Elsevier

Life Sciences

Volume 247, 15 April 2020, 117443
Life Sciences

Review article
The mechanisms of lysophosphatidylcholine in the development of diseases

https://doi.org/10.1016/j.lfs.2020.117443Get rights and content

Abstract

Lysophosphatidylcholine (LPC) is the main component of oxidatively damaged low-density lipoprotein (oxLDL). LPC originates from the cleavage of phosphatidylcholine by phospholipase A2 (PLA2). LPC plays a biological role by binding to G protein-coupled receptors and Toll-like receptors. LPC can induce the migration of lymphocytes and macrophages, increase the production of pro-inflammatory cytokines, induce oxidative stress, and promote apoptosis, which can aggregate inflammation and promote the development of diseases. The effects of LPC on endothelial cells, vascular smooth muscle cells and arteries play a vital role in the progression of atherosclerosis and other cardiovascular diseases. In addition, the regulation of inflammation by LPC plays different roles in inflammatory and infectious diseases. In diabetes, LPC can induce insulin resistance. On the other hand, it can decrease blood glucose. The concentration of LPC varies in different tumours. LPC plays an important role in the invasion, metastasis and prognosis of tumours. Therefore, targeting LPC and lipid metabolism might be a potential therapeutic method for inflammation-related diseases.

Graphical abstract

The effect of LPC on cells

LPC can exert its biological function by activating ion channels, increasing the release of inflammatory factors and the expression of adhesion molecule, inducing apoptosis and oxidative stress. LPC can induce intracellular calcium mobilization via increasing external Ca(2+) influx and Ca(2+) release from endoplasmic reticulum Ca(2+) stores. Besides, LPC can affect the concentration of intracellular Na(+) and K(+), affecting the normal physiological function of cells. Importantly, LPC can bind to G protein coupled receptors (GPCRs) and Toll-like receptors (TLRs), activate transcription factors, stable mRNA and then increase the expression of target genes through Ca(2+)-mediated second messenger or directly inducing downstream inflammatory signalling pathways, which could increase the release of inflammatory factors, or induce the expression of adhesion molecules. In addition, there are many mechanisms by which LPC induces apoptosis, such as caspase activation, calcium influx, the release of cytochrome C, and the mitochondrial pathway. LPC increases ROS generation and enhances oxidative stress. Therefore, LPC plays a variety of biological functions in cells, so targeting LPC may be a potential method for the treatment of inflammation-related diseases.

Unlabelled Image
  1. Download : Download high-res image (208KB)
  2. Download : Download full-size image

Introduction

Lysophosphatidylcholine (LPC) is the main component of oxidatively damaged low-density lipoprotein (oxLDL) [1]. A high level of LPC, up to 100 μM, has been found in healthy subjects [2]. However, the concentration of LPC varies in different diseases. Researchers have found increased levels of LPC in atherosclerosis [3], inflammatory disease [4], diabetes [5], adrenoleukodystrophy [[6], [7], [8]], and squamous cervical cancer [9], while lower concentrations of LPC have been reported in infectious diseases [10,11], ovarian cancer [12], and colorectal cancer [13]. LPC can exert its biological function by inducing cell division, the release of inflammatory factors and oxidative stress. Our paper reviews the mechanism by which LPC affects different cells and its important role in the development of several diseases.

The source of saturated LPC, palmitoyl (16:0) LPC, is phosphatidylcholine (PC) [[14], [15], [16]], the major component of the cell membrane, and is produced by the cleavage of PC by phospholipase A2 (PLA2) [17]. In addition, the transfer of fatty acids to free cholesterol by lecithin-cholesterol acyltransferase (LCAT) can also generate saturated LPC. Importantly, LPC can be converted to PC via lysophosphatidylcholine acyltransferase (LPCAT) in the presence of Acyl-CoA. LPC can be catalysed by lysophospholipases A1, C and D (Fig. 1) [18]. Autotaxin (ATX) has lysophospholipase D activity, which could degrade LPC and produce lysophosphatidic acid (LPA),a mediator that is highly related with skin diseases and cancers [19]. Another sources of LPC are endothelial lipase (EL) and hepatic lipase (HL), and are generated by the cleavage of HDL-PC, which could produce amounts of unsaturated LPC, like oleoyl-lysophosphatidylcholine (LPC 18:1), linoleoyl-lysophosphatidylcholine (LPC 18:2), arachidonoyl-lysophosphatidylcholine (LPC 20:4), and so on (Fig. 2) [20].

The reported LPC receptors are mainly G protein coupled receptors (GPCRs) and Toll-like receptors (TLRs) (Graphical abstract). LPC is the ligand of GPR132 (G2A) and GPR4, and its affinity for G2A is remarkably higher than that for GPR4. G2A is mainly expressed on lymphocytes and macrophages, and G2A deficiency results in the hyperproliferation of T cells and induces advanced autoimmune diseases. LPC can bind to G2A and then activate ERK mitogen-activated protein kinase, inducing the migration of T lymphocytes [2,21]. LPC plays a key role in the development of atherosclerosis and inflammatory diseases through the role of the G2A receptor. The role of LPC mediated by GPR4 is described in detail below.

TLR2 and TLR4 are major Toll-like receptors that mediate LPC function. LPC can activate the NF-kB, p38 MAPK, and JUN signalling pathways by combining the TLR2 and TLR4 receptors [22,23]. The activation of these pathways can induce the production of pro-inflammatory factors, regulating inflammatory and infectious diseases.

The change in intracellular ion concentration by LPC can regulate many physiological functions (Graphical abstract). LPC can induce intracellular calcium mobilization [[24], [25], [26]]. The increased level of calcium mainly originates from external Ca(2+) influx and Ca(2+) release from endoplasmic reticulum Ca(2+) stores [27]. At the same time, intracellular calcium can be used as a second messenger to activate downstream signalling pathways, such as the p38 MAPK pathway [28]. The concentration of Na(+) and K(+) affect the membrane potential of myocardial cells and ensure normal heart function. LPC can affect the concentration of Na(+) in myocardial cells and induce arrhythmogenesis [29]. Na(+)-H+ exchange inhibition can protect the myocardium from injury induced by LPC [30]. In addition, LPC can also affect K(+) currents through the PKC and Rho-kinase pathways [31].

LPC has many biological functions in organisms, such as pro-inflammatory, oxidative stress, apoptosis induction and anti-infective effects (Graphical abstract). Caspase-1, belonging to the family of pro-inflammatory caspases, can activate biologically inactive pro-cytokines, such as IL-1β and IL-18, and plays an important role in inflammation. Researchers have found that LPC can activate caspase-1 and the production of ROS, which is Na(+)-dependent [[32], [33], [34]]. In addition, LPC can increase the generation of chemokines to attract inflammatory cells and increase the release of inflammatory factors, such as IL-1β, IL-8, IFN-γ, IL-6 and IL-5 [35]. This pro-inflammatory effect of LPC plays a critical role in the pathogenesis of diseases. We will introduce this effect in the following sections.

There are many mechanisms by which LPC induces apoptosis, such as caspase activation, calcium influx, the release of cytochrome C, and the mitochondrial pathway [36,37]. In addition, LPC can also induce apoptosis by increasing the expression of FasL by activating the NF-κB signalling pathway [38].

LPC plays a protective role in infectious diseases. One of the mechanisms is that LPC decreases the release of LPS-induced high mobility group box 1 (HMGB1) via the G2A/Ca(2+)/AMPK signalling pathway [39,40] and then plays an anti-infective role. LPC acts on a variety of immune cells and plays an important role in the process of disease. Next, we will elaborate.

Section snippets

Lymphocyte and neutrophils

The effects of LPC on immune cells include the induction of chemotaxis and alterations in immune function. LPC has a strong chemotaxic effect on thymic lymphoma lymphocytes from the mouse spleen and NK T cells (Table 1) [41,42]. Jurkat cells (a human CD4 T-cell line) exposed to LPC exhibit significantly increased expression of the chemokine receptors CXCR4 and CCR5 [43]. The main mechanism of this chemotaxis is dependent on a signalling pathway mediated by G2A, a receptor that is widely

Cardiovascular diseases (atherosclerosis)

LPC, the main component of oxLDL, has a variety of biological functions in cardiovascular diseases and can be used as a biomarker in some diseases [3]. LPC induces remarkable pro-inflammatory effects and vascular dysfunction in atherosclerosis and other cardiovascular diseases. Researchers have found that the concentration of LPC in plasma correlates with vascular damage and heart rate [99] and that the level of LPC in atherosclerotic plaques is markedly related to inflammatory factors, such as

Conclusion

LPC activates a variety of downstream signalling pathways, such as the MAPK and NF-κB pathways, through G protein coupled receptors and Toll-like receptors and mediates multiple biological functions, including the induction of chemotaxis, the release of inflammatory factors, oxidative stress, and apoptosis. The biological function of LPC in different cells promotes the formation of atherosclerotic plaques, aggravates inflammation, enhances anti-infective responses, regulates blood glucose, and

Acknowledgements

This work was supported by 81830096 from the key project of the National Science Foundation and supported Grant No.81773341 by National Natural Science Foundation of China.

Declaration of competing interest

The authors declare no conflict of interest.

References (184)

  • H. Jeong et al.

    TRPM2 contributes to LPC-induced intracellular Ca(2+) influx and microglial activation

    Biochem. Biophys. Res. Commun.

    (2017)
  • W.G. Ding et al.

    Lysophosphatidylcholine enhances I(Ks) currents in cardiac myocytes through activation of G protein, PKC and Rho signaling pathways

    J. Mol. Cell. Cardiol.

    (2011)
  • T. Schilling et al.

    Sodium dependence of lysophosphatidylcholine-induced caspase-1 activity and reactive oxygen species generation

    Immunobiology

    (2011)
  • T. Schilling et al.

    Importance of lipid rafts for lysophosphatidylcholine-induced caspase-1 activation and reactive oxygen species generation

    Cell. Immunol.

    (2010)
  • Y. Sun et al.

    Lysophosphatidylcholine-induced apoptosis in H19-7 hippocampal progenitor cells is enhanced by the upregulation of Fas Ligand

    Biochim. Biophys. Acta

    (2009)
  • H. Quan et al.

    Stearoyl lysophosphatidylcholine inhibits LPS-induced extracellular release of HMGB1 through the G2A/calcium/CaMKKbeta/AMPK pathway

    Eur. J. Pharmacol.

    (2019)
  • J.M. Kim et al.

    Stearoyl lysophosphatidylcholine prevents lipopolysaccharide-induced extracellular release of high mobility group box-1 through AMP-activated protein kinase activation

    Int. Immunopharmacol.

    (2015)
  • J. Rolin et al.

    Oxidized lipids and lysophosphatidylcholine induce the chemotaxis and intracellular calcium influx in natural killer cells

    Immunobiology

    (2013)
  • H. Hasegawa et al.

    Lysophosphatidylcholine enhances the suppressive function of human naturally occurring regulatory T cells through TGF-beta production

    Biochem. Biophys. Res. Commun.

    (2011)
  • D.H. Chang et al.

    Inflammation-associated lysophospholipids as ligands for CD1d-restricted T cells in human cancer

    Blood

    (2008)
  • L.V. Yang et al.

    Gi-independent macrophage chemotaxis to lysophosphatidylcholine via the immunoregulatory GPCR G2A

    Blood

    (2005)
  • H. Quan et al.

    Stearoyl lysophosphatidylcholine enhances the phagocytic ability of macrophages through the AMP-activated protein kinase/p38 mitogen activated protein kinase pathway

    Int. Immunopharmacol.

    (2016)
  • X. Qin et al.

    Lysophosphatidylcholine perpetuates macrophage polarization toward classically activated phenotype in inflammation

    Cell. Immunol.

    (2014)
  • M. Feng et al.

    Gene transfer of dimethylarginine dimethylaminohydrolase-2 improves the impairments of DDAH/ADMA/NOS/NO pathway in endothelial cells induced by lysophosphatidylcholine

    Eur. J. Pharmacol.

    (2008)
  • G.P. Campos-Mota et al.

    Role of ERK1/2 activation and nNOS uncoupling on endothelial dysfunction induced by lysophosphatidylcholine

    Atherosclerosis

    (2017)
  • H. Cheng et al.

    Lysophosphatidylcholine activates the Akt pathway to upregulate extracellular matrix protein production in human aortic valve cells

    J. Surg. Res.

    (2017)
  • J. Xu et al.

    Microglia colonization of developing zebrafish midbrain is promoted by apoptotic neuron and lysophosphatidylcholine

    Dev. Cell

    (2016)
  • H. Scholz et al.

    Lysophosphatidylcholine activates caspase-1 in microglia via a novel pathway involving two inflammasomes

    J. Neuroimmunol.

    (2017)
  • D.F. Schaeffer et al.

    LOX-1 augments oxLDL uptake by lysoPC-stimulated murine macrophages but is not required for oxLDL clearance from plasma

    J. Lipid Res.

    (2009)
  • K.E. Olofsson et al.

    Nanomolar concentrations of lysophosphatidylcholine recruit monocytes and induce pro-inflammatory cytokine production in macrophages

    Biochem. Biophys. Res. Commun.

    (2008)
  • K. Paapstel et al.

    Inverse relations of serum phosphatidylcholines and lysophosphatidylcholines with vascular damage and heart rate in patients with atherosclerosis

    Nutr. Metab. Cardiovasc. Dis.

    (2018)
  • J.H. Kabarowski et al.

    Xu, Y. Lysophosphatidylcholine as a ligand for the immunoregulatory receptor G2A

    Science

    (2001)
  • Zhou, X.; Wang, R.; Zhang, T.; Liu, F.; Zhang, W.; Wang, G.; Gu, G.; Han, Q.; Xu, D.; Yao, C., et al. Identification of...
  • N.D. Hung et al.

    Prevention of 1-palmitoyl lysophosphatidylcholine-induced inflammation by polyunsaturated acyl lysophosphatidylcholine

    Inflamm. Res.

    (2012)
  • M.Z. Yin et al.

    Identification of phosphatidylcholine and lysophosphatidylcholine as novel biomarkers for cervical cancers in a prospective cohort study

    Tumour Biol.

    (2016)
  • W.G. Ahn et al.

    Alteration of lysophosphatidylcholine-related metabolic parameters in the plasma of mice with experimental Sepsis

    Inflammation

    (2017)
  • W.H. Cho et al.

    Clinical significance of enzymatic lysophosphatidylcholine (LPC) assay data in patients with sepsis

    Eur. J. Clin. Microbiol. Infect. Dis.

    (1805-1810)
  • S.C. Kim et al.

    Differential levels of L-homocysteic acid and lysophosphatidylcholine (16:0) in sera of patients with ovarian cancer

    Oncol. Lett.

    (2014)
  • Z. Zhao et al.

    Plasma lysophosphatidylcholine levels: potential biomarkers for colorectal cancer

    J. Clin. Oncol.

    (2007)
  • T. Kuhn et al.

    Higher plasma levels of lysophosphatidylcholine 18:0 are related to a lower risk of common cancers in a prospective metabolomics study

    BMC Med.

    (2016)
  • T.A.P. Lagace

    greasing the cholesterol transport machinery

    Lipid Insights

    (2015)
  • C.R. McMaster

    From yeast to humans - roles of the Kennedy pathway for phosphatidylcholine synthesis

    FEBS Lett.

    (2018)
  • D. Mannheim et al.

    Enhanced expression of Lp-PLA2 and lysophosphatidylcholine in symptomatic carotid atherosclerotic plaques

    Stroke

    (2008)
  • S. Xu et al.

    The identification of a phospholipase B precursor in human neutrophils

    FEBS J.

    (2009)
  • C.G. Radu et al.

    T cell chemotaxis to lysophosphatidylcholine through the G2A receptor

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

    (2004)
  • A.B. Carneiro et al.

    Lysophosphatidylcholine triggers TLR2- and TLR4-mediated signaling pathways but counteracts LPS-induced NO synthesis in peritoneal macrophages by inhibiting NF-kappaB translocation and MAPK/ERK phosphorylation

    PLoS One

    (2013)
  • K. Magalhaes et al.

    Schistosomal-derived lysophosphatidylcholine are involved in eosinophil activation and recruitment through Toll-like receptor-2-dependent mechanisms

    J. Infect. Dis.

    (2010)
  • X. Zhu et al.

    Regulation of eosinophil adhesion by lysophosphatidylcholine via a non-store-operated Ca2+ channel

    Am. J. Respir. Cell Mol. Biol.

    (2007)
  • C.L. Watson et al.

    Lysophosphatidylcholine modulates cardiac I(Na) via multiple protein kinase pathways

    Circ. Res.

    (1997)
  • A.N. Hoque et al.

    Na(+)-H+ exchange inhibition protects against mechanical, ultrastructural, and biochemical impairment induced by low concentrations of lysophosphatidylcholine in isolated rat hearts

    Circ. Res.

    (1997)
  • Cited by (227)

    View all citing articles on Scopus
    View full text