Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 24.
Published in final edited form as: Pharmacol Ther. 2023 Apr 13;245:108414. doi: 10.1016/j.pharmthera.2023.108414

Emerging roles of lysophosphatidic acid receptor subtype 5 (LPAR5) in inflammatory diseases and cancer

Mélanie A Dacheux 1, Derek D Norman 1, Gábor J Tigyi 1, Sue Chin Lee 1,*
PMCID: PMC10290275  NIHMSID: NIHMS1907166  PMID: 37061203

Abstract

Lysophosphatidic acid (LPA) is a bioactive lipid mediator that regulates a variety of cellular functions such as cell proliferation, migration, survival, calcium mobilization, cytoskeletal rearrangements, and neurite retraction. The biological actions of LPA are mediated by at least six G protein-coupled receptors known as LPAR1–6. Given that LPAR1–3 were among the first LPARs identified, the majority of research efforts have focused on understanding their biology. This review provides an in-depth discussion of LPAR5, which has recently emerged as a key player in regulating normal intestinal homeostasis and modulating pathological conditions such as pain, itch, inflammatory diseases, and cancer. We also present a chronological overview of the efforts made to develop compounds that target LPAR5 for use as tool compounds to probe or validate LPAR5 biology and therapeutic agents for the treatment of inflammatory diseases and cancer.

Keywords: Lysophosphatidic acid, Lysophosphatidic acid receptor, Cancer, Inflammation, Tumor immunity, Wound healing

1. Introduction

Lysophosphatidic acid (LPA) is a simple lipid molecule comprised of a phosphate or a cyclic phosphate head group (designated as cyclic phosphatidic acid), a glycerol backbone, and a fatty acid or fatty alcohol chain. The ether analog is designated as alkyl glycerophosphate in IUPHAR nomenclature but commonly referred to as alkyl-LPA. LPA was originally discovered in urine as the Darmstoff and brain extracts in late 1950s (Kirschner & Vogt, 1961; Vogt, 1957). However, it was not until the late 1980s that LPA research gained traction, when it was shown to regulate blood pressure in vivo, elicit calcium transients in fibroblasts, promote cell proliferation, activate oscillatory chloride current via a putative plasma membrane receptor in Xenopus oocytes, and induce neurite retraction in vitro (Jalink, van Corven, & Moolenaar, 1990; Moolenaar et al., 1986; Tigyi & Miledi, 1992; Tokumura, Fukuzawa, & Tsukatani, 1978; van Corven, Groenink, Jalink, Eichholtz, & Moolenaar, 1989).

Around the same time, evidence supporting a role for LPA in regulating wound healing began to emerge. The production of LPA was linked to platelet activation, leading to the presumption that it was released by thrombin-activated platelets (Eichholtz, Jalink, Fahrenfort, & Moolenaar, 1993; Gerrard & Robinson, 1989). Elevated levels of LPA were detected in the aqueous humor, brain, and cerebrospinal fluid (CSF) following corneal or cerebrovascular injury and in skin blisters (Crack et al., 2014; Liliom et al., 1998; Mazereeuw-Hautier et al., 2005; Tigyi et al., 1995). Furthermore, several studies showed that LPA treatment accelerated the closure of skin wounds in mice and rats and repaired intestinal epithelium damaged by either experimentally induced colitis or radiation (Balazs, Okolicany, Ferrebee, Tolley, & Tigyi, 2001; Demoyer, Skalak, & Durieux, 2000; Deng et al., 2002; Sturm, Sudermann, Schulte, Goebell, & Dignass, 1999). A growing list of cellular responses is mediated by LPA following tissue injury, including the proliferation and recruitment of fibroblasts and epithelial cells, vascular and extracellular matrix (ECM) remodeling, and modulation of the immune response (Balazs et al., 2001; Demoyer et al., 2000; English, Garcia, & Brindley, 2001; Goetzl & Rosen, 2004; Hadidi & Athanasiou, 2013; Jalink, Hordijk, & Moolenaar, 1994; Lee, Goetzl, & An, 2000; Olorundare, Peyruchaud, Albrecht, & Mosher, 2001; Panetti et al., 2001; Panetti, Chen, Misenheimer, Getzler, & Mosher, 1997; Sauer et al., 2004; Stortelers, Kerkhoven, & Moolenaar, 2008; Sturm et al., 1999; Tigyi, Dyer, & Miledi, 1994; Xu, Yin, & Yu, 2007; Zhao & Natarajan, 2013). These studies further establish LPA as a master regulator of the natural healing process.

Given the importance of LPA in wound healing, it is not surprising that dysregulation of this signaling pathway can cause chronic inflammation and promote cancer progression. Increased levels of LPA are found in the biological fluids of patients with ovarian or gastric cancer, idiopathic pulmonary fibrosis, rheumatoid arthritis, chronic liver or renal diseases, systemic sclerosis, multiple sclerosis, cardiovascular diseases and neuropathic pain (Baker et al., 2002; Balood et al., 2014; Kremer et al., 2010; Kurano et al., 2015; Kuwajima et al., 2018; Nochi et al., 2008; Sasagawa, Suzuki, Shiota, Kondo, & Okita, 1998; Tager et al., 2008; Tokumura et al., 2009; Wang et al., 2019; Watanabe et al., 2007; Xu et al., 1995; Zeng et al., 2017; Zhang et al., 2016). In each of these studies, the presence of high LPA levels has been linked to the pathological progression of the disease.

How LPA modulates such a broad spectrum of physiological and pathological responses has long piqued the interest of researchers. Over the last four decades, we learned that the biological responses of LPA are mediated through at least six G protein-coupled receptors (GPCRs) termed LPA receptors 1–6 (LPAR1–6), and intracellularly via the nuclear receptor peroxisome proliferator activated receptor gamma (PPARγ). LPAR1–3 belong to the endothelial gene family of GPCRs, while LPAR4–6 are members of the P2Y purinergic family of GPCRs (Geraldo et al., 2021; Xiang, Lu, Shao, & Wu, 2020; Yung, Stoddard, & Chun, 2014). Different molecular species of LPA exist in biological fluids, each with varying fatty acid chain length and degree of saturation, with the fatty acid moieties able to be linked to the glycerol backbone at either the sn-1 or sn-2 position, resulting in 1-acyl- or 2-acyl-LPA species, respectively (Bandoh et al., 2000; Gerrard & Robinson, 1989; Tigyi & Miledi, 1992). Alternatively, an ether linkage, rather than an ester bond, can form at the sn-1 position of the glycerol backbone, giving rise to 1-alkyl-LPA and 1-alkenyl-LPA (Fischer et al., 1998; Sugiura et al., 1999). Structure-activity relationship (SAR) studies revealed that LPARs have distinct preferences for different LPA species. When expressed in mammalian cells, LPAR1–3 showed preferential activation by 1-acyl-LPA species containing unsaturated fatty acids over their saturated analogs of the same hydrocarbon chain length (Fujiwara et al., 2005). Studies have proposed that 2-acyl-LPA with unsaturated fatty acids could be the preferred ligand for LPAR6, whereas alkyl-LPA is a more potent ligand at LPAR5 and PPARγ than acyl-LPA species (Davies et al., 2001; Inoue et al., 2011; Tsukahara et al., 2006; Williams et al., 2009; Yanagida et al., 2009). Not only do LPARs differ in their ligand specificities, but they also couple to different G proteins such as Gαs, Gαi, Gαq and Gα12/13 (Fig. 1). Moreover, the expression profile of LPARs is cell type-specific and spatiotemporally regulated during embryonic development and pathological conditions (Geraldo et al., 2021; Xiang et al., 2020; Yung et al., 2014). These differences enable LPA to mediate a wide range of physiological and pathological responses.

Fig. 1.

Fig. 1.

LPAR signaling pathway. LPA signaling is mediated through at least six GPCRs (LPAR1–6). LPAR1–6 can couple to different G protein family members (Gα12/13, Gαq/11, Gαi/0, or Gαs) to activate different downstream pathways, resulting in a variety of cellular responses. The G proteins with which LPAR1 interacts are represented by blue arrows, whereas those interacting with LPAR2, LPAR3, LPAR4, LPAR5 and LPAR6 are indicated by red, green, orange, pink and gray arrows, respectively. Abbreviations: AC, adenylate cyclase; ATX, autotaxin; LPC, lysophosphatidylcholine; PI3K, phosphatidylinositol 3-kinase; PLC, phospholipase C; Ras, rat sarcoma protein; Rho, Ras homolog protein.

In this review, we provide a brief overview of the pathways that regulate LPA production and degradation, followed by an in-depth discussion on the LPAR subtype 5 (LPAR5), which has recently emerged as a key player in modulating pathological conditions including pain, itch, cerebral inflammation, and cancer. Finally, we discuss some of the most recent developments on promising compounds that target LPAR5 as a therapeutic approach for treating inflammatory diseases and cancer.

2. LPA production and degradation pathways

Several pathways are involved in the biosynthesis of LPA (Fig. 2). First, lysophosphatidylcholine (LPC) is hydrolyzed by autotaxin (ATX), a secreted enzyme with lysophospholipase D activity, to generate LPA. The bulk of LPA in biological fluids is generated through this pathway, as mice heterozygous for the ATX gene (Enpp2+/−) exhibit a 50% reduction in plasma LPA levels (van Meeteren et al., 2006). The second pathway involves the conversion of phosphatidic acid (PA) to LPA by membrane-associated PA-selective phospholipase A1 enzymes (PA-PLA1α and PA-PLA1β) (Aikawa, Hashimoto, Kano, & Aoki, 2015). Relatively little is known about the cellular actions of LPA produced by PA-PLA1α and PA-PLA1β. Only two studies show that PA-PLA1α preferentially generates 2-acyl LPA and is involved in hair follicle formation, and that PA-PLA1β is abundantly expressed in Ewing family tumors (Foell et al., 2008; Inoue et al., 2011). Additional intracellular enzymes involved in LPA production include glycerol-3 phosphate acyltransferase (GPAT), which converts glycerol-3 phosphate to LPA, and acylglycerol kinase (AGK), which converts monoacylglycerol (MAG) to LPA (Bektas et al., 2005).

Fig.2.

Fig.2.

LPA biosynthesis and degradation pathways. LPA can be synthesized from lysophospholipids, phosphatidic acid (PA), glycerol-3 phosphate, or monoacylglycerol by autotaxin (ATX), PA-selective phospholipase A1α (PA-PLA1α) or β (PA-PLA1β), glycerol-3 phosphate acyltransferase (GPAT), and acylglycerol kinase (AGK), respectively. Conversely, LPA can be converted to PA by LPA acyltransferase (LPAAT), to glycerol-3 phosphate by lysophospholipase (LPL), and to monoacylglycerol by lipid phosphate phosphatase (LPP) or phosphatidate phosphatase (PAP). Abbreviations: G3P, glycerol-3 phosphate; DAG lipase, diacylglycerol lipase; MGAT, monoacylglycerol acyltransferase; PLA1, phospholipase A1; PLA2, phospholipase A2; PLD, phospholipase D.

Likewise, the breakdown of LPA is also mediated by several classes of enzymes, the most important of which are the lipid phosphate phosphatases (LPPs). Thus far, only three members of the LPP family of enzymes (LPP1–3) have been identified. They are localized to the plasma membrane, and membranes of the Golgi apparatus and endoplasmic reticulum, allowing them to hydrolyze both extra- and intracellular lipid phosphates (Tang & Brindley, 2020). LPA can also be converted to PA by LPA acyltransferases (LPAAT) and to glycero-3 phosphate by lysophospholipases (Baker & Chang, 1999; Thompson & Clark, 1994).

LPA levels are strictly regulated during embryonic development. In mice, deletion of enzymes such as ATX or LPP3 that are involved in the production or degradation of LPA resulted in embryonic death due to abnormal vascular and neural development (Escalante-Alcalde et al., 2003; Moolenaar, Houben, Lee, & van Meeteren, 2013; Tanaka et al., 2006; van Meeteren et al., 2006). Furthermore, overexpression of ATX can cause lethal vascular defects in mice during embryogenesis (Yukiura, Kano, Kise, Inoue, & Aoki, 2015). Interestingly, the majority of ATX activity in adult mice is not required to maintain normal physiological homeostasis. Specifically, genetic deletion of Enpp2 in adult mice resulted in >80% reduction in plasma LPA levels, which was well-tolerated (Katsifa et al., 2015). This finding is consistent with the lack of an obvious phenotype in Enpp2+/− heterozygous mice that exhibit a 50% reduction in plasma LPA levels (van Meeteren et al., 2006). It is possible that residual levels of ATX-derived LPA, combined with LPA produced by other biosynthetic pathways, are sufficient to sustain normal tissue homeostasis (Ninou, Magkrioti, & Aidinis, 2018).

It is worth noting that LPA can inhibit ATX expression, implying the presence of a self-regulating mechanism to ensure that LPA levels do not fluctuate aberrantly (Benesch, Zhao, Curtis, McMullen, & Brindley, 2015). Nevertheless, the presence of inflammatory factors can override this self-regulating mechanism. For example, ATX expression can be induced by lipopolysaccharide (LPS), tumor necrosis factor (TNFα), interleukins (IL-1β or IL-6) and type 1 interferons (IFNα or IFNβ), which could explain the elevated levels of ATX and LPA observed during inflammation (Benesch et al., 2015; Castelino et al., 2016; Li & Zhang, 2009; Nikitopoulou et al., 2012; Song, Guan, Zhao, & Zhang, 2015; Wu et al., 2010). If left unresolved, this could lead to the development of chronic inflammatory diseases and promote cancer progression. Indeed, aberrant expression of ATX, LPARs and/or LPPs is linked to the pathological progression of chronic inflammatory diseases and a variety of cancers including ovarian, breast, lung, prostate, pancreas, colon, melanoma, liver, and brain (Balijepalli, Sitton, & Meier, 2021; Magkrioti et al., 2019).

3. LPAR5 signaling

The majority of research efforts have been devoted to understanding the biology of LPAR1–3, which were among the first LPARs to be identified, as reviewed by (Geraldo et al., 2021; Xiang et al., 2020; Yung et al., 2014). However, growing evidence from the last decade suggests important roles for LPAR5 in modulating inflammatory diseases and cancer. LPAR5 was discovered in 2006 by two independent groups using distinct screening approaches that resulted in the deorphanization of GPR92. They found that LPAR5 shares 22% amino acid identity with LPAR1–3 and ~35% similarity with LPAR4 (Kotarsky et al., 2006; Lee, Rivera, Gardell, Dubin, & Chun, 2006). LPAR5 can be activated by not only LPA, but also natural lipid analogs including cyclic phosphatidic acid, farnesyl phosphate, farnesyl pyrophosphate, and anandamide phosphate. SAR studies highlighted that the ligand preferences of LPAR5 are different than those of LPAR1–3, with the order of potency of LPAR5 ligands being alkyl-LPA > acyl-LPA > farnesyl phosphates ≥ N-arachidonylglycine (Williams et al., 2009). In humans, LPAR5 is predominantly expressed in the heart, placenta, spleen, brain, lung, and gut, while in mice, it is expressed in the small intestine, stomach, spleen, skin, thymus, lung, and the developing brain up to embryonic day E12.5 (Kotarsky et al., 2006; Lee et al., 2006; Oh et al., 2008; Ohuchi et al., 2008). However, LPAR5 expression in the mouse brain was not detected between days E16 and postnatal day 30 (Suckau et al., 2019). Whether this temporal expression of LPAR5 in the developing brain has any biological significance remains to be investigated.

In terms of cellular functions, LPAR5 activates Gαq/11 to promote Ca2+ mobilization and inositol phosphate production and Gα12/13 to regulate neurite retraction and stress fiber formation (Lee et al., 2006; Oh et al., 2008). Activation of LPAR5 also increases cAMP levels, although this appears to be mediated by the G protein βγ subunits rather than the Gαs subunits (Kotarsky et al., 2006; Lee et al., 2006; Oh et al., 2008). LPAR5 has been shown to induce β-Arrestin recruitment. Whether the recruitment of β-Arrestin by LPAR5 participates in additional downstream signaling events other than signal termination via receptor internalization remains to be determined (Kroeze et al., 2015; Yin et al., 2009). Interestingly, Lpar5−/− knockout (KO) mice develop normally but exhibit behavioral abnormalities relative to wildtype (WT) mice. The Lpar5−/− KO mice were less anxious, displayed reduced fear responses, and were hyperactive at night. Lpar5−/− KO mice also outperformed WT mice in several motivational-driven tasks, suggesting that LPAR5 signaling may be involved in psychopathology (Callaerts-Vegh, Leo, Vermaercke, Meert, & D’Hooge, 2012).

4. Role of LPAR5 in modulating platelet function

In response to injury, platelets are among the first cells to become activated to initiate hemostasis and tissue repair (Etulain, 2018). As noted previously, LPA was presumed to be released when platelets are activated (Eichholtz et al., 1993; Gerrard & Robinson, 1989). However, activated platelets actually generate LPA in a two-step process: first by secreting phospholipases including group IIA secretory PLA2 (sPLA2-IIA) and phosphatidylserine-specific PLA1 (PS-PLA1) that convert membrane phosphospholipids to various lysophospholipids including LPC, lysophosphatidylethanolamine, and lysophosphatidylserine. Second, these lysophospholipids are hydrolyzed by ATX to produce LPA (Aoki et al., 2002; Bolen et al., 2011; Sano et al., 2002).

LPA induces human platelet shape change, aggregation, and co-adhesion to monocytes, suggesting that LPA signaling may be involved in the pathological activation of platelets during thrombosis (Haseruck et al., 2004; Simon, Chap, & Douste-Blazy, 1982; Tokumura et al., 2002). This is supported by the discovery that LPA is produced during the mild oxidization of low-density lipoprotein (Kurano et al., 2015; Siess et al., 1999). High levels of LPA were detected subsequently in the lipid-rich core of human and mouse atherosclerotic plaques, with alkyl-LPA species accounting for 20% of total LPA species detected (Bot et al., 2010; Rother et al., 2003; Siess et al., 1999). Moreover, platelets isolated from patients with coronary artery disease were more responsive to LPA than were those from healthy donors. However, the effect of LPA on platelet aggregation in healthy donors varies, with approximately 20% of donors being nonresponsive to LPA stimulation (Haseruck et al., 2004; Pamuklar et al., 2008). Gene expression profiling showed that LPAR4, LPAR5 and PPARγ were expressed abundantly in human platelets, whereas LPAR1 and LPAR2 were less abundantly expressed (Amisten, Braun, Bengtsson, & Erlinge, 2008; Rowley et al., 2011; Spinelli et al., 2008). Interestingly, increased expression of LPAR4 and PPARγ was detected in platelets from nonresponsive donors, suggesting that LPA may act via LPAR4 and/or PPARγ to mediate inhibitory signals that block platelet aggregation in humans (Pamuklar et al., 2008). In fact, activation of PPARγ by both synthetic and endogenous ligands such as rosiglitazone and 15d-PGJ2, respectively, has been shown to block activation of human platelets by collagen (Moraes et al., 2010).

The discovery that human platelets express LPAR5 and that alkyl-LPA is more potent than acyl-LPA in inducing platelet aggregation suggests that LPAR5 is most likely the receptor involved in platelet activation (Simon et al., 1982; Tokumura et al., 2002). Indeed, siRNA-mediated knockdown of LPAR5 in two human megakaryocytic cell lines abolished LPA-induced platelet shape change and aggregation (Khandoga, Pandey, Welsch, Brandl, & Siess, 2011). Similar findings were obtained when isolated human platelets were treated with LPAR5 antagonists (H2L 5987411 and H2L 5765834) (Williams et al., 2009). Although these studies confirmed that LPAR5 is the receptor responsible for LPA-mediated activation of human platelets, this is not the case in mouse platelets. LPA failed to induce murine platelet aggregation and prevented platelet aggregation induced by other agonists such as thrombin and adenosine diphosphate. Moreover, transgenic mice that overexpress ATX (Enpp2-Tg) exhibit elevated levels of circulating LPA and are more prone to bleeding than WT mice (Pamuklar et al., 2009). Transcriptomic analysis revealed that murine platelets express LPAR4 and do not express LPAR1–3, LPAR5, or PPARγ (Rowley et al., 2011). This may in part explain the observed differences in regulation of platelets by LPA in humans and mice (Fig. 3). These distinctions should be considered when using mouse models to study the role of LPA in regulating platelet function in cardiovascular diseases, inflammatory diseases, and cancer.

Fig. 3.

Fig. 3.

Role of LPAR5 in regulating platelet function. LPA induces human platelet activation and aggregation through LPAR5. One study proposed the potential involvement of LPAR4 and PPARγ in inhibiting platelet aggregation as their expression was elevated in platelets that were nonresponsive to LPA stimulation (Pamuklar et al., 2008). While the involvement of PPARγ in inhibiting platelet function has been demonstrated with other PPARγ ligands, more research is needed to confirm the role of LPAR4 in modulating platelet function. Murine platelets express LPAR4 rather than LPAR5, which could explain in part why LPA does not induce platelet activation and aggregation in mice.

5. Role of LPAR5 in regulating neuropathic pain

The role of LPA signaling in modulating neuropathic pain was first reported to be mediated by LPAR1. Lpar1−/− KO mice were protected from neuropathic pain caused either by a single intrathecal injection of LPA or by partial sciatic nerve ligation (PSNL). Specifically, WT mice exhibited demyelination of the dorsal root ganglion (DRG) neurons, mechanical allodynia, and hyperalgesia while Lpar1−/− KO mice did not (Inoue et al., 2004). Subsequently, it was discovered that Lpar3−/− KO mice exhibited a blockage in the feed-forward mechanism of LPA accumulation following nerve injury and were thereby protected from neuropathic pain (Ma et al., 2009; Ueda, 2020). This is consistent with the observation that increased levels of LPA correlate significantly with pain intensity in patients with neuropathic pain (Kuwajima et al., 2018).

LPAR5 is also abundantly expressed in the DRG and dorsal horn neurons, where it may play a role in modulating pain signals (Lin, Rivera, & Chun, 2012; Oh et al., 2008). Indeed, Lpar5−/− KO mice were protected from developing neuropathic pain caused by PSNL and multiple sclerosis (Lin et al., 2012; Tsukahara et al., 2018). Interestingly, the mechanism by which LPAR5 modulate pain signals is distinct from that of LPAR1. In response to nerve injury, LPAR1 causes demyelination of the DRG neurons, upregulation of neuropathic pain markers (calcium channel Caα2δ1, ephrinB1, and PKCγ), and reorganization of Aβ fiber-mediated signal transmission (Inoue et al., 2004; Lin et al., 2012; Xie, Matsumoto, Chun, & Ueda, 2008). These events were not observed in Lpar5−/− KO mice subjected to PSNL. Instead, nerve injury in the spinal cord dorsal horn of Lpar5−/− KO mice resulted in significant reduction in the phosphorylation of cAMP response element binding protein (pCREB), a known mediator of neuropathic pain. Since LPAR1 and LPAR5 have opposing effects on cAMP signaling, it is possible that an increase in cAMP accumulation via LPAR5 could result in pCREB activation in this scenario, leading to the development of neuropathic pain (Lin et al., 2012) (Fig. 4).

Fig. 4.

Fig. 4.

Role of LPAR in neuropathic pain. Upon nerve injury, LPA can induce neuropathic pain via two pathways. The first involves activation of LPAR1, which upregulates neuropathic pain markers such as calcium channel Caα2δ1, ephrinB1 and PKCγ, resulting in the demyelination of DRG neurons. The second pathway involves activation of LPAR5, which increases phosphorylation of cAMP and pCREB activation.

Aside from neuropathic pain, Lpar5−/− KO mice exhibit some degree of reduced sensitivity to acute pain compared to WT mice. However, significance differences in pain sensitivity between genotypes were observed in the tail withdrawal latency test, but not in the hot plate and Hargreaves’ tests. Lpar5−/− KO mice also recovered faster from inflammatory pain caused by complete Freund’s adjuvant, whereas no differences were observed when formalin or carrageenan were used as pain-inducing stimuli (Callaerts-Vegh et al., 2012). This is in contrast to the results of a recent study in which induction of pain by formalin, carrageenan, and prostaglandin E2 (PGE2) was significantly reduced by the LPAR5 antagonist Cpd3/UA02085 (Langedijk et al., 2022). Whether the differences between the two studies were due to the different mouse strains used in the in vivo experiments remains to be determined.

6. Role of LPAR5 in regulating itch

LPA was identified in the serum of patients with cholestatic pruritus as an itch factor that induces mobilization of intracellular Ca2+ in neuronal cells. The levels of ATX in cholestatic serum was found to correlate positively with itch intensity (Kremer et al., 2010). Furthermore, intradermal injection of LPA induced scratch responses in mice in a dose-dependent manner (Hashimoto, Ohata, & Momose, 2004; Shimizu et al., 2014). However, the mechanism by which LPA causes itch remains unclear. In one study, this effect was mediated in part by activation of LPAR1 (Shimizu et al., 2014), while another showed that genetic silencing or pharmacological inhibition of LPAR5 with the antagonist TCLPA5–4 abolished LPA-induced Ca2+ mobilization in neuronal cells. The authors found that LPAR5 was involved in a feed forward mechanism that promotes the intracellular production of LPA via the actions of phospholipase D and Ca2+-independent iPLA2. The resultant intracellular LPA activates transient receptor potential ankyrin 1 (TRPA1) and vanilloid 1 (TRPV1) to induce itch sensations (Kittaka, Uchida, Fukuta, & Tominaga, 2017). This mechanism differs from previous research that found that extracellular LPA was transported intracellularly to activateTRPV1 to induce pain (Nieto-Posadas et al., 2011). Interestingly, both LPAR5 and LPAR1 colocalize with TRPV1 in DRG neurons and activation of LPAR1 potentiated TRPV1 signals via a PKCε-dependent pathway (Oh et al., 2008; Pan, Zhang, & Zhao, 2010). Thus, further studies are needed to clarify the crosstalk between the LPAR1, LPAR5, and TRPV1 signaling pathways that mediates the sensations of itch and pain (Fig. 5).

Fig. 5.

Fig. 5.

Role of LPAR in itch. LPA signaling has been reported to regulate itch sensation via activation of LPAR1 and/or LPAR5. However, the downstream signaling pathway(s) linking LPAR1 to itch has not been elucidated (gray box). Although LPAR1 has been shown to activate PKCϵ to potentiate TRPV1 activity leading to pain sensation, whether this pathway is involved in mediating itch sensation remains unknown (Pan et al., 2010). Likewise, activation of LPAR5 has been shown to induce itch via PLD-iPLA2 generation of intracellular LPA, which in turns activates TRPA1 and TRPV1. However, experimental evidence linking LPAR5 to direct PLD activation is currently lacking (denoted as question mark).

Unexpectedly, treatment of mice with the LPAR5 antagonist Cpd3/UA08125 increased their scratch responses to itch in a manner that was independent of TRPA1 and TRPV1 activation (Langedijk et al., 2022). Although Cpd3/UA08025 was previously reported to be a selective antagonist of LPAR5, we found it to be also a potent antagonist of LPAR4 (Kozian et al., 2016; Lee et al., 2020). Thus, it is unclear whether the itch sensation observed in mice treated with Cpd3/UA08025 is caused by inhibition of LPAR5 or LPAR4. More research is needed to address the disparities in the findings of these studies.

7. Role of LPAR5 in regulating cerebral inflammation

As noted previously, LPA levels are increased in the CSF of patients with traumatic brain injury and in the plasma of patients with ischemic cerebrovascular disease (Crack et al., 2014; Li et al., 2008). In mice challenged with transient middle cerebral artery occlusion (tMCAO), increased expression of LPAR5 was observed in activated microglia in the ischemic core (Sapkota, Lee, Park, & Choi, 2020). Microglia are macrophage-like cells that reside in the central nervous system (CNS) and play essential roles in brain development, immune surveillance, and tissue repair. Like peripheral macrophages, microglia have both pro- and anti-inflammatory activities and their dysfunction is linked to neuroinflammatory and neurodegenerative diseases (Bachiller et al., 2018). Activation of LPAR5 induces a proinflammatory and promigratory signature in microglia that is potentially neurotoxic, which may contribute to the pathogenesis of CNS disorders (Plastira et al., 2016; Plastira et al., 2017; Sapkota et al., 2020). Specifically, LPAR5 activates the protein kinase D pathway, resulting in the phosphorylation of downstream transcription factors such as NFκB, STAT1, STAT3, and c-Jun. The majority of these transcription factors drive inflammation by increasing the expression of cytokines (TNFα, IL-1β, and IL-6) and chemokines (CXCL10, CCL5 and CXCL2) and by producing nitric oxide and reactive oxygen species (Plastira et al., 2016; Plastira et al., 2017; Plastira et al., 2019; Plastira et al., 2020). Following brain injury or cerebral ischemia, persistent accumulation of these factors causes neurotoxicity and brain damage (Fig. 6).

Fig. 6.

Fig. 6.

Role of LPAR5 in regulating cerebral inflammation. In microglia, LPAR5 activates the protein kinase D pathway, resulting in the phosphorylation of transcription factors such as NFκB, STAT1, STAT3 and c-Jun. This event causes an increase in proinflammatory factors such as TNFα, IL-1β, IL-6, CXCL10, CCL5, CXCL12, nitric oxide (NO) and reactive oxigen species (ROS), all of which cause cerebral inflammation.

Indeed, when mice challenged with tMCAO were treated with the LPAR5 antagonist TCLPA5–4, they exhibited reduced accumulation of proinflammatory microglia in the ischemic core. An overall reduction in neuroinflammation and neuronal cell death was also observed in mice treated with TCLPA5–4 (Sapkota et al., 2020). The role of LPAR5 in promoting neuroinflammation caused by LPS-induced peripheral endotoxemia was investigated in mice treated with the novel LPAR5 antagonist AS2717638, which were protected from neuroinflammation (Joshi et al., 2021). This observation was further validated in Lpar5−/− KO mice, which exhibited circulating levels of TNFα and IL-1β that were significantly lower than those in WT mice. Moreover, the expression of IL-6 and CXCL2 was significantly reduced in the brain of Lpar5−/− KO mice treated with LPS (Joshi et al., 2022). The discovery that LPAR5 signaling plays an important role in modulating microglia function may open up new avenues of research in areas such as neurodegenerative diseases in which microglia dysfunction has been implicated.

8. Role of LPAR5 in cancer

LPAR5 regulates cancer cell proliferation, migration, invasion, and tumor immunity. LPAR5 expression is upregulated in human non-small-cell lung carcinoma (NSCLC) and human papillary thyroid carcinoma (PTC) compared to their respective normal tissues (Wu et al., 2020; Zhang, Chen, & Xu, 2020; Zhao et al., 2021). Overexpression of LPAR5 in NSCLC positively correlates with a higher incidence of metastasis and poor prognosis. ShRNA-mediated knockdown of LPAR5 in several human NSCLC cell lines reduced cell proliferation and migration in vitro and tumor growth in vivo, further confirming the protumorigenic role of LPAR5 in NSCLC (Zhang et al., 2020). Likewise, LPAR5 promotes the proliferation, migration, and invasion of human PTC cells in vitro and treatment with TCLPA5–4 significantly decreases growth of PTC xenografts in nude mice (Wu et al., 2020; Zhao et al., 2021). Recently, LPAR5 was identified as a key regulator of radiation-induced epithelial-to-mesenchymal transition (EMT) in human cervical (HeLa) and lung (A549) cancer cells. Specifically, radiation upregulated LPAR5 expression in a bidirectional manner, with downregulation occurring between 2 and 4 h after irradiation and upregulation occurring 24 h later. Upregulation of LPAR5 at the 24-h time point was linked to increased expression of EMT markers such as Snail, matrix metalloproteinase 1 (MMP1) and MMP9, while siRNA-targeted knockdown of LPAR5 abolished their expression. The authors proposed that the increase in EMT markers induced by LPAR5 could make cancer cells more radioresistant (Sun et al., 2022). Contrary to these findings, LPAR5 serves as a negative regulator of cell migration and invasion in human sarcoma cell lines (HOSL5 and HT1080L5) by a mechanism that involves the downregulation of MMP2 and MMP9 (Dong et al., 2014). Likewise, activation of LPAR5 by LPA inhibits the migration and invasion of murine B16-F10 melanoma cells in vitro (Jongsma, MatasRico, Rzadkowski, Jalink, & Moolenaar, 2011; Lee et al., 2015). The underlying mechanism is mediated by decreased PIP3 signaling together with an increase in cAMP accumulation and activation of the protein kinase A pathway (Jongsma et al., 2011). Taken together, these findings suggest that LPAR5 signaling can mediate opposing cellular effects depending on the type of cancer.

Perhaps the most intriguing aspect of LPAR5 signaling in cancer is its role in regulating tumor immunity. Early reports revealed that CD8 T cells isolated from the small intestines of mice primarily expressed LPAR5, followed by LPAR2 and LPAR1 (Kotarsky et al., 2006). Subsequently, naïve CD8 T cells isolated from mouse spleen or human blood were found to express LPAR5, LPAR6 and LPAR2 (Mathew et al., 2019; Oda et al., 2013). Interestingly, LPAR5 was found to serve as an inhibitory receptor that repressed T cell receptor (TCR) signaling in human and mouse cells. In particular, activation of LPAR5 by LPA impeded antigen specific TCR-induced Ca2+ mobilization, T cell activation, proliferation, and cytotoxic effector functions (Mathew et al., 2019; Oda et al., 2013). LPA also hampered the formation and function of immunological synapses by reprogramming the cytoskeletal dynamics and disrupting the localization of at least three key signaling molecules, namely RhoA, mDia1, and IP3R1. However, the LPAR responsible for mediating this process remains unknown (Kremer et al., 2022). Nevertheless, all of these events could lead to an impaired antitumor immune response. This hypothesis was validated in several in vivo experiments showing firstly that CD8 T cells lacking LPAR5 expression were more effective at reducing the growth rate of B16-F10 melanoma and EG7 lymphoma tumors in mice than WT CD8 T cells (Mathew et al., 2019; Oda et al., 2013). Secondly, Lpar5 −/− KO mice exhibited an 85% reduction in B16-F10-derived lung metastasis relative to their WT littermates (Lee et al., 2015). Notably, robust CD8 T cell infiltration was observed in the few lung metastasis that developed in Lpar5−/− KO mice (Lee et al., 2020). These studies highlight a distinct role for LPAR5 as an immune checkpoint receptor expressed in CD8 T cells to modulate tumor immunity (Fig. 7).

Fig. 7.

Fig. 7.

Role of LPAR5 in cancer. Depending on the type of cancer, LPAR5 can mediate pro- or anti-tumorigenic functions. In terms of the role of LPAR5 in immune cells, LPAR5 expression on TAM correlates positively with markers of phagocytosis and antigen presentation, whereas LPAR5 expression on CD8 T cells inhibits TCR induced Ca mobilization, T cell activation, and impair the release of perforin and granzymes. Furthermore, LPA signaling impairs the formation and function of immunological synapses in CD8 T cells by altering the localization of RhoA, mDia1, and IP3R1. The question mark indicates that the LPAR responsible for mediating this effect has not been experimentally determined.

Further transcriptomic analysis of immune cells isolated from the ascites fluid of ovarian cancer patients revealed that tumor associated macrophages (TAM) express ATX, LPAR5 and LPAR6 (Reinartz et al., 2019). TAM in osteosarcoma tumors also express high levels of LPAR5, which positively correlates with expression markers for phagocytosis and antigen presentation (He et al., 2022). Although LPA promotes the conversion of circulating monocytes to macrophages via activation of PPARγ, the role of LPAR5 in regulating the function of macrophages or TAM in the context of cancer is largely unknown (Ray & Rai, 2017). However, an in vivo model of murine psoriasis was used to show that LPAR5 induces activation of the NLRP3 inflammasome in macrophages, causing the production of proinflammatory cytokines. Treatment with TCLPA5–4 improved psoriasis symptoms and reduced the infiltration of macrophages into skin lesions (Gaire et al., 2020).

LPAR5 is also highly expressed in human mast cells. Activation of LPAR5 by LPA causes mast cells to release macrophage inflammatory protein-1β (MIP-1β), a potent activator and chemoattractant for many immune cells including monocytes, macrophages, dendritic cells, and lymphocytes (Lundequist & Boyce, 2011). Likewise, LPAR5 and LPAR2 are abundantly expressed in B cells. Activation of LPAR5 by LPA inhibits antigen specific BCR-induced Ca2+ mobilization and subsequent antibody production, suggesting that LPAR5 signaling may be involved in impeding humoral immune responses (Hu et al., 2014). While the observation that LPAR5 is abundantly expressed in many different immune cell types is certainly exciting, more research is needed to fully comprehend its significance in the context of inflammatory diseases and tumor immunity.

9. Role of LPAR5 in regulating intestinal epithelial cell repair

The gastrointestinal (GI) tract is lined with a layer of enterocytes responsible for nutrient absorption and formation of a physical and chemical barrier to protect the host from harmful contents in the GI lumen. LPAR1, LPAR2 and LPAR5 are abundantly expressed in the intestinal epithelium, where they regulate a wide range of cellular functions (Lee et al., 2013). For example, LPAR1 promotes the proliferation and migration of rat intestinal epithelial cells (IEC-6) via a PLCβ-dependent pathway. The authors found that both the number of proliferating enterocytes in the crypts and the rate of enterocytes migration along the crypt-villus axis were significantly reduced in Lpar1−/− KO mice relative to WT or Lpar2−/− KO mice. Furthermore, LPA treatment accelerated intestinal wound closure in WT mice but not in Lpar1−/− KO mice, suggesting that LPAR1 may play an important role in wound repair (Lee et al., 2013).

LPA also protects IEC-6 cells from apoptosis induced by serum withdrawal, chemotherapy, and radiation (Deng et al., 2002). The protection from radiation-induced apoptosis is mediated by LPAR2, since Lpar2−/− KO mice were more susceptible to radiation-induced death than WT or Lpar1−/− KO mice (Deng et al., 2007). At least four key events occur upon activation of LPAR2 (Fig. 8A). First, the unique C terminus of LPAR2 contains LIM- and PDZ-binding domains that form a ternary macromolecular complex with thyroid receptor-interacting protein 6 (TRIP6) and Na\\H exchange response factor (NHERF2). This macromolecular signaling complex promotes the long-lasting activation of prosurvival signaling pathways such as ERK1/2 and AKT. Second, LPAR2 can bind to the pro-apoptotic mediator SIVA1 and target it for polyubiquitination and subsequent proteasomal degradation (E, et al., 2009). Third, radiation increases the expression of ATX and LPAR2, indicating the presence of a feed-forward protective mechanism that amplifies LPA signaling. Fourth, activation of LPAR2 accelerates the resolution of phosphorylated γH2AX, which aids in DNA repair (Balogh et al., 2015). All four events contribute to the protection of cells from apoptosis caused by genotoxic stress. Recently, we found that the Lgr5-positive intestinal stem cells (Lgr5+ ISC) that maintain the continuous renewal of the intestinal epithelium express LPAR2. We showed that the LPAR2 agonist radioprotectin-1 protected Lgr5+ ISC from radiation-induced apoptosis in vitro and protected mice from radiation-induced death (Kuo et al., 2018).

Fig. 8.

Fig. 8.

Role of LPAR in regulating intestinal epithelium repair and homeostasis. (A) LPAR2 protects enterocytes from radiation-induced apoptosis by promoting the proteasomal degradation of pro-apoptotic SIVA1, activation of ERK1/2 and AKT prosurvival pathways, and repairing DNA damage. Figure adapted from (Tigyi et al., 2019) (B) Although inducible Lpar5iKO mice show crypt ablation, intestinal inflammation, weight loss, and mortality by day 11, (C) constitutive Lpar5cKO mice show no GI abnormalities under physiological conditions. In the enterocytes of constitutive Lpar5cKO mice, a compensatory upregulation of LPAR2 was observed, which was responsible for maintaining normal GI functions in these mice. Similarly, LPAR5 was found to be upregulated in the enterocytes of germline Lpar2−/− KO mice in order to maintain normal GI functions under physiological conditions.

Although LPAR5 is highly expressed in the intestinal epithelium, its role in regulating GI functions has been investigated only recently. Surprisingly, inducing global deletion of Lpar5 (Lpar5iKO) in adult mice causes significant intestinal inflammation, crypt ablation, weight loss, diarrhea, and mortality by day 11 (Liang, He, Han, & Yun, 2022) (Fig. 8B). The loss of intestinal crypts in Lpar5iKO mice was further confirmed using cultured enteroids, in which deletion of Lpar5 results in the loss of epithelial integrity, increased enterocyte apoptosis, and an overall reduction in growth compared to enteroids from WT mice. Similar results were obtained on treatment of WT enteroids with TCLPA5–4, suggesting that LPAR5 plays an essential role in the survival and growth of enteroids. Moreover, deletion of Lpar5 in Lgr5+ ISC significantly decreased the number of crypts in vivo, suggesting that LPAR5 is required for the maintenance and renewal of ISC (Liang & Yun, 2022).

Nonetheless, the severe GI phenotype observed in Lpar5iKO is strikingly different from that seen in constitutive or germline Lpar5−/− KO mice whose GI functions are normal. This suggests that a compensatory mechanism may be present in Lpar5−/− KO mice to maintain normal GI functions. Using a model in which Lpar5 is constitutively deleted only in enterocytes (i.e., Villin-Cre; Lpar5f/f or Lpar5cKO mice), the authors found that LPAR2 and LPAR3 were significantly upregulated in Lpar5cKO enterocytes. Pharmacological inhibition of LPAR2 with H2L 5186303 significantly impaired the growth of Lpar5cKO enteroids. However, no effect was observed when LPAR3 was inhibited by the LPAR1/3 antagonist Ki16425, suggesting that LPAR2 compensates for the loss of LPAR5 function (Liang & Yun, 2022). Akt phosphorylation decreases after LPAR2 inhibition in Lpar5cKO enteroids, implying that LPAR2 may maintain the growth of Lpar5cKO enteroids via the PI3K/AKT pathway. These in vitro findings were confirmed in vivo when Lpar5cKO mice were treated with the LPAR2 antagonist H2L 5186303, which increased enterocyte apoptosis. Interestingly, expression of LPAR5 and LPAR3 is upregulated in enteroids isolated from Lpar2−/− KO mice, suggesting that a similar compensatory mechanism also may exist in Lpar2−/− KO mice. Indeed, pharmacological inhibition of LPAR5 with AS2717638 substantially reduces the growth of enteroids from Lpar2−/− KO mice, whereas a moderate effect was observed when the LPAR1/3 antagonist Ki16425 was used (Liang & Yun, 2022). It is worth noting that the majority of these studies were conducted under basal or normal conditions. Nevertheless, the discovery that LPAR2 and LPAR5 may have overlapping functions in the basal maintenance of intestinal epithelium certainly raises the question of whether this compensatory mechanism exists during genotoxic insults or pathological conditions (Fig. 8C). Although H2L 5186303 is a more potent antagonist of LPAR2, it also can inhibit LPAR3 at low micromolar concentrations (Fells et al., 2008). Furthermore, we recently verified that H2L 5186303 also blocked LPAR5-mediated Ca2+ mobilization at low micromolar concentrations (unpublished data). More research will be needed to investigate this phenomenon.

10. Development of antagonist targeting LPAR5

Based on the mounting evidence that implicates LPAR5 in various inflammatory diseases and cancer, considerable efforts have been made to develop LPAR5 antagonists. Two compounds, H2L 5987411 and H2L 5765834 were identified from an in-silico screening of the Hit2Lead database using a pharmacophore model of LPAR5. H2L 5987411 served as a partial antagonist of both LPAR5 and LPAR4, whereas H2L 5765834 had antagonistic effects on LPAR1 > LPAR5 > LPAR3. Both compounds inhibit LPA-induced human platelet aggregation in vitro (Williams et al., 2009).

Subsequently, the non-lipid small molecule inhibitor diphenyl pyrazole carboxylic acid (TCLPA5–4) was identified by a high-throughput screening campaign as a selective antagonist of LPAR5. TCLPA5–4 inhibits LPA-mediated Ca2+ mobilization in RH7777 cells that stably express LPAR5 and inhibits LPA-induced aggregation of human platelets. Moreover, TCLPA5–4 exhibits no off target effects at high micromolar concentrations when screened against >80 other drug targets, with the notable exception of one protease (Kozian et al., 2012).

A more potent derivative of TCLPA5–4 was later synthesized and designated Cpd3/UA08025. This compound inhibits LPA-mediated Ca2+ mobilization and the release of monocyte chemoattractant protein 1 from the HMC-1 human mast cell line. Although the authors reported that this compound had no effect when screened against LPAR1–3 at 10 μM, no detailed information regarding the type of cells or the assay was provided (Kozian et al., 2016). As mentioned previously, we found that Cpd3/UA08025 is a more potent antagonist of LPAR4 than LPAR5 when tested in the Ca2+ mobilization assay using cells that stably express the respective receptor (Lee et al., 2020).

An orally bioavailable selective LPAR5 antagonist termed AS2717638 was identified. This compound inhibited LPA-induced accumulation of cAMP in CHO cells that stably express human LPAR5. The selectivity of AS2717638 for LPAR1–3 was determined in cells that overexpress the respective human LPAR using Ca2+ assays. However, its activity was not verified against LPAR4 and LPAR6, which share higher homology with LPAR5 than with LPAR1–3. Nonetheless, the therapeutic utility of AS2717638 as an analgesic was evaluated in both mice and rats in which mechanical allodynia was induced by intrathecal injections of LPA, PGE2 or (S)-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). In all of the pain models tested, AS2717638 significantly reduced allodynia in WT but not in Lpar5−/− KO mice, confirming that the effect is mediated by LPAR5 (Murai et al., 2017). A summary of the IC50 values along with the assays that were used for target validation are listed for each compound in Table 1.

Table 1.

List of published LPAR5 antagonists. No effect (NE).

Compound IC50 at LPAR5 Selectivity References

H2L 5987411 3.5 μM [Ca2+ assay, RH7777 cells] NE at LPAR1–3
IC50 1.4 μM at LPAR4 [Ca2+ assay]
(Williams et al., 2009)
H2L 5765834 0.46 μM [Ca2+ assay RH7777 cells] NE at LPAR2 and LPAR4
IC50 0.09 μM at LPAR1 [Ca2+ assay]
IC50 0.75 μM at LPAR3 [Ca2+ assay]
(Williams et al., 2009)
TCLPA5–4 0.8 μM [Ca2+ assay]
2.2 μM [human platelets]
NE at >80 drug targets (Kozian et al., 2012)
Cpd3/UA08025 0.14 μM [MCP-1 release, HMC-1 cells]
0.14 μM [Ca2+ assay, HMC-1 cells]
0.73 μM [Ca2+ assay, BV-2 cells]
3 μM [Ca2+ assay, B103 cells]
16.9 μM [β-arrestin assay]
IC50 > 10 μM at LPAR1–3
IC50 0.9 μM at LPAR4 [Ca2+ assay]
(Kozian et al., 2016; Lee et al., 2020)
AS2717638 0.04 μM [cAMP assay, CHO cells]
0.33 μM [TGFα shedding assay, HEK cells; unpublished]
IC50 > 10 μM at LPAR1–3 [Ca2+ assay]
NE at 10 μM on binding of 22 other targets (receptors/channels)
(Murai et al., 2017)

11. Conclusions

Despite significant advances in our understanding of LPAR5 biology in both physiology and pathophysiology, many of the molecular mechanisms underlying the cellular functions mediated by LPAR5 remain unknown. While we are beginning to comprehend the role of LPAR5 in modulating the immune response of T lymphocytes in the context of cancer, its role in inflammatory diseases has not yet been evaluated. Similarly, while LPAR5 appears to modulate macrophage and microglia functions by driving proinflammatory signals in these cells and affecting the healing process of skin lesions and brain injury, respectively, their role in cancer remains unknown. There is a gap in our knowledge as to whether LPAR5 is expressed by other immune cell types, such as dendritic cells, natural killer T cells, neutrophils, and myeloid-derived suppressor cells and if so, what its function(s) may be. In light of the recent discovery that LPAR2 and LPAR5 may compensate for one another in maintaining intestinal homeostasis, this begs the question of whether such overlapping function with other LPARs exists under physiological and pathophysiological conditions. Although germline or constitutive gene knockout mice models are useful for identifying genes required for embryo development and determining the functional roles of specific genes in pathophysiology, the fact that these models can potentially mask compensatory mechanisms in animals with no overt phenotype reminds us that all models have limitations and that the inclusion of inducible and/or conditional knockout models when available is critical.

Acknowledgments

All colored figures are original and were created with BioRender. com. The authors wish to thank Dr. Kyle Johnson Moore of the UTHSC Office of Scientific Writing for editing of the manuscript.

Funding

This work was supported by the William and Ella Owens Medical Research Foundation (SCL), the University of Tennessee CORNET grant (SCL), the National Cancer Institute grant CA092160 (GT), the National Cancer Institute grant CA266207 (GT and SCL), and the Harriet Van Vleet endowment (GT).

Abbreviations:

ATX

autotaxin

cAMP

adenosine 3′,5’-cyclic monophosphate

DRG

dorsal root ganglion

EMT

epithelial to mesenchymal transition

Enpp2

ectonucleotide pyrophosphatase phosphodiesterase 2

GPAT

glycerol-3 phosphate acyltransferase

GPCR

G protein-coupled receptor

HMC-1

human mast cells

ISC

intestinal stem cells

LPA

lysophosphatidic acid

LPAR

lysophosphatidic acid receptor

MMP

matrix metalloproteinase

NSCLC

non-small-cell lung carcinoma

pCREB

cAMP response element binding protein

PLA1

phospholipase A1

PLA2

phospholipase A2

PLD

phospholipase D

PPARγ

peroxisome proliferator activated receptor gamma

PSNL

partial sciatic nerve ligation

PTC

papillary thyroid carcinoma

tMCAO

transient middle cerebral artery occlusion

TRPV1

transient receptor potential vanilloid 1

Footnotes

Declaration of Competing Interest

The authors declare no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Data availability

No data was used for the research described in the article.

References

  1. Aikawa S, Hashimoto T, Kano K, & Aoki J (2015). Lysophosphatidic acid as a lipid mediator with multiple biological actions. Journal of Biochemistry 157, 81–89. [DOI] [PubMed] [Google Scholar]
  2. Amisten S, Braun OO, Bengtsson A, & Erlinge D (2008). Gene expression profiling for the identification of G-protein coupled receptors in human platelets. Thrombosis Research 122, 47–57. [DOI] [PubMed] [Google Scholar]
  3. Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishimoto T, … Arai H. (2002). Serum lysophosphatidic acid is produced through diverse phospholipase pathways. The Journal of Biological Chemistry 277, 48737–48744. [DOI] [PubMed] [Google Scholar]
  4. Bachiller S, Jimenez-Ferrer I, Paulus A, Yang Y, Swanberg M, Deierborg T, & Boza-Serrano A (2018). Microglia in neurological diseases: A road map to brain-disease dependent-inflammatory response. Frontiers in Cellular Neuroscience 12, 488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Baker DL, Morrison P, Miller B, Riely CA, Tolley B, Westermann AM, … Tigyi G. (2002). Plasma lysophosphatidic acid concentration and ovarian cancer. JAMA 287, 3081–3082. [DOI] [PubMed] [Google Scholar]
  6. Baker RR, & Chang HY (1999). Evidence for two distinct lysophospholipase activities that degrade lysophosphatidylcholine and lysophosphatidic acid in neuronal nuclei of cerebral cortex. Biochimica et Biophysica Acta 1438, 253–263. [DOI] [PubMed] [Google Scholar]
  7. Balazs L, Okolicany J, Ferrebee M, Tolley B, & Tigyi G (2001). Topical application of the phospholipid growth factor lysophosphatidic acid promotes wound healing in vivo. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 280, R466–R472. [DOI] [PubMed] [Google Scholar]
  8. Balijepalli P, Sitton CC, & Meier KE (2021). Lysophosphatidic acid signaling in Cancer cells: What makes LPA so special? Cells 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Balogh A, Shimizu Y, Lee SC, Norman DD, Gangwar R, Bavaria M, … Tigyi G. (2015). The autotaxin-LPA2 GPCR axis is modulated by gamma-irradiation and facilitates DNA damage repair. Cellular Signalling 27, 1751–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Balood M, Zahednasab H, Siroos B, Mesbah-Namin SA, Torbati S, & Harirchian MH (2014). Elevated serum levels of lysophosphatidic acid in patients with multiple sclerosis. Human Immunology 75, 411–413. [DOI] [PubMed] [Google Scholar]
  11. Bandoh K, Aoki J, Taira A, Tsujimoto M, Arai H, & Inoue K (2000). Lysophosphatidic acid (LPA) receptors of the EDG family are differentially activated by LPA species. Structure-activity relationship of cloned LPA receptors. FEBS Letters 478, 159–165. [DOI] [PubMed] [Google Scholar]
  12. Bektas M, Payne SG, Liu H, Goparaju S, Milstien S, & Spiegel S (2005). A novel acylglycerol kinase that produces lysophosphatidic acid modulates cross talk with EGFR in prostate cancer cells. The Journal of Cell Biology 169, 801–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Benesch MG, Zhao YY, Curtis JM, McMullen TP, & Brindley DN (2015). Regulation of autotaxin expression and secretion by lysophosphatidate and sphingosine 1-phosphate. Journal of Lipid Research 56, 1134–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bolen AL, Naren AP, Yarlagadda S, Beranova-Giorgianni S, Chen L, Norman D, … Tigyi G. (2011). The phospholipase A1 activity of lysophospholipase A-I links platelet activation to LPA production during blood coagulation. Journal of Lipid Research 52, 958–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bot M, Bot I, Lopez-Vales R, van de Lest CH, Saulnier-Blache JS, Helms JB, … Biessen EA. (2010). Atherosclerotic lesion progression changes lysophosphatidic acid homeostasis to favor its accumulation. The American Journal of Pathology 176, 3073–3084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Callaerts-Vegh Z, Leo S, Vermaercke B, Meert T, & D’Hooge R (2012). LPA5 receptor plays a role in pain sensitivity, emotional exploration and reversal learning. Genes, Brain, and Behavior 11, 1009–1019. [DOI] [PubMed] [Google Scholar]
  17. Castelino FV, Bain G, Pace VA, Black KE, George L, Probst CK, … Tager AM. (2016). An Autotaxin/lysophosphatidic acid/Interleukin-6 amplification loop drives scleroderma fibrosis. Arthritis & Rhematology 68, 2964–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. van Corven EJ, Groenink A, Jalink K, Eichholtz T, & Moolenaar WH (1989). Lysophosphatidate-induced cell proliferation: Identification and dissection of signaling pathways mediated by G proteins. Cell 59, 45–54. [DOI] [PubMed] [Google Scholar]
  19. Crack PJ, Zhang M, Morganti-Kossmann MC, Morris AJ, Wojciak JM, Fleming JK, … Pebay A. (2014). Anti-lysophosphatidic acid antibodies improve traumatic brain injury outcomes. Journal of Neuroinflammation 11, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Davies SS, Pontsler AV, Marathe GK, Harrison KA, Murphy RC, Hinshaw JC, … McIntyre TM. (2001). Oxidized alkyl phospholipids are specific, high affinity peroxisome proliferator-activated receptor gamma ligands and agonists. The Journal of Biological Chemistry 276, 16015–16023. [DOI] [PubMed] [Google Scholar]
  21. Demoyer JS, Skalak TC, & Durieux ME (2000). Lysophosphatidic acid enhances healing of acute cutaneous wounds in the mouse. Wound Repair and Regeneration 8, 530–537. [DOI] [PubMed] [Google Scholar]
  22. Deng W, Balazs L, Wang DA, Van Middlesworth L, Tigyi G, & Johnson LR (2002). Lysophosphatidic acid protects and rescues intestinal epithelial cells from radiation- and chemotherapy-induced apoptosis. Gastroenterology 123, 206–216. [DOI] [PubMed] [Google Scholar]
  23. Deng W, Shuyu E, Tsukahara R, Valentine WJ, Durgam G, Gududuru V, … Tigyi G. (2007). The lysophosphatidic acid type 2 receptor is required for protection against radiation-induced intestinal injury. Gastroenterology 132, 1834–1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Dong Y, Hirane M, Araki M, Fukushima N, Honoki K, & Tsujiuchi T (2014). Lysophosphatidic acid receptor-5 negatively regulates cell motile and invasive activities of human sarcoma cell lines. Molecular and Cellular Biochemistry 393, 17–22. [DOI] [PubMed] [Google Scholar]
  25. Eichholtz T, Jalink K, Fahrenfort I, & Moolenaar WH (1993). The bioactive phospholipid lysophosphatidic acid is released from activated platelets. The Biochemical Journal 291(Pt 3), 677–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. English D, Garcia JG, & Brindley DN (2001). Platelet-released phospholipids link haemostasis and angiogenesis. Cardiovascular Research 49, 588–599. [DOI] [PubMed] [Google Scholar]
  27. Escalante-Alcalde D, Hernandez L, Le Stunff H, Maeda R, Lee HS Jr., Gang C, … Stewart CL. (2003). The lipid phosphatase LPP3 regulates extra-embryonic vasculogenesis and axis patterning. Development 130, 4623–4637. [DOI] [PubMed] [Google Scholar]
  28. Etulain J (2018). Platelets in wound healing and regenerative medicine. Platelets 29, 556–568. [DOI] [PubMed] [Google Scholar]
  29. Fells JI, Tsukahara R, Fujiwara Y, Liu J, Perygin DH, Osborne DA, … Parrill AL. (2008). Identification of non-lipid LPA3 antagonists by virtual screening. Bioorganic & Medicinal Chemistry 16, 6207–6217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fischer DJ, Liliom K, Guo Z, Nusser N, Virag T, Murakami-Murofushi K, … Tigyi G. (1998). Naturally occurring analogs of lysophosphatidic acid elicit different cellular responses through selective activation of multiple receptor subtypes. Molecular Pharmacology 54, 979–988. [DOI] [PubMed] [Google Scholar]
  31. Foell JL, Hesse M, Volkmer I, Schmiedel BJ, Neumann I, & Staege MS (2008). Membrane-associated phospholipase A1 beta (LIPI) is an Ewing tumour-associated cancer/testis antigen. Pediatric Blood & Cancer 51, 228–234. [DOI] [PubMed] [Google Scholar]
  32. Fujiwara Y, Sardar V, Tokumura A, Baker D, Murakami-Murofushi K, Parrill A, & Tigyi G (2005). Identification of residues responsible for ligand recognition and regioisomeric selectivity of lysophosphatidic acid receptors expressed in mammalian cells. The Journal of Biological Chemistry 280, 35038–35050. [DOI] [PubMed] [Google Scholar]
  33. Gaire BP, Lee CH, Kim W, Sapkota A, Lee DY, & Choi JW (2020). Lysophosphatidic acid receptor 5 contributes to Imiquimod-induced psoriasis-like lesions through NLRP3 Inflammasome activation in macrophages. Cells 9, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Geraldo LHM, Spohr T, Amaral RFD, Fonseca A, Garcia C, Mendes FA, … Lima, F. R. S. (2021). Role of lysophosphatidic acid and its receptors in health and disease: Novel therapeutic strategies. Signal Transduction and Targeted Therapy 6, 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Gerrard JM, & Robinson P (1989). Identification of the molecular species of lysophosphatidic acid produced when platelets are stimulated by thrombin. Biochimica et Biophysica Acta 1001, 282–285. [DOI] [PubMed] [Google Scholar]
  36. Goetzl EJ, & Rosen H (2004). Regulation of immunity by lysosphingolipids and their G protein-coupled receptors. The Journal of Clinical Investigation 114, 1531–1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Hadidi P, & Athanasiou KA (2013). Enhancing the mechanical properties of engineered tissue through matrix remodeling via the signaling phospholipid lysophosphatidic acid. Biochemical and Biophysical Research Communications 433, 133–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Haseruck N, Erl W, Pandey D, Tigyi G, Ohlmann P, Ravanat C, … Siess W. (2004). The plaque lipid lysophosphatidic acid stimulates platelet activation and platelet-monocyte aggregate formation in whole blood: Involvement of P2Y1 and P2Y12 receptors. Blood 103, 2585–2592. [DOI] [PubMed] [Google Scholar]
  39. Hashimoto T, Ohata H, & Momose K (2004). Itch-scratch responses induced by lysophosphatidic acid in mice. Pharmacology 72, 51–56. [DOI] [PubMed] [Google Scholar]
  40. He Y, Zhou H, Huang X, Qu Y, Wang Y, Pei W, … You H. (2022). Infiltration of LPAR5 (+) macrophages in osteosarcoma tumor microenvironment predicts better outcomes. Frontiers in Immunology 13, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Hu J, Oda SK, Shotts K, Donovan EE, Strauch P, Pujanauski LM, … Torres RM. (2014). Lysophosphatidic acid receptor 5 inhibits B cell antigen receptor signaling and antibody response. Journal of Immunology 193, 85–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Inoue A, Arima N, Ishiguro J, Prestwich GD, Arai H, & Aoki J (2011). LPA-producing enzyme PA-PLA(1)alpha regulates hair follicle development by modulating EGFR signalling. The EMBO Journal 30, 4248–4260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Inoue M, Rashid MH, Fujita R, Contos JJ, Chun J, & Ueda H (2004). Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling. Nature Medicine 10, 712–718. [DOI] [PubMed] [Google Scholar]
  44. Jalink K, Hordijk PL, & Moolenaar WH (1994). Growth factor-like effects of lysophosphatidic acid, a novel lipid mediator. Biochimica et Biophysica Acta 1198, 185–196. [DOI] [PubMed] [Google Scholar]
  45. Jalink K, van Corven EJ, & Moolenaar WH (1990). Lysophosphatidic acid, but not phosphatidic acid, is a potent Ca2(+)-mobilizing stimulus for fibroblasts. Evidence for an extracellular site of action. The Journal of Biological Chemistry 265, 12232–12239. [PubMed] [Google Scholar]
  46. Jongsma M, Matas-Rico E, Rzadkowski A, Jalink K, & Moolenaar WH (2011). LPA is a chemorepellent for B16 melanoma cells: Action through the cAMP-elevating LPA5 receptor. PLoS One 6, Article e29260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Joshi L, Plastira I, Bernhart E, Reicher H, Koshenov Z, Graier WF, … Sattler W. (2022). Lysophosphatidic acid receptor 5 (LPA(5)) knockout ameliorates the neuroinflammatory response in vivo and modifies the inflammatory and metabolic landscape of primary microglia in vitro. Cells 11, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Joshi L, Plastira I, Bernhart E, Reicher H, Triebl A, Kofeler HC, & Sattler W (2021). Inhibition of Autotaxin and lysophosphatidic acid receptor 5 attenuates neuroinflammation in LPS-activated BV-2 microglia and a mouse endotoxemia model. International Journal of Molecular Sciences 22, 1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Katsifa A, Kaffe E, Nikolaidou-Katsaridou N, Economides AN, Newbigging S, McKerlie C, & Aidinis V (2015). The bulk of Autotaxin activity is dispensable for adult mouse life. PLoS One 10, Article e0143083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Khandoga AL, Pandey D, Welsch U, Brandl R, & Siess W (2011). GPR92/LPA (5) lysophosphatidate receptor mediates megakaryocytic cell shape change induced by human atherosclerotic plaques. Cardiovascular Research 90, 157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Kirschner H, & Vogt W (1961). Pharmacologically active lipidsoluble acids in brain extracts: Isolation of lysophosphatidic acid and ganglioside. Biochemical Pharmacology 8, 224–234. [DOI] [PubMed] [Google Scholar]
  52. Kittaka H, Uchida K, Fukuta N, & Tominaga M (2017). Lysophosphatidic acid-induced itch is mediated by signalling of LPA(5) receptor, phospholipase D and TRPA1/TRPV1. The Journal of Physiology 595, 2681–2698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Kotarsky K, Boketoft A, Bristulf J, Nilsson NE, Norberg A, Hansson S, … Olde B. (2006). Lysophosphatidic acid binds to and activates GPR92, a G protein-coupled receptor highly expressed in gastrointestinal lymphocytes. The Journal of Pharmacology and Experimental Therapeutics 318, 619–628. [DOI] [PubMed] [Google Scholar]
  54. Kozian DH, Evers A, Florian P, Wonerow P, Joho S, & Nazare M (2012). Selective non-lipid modulator of LPA5 activity in human platelets. Bioorganic & Medicinal Chemistry Letters 22, 5239–5243. [DOI] [PubMed] [Google Scholar]
  55. Kozian DH, von Haeften E, Joho S, Czechtizky W, Anumala UR, Roux P, … Nazare M. (2016). Modulation of Hexadecyl-LPA-mediated activation of mast cells and microglia by a chemical probe for LPA5. Chembiochem 17, 861–865. [DOI] [PubMed] [Google Scholar]
  56. Kremer AE, Martens JJ, Kulik W, Rueff F, Kuiper EM, van Buuren HR, … Oude Elferink RP. (2010). Lysophosphatidic acid is a potential mediator of cholestatic pruritus. Gastroenterology 139, 1008–1018. [DOI] [PubMed] [Google Scholar]
  57. Kremer KN, Buser A, Thumkeo D, Narumiya S, Jacobelli J, Pelanda R, & Torres RM (2022). LPA suppresses T cell function by altering the cytoskeleton and disrupting immune synapse formation. Proceedings of the National Academy of Sciences of the United States of America 119, Article e2118816119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kroeze WK, Sassano MF, Huang XP, Lansu K, McCorvy JD, Giguere PM, … Roth BL. (2015). PRESTO-tango as an open-source resource for interrogation of the druggable human GPCRome. Nature Structural & Molecular Biology 22, 362–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kuo B, Szabo E, Lee SC, Balogh A, Norman D, Inoue A, … Tigyi G. (2018). The LPA (2) receptor agonist Radioprotectin-1 spares Lgr5-positive intestinal stem cells from radiation injury in murine enteroids. Cellular Signalling 51, 23–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kurano M, Suzuki A, Inoue A, Tokuhara Y, Kano K, Matsumoto H, … Yatomi Y. (2015). Possible involvement of minor lysophospholipids in the increase in plasma lysophosphatidic acid in acute coronary syndrome. Arteriosclerosis, Thrombosis, and Vascular Biology 35, 463–470. [DOI] [PubMed] [Google Scholar]
  61. Kuwajima K, Sumitani M, Kurano M, Kano K, Nishikawa M, Uranbileg B, Tsuchida R, Ogata T, Aoki J, Yatomi Y, & Yamada Y (2018). Lysophosphatidic acid is associated with neuropathic pain intensity in humans: An exploratory study. PLoS One 13, Article e0207310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Langedijk J, Araya EI, Barroso AR, Tolenaars D, Nazare M, Belabed H, … Oude Elferink R. (2022). An LPAR (5) -antagonist that reduces nociception and increases pruriception. Front Pain Res (Lausanne) 3, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Lee CW, Rivera R, Gardell S, Dubin AE, & Chun J (2006). GPR92 as a new G12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. The Journal of Biological Chemistry 281, 23589–23597. [DOI] [PubMed] [Google Scholar]
  64. Lee H, Goetzl EJ, & An S (2000). Lysophosphatidic acid and sphingosine 1-phosphate stimulate endothelial cell wound healing. American Journal of Physiology. Cell Physiology 278, C612–C618. [DOI] [PubMed] [Google Scholar]
  65. Lee SC, Dacheux MA, Norman DD, Balazs L, Torres RM, Augelli-Szafran CE, & Tigyi GJ (2020). Regulation of tumor immunity by lysophosphatidic acid. Cancers (Basel) 12, 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Lee SC, Fujiwara Y, Liu J, Yue J, Shimizu Y, Norman DD, … Tigyi GJ. (2015). Autotaxin and LPA1 and LPA5 receptors exert disparate functions in tumor cells versus the host tissue microenvironment in melanoma invasion and metastasis. Molecular Cancer Research 13, 174–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Lee SJ, Leoni G, Neumann PA, Chun J, Nusrat A, & Yun CC (2013). Distinct phospholipase C-beta isozymes mediate lysophosphatidic acid receptor 1 effects on intestinal epithelial homeostasis and wound closure. Molecular and Cellular Biology 33, 2016–2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Li S, & Zhang J (2009). Lipopolysaccharide induces autotaxin expression in human monocytic THP-1 cells. Biochemical and Biophysical Research Communications 378, 264–268. [DOI] [PubMed] [Google Scholar]
  69. Li ZG, Yu ZC, Wang DZ, Ju WP, Zhan X, Wu QZ, … Man HH. (2008). Influence of acetylsalicylate on plasma lysophosphatidic acid level in patients with ischemic cerebral vascular diseases. Neurological Research 30, 366–369. [DOI] [PubMed] [Google Scholar]
  70. Liang Z, He P, Han Y, & Yun CC (2022). Survival of stem cells and progenitors in the intestine is regulated by LPA(5)-dependent signaling. Cellular and Molecular Gastroenterology and Hepatology 14, 129–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Liang Z, & Yun CC (2022). Compensatory upregulation of LPA(2) and activation of the PI3K-Akt pathway prevent LPA(5)-dependent loss of intestinal epithelial cells in intestinal organoids. Cells 11, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Liliom K, Guan Z, Tseng JL, Desiderio DM, Tigyi G, & Watsky MA (1998). Growth factor-like phospholipids generated after corneal injury. The American Journal of Physiology 274, C1065–C1074. [DOI] [PubMed] [Google Scholar]
  73. Lin ME, Rivera RR, & Chun J (2012). Targeted deletion of LPA5 identifies novel roles for lysophosphatidic acid signaling in development of neuropathic pain. The Journal of Biological Chemistry 287, 17608–17617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lundequist A, & Boyce JA (2011). LPA5 is abundantly expressed by human mast cells and important for lysophosphatidic acid induced MIP-1beta release. PLoS One 6, Article e18192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Ma L, Uchida H, Nagai J, Inoue M, Chun J, Aoki J, & Ueda H (2009). Lysophosphatidic acid-3 receptor-mediated feed-forward production of lysophosphatidic acid: An initiator of nerve injury-induced neuropathic pain. Molecular Pain 5, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Magkrioti C, Galaris A, Kanellopoulou P, Stylianaki EA, Kaffe E, & Aidinis V (2019). Autotaxin and chronic inflammatory diseases. Journal of Autoimmunity 104, 1–10. [DOI] [PubMed] [Google Scholar]
  77. Mathew D, Kremer KN, Strauch P, Tigyi G, Pelanda R, & Torres RM (2019). LPA5 is an inhibitory receptor that suppresses CD8 T-cell cytotoxic function via disruption of early TCR signaling. Frontiers in Immunology 10, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Mazereeuw-Hautier J, Gres S, Fanguin M, Cariven C, Fauvel J, Perret B, … Saulnier-Blache JS. (2005). Production of lysophosphatidic acid in blister fluid: Involvement of a lysophospholipase D activity. The Journal of Investigative Dermatology 125, 421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradere JP, … Jonkers J. (2006). Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Molecular and Cellular Biology 26, 5015–5022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Moolenaar WH, Houben AJ, Lee SJ, & van Meeteren LA (2013). Autotaxin in embryonic development. Biochimica et Biophysica Acta 1831, 13–19. [DOI] [PubMed] [Google Scholar]
  81. Moolenaar WH, Kruijer W, Tilly BC, Verlaan I, Bierman AJ, & de Laat SW (1986). Growth factor-like action of phosphatidic acid. Nature 323, 171–173. [DOI] [PubMed] [Google Scholar]
  82. Moraes LA, Spyridon M, Kaiser WJ, Jones CI, Sage T, Atherton RE, & Gibbins JM (2010). Non-genomic effects of PPARgamma ligands: Inhibition of GPVI-stimulated platelet activation. Journal of Thrombosis and Haemostasis 8, 577–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Murai N, Hiyama H, Kiso T, Sekizawa T, Watabiki T, Oka H, & Aoki T (2017). Analgesic effects of novel lysophosphatidic acid receptor 5 antagonist AS2717638 in rodents. Neuropharmacology 126, 97–107. [DOI] [PubMed] [Google Scholar]
  84. Nieto-Posadas A, Picazo-Juarez G, Llorente I, Jara-Oseguera A, Morales-Lazaro S, Escalante-Alcalde D, … Rosenbaum T. (2011). Lysophosphatidic acid directly activates TRPV1 through a C-terminal binding site. Nature Chemical Biology 8, 78–85. [DOI] [PubMed] [Google Scholar]
  85. Nikitopoulou I, Oikonomou N, Karouzakis E, Sevastou I, Nikolaidou-Katsaridou N, Zhao Z, … Aidinis V. (2012). Autotaxin expression from synovial fibroblasts is essential for the pathogenesis of modeled arthritis. The Journal of Experimental Medicine 209, 925–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Ninou I, Magkrioti C, & Aidinis V (2018). Autotaxin in pathophysiology and pulmonary fibrosis. Front Med (Lausanne) 5, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Nochi H, Tomura H, Tobo M, Tanaka N, Sato K, Shinozaki T, … Tamoto K. (2008). Stimulatory role of lysophosphatidic acid in cyclooxygenase-2 induction by synovial fluid of patients with rheumatoid arthritis in fibroblast-like synovial cells. Journal of Immunology 181, 5111–5119. [DOI] [PubMed] [Google Scholar]
  88. Oda SK, Strauch P, Fujiwara Y, Al-Shami A, Oravecz T, Tigyi G, … Torres RM. (2013). Lysophosphatidic acid inhibits CD8 T cell activation and control of tumor progression. Cancer Immunology Research 1, 245–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Oh DY, Yoon JM, Moon MJ, Hwang JI, Choe H, Lee JY, … Seong JY. (2008). Identification of farnesyl pyrophosphate and N-arachidonylglycine as endogenous ligands for GPR92. The Journal of Biological Chemistry 283, 21054–21064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Ohuchi H, Hamada A, Matsuda H, Takagi A, Tanaka M, Aoki J, … Noji S. (2008). Expression patterns of the lysophospholipid receptor genes during mouse early development. Developmental Dynamics 237, 3280–3294. [DOI] [PubMed] [Google Scholar]
  91. Olorundare OE, Peyruchaud O, Albrecht RM, & Mosher DF (2001). Assembly of a fibronectin matrix by adherent platelets stimulated by lysophosphatidic acid and other agonists. Blood 98, 117–124. [DOI] [PubMed] [Google Scholar]
  92. Pamuklar Z, Federico L, Liu S, Umezu-Goto M, Dong A, Panchatcharam M, … Smyth SS (2009). Autotaxin/lysopholipase D and lysophosphatidic acid regulate murine hemostasis and thrombosis. The Journal of Biological Chemistry 284, 7385–7394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Pamuklar Z, Lee JS, Cheng HY, Panchatcharam M, Steinhubl S, Morris AJ, … Smyth SS. (2008). Individual heterogeneity in platelet response to lysophosphatidic acid: Evidence for a novel inhibitory pathway. Arteriosclerosis, Thrombosis, and Vascular Biology 28, 555–561. [DOI] [PubMed] [Google Scholar]
  94. Pan HL, Zhang YQ, & Zhao ZQ (2010). Involvement of lysophosphatidic acid in bone cancer pain by potentiation of TRPV1 via PKCepsilon pathway in dorsal root ganglion neurons. Molecular Pain 6, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Panetti TS, Chen H, Misenheimer TM, Getzler SB, & Mosher DF (1997). Endothelial cell mitogenesis induced by LPA: Inhibition by thrombospondin-1 and thrombospondin-2. The Journal of Laboratory and Clinical Medicine 129, 208–216. [DOI] [PubMed] [Google Scholar]
  96. Panetti TS, Magnusson MK, Peyruchaud O, Zhang Q, Cooke ME, Sakai T, & Mosher DF (2001). Modulation of cell interactions with extracellular matrix by lysophosphatidic acid and sphingosine 1-phosphate. Prostaglandins & Other Lipid Mediators 64, 93–106. [DOI] [PubMed] [Google Scholar]
  97. Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, … Sattler W. (2017). Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. Journal of Neuroinflammation 14, 1–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Plastira I, Bernhart E, Goeritzer M, Reicher H, Kumble VB, Kogelnik N, … Sattler W. (2016). 1-Oleyl-lysophosphatidic acid (LPA) promotes polarization of BV-2 and primary murine microglia towards an M1-like phenotype. Journal of Neuroinflammation 13, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Plastira I, Bernhart E, Joshi L, Koyani CN, Strohmaier H, Reicher H, … Sattler W. (2020). MAPK signaling determines lysophosphatidic acid (LPA)-induced inflammation in microglia. Journal of Neuroinflammation 17, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Plastira I, Joshi L, Bernhart E, Schoene J, Specker E, Nazare M, & Sattler W (2019). Small-molecule lysophosphatidic acid receptor 5 (LPAR5) antagonists: Versatile pharmacological tools to regulate inflammatory signaling in BV-2 microglia cells. Frontiers in Cellular Neuroscience 13, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Ray R, & Rai V (2017). Lysophosphatidic acid converts monocytes into macrophages in both mice and humans. Blood 129, 1177–1183. [DOI] [PubMed] [Google Scholar]
  102. Reinartz S, Lieber S, Pesek J, Brandt DT, Asafova A, Finkernagel F, … Muller R. (2019). Cell type-selective pathways and clinical associations of lysophosphatidic acid biosynthesis and signaling in the ovarian cancer microenvironment. Molecular Oncology 13, 185–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, Tigyi G, & Siess W (2003). Subtype-selective antagonists of lysophosphatidic acid receptors inhibit platelet activation triggered by the lipid core of atherosclerotic plaques. Circulation 108, 741–747. [DOI] [PubMed] [Google Scholar]
  104. Rowley JW, Oler AJ, Tolley ND, Hunter BN, Low EN, Nix DA, … Weyrich AS. (2011). Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood 118, e101–e111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sano T, Baker D, Virag T, Wada A, Yatomi Y, Kobayashi T, … Tigyi G. (2002). Multiple mechanisms linked to platelet activation result in lysophosphatidic acid and sphingosine 1-phosphate generation in blood. The Journal of Biological Chemistry 277, 21197–21206. [DOI] [PubMed] [Google Scholar]
  106. Sapkota A, Lee CH, Park SJ, & Choi JW (2020). Lysophosphatidic acid receptor 5 plays a pathogenic role in brain damage after focal cerebral ischemia by modulating neuroinflammatory responses. Cells 9, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Sasagawa T, Suzuki K, Shiota T, Kondo T, & Okita M (1998). The significance of plasma lysophospholipids in patients with renal failure on hemodialysis. Journal of Nutritional Science and Vitaminology (Tokyo) 44, 809–818. [DOI] [PubMed] [Google Scholar]
  108. Sauer B, Vogler R, Zimmermann K, Fujii M, Anzano MB, Schafer-Korting M, … Kleuser B. (2004). Lysophosphatidic acid interacts with transforming growth factor-beta signaling to mediate keratinocyte growth arrest and chemotaxis. The Journal of Investigative Dermatology 123, 840–849. [DOI] [PubMed] [Google Scholar]
  109. Shimizu Y, Morikawa Y, Okudaira S, Kimoto S, Tanaka T, Aoki J, & Tokumura A (2014). Potentials of the circulating pruritogenic mediator lysophosphatidic acid in development of allergic skin inflammation in mice: Role of blood cell-associated lysophospholipase D activity of autotaxin. The American Journal of Pathology 184, 1593–1603. [DOI] [PubMed] [Google Scholar]
  110. Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, Corrinth C, … Aepfelbacher M. (1999). Lysophosphatidic acid mediates the rapid activation of platelets and endothelial cells by mildly oxidized low density lipoprotein and accumulates in human atherosclerotic lesions. Proceedings of the National Academy of Sciences of the United States of America 96, 6931–6936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Simon MF, Chap H, & Douste-Blazy L (1982). Human platelet aggregation induced by 1-alkyl-lysophosphatidic acid and its analogs: A new group of phospholipid mediators? Biochemical and Biophysical Research Communications 108, 1743–1750. [DOI] [PubMed] [Google Scholar]
  112. Song J, Guan M, Zhao Z, & Zhang J (2015). Type I interferons function as autocrine and paracrine factors to induce Autotaxin in response to TLR activation. PLoS One 10, Article e0136629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Spinelli SL, O’Brien JJ, Bancos S, Lehmann GM, Springer DL, Blumberg N, … Phipps RP. (2008). The PPAR-platelet connection: Modulators of inflammation and potential cardiovascular effects. PPAR Research 2008, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Stortelers C, Kerkhoven R, & Moolenaar WH (2008). Multiple actions of lysophosphatidic acid on fibroblasts revealed by transcriptional profiling. BMC Genomics 9, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Sturm A, Sudermann T, Schulte KM, Goebell H, & Dignass AU (1999). Modulation of intestinal epithelial wound healing in vitro and in vivo by lysophosphatidic acid. Gastroenterology 117, 368–377. [DOI] [PubMed] [Google Scholar]
  116. Suckau O, Gross I, Schrotter S, Yang F, Luo J, Wree A, … Brauer AU. (2019). LPA(1), LPA(2), LPA(4), and LPA(6) receptor expression during mouse brain development. Developmental Dynamics 248, 375–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Sugiura T, Nakane S, Kishimoto S, Waku K, Yoshioka Y, Tokumura A, & Hanahan DJ (1999). Occurrence of lysophosphatidic acid and its alkyl ether-linked analog in rat brain and comparison of their biological activities toward cultured neural cells. Biochimica et Biophysica Acta 1440, 194–204. [DOI] [PubMed] [Google Scholar]
  118. Sun XY, Li HZ, Xie DF, Gao SS, Huang X, Guan H, … Zhou PK. (2022). LPAR5 confers radioresistance to cancer cells associated with EMT activation via the ERK/Snail pathway. Journal of Translational Medicine 20, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, … Luster AD. (2008). The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nature Medicine 14, 45–54. [DOI] [PubMed] [Google Scholar]
  120. Tanaka M, Okudaira S, Kishi Y, Ohkawa R, Iseki S, Ota M, … Arai H. (2006). Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophosphatidic acid. The Journal of Biological Chemistry 281, 25822–25830. [DOI] [PubMed] [Google Scholar]
  121. Tang X, & Brindley DN (2020). Lipid phosphate phosphatases and Cancer. Biomolecules 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Thompson FJ, & Clark MA (1994). Purification of a lysophosphatidic acid-hydrolysing lysophospholipase from rat brain. The Biochemical Journal 300(Pt 2), 457–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Tigyi G, Dyer DL, & Miledi R (1994). Lysophosphatidic acid possesses dual action in cell proliferation. Proceedings of the National Academy of Sciences of the United States of America 91, 1908–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Tigyi G, Hong L, Yakubu M, Parfenova H, Shibata M, & Leffler CW (1995). Lysophosphatidic acid alters cerebrovascular reactivity in piglets. The American Journal of Physiology 268, H2048–H2055. [DOI] [PubMed] [Google Scholar]
  125. Tigyi G, & Miledi R (1992). Lysophosphatidates bound to serum albumin activate membrane currents in Xenopus oocytes and neurite retraction in PC12 pheochromocytoma cells. The Journal of Biological Chemistry 267, 21360–21367. [PubMed] [Google Scholar]
  126. Tigyi GJ, Johnson LR, Lee SC, Norman DD, Szabo E, Balogh A, … McCool WS. (2019). Lysophosphatidic acid type 2 receptor agonists in targeted drug development offer broad therapeutic potential. Journal of Lipid Research 60, 464–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Tokumura A, Carbone LD, Yoshioka Y, Morishige J, Kikuchi M, Postlethwaite A, & Watsky MA (2009). Elevated serum levels of arachidonoyl-lysophosphatidic acid and sphingosine 1-phosphate in systemic sclerosis. International Journal of Medical Sciences 6, 168–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Tokumura A, Fukuzawa K, & Tsukatani H (1978). Effects of synthetic and natural lysophosphatidic acids on the arterial blood pressure of different animal species. Lipids 13, 572–574. [DOI] [PubMed] [Google Scholar]
  129. Tokumura A, Sinomiya J, Kishimoto S, Tanaka T, Kogure K, Sugiura T, … Fukuzawa K. (2002). Human platelets respond differentially to lysophosphatidic acids having a highly unsaturated fatty acyl group and alkyl ether-linked lysophosphatidic acids. The Biochemical Journal 365, 617–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Tsukahara R, Yamamoto S, Yoshikawa K, Gotoh M, Tsukahara T, Neyama H, … Ueda H. (2018). LPA5 signaling is involved in multiple sclerosis-mediated neuropathic pain in the cuprizone mouse model. Journal of Pharmacological Sciences 136, 93–96. [DOI] [PubMed] [Google Scholar]
  131. Tsukahara T, Tsukahara R, Yasuda S, Makarova N, Valentine WJ, Allison P, … Tigyi G. (2006). Different residues mediate recognition of 1-O-oleyllysophosphatidic acid and rosiglitazone in the ligand binding domain of peroxisome proliferator-activated receptor gamma. The Journal of Biological Chemistry 281, 3398–3407. [DOI] [PubMed] [Google Scholar]
  132. Ueda H (2020). LPA receptor signaling as a therapeutic target for radical treatment of neuropathic pain and fibromyalgia. Pain Manag 10, 43–53. [DOI] [PubMed] [Google Scholar]
  133. Vogt W (1957). The chemical nature of Darmstoff. The Journal of Physiology 137, 154–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Wang H, Tu S, Yang S, Shen P, Huang Y, Ba X, … Chen, Z. (2019). Berberine modulates LPA function to inhibit the proliferation and inflammation of FLS-RA via p38/ERK MAPK pathway mediated by LPA(1). Evidence-based Complementary and Alternative Medicine 2019, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Watanabe N, Ikeda H, Nakamura K, Ohkawa R, Kume Y, Aoki J, … Yatomi Y. (2007). Both plasma lysophosphatidic acid and serum autotaxin levels are increased in chronic hepatitis C. Journal of Clinical Gastroenterology 41, 616–623. [DOI] [PubMed] [Google Scholar]
  136. Williams JR, Khandoga AL, Goyal P, Fells JI, Perygin DH, Siess W, … Fujiwara Y. (2009). Unique ligand selectivity of the GPR92/LPA5 lysophosphatidate receptor indicates role in human platelet activation. The Journal of Biological Chemistry 284, 17304–17319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wu CY, Zheng C, Xia EJ, Quan RD, Hu J, Zhang XH, & Hao RT (2020). Lysophosphatidic acid receptor 5 (LPAR5) plays a significance role in papillary thyroid cancer via phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway. Medical Science Monitor 26, Article e919820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Wu JM, Xu Y, Skill NJ, Sheng H, Zhao Z, Yu M, … Maluccio MA. (2010). Autotaxin expression and its connection with the TNF-alpha-NF-kappaB axis in human hepatocellular carcinoma. Molecular Cancer 9, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Xiang H, Lu Y, Shao M, & Wu T (2020). Lysophosphatidic acid receptors: Biochemical and clinical implications in different diseases. Journal of Cancer 11, 3519–3535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Xie W, Matsumoto M, Chun J, & Ueda H (2008). Involvement of LPA1 receptor signaling in the reorganization of spinal input through Abeta-fibers in mice with partial sciatic nerve injury. Molecular Pain 4, 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Xu KP, Yin J, & Yu FS (2007). Lysophosphatidic acid promoting corneal epithelial wound healing by transactivation of epidermal growth factor receptor. Investigative Ophthalmology & Visual Science 48, 636–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Xu Y, Gaudette DC, Boynton JD, Frankel A, Fang XJ, Sharma A, … Mellors A., et al. (1995). Characterization of an ovarian cancer activating factor in ascites from ovarian cancer patients. Clinical Cancer Research 1, 1223–1232. [PubMed] [Google Scholar]
  143. Yanagida K, Masago K, Nakanishi H, Kihara Y, Hamano F, Tajima Y, … Ishii S. (2009). Identification and characterization of a novel lysophosphatidic acid receptor, p2y5/LPA6. The Journal of Biological Chemistry 284, 17731–17741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Yin H, Chu A, Li W, Wang B, Shelton F, Otero F, … Chen YA. (2009). Lipid G protein-coupled receptor ligand identification using beta-arrestin PathHunter assay. The Journal of Biological Chemistry 284, 12328–12338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Yukiura H, Kano K, Kise R, Inoue A, & Aoki J (2015). Autotaxin overexpression causes embryonic lethality and vascular defects. PLoS One 10, Article e0126734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Yung YC, Stoddard NC, & Chun J (2014). LPA receptor signaling: Pharmacology, physiology, and pathophysiology. Journal of Lipid Research 55, 1192–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Zeng R, Li B, Huang J, Zhong M, Li L, Duan C, … Dai Y. (2017). Lysophosphatidic acid is a biomarker for peritoneal carcinomatosis of gastric cancer and correlates with poor prognosis. Genetic Testing and Molecular Biomarkers 21, 641–648. [DOI] [PubMed] [Google Scholar]
  148. Zhang HP, Chen QK, & Xu JF (2020). LPAR5 stimulates the malignant progression of non-small-cell lung carcinoma by upregulating MLLT11. European Review for Medical and Pharmacological Sciences 24, 8902–8910. [DOI] [PubMed] [Google Scholar]
  149. Zhang ZH, Chen H, Vaziri ND, Mao JR, Zhang L, Bai X, & Zhao YY (2016). Metabolomic signatures of chronic kidney disease of diverse etiologies in the rats and humans. Journal of Proteome Research 15, 3802–3812. [DOI] [PubMed] [Google Scholar]
  150. Zhao WJ, Zhu LL, Yang WQ, Xu SJ, Chen J, Ding XF, … Chen G. (2021). LPAR5 promotes thyroid carcinoma cell proliferation and migration by activating class IA PI3K catalytic subunit p110beta. Cancer Science 112, 1624–1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Zhao Y, & Natarajan V (2013). Lysophosphatidic acid (LPA) and its receptors: Role in airway inflammation and remodeling. Biochimica et Biophysica Acta 1831, 86–92. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was used for the research described in the article.

RESOURCES