided by the group of David Mangelsdorf in 1998, when they described the regulation of the gene CYP7A1 (an enzyme responsible for bile acid synthesis) by LXRα in mice [3,7]. This study showed that high cellular cholesterol levels are necessary to induce the upregulation of CYP7A1 expression via LXRα, supporting that the endogenous ligands of LXRα are derivate forms of cholesterol, as it was previously shown in vitro [8]. Over

Anti-inflammatory response via LXR-transcriptional regulation

Several studies have explored the role of LXRs in macrophage inflammatory pathways, showing a reciprocal relationship between lipid metabolism and inflammatory gene expression in macrophages [35,36,37]. The first studies on LXRs and inflammation showed that the activation of LXR by their ligands inhibit the expression of inflammatory mediators (iNOS, Il-6) in macrophages in response to bacterial infection or LPS stimulation in vitro [38]. In vivo, LXR agonists reduce inflammation in

Roles of LXR in macrophage development and differentiation

Over the last decade, our view of tissue-resident macrophage development has evolved from the general dogma that all tissue macrophages develop through differentiation from monocyte precursors [56,57]. Several tissue-resident macrophage populations, such as microglia in the central nervous system and Langerhans cells in the skin, are established embryonically and derived from yolk sac progenitors that self-maintain in the adult through proliferation [58]. Furthermore, the capacity to

Conclusions and future perspectives

Macrophages have the great ability to adopt different phenotypes and functions in response to environmental stimuli. This immune plasticity is modulated through the regulation of numerous signaling pathways and metabolic routes. The LXR nuclear receptors have arisen as key modulators of the immune phenotype and functions in macrophages using several mechanisms in health and disease. For many years the dual role of LXRs in macrophages regulating cholesterol metabolism and modulating the

Conflict of interest statement

  • All authors have participated in (a) conception and design, or analysis and interpretation of the data; (b) drafting the article or revising it critically for important intellectual content; and (c) approval of the final version.

  • This manuscript has not been submitted to, nor is under review at, another journal or other publishing venue.

  • The authors have no affiliation with any organization with a direct or indirect financial interest in the subject matter discussed in the manuscript.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

CRediT authorship contribution statement

Sophia Leussink: Writing - original draft. Irene Aranda-Pardos: Writing - original draft. Noelia A-Gonzalez: Writing - original draft, Supervision.

References (74)

  • S.B. Joseph et al.

    LXR-dependent gene expression is important for macrophage survival and the innate immune response

    Cell

    (2004)
  • A.F. Valledor et al.

    Activation of liver X receptors and retinoid X receptors prevents bacterial-induced macrophage apoptosis

    Proc Natl Acad Sci U S A

    (2004)
  • J. Matalonga et al.

    The nulear receptor LXR limits bacterial infection of host macrophages through a mechanism that impacts cellular NAD metabolism

    Cell Rep

    (2017)
  • N. A-Gonzalez et al.

    Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR

    Immunity

    (2009)
  • C. Hong et al.

    Coordinate regulation of neutrophil homeostasis by liver X receptors in mice

    J Clin Invest

    (2012)
  • M.J. Kim et al.

    Apoptotic cells trigger the ABCA1/STAT6 pathway leading to PPAR-γ expression and activation in macrophages

    J Leukoc Biol

    (2018)
  • E.D. Muse et al.

    Cell-specific discrimination of desmosterol and desmosterol mimetics confers selective regulation of LXR and SREBP in macrophages

    Proc Natl Acad Sci U S A

    (2018)
  • C.Z. Han et al.

    Metabolic connections during apoptotic cell engulfment

    Cell

    (2011)
  • M. Guilliams et al.

    Developmental and functional heterogeneity of monocytes

    Immunity

    (2018)
  • W. T’Jonck et al.

    Niche signals and transcription factors involved in tissue-resident macrophage development

    Cell Immunol

    (2018)
  • K.M. Robertson Remen et al.

    Activation of liver X receptor (LXR) inhibits receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation in an LXRβ-dependent mechanism

    J Biol Chem

    (2011)
  • J.M.M. Den Haan et al.

    Innate immune functions of macrophage subpopulations in the spleen

    J Innate Immun

    (2012)
  • T.B.H. Geijtenbeek et al.

    Marginal zone macrophages express a murine homologue of DC-SIGN that captures blood-borne antigens in vivo

    Blood

    (2002)
  • H. Borges Da Silva et al.

    Splenic macrophage subsets and their function during blood-borne infections

    Front Immunol

    (2015)
  • G. Bories et al.

    Liver X receptor activation stimulates iron export in human alternative macrophages

    Circ Res

    (2013)
  • M.C. Gage et al.

    Disrupting LXRα phosphorylation promotes FoxM1 expression and modulates atherosclerosis by inducing macrophage proliferation

    Proc Natl Acad Sci U S A

    (2018)
  • I.G. Schulman

    Liver X receptors link lipid metabolism and inflammation

    FEBS Lett

    (2017)
  • R. Komati et al.

    Ligands of therapeutic utility for the liver X receptors

    Molecules

    (2017)
  • N. A-Gonzalez et al.

    The nuclear receptor LXRα controls the functional specialization of splenic macrophages

    Nat Immunol

    (2013)
  • D. Duc et al.

    Oxysterols in autoimmunity

    Int J Mol Sci

    (2019)
  • J.M. Lehmann et al.

    Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway

    J Biol Chem

    (1997)
  • B. Wang et al.

    Liver X receptors in lipid signalling and membrane homeostasis

    Nat Rev Endocrinol

    (2018)
  • L. Zhang et al.

    Feedback regulation of cholesterol uptake by the LXR-IDOL- LDLR axis

    Arterioscler Thromb Vasc Biol

    (2012)
  • C.R. Jefcoate et al.

    Cholesterol signaling in single cells: lessons from STAR and sm-FISH

    J Mol Endocrinol

    (2018)
  • T. Yoshikawa et al.

    Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter

    Mol Cell Biol

    (2001)
  • C. Thomas et al.

    LPCAT3 deficiency in hematopoietic cells alters cholesterol and phospholipid homeostasis and promotes atherosclerosis

    Atherosclerosis

    (2018)
  • J.J. Repa et al.

    Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers

    Science (80-)

    (2000)
  • Cited by (0)

    3

    These authors contributed equally.

    View full text