Review
Liver X receptor biology and pharmacology: new pathways, challenges and opportunities

https://doi.org/10.1016/j.tips.2012.03.013Get rights and content

Nuclear receptors (NRs) are master regulators of transcriptional programs that integrate the homeostatic control of almost all biological processes. Their direct mode of ligand regulation and genome interaction is at the core of modern pharmacology. The two liver X receptors LXRα and LXRβ are among the emerging newer drug targets within the NR family. LXRs are best known as nuclear oxysterol receptors and physiological regulators of lipid and cholesterol metabolism that also act in an anti-inflammatory way. Because LXRs control diverse pathways in development, reproduction, metabolism, immunity and inflammation, they have potential as therapeutic targets for diseases as diverse as lipid disorders, atherosclerosis, chronic inflammation, autoimmunity, cancer and neurodegenerative diseases. Recent insights into LXR signaling suggest future targeting strategies aiming at increasing LXR subtype and pathway selectivity. This review discusses the current status of our understanding of LXR biology and pharmacology, with an emphasis on the molecular aspects of LXR signaling that constitute the potential of LXRs as drug targets.

Section snippets

The oxysterol receptors LXRα and LXRβ are members of the NR family of transcription factors

The 48 members of the NR family in humans are master regulators of transcriptional programs that integrate the homeostatic control of almost all biological processes including development, reproduction, cell growth, metabolism, immunity and inflammation. Most NRs appear to regulate gene expression in response to small lipophilic compounds such as steroids, thyroid hormone, fatty acids and cholesterol metabolites. As ligands, these compounds directly bind to the NR ligand-binding domain (LBD)

LXRs as regulators of metabolic function

LXRβ is ubiquitously expressed at a moderate level in most physiological systems, whereas LXRα expression is mostly restricted to metabolically active tissues (www.nursa.org). It is important to note that this is merely based on quantification of mRNA levels and that protein expression levels (and protein stability) are largely unknown. The first studies on the physiological roles of LXR revealed key functions in the control of cholesterol metabolism [14] and lipogenesis [15] in the liver, a

LXRs regulate inflammatory responses and immunity

Considerable evidence has identified both LXRα and LXRβ as anti-inflammatory transcription factors and physiological regulators of innate and adaptive immune responses, apoptosis and phagocytosis. The first studies linking LXRs to inflammatory responses revealed that LXRs antagonized cytokine-mediated expression of proinflammatory genes in macrophages; it was suggested that this is a consequence of transcriptional silencing of the proinflammatory transcription factor nuclear factor (NF)-κB [6].

LXR pathways in cancer cells

LXRs appear to have dual roles in cancer biology. First, LXRs suppress the proliferation of a variety of human cancer cells [86], and second, tumors produce LXR agonists (oxysterols) that inhibit a robust immune response as a mechanism of tumor escape from immune surveillance [55]. At the molecular level, LXRs target the cell cycle at several points. LXRs reduce the expression of positive cell-cycle regulators, whereas they increase the expression of cell-cycle inhibitors. Although LXRs control

Molecular mechanisms of LXR action: focus on cistromes, PTMs and transrepression

According to conventional models, it is assumed that unliganded LXRs are constitutively nuclear and interact with corepressors such as NCoR/SMRT (nuclear receptor corepressor/silencing mediator of retinoic acid and thyroid receptor) on DNA [92]. On binding of ligand to LXRs, the corepressor complex dissociates and coactivators are recruited, leading to activation of transcription (Figure 3a). Recent experiments using chromatin-immunoprecipitation (ChIP)-based techniques, including

Perspectives

The LXR involvement demonstrated in various metabolic, inflammatory and proliferative disease pathways makes them highly interesting pharmaceutical targets for novel therapies (Figure 4). However, current synthetic agonists cause activation of hepatic lipogenic enzymes via LXRα, thereby increasing triglyceride levels, a known risk factor for cardiovascular disease [105]. Clinical trials in humans using the LXR-623 agonist, which appears to activate LXR without causing hepatic lipogenesis, were

Concluding remarks

The recent discoveries of LXR-regulated pathways in inflammation and proliferation, in addition to the well-established role of LXRs in metabolism, have ignited new promise for LXRs as drug targets for metabolic disorders, chronic inflammatory diseases, cancer and neurodegenerative diseases. However, the broad physiological roles of LXRs involve a high risk of unwanted side effects, the most significant being an increased lipogenic profile. This suggests that successful drug development must

Acknowledgments

This study was supported by grants from the Center for Biosciences (E.T., J-Å.G), the Novo Nordisk Foundation (E.T.), the Swedish Research Council (E.T., J-Å.G., K.R.S.), the Welch Foundation (J-Å.G.) and the Emerging Technology Fund of Texas (J-Å.G.). K.R.S. holds a research assistant professorship from the Swedish Research Council (contract no. 522-2008-3745).

References (130)

  • O. Cheng

    Activation of liver X receptor reduces global ischemic brain injury by reduction of nuclear factor-κB

    Neuroscience

    (2010)
  • J.H. Lee

    Differential SUMOylation of LXRα and LXRβ mediates transrepression of STAT1 inflammatory signaling in IFN-γ-stimulated brain astrocytes

    Mol. Cell

    (2009)
  • C.X. Zhang-Gandhi et al.

    Liver X receptor and retinoid X receptor agonists inhibit inflammatory responses of microglia and astrocytes

    J. Neuroimmunol.

    (2007)
  • L. Chang

    Liver-X-receptor activator prevents homocysteine-induced production of IgG antibodies from murine B lymphocytes via the ROS–NF-κB pathway

    Biochem. Biophys. Res. Commun.

    (2007)
  • A. Mallat et al.

    The liver X receptor in hepatic stellate cells: a novel antifibrogenic target?

    J. Hepatol.

    (2011)
  • S.W. Beaven

    Liver X receptor signaling is a determinant of stellate cell activation and susceptibility to fibrotic liver disease

    Gastroenterology

    (2011)
  • K.M. Remen

    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)
  • R. Geyeregger

    Liver X receptors regulate dendritic cell phenotype and function through blocked induction of the actin-bundling protein fascin

    Blood

    (2007)
  • E. Kiss

    Suppression of chronic damage in renal allografts by liver X receptor (LXR) activation relevant contribution of macrophage LXRα

    Am. J. Pathol.

    (2011)
  • M.A. Birrell

    Novel role for the liver X nuclear receptor in the suppression of lung inflammatory responses

    J. Biol. Chem.

    (2007)
  • H. Gong

    Activation of the liver X receptor prevents lipopolysaccharide-induced lung injury

    J. Biol. Chem.

    (2009)
  • L.A. Collins-Racie

    Global analysis of nuclear receptor expression and dysregulation in human osteoarthritic articular cartilage: reduced LXR signaling contributes to catabolic metabolism typical of osteoarthritis

    Osteoarthr. Cartil.

    (2009)
  • R. Koldamova

    The role of ATP-binding cassette transporter A1 in Alzheimer's disease and neurodegeneration

    Biochim. Biophys. Acta

    (2010)
  • K. Saijo

    Nuclear receptors, inflammation, and neurodegenerative diseases

    Adv. Immunol.

    (2010)
  • P. Sacchetti

    Liver X receptors and oxysterols promote ventral midbrain neurogenesis in vivo and in human embryonic stem cells

    Cell Stem Cell

    (2009)
  • C.P. Chuu

    Modulation of liver X receptor signaling as a prevention and therapy for colon cancer

    Med. Hypothese

    (2011)
  • S.J. Bensinger

    LXR signaling couples sterol metabolism to proliferation in the acquired immune response

    Cell

    (2008)
  • S. Heinz

    Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities

    Mol. Cell

    (2010)
  • T. Jakobsson

    GPS2 is required for cholesterol efflux by triggering histone demethylation, LXR recruitment, and coregulator assembly at the ABCG1 locus

    Mol. Cell

    (2009)
  • X. Li

    SIRT1 deacetylates and positively regulates the nuclear receptor LXR

    Mol. Cell

    (2007)
  • M. Chen

    Phosphorylation of the liver X receptors

    FEBS Lett.

    (2006)
  • N.J. McKenna

    Evolution of NURSA, the nuclear receptor signaling atlas

    Mol. Endocrinol.

    (2009)
  • H. Gronemeyer

    Principles for modulation of the nuclear receptor superfamily

    Nat. Rev. Drug Discov.

    (2004)
  • C. Thomas et al.

    The different roles of ER subtypes in cancer biology and therapy

    Nat. Rev. Cancer

    (2011)
  • J.T. Moore

    The nuclear receptor superfamily and drug discovery

    ChemMedChem

    (2006)
  • N. Zelcer et al.

    Liver X receptors as integrators of metabolic and inflammatory signaling

    J. Clin. Invest.

    (2006)
  • R. Apfel

    A novel orphan receptor specific for a subset of thyroid hormone-responsive elements and its interaction with the retinoid/thyroid hormone receptor subfamily

    Mol. Cell. Biol.

    (1994)
  • M. Teboul

    OR-1, a member of the nuclear receptor superfamily that interacts with the 9-cis-retinoic acid receptor

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

    (1995)
  • S. Svensson

    Crystal structure of the heterodimeric complex of LXRα and RXRβ ligand-binding domains in a fully agonistic conformation

    EMBO J.

    (2003)
  • J.J. Repa

    Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRα and LXRβ

    Genes Dev.

    (2000)
  • S. Alberti

    Hepatic cholesterol metabolism and resistance to dietary cholesterol in LXRβ-deficient mice

    J. Clin. Invest.

    (2001)
  • L. Yvan-Charvet

    Combined deficiency of ABCA1 and ABCG1 promotes foam cell accumulation and accelerates atherosclerosis in mice

    J. Clin. Invest.

    (2007)
  • X. Wang

    Macrophage ABCA1 and ABCG1, but not SR-BI, promote macrophage reverse cholesterol transport in vivo

    J. Clin. Invest.

    (2007)
  • S.U. Naik

    Pharmacological activation of liver X receptors promotes reverse cholesterol transport in vivo

    Circulation

    (2006)
  • L.R. Brunham

    Tissue-specific induction of intestinal ABCA1 expression with a liver X receptor agonist raises plasma HDL cholesterol levels

    Circ. Res.

    (2006)
  • J.E. Feig

    LXR promotes the maximal egress of monocyte-derived cells from mouse aortic plaques during atherosclerosis regression

    J. Clin. Invest.

    (2010)
  • N. Zelcer

    LXR regulates cholesterol uptake through Idol-dependent ubiquitination of the LDL receptor

    Science

    (2009)
  • M. Korach-Andre

    Separate and overlapping metabolic functions of LXRα and LXRβ in C57Bl/6 female mice

    Am. J. Physiol. Endocrinol. Metab.

    (2010)
  • M. Korach-Andre

    Both liver-X receptor (LXR) isoforms control energy expenditure by regulating brown adipose tissue activity

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

    (2011)
  • K.R. Steffensen

    Genome-wide expression profiling; a panel of mouse tissues discloses novel biological functions of liver X receptors in adrenals

    J. Mol. Endocrinol.

    (2004)
  • Cited by (260)

    • Receptor biology: Challenges and opportunities

      2023, Progress in Molecular Biology and Translational Science
    View all citing articles on Scopus
    View full text