Vol. 55 No. 2/2008, 227–240

on-line at: www.actabp.pl Review

Lysophosphatidic acids, cyclic phosphatidic acids and autotaxin as promissing targets in therapies of cancer and other diseases

Edyta Gendaszewska-Darmach

Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Technical University of Łódź, Łódź, Poland

Received: 17 March, 2008; revised: 20 May, 2008; accepted: 30 May, 2008 available on-line: 14 June, 2008

Lysophospholipids have long been recognized as membrane phospholipid metabolites, but only recently lysophosphatidic acids (LPA) have been demonstrated to act on specific G protein-cou- pled receptors. The widespread expression of LPA receptors and coupling to several classes of G proteins allow LPA-dependent regulation of numerous processes, such as vascular development, neurogenesis, wound healing, immunity, and cancerogenesis. Lysophosphatidic acids have been found to induce many of the hallmarks of cancer including cellular processes such as prolifera- tion, survival, migration, invasion, and neovascularization. Furthermore, autotaxin (ATX), the main converting lysophosphatidylcholine into LPA was identified as a tumor cell - auto crine motility factor. On the other hand, cyclic phosphatidic acids (naturally occurring analogs of LPA generated by ATX) have anti-proliferative activity and inhibit tumor cell invasion and metastasis. Research achievements of the past decade suggest implementation of preclinical and clinical evaluation of LPA and its analogs, LPA receptors, as well as autotaxin as potential thera- peutic targets.

Keywords: autotaxin/NPP2, , cyclic phosphatidic acid, G protein-coupled receptors

INTRODUCTION small amounts of LPA associated with membrane biosynthesis, some cellular sources (such as activat- Lysophosphatidic acids (LPA; 1-acyl-2-hy- ed platelets) can produce significant amounts of ex- droxy-sn-glycero-3-phosphates) (Fig. 1) are simple tracellular LPA, which account for the LPA found in phospholipids consisting of various compounds serum (Eichholtz et al., 1993). Although a signaling bearing both saturated (16:0, 18:0) and unsaturated role of lysophosphatidic acids has been recognized, (18:1, 18:2, 20:4) fatty acid chains. They have been the identification and cloning of cell surface Gpro- recognized for decades as components of biologi- tein-coupled receptors (GPCRs) having high affinity cal membranes. However, they are also ubiquitous for LPA (LPA1–LPA5) in the last several years has bioactive molecules influencing a broad variety of dramatically improved our understanding of the cellular processes, particularly proliferation, differ- diverse roles of LPA in biological processes (Meyer entiation, and migration. Although all cells contain zu Heringdorf & Jacobs, 2007). Recently, peroxisome

Corresponding author: Edyta Gendaszewska-Darmach, Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Technical University of Łódź, Stefanowskiego 4/10, 90-924 Łódź, Poland; e-mail: [email protected] 2+ 2+ Abbreviations: AC, adenyl cyclase; ATX, autotaxin; [Ca ]i, intracellular free Ca concentration; cPA, cyclic phosphatidic acids; EGF, epidermal growth factor; GPCR, G protein-coupled receptor; LDL, low-density lipoproteins; LPA, lysophos- phatidic acids; LPA1-5, LPA receptor type 1-5; LPAAT, LPA acyl ; LPC, lysophosphatidylcholine; LPE, lyso- phosphatidylethanolamine; LPI, lysophosphatidylinositol; LPLs, lysophosholipids; LPPs, lipid phosphate ; LPS, lysophosphatidylserine; lysoPLD, D; MAG, monoacylglycerol; moxLDL, mild oxidation of low density lipoproteins; NPP, nucleotide pyrophosphatase/; PA, phosphatidic acids; PKCγ, protein kinase C γ; PPARγ, peroxisome proliferator-activated receptor γ; S1P, sphingosine-1-phosphate; SPC, sphingosyl-phosphorylcho- line; TZDs, thiazolidinediones; VEGF, vascular endothelial growth factor; VSMCs, vascular smooth muscle cells. 228 E. Gendaszewska-Darmach 2008

mainly LPA metabolism and LPA-mediated signal- . ing (van Meeteren & Moolenaar, 2007).

O O O O O LPA RECEPTORS R C O OH R C O O P P O O O O

LPA cPA Cell surface receptors in LPA signaling

An extracellular action of LPA was first men- tioned in the 1970s when Tokumura et al. (1978) described the effect of LPA on blood pressure. However, clear evidence of LPA as an extracellu- O O C H lar signaling molecule only came in the late 1980s 6 13 C O O P O (van Corven et al., 1989). The first for an LPA O receptor (LPA1) was identified in 1996 during a PHYLPA search for with predominant expression in the ventricular zone of the cerebral cortex. Hecht et al. (1996) identified ventricular zone gene 1(VZG-1) that was shown to encode a high-affinity GPCR for LPA (further renamed EDG2 based on its similarity to “endothelium differentiation genes“ — EDGs). Sub- R=saturated or unsaturated fatty acid chain . sequently, sequence similarities allowed rapid iden- Figure 1. Structures of lysophosphatidic acid (LPA), cy- tification of further cognate LPA receptors (Contos clic phosphatidic acid (cPA), and PHYLPA. et al., 2000). The second LPA receptor gene, EDG4, was identified by An et al. (1998) on the basis of functional studies. The encoded protein was deter- proliferator-activated receptor γ (PPARγ) has been mined to be another LPA receptor (LPA2). In 1999 also identified as an intracellular receptor for LPA Bandoh and coworkers isolated a human cDNA

(McIntyre et al., 2003). The widespread expression encoding the third LPA receptor (LPA3) and desig- of LPA receptors and coupling to several classes of nated it EDG7. Because of the inconsistency of the G proteins allow regulation of numerous processes, LPA receptors’ nomenclature, in 2002 EDG2/VZG-1, such as vascular development, neurogenesis, immu- EDG4 and EDG7 genes were renamed as LPA1, LPA2 nity, cancerogenesis (Gardell et al., 2006) and wound and LPA3 following the guidelines of IUPHAR (In- healing (Watterson et al., 2007). ternational Union of Pharmacology). Then, in 2003 It is worth to mention that among the most Noguchi and coworkers identified p2y(9)/GPR23 as intensively studied lysophospholipids there is not the fourth LPA receptor, LPA4. With only 20–24% only LPA but also sphingosine-1-phosphate (S1P). amino-acid homology with Edg-2/LPA1, Edg-4/LPA2, This molecule has diverse signaling functions me- and Edg-7/LPA3, LPA4 is evolutionarily distant from diated by five cell membrane receptors (S1P1 to other LPA receptors, which share 50–57% identity S1P5). One of S1P analogs (FTY720 or 2-amino-2[2- of their amino-acid sequences. Recently, Lee et al. (4-octylphenyl)ethyl]-1,3-propanediol also known as (2006a) demonstrated that the orphan GPR92 is acti- fingolimod) acts as an agonist of S1P and is currently vated by LPA, and named it LPA5. A comparison of tested in Phase III clinical trials for multiple sclerosis, amino-acid sequences showed that this receptor had and, independently, in Phase III as a novel immu- approx. 35% amino-acid identity with the LPA4 re- nosuppressant for kidney transplantation (Gardell et ceptor (Lee et al., 2006a) and only between 21.3 and al., 2006). 22.6% homology with the LPA1-3 family of receptors The studies on clinical evaluation of S1P as a (Kotarsky et al., 2006). therapeutic target have indicated that interventions LPA receptors couple to members of three in LPA signaling might also provide new therapeu- major G protein families, Gi, Gq, and G12 (Table 1). tic approaches for the treatment of human diseases. LPA1 and LPA2 are known to interact with mem- The pleiotropic roles of lysophospatidic acids in can- bers of all three aforementioned G protein families, cer behavior suggest that novel LPA-related drugs while LPA3 interacts with Gi and Gq proteins only. would have wide applicability in the treatment of LPA4 and LPA5 elevate cAMP levels, but their cou- many types of tumors. A significant progress in pling to Gs proteins has yet to be proven. The LPA5 this field as well as unexpected results were -recent receptor additionally interacts with Gq and G12 pro- ly achieved and therefore this review will describe teins (Meyer zu Heringdorf & Jacobs, 2007). The Vol. 55 Biological activity of LPA and cPA 229

Table 1. LPA receptors-mediated signaling pathways

Receptor Expression in human organs G family Effector References

LPA1 Widely expressed; low expression in skeletal musc- Gi, Gq, G12 AC↓ An et al. (1998), le, kidney, lung and thymus; not detected in liver ERK↑ Anliker et al. (2004), and peripheral blood leukocytes Akt↑ Meyer zu Heringdorf & Rho↑ Jacobs (2007) PLC↑ 2+ [Ca ]i↑

LPA2 High level in testis and leukocytes; moderate in Gi, Gq, G12 Rho↑ An et al. (1998) pancreas, thymus, spleen and prostate; little or no PLC↑ Anliker et al. (2004), 2+ expression in brain, heart, lung, liver, kidney, mu- [Ca ]i↑ Meyer zu Heringdorf & scle, ovary, placenta, intestine and colon ERK↑ Jacobs (2007) Akt↑ AC↓

LPA3 High level in heart, prostate, brain, pancreas and Gi, Gq PLC↑ Bandoh et al. (1999) 2+ testis; moderate level in lung and ovary [Ca ]i↑ Anliker et al. (2004), ERK↑ Meyer zu Heringdorf & AC↓ Jacobs (2007)

LPA4 Weakly expressed (high level only in ovary) AC↑ Noguchi et al. (2003) 2+ [Ca ]i↑

LPA5 Weakly expressed in most organs; strong expres- Gq, G12 AC↑ Lee et al. (2006a) 2+ sion in small intestine, spleen, dorsal root ganglion [Ca ]i↑ cells and embryonic stem cells major effector systems of LPA receptors include tors increases insulin sensitivity, and therefore TZDs phoshatidylinositol-3-kinase (PI3K), Gi — Ras — are frequently administered to patients with insulin extracellular signal regulated kinase (ERK), Gq — resistance associated with type II diabetes. C (PLC), and a small GTPase (Rho). McIntyre et al. (2003) showed that LPA, but Activation of PLC, ERK or Rho via LPA receptors not their precursors (phosphatidic acids) displaced results in cell proliferation as well as survival and rosiglitazone from the ligand-binding pocket of a changes in cell morphology. purified PPARγ protein. Using RAW 264.7 mono- One cannot exclude that some other LPA re- cytic cells transfected with a luciferase gene con- ceptors (membrane or intracellular) may be identi- trolled by peroxisome proliferator response element, fied in the future. One of the candidates for intracel- they showed that LPA stimulated transcription of lular LPA receptors appears to be peroxisome prolif- the reporter gene. However, unlike rosiglitazone (a erator-activated receptor γ (PPARγ) (McIntyre et al., PPARγ agonist), LPA were unable to induce PPARγ 2003). activity in adipocytes (Simon et al., 2005). Although LPA were demonstrated to bind to PPARγ in an Interaction of LPA with peroxisome proliferator-acti- in vitro assay (McIntyre et al., 2003), their ability vated receptor γ (PPARγ) to activate PPARγ appears to be dependent on the cell type. In adipocytes, activity of high ecto-lipid The peroxisome proliferator-activated recep- phosphate phosphohydrolase was observed, which tors (PPARs) are nuclear fatty acid receptors that dephosphorylates and inactivates LPA (Simon et have been implicated to play an important role in al., 2002). This finding also suggests that ecto-lip- obesity-related metabolic diseases such as hyperlip- id phosphate phosphohydrolase activity could be idemia, insulin resistance, and coronary artery dis- weaker in monocytes than in adipocytes, allowing ease (Lee et al., 2003a). PPARγ controls transcrip- a higher amount of LPA to enter into the cells and tion of genes that in general are involved in energy activate PPARγ. metabolism (glucose and fatty acids), is essential for adipocyte differentiation and also modulates anti- inflammatory as well as antineoplastic reactions. LPA METABOLISM PPARγ is activated after binding natural ligands such as polyunsaturated fatty acids or prostaglandin metabolites. It can also be activated by synthetic li- involved in LPA production gands such as thiazolidinediones (TZDs, e.g. rosigli- tazone, pioglitazone or troglitazone) (Lehmann et al., LPA have long been known as products of 1995). Most of them bind predominantly to PPARγ the lipid synthetic pathways generated from glycer- in adipose cells. The binding of TZDs to their recep- ol-3-phosphate and acyl-CoA by glycerolphosphate 230 E. Gendaszewska-Darmach 2008

Figure 2. Synthesis of LPA in serum. In a platelet-dependent pathway, acti- vated platelets release large amounts of phospholipids which are then converted

by (PLA1) and phos- pholipase A2 (PLA2) to lysophospholip- ids (LPLs) such as lysophosphatidylcho- line (LPC), lysophosphatidylethanolamine (LPE), and lysophosphatidylserine (LPS). Subsequently LPA is generated from LPLs by a plasma enzyme lysophospholipase D (lysoPLD). About half of serum LPA is produced by a platelet-independent path- way from lysophosphatidylcholine. Plasma LPC is synthesized mainly by lecithin-cho- lesterol acyltransferase (LCAT) which cata- lyzes transesterification of phosphatidyl- choline and free cholesterol. acyl . The conversion occurs in the endo- antagonists of platelet LPA receptors might pro- plasmic reticulum and mitochondria (Aoki, 2004). vide a new strategy to prevent thrombus forma- However, new mechanisms of LPA production have tion in patients with cardiovascular diseases. also been identified both in cells and in biological In serum and plasma LPA are mainly pro- fluids, where multiple pathways occur under vari- duced from lysophospholipids. By contrast, in plate- ous conditions. LPA are produced extracellularly by lets and some cancer cells they are created from lipoprotein oxidation (Siess et al., 1999) or by the ac- phosphatidic acids (PA). Serum is the best charac- tion of secretory on microvesicles terized source of LPA. In serum incubated for 1 h, released from activated cells (Fourcade et al., 1995). total LPA levels are significantly higher (0.85–1.5 LPA are also produced in plasma by thrombin- μM) than those measured in corresponding plasma activated platelets through the stimulated release samples (0.6–0.7 μM). The overall increase of total of phospholipase A1, phospholipase A2 (Sano et al., LPA content in serum versus EDTA-anticoagulated 2002), and lysophospholipase D (autotaxin) (Toku- plasma is due to the contribution of platelet-derived mura et al., 2002; Umezu-Goto et al., 2002), which act LPA, which was determined to be 0.6 μM following on plasma lipids or secreted lysophosphatidylcho- a 1-hour incubation at 25°C (Baker et al., 2001). It line (Fig. 2). has been proposed that LPA in serum are produced Mild oxidation of low density proteins as a result of blood coagulation and, therefore, plate- (moxLDL) catalyzed by Cu2+ changes the biologi- lets are involved, in part, in the production of these cal properties of LDL making them prothrombotic phospholipids in serum. Activated platelets pro- and proatherogenic. Siess et al. (1999) discovered duce and release a large amount of lysophosholipids that lysophosphatidic acids are formed during (LPLs) such as lysophosphatidylcholine (LPC), lyso- mild oxidation of LDL and are responsible for phosphatidylethanolamine (LPE), and lysophosphati- platelet activation induced by moxLDL and mini- dylserine (LPS). Aoki et al. (2002) propose that in a mally modified LDL. They also found that LPA ac- platelet-dependent pathway platelets supply LPLs cumulate in the intima of human atherosclerotic which are produced by phospholipase A1 (PLA1) hy- lesions. LPA and moxLDL activate Rho and Rho- drolysing fatty acids at the sn-1 position and phos- kinase through G12/13 and this pathway mediates pholipase A2 (PLA2) hydrolysing them at the sn-2 the reorganization of the actin cytoskeleton un- position. Subsequently, these LPLs are converted derlying platelet shape change (Retzer & Essler, to LPA by a plasma enzyme lysophospholipase D 2000). In addition, LPA and moxLDL stimulate a (Fig. 2). This pathway requires activation of plate- different pathway during shape change. Thus the lets. Thus, LPA are produced through this pathway activation of the Src family of tyrosine kinases under pathological conditions such as injury, inflam- and the tyrosine kinase Syk occurs, which medi- mation or artherosclerosis. ates the exposure of fibrinogen-binding sites on About half of serum LPA are produced by a integrin αIIbβ3 during a shape change, which is a platelet-independent pathway. Their levels in freshly prerequisite for platelet aggregation (Bauer et al., prepared plasma or serum are much lower than in 2001). LPA molecules present in the core region of samples incubated for 24 h at 25°C (about 1 μM and atherosclerotic plaques trigger rapid platelet acti- 5 μM, respectively) (Baker et al., 2001), indicating vation through the stimulation of LPA1 and LPA3 that lysophosphatidic acids are also produced in a receptors (Rother et al., 2003). Finding of efficient cell-free system. Tokumura et al. (1986) first showed Vol. 55 Biological activity of LPA and cPA 231 that LPA are created in plasma from lysophosphati- pected discovery that the lysophospholipase D pro- dylcholines by lysophosphospholipase D (lysoPLD). ducing LPA in serum was identical with autotaxin This activity has been detected in plasma and serum (ATX, also known as NPP2), a widely expressed nu- in a wide variety of mammalian species including cleotide pyrophosphatase/phosphodiesterase (NPP) rat, mouse, cattle, and humans (Aoki, 2004). Initially, (Umezu-Goto et al., 2002; Tokumura et al., 2002). Au- only LPC was considered as a substrate for serum totaxin has been known since the early 1990s as an lysoPLD. However, the enzyme also acts on other ecto-nucleotide pyrophosphatase/phosphodiesterase LPLs including LPE, LPS and LPI (lysophosphatidyl­ that stimulates the motility of A2058 melanoma cells inositols) (Aoki et al., 2002). LysoPLD is specific for in a pertussis toxin-sensitive manner (Stracke et al., the “lyso” forms of phospholipids; this means that 1992). Molecular cloning of ATX has revealed that it the enzyme does not act on is a member of the NPP family which also includes (PC) or other diacyl phospholipids. Plasma lysoPLD NPP1/PC-1, gp130RB13-6/NPP3, NPP4, NPP5, NPP6 was purified in 2002 and it was found to be identi- and NPP7 (Stefan et al., 2005). NPP1-3 hydrolyzes cal with autotaxin (ATX) — an earlier identified cell nucleotides and some of their derivatives with the motility-stimulating factor (Stracke et al., 1992; Ume- formation of nucleoside-5’-monophosphates. The zu-Goto et al., 2002; Tokumura et al., 2002). Since lys- phosphodiesterase activity of ATX/NPP2 could not oPLD/ATX is a key enzyme of significant biological explain the involvement of the enzyme in the stimu- importance, its detailed characterization will be fur- lation of melanoma cells and tumor progression. It ther discussed (see next chapter). Lysophosphatidyl- was difficult to envision how altered nucleotide pro- cholines — one of the main substrates for lysoPLD cessing in the extracellular environment could ac- — exist in plasma at a concentration of 125–150 μM, count for these biological effects of ATX. Finally, the making them the most abundant lysophospholipids identification of ATX/NPP2 as a lysophospholipase in plasma (Tigyi & Parrill, 2003). Plasma LPC are D allowed the previously unconnected areas of cell synthesized mainly by two enzymes: liver PLA1 and biology to be linked: the biological activities of the lecithin-cholesterol acyltransferase (LCAT), which NPP family members and LPA synthesis and signal- catalyze transesterification of phosphatidylcholines ing. ATX/NPP2 is unique in that it is the only lyso- and free cholesterol. PLD within the NPP family. However, it was found

Lysophosphatidic acids are also produced that the Km values for LPC and nucleotide substrate and released from many types of cells. However, are 100–150 μM and 0.9–1.0 mM, respectively. This in contrast to the mechanism of LPA production indicates that although ATX/lysoPLD is capable of in serum and plasma, the mechanisms of their hydrolyzing nucleotides in vitro, it primarily func- synthesis in cells are still ambiguous. Fourcade tions as a lipid phosphodiesterase (van Meeteren et et al. (1995) first demonstrated LPA production al., 2005). from microvesicles shed by activated cells, such ATX/lysoPLD has no homology to previously as erythrocytes, platelets and white blood cells. characterized . Instead, similarly to LPA are generated as a result of cellular activation its closest relatives (NPP1 and NPP3), ATX has a induced by various stimuli (e.g. phorbol esters, domain structure consisting of two N-terminal so- bombesin, ATP, and LPA itself) (Aoki, 2004). The matomedin B-like (SMB) domains, a central catalytic major pathway consists of two steps: production phosphodiesterase (PDE) domain and a C-terminal of phosphatidic acids (PA) and subsequent conver- -like domain (Stefan et al., 2005). Amino ac- sion of PA to LPA. The first step of this pathway, ids from all these domains form a lysophospholipid- conversion of diacyl phospholipids (for example: binding pocket. The central catalytic domain of ATX PC) to phosphatidic acids, is mainly catalyzed by (approx. 400 amino acids) is, however, structurally intracellular (PLD) (Cummings et unrelated to those found in typical phospholipases; al., 2002). The next step, the conversion of PA to it has been predicted to fold similarly to the cata-

LPA, is catalyzed by PLA1 and PLA2 phospholi- lytic domains of alkaline phosphatases. The C-ter- pases. minal domain of NPP1-3 is structurally related to The co-existence of the various pathways re- non-specific DNA or RNA , however, sponsible for LPA synthesis suggests that this proc- it lacks the key residues required for enzymatic ac- ess has to be highly regulated under in vivo condi- tivity. An intact nuclease-like domain is needed for tions, but details of this regulation remain to be ex- proper folding, subcellular localization and secretion. plained. Full-length ATX is synthesized as a pre-pro-enzyme and its secretion requires specific proteolytic cleav- Autotaxin/NPP2 — a secreted lysophospholipase D age. Following the removal of a 27-amino-acid frag- ment by the signal peptidase, NPP2 is subsequently A major step towards understanding of the cleaved by proprotein convertases (PCs). Then it biological activities of LPA was made by the unex- traffics along the classical export pathway and is se- 232 E. Gendaszewska-Darmach 2008 creted as an active glycoprotein (Jansen et al., 2005). (Tigyi & Parrill, 2003). Therefore, why do plasma ATX is N-glycosylated on three sites (N52, N410 and LPA levels reach only 100 nM (Aoki et al., 2002)? N524). Mutagenesis studies showed that the glyco- van Meeteren et al. showed that ATX is a subject of sylation of N524, a conserved residue in the catalytic feedback inhibition by enzymatic hydrolysis prod- domain, was absolutely required for the activity of ucts, LPA and sphingosine 1-phosphate (2005). This ATX (Jansen et al., 2007). phenomenon offers the most likely explanation of The human ATX-encoding gene, ENPP2, is the low plasma LPA levels and provides insight into organized in 27 exons. Alternative splicing results modulation of ATX activity. in generation of three different isoforms (van Mee- teren & Moolenaar, 2007). The shortest and the most Cyclic phosphatidic acids — naturally occurring abundant form (ATXter), having 863 amino acids, analogs of LPA generated by ATX was initially cloned from a teratocarcinoma cell line and is identical with plasma lysoPLD. The melano- There are several lines of evidence that ATX ma-derived isoform (ATXmel; 915 amino acids) con- is capable of producing not only LPA, but also cy- tains a highly basic insertion (52 residues) in the clic phosphatidic acids (cPA) (Fig. 1) which contain catalytic domain. A less frequent, ‘brain-specific’ iso- a dioxaphospholane ring spanning the sn-2 and sn-3 form (originally termed PD-1α) is expressed mainly positions of glycerol. The first cPA family member in oligodendrocytes and contains a 24-residue inser- was originally isolated from myxoamoebae of a true tion close to the nuclease-like domain. slime mold, Physarum polycephalum, and designated NPP2 is expressed in many cell lines and as PHYLPA (Murakami-Murofushi et al., 1992). This tissues with the highest mRNA levels detected in compound had a cyclopropane-containing fatty acyl brain, ovary, lung, intestine and kidney. As a se- chain and a cyclic phosphate joining the sn-2 and sn- creted protein, NPP2 accumulates in body fluids 3 positions of glycerol (Fig. 1). PHYLPA reversibly such as plasma and cerebrospinal fluid. In addi- inhibited activity of eukaryotic DNA polymerases of tion, its elevated expression has been observed α-family (Murakami-Murofushi et al., 1992) and cell during tumor progression, and this upregulation proliferation (Murakami-Murofushi et al., 1993). correlates with the invasiveness of the cancer. Cyclic phosphatidic acids were also detected ATX was originally identified as a tumor cell mo- as physiological constituents of human serum (Ko- tility factor of A2058 melanoma cells (Stracke et bayashi et al., 1999). Recently it was found that ATX al., 1992). This protein is overexpressed in several is largely responsible for their generation in mam- human cancers, including glioblastoma, lung and malian serum (Tsuda et al., 2006). ATX has the abil- breast cancer, renal cell carcinoma, neuroblastoma, ity to synthesize cPA by catalysis of intramolecular thyroid carcinoma and Hodgkin lymphoma (Kishi transphosphatidylation in LPC molecules (Fig. 3). et al., 2006). The highest ATX expression is de- Although the structure of cPA is similar to that of tected in glioblastoma multiforme (GBM), a very lysophosphatidic acids, their biological activities are malignant cancer with high infiltration rate. Au- apparently distinct from those of LPA. Cyclic phos- totaxin stimulates cell survival, proliferation, con- phatidic acids have anti-proliferative activity (Mu- traction and migration, and these effects can all be rakami-Murofushi et al., 1992) and inhibit tumor due to its ability to generate signaling molecules cell invasion and metastasis both in vitro and in vivo such as lysophosphatidic acids. The formation of (Ishihara et al., 2004) (Table 2). Previous reports on LPA can also account for the ability of ATX to the anti-invasive properties of cPA suggested their promote tumor progression, angiogenesis and me- role in increasing cellular cAMP levels (Murakami- tastasis. In addition to converting LPC into LPA, Murofushi et al., 1993) and subsequent RhoA inacti- ATX can also hydrolyze sphingosyl-phosphoryl- vation (Mukai et al., 2000). These observations impli- choline (SPC) to yield sphingosine 1-phosphate cated cell surface GPCR activation as the response. (S1P). However, the physiological significance of However, natural cPA isoforms as well as various the SPC-to-S1P conversion is doubtful, since plas- ma levels of SPC are >1 000-fold lower than those Table 2. Comparison of biological activities of lyso- of LPC, and in addition ATX hydrolyzes SPC phosphatidic acids (LPA) and cyclic phosphatidic ac- much less efficiently than LPC (Clair et al., 2003). ids (cPA) Although many problems were solved when Biological response LPA cPA the biology associated with ATX was linked to the Cell proliferation generally inhibition formation of LPA, one significant question remained, activation namely, how are plasma LPA levels maintained in Tumor cell invasion and generally inhibition such a low nanomolar range? ATX is a ubiquitous metastasis activation 2+ plasma protein, and the substrate, LPC, is abundant- [Ca ]i increased increased ly present in plasma at a 125–150 μM concentration cAMP level decreased increased Vol. 55 Biological activity of LPA and cPA 233 cPA analogs were found to be poor activators of the dephosphorylate a variety of phospholipid sub-

LPA1, LPA2, LPA3, and LPA4 receptors (Baker et al., strates. Three mammalian LPP isoforms, termed 2006). On the other hand, it has been shown that cPA LPP1, LPP2 and LPP3, have been cloned. In addi- and their analogs are potent inhibitors of ATX activ- tion, a catalytically active splice variant of LPP1, ity and LPA production and thus these compounds termed LPP1a, and truncated catalytically inactive can block cancer cell invasion in vitro and tumor cell forms, termed LPP1b and LPP1c, have been iden- metastasis in vivo (Baker et al., 2006; Uchiyama et tified, although their physiological relevance is not al., 2007). Those results raised the possibility for the yet known (Pyne et al., 2004). Various reports of use of cPA as a potential target in cancer therapy. their substrate specificity suggest that, in general, However, it should be remembered that hydrolytic LPP1 and LPP3 can readily hydrolyze PA and LPA cleavage of the cyclic phosphate ring will lead to the thereby producing diacylglycerols and monoacyl­ formation of LPA which are well known to augment glycerols, whereas LPP2 is most active against LPA tumor invasion and metastasis. Furthermore, cleav- and S1P, producing monoacylglycerols and sphin- age of the fatty acid by phospholipases may limit gosine. the effective half-life of cPA. Hence, different ana- The second LPA conversion pathway involves logs of cPA were recently synthesized rather than the action of lysophosphatidic acid acyltransferases unmodified cPA. Among these novel compounds, (LPAAT). These enzymes catalyze the transfer of an so-called carba derivatives of cPA (ccPA), in which acyl group from acyl-CoA to LPA to form PA. Five the phosphate oxygen was replaced with a methy- members of the LPAAT family have been identi- lene group at either the sn-2 or the sn-3 position, fied and sequenced: LPAATα, LPAATβ, LPAATγ, showed a much more potent inhibitory effect on the LPAATδ and LPAATε (Tigyi & Parrill, 2003). Two migration of cancer cells in vitro and metastasis in of them, LPAATα and LPAATβ, appear responsible vivo than the natural cPA (Uchiyama et al., 2007). for most of the LPAAT activity in cells due to their higher catalytic potency in relation to other family Degradation of LPA members. LPAATα shows a marked preference for LPA over other acyl acceptors, including LPC, LPE In view of the pleiotropic effects generated and LPI. by LPA, not only their synthesis but also deg- radation is a precisely regulated multistep proc- ess. The degradation of extracellular LPA can be BIOLOGICAL ACTIVITIES OF LPA mainly attributed to the ectophosphatase activity of plasma membrane lipid phosphate phosphatases Lysophosphatidic acids are required to main- (LPPs). LPPs are integral membrane proteins that tain homeostasis of many physiological processes,

CH3 O + O O N CH R C O OH 3 P CH3 O O

Lysophosphatidylcholines hydrolysis

H2O O O O O R C O HO O O P R C O OH P O O lysoPLD O O Figure 3. Synthesis of LPA Intermediate and cPA by lysophospholi- LPA pase D. + LysoPLD catalyzes hydroly- Choline sis of LPC that produces LPA transphosphatidylation and choline. The enzyme also attacks the substrate to form O O a transient lysophosphatidyl- R C O O P lysoPLD intermediate. Then, O intramolecular transphosphati- O dylation occurs with the hy- droxyl group at the sn-2 posi- cyclic phosphatidic acids R = saturated or unsaturated fatty acid chain tion of LPC to form cPA. + Choline 234 E. Gendaszewska-Darmach 2008 including reproduction, vascular development, and tion of fibroblasts (Pilquil et al., 2006). LPA can also functioning of the nervous system. However, they stimulate fibroblast-collagen matrix contraction (Lee may also trigger the development and expansion of et al., 2003b). They show vasoconstrictive actions and pathological processes, including cancers. Since their enhance the production of matrix metalloproteina­ initial characterization as growth factors, the list of ses (Wu et al., 2005), which all are important events cellular responses to LPA has expanded consider- in tissue repair. Moreover, platelets themselves are ably and now includes also many non-proliferative activated by LPA which adds an element of posi- effects, ranging from stimulation of cell migration tive feedback in the wound healing response. When and survival to neurite retraction and gap junction applied topically, LPA promote wound healing in closure. skin (Balazs et al., 2001), while applied rectally they stimulate intestinal epithelial wound healing in rats The role of LPA in the cardiovascular system (Sturm & Dignass, 2002). Lysophosphatidic acids may also stimulate wound healing in the upper di- Lysophosphatidic acids are constitutively gestive organs, as they are present in the saliva at present in serum at physiological concentrations of physiologically relevant concentrations (close to about 1–5 μM. Under pathological conditions, LPA 1 μM). LPA markedly enhance the growth of cells is released to serum from platelets in response to in- of esophagus, pharynx, and tongue origin in vitro jury and thrombosis (Aoki et al., 2002) as well as to (Sugiura et al., 2002). Lysophosphatidic acids also myocardial infarction (Chen et al., 2003), which indi- stimulate the healing responses of human periodon- cates a role of these phospholipids in cardiac patho- tal ligament fibroblasts and interact positively with physiology. LPA may be involved in process of an- PDGF (Cerutis et al., 2007). giogenesis and atherosclerosis, as well as regulation Lysophosphatidic acids stimulate endothelial of blood vessel tone. The oxidation of low-density cell migration which is a critical feature of several lipoprotein (LDL) is thought to contribute to athero- physiological and pathological processes, including genesis, which is an inflammatory disease involving angiogenesis and metastasis. Neovascularization (an- activation of phagocytic cells (Dever et al., 2006). Ly- giogenesis) as the process of restoring of the vascu- sophosphatidic acids formed during mild oxidation lar network is an essential part of repair processes. of LDL initiate platelet activation and stimulate en- LPA enhance the expression of genes that promote dothelial cell stress-fiber and gap formation (Siess et angiogenesis, including those for vascular endothe- al., 1999). Moreover, LPA accumulate in and are the lial growth factor (VEGF) (Hu et al., 2001) and in- primary platelet-activating lipid of atherosclerotic terleukin-8 (IL-8) (So et al., 2004). Recently, it has plaques, as observed in human carotid artery speci- been proposed that LPA stimulate the expression of mens where LPA level was highest in the lipid-rich vascular endothelial growth factor (VEGF) through core, the region that is most thrombogenic and prone hypoxia-inducible factor-1 (HIF-1α) activation (Lee to rupture. An early change in atherosclerosis is also et al., 2006b). Upon binding to the hypoxia-respon- characterized by neointima formation resulting from sive element within the target gene, HIF-1 activates the proliferation and migration of dedifferentiated transcription of various hypoxia-inducible genes like vascular smooth muscle cells (VSMCs). Unsaturated angiogenic factors, including VEGF. LPA have been LPAs strongly induce VSMC dedifferentiation via the found to stabilize endothelial monolayer barriers, a coordinated activation of the extracellular signal-reg- later event in angiogenesis (English et al., 1999). Tak- ulated kinase (ERK) and p38 mitogen-activated pro- ing into account these data, LPA are potent media- tein kinase (p38MAPK), resulting in the proliferation tors of tissue repair and wound healing. and migration of dedifferentiated VSMCs (Yoshida et al., 2003). Zhang et al. (2004) have shown that LPA LPA and autotaxin in tumor progression containing unsaturated acyl elicit progressive and long-lasting neointima formation in the rat carotid The formation of vasculature by angiogenesis artery, which requires activation of PPARγ. All these is essential not only for embryonic development or findings suggest that in vivo LPA may act as endog- proper wound healing but also for the unrestrained enous atherogenic factors. growth of tumors. LPA and their receptors play an important role in the development of ovarian, pros- LPA in wound healing tate, breast, head and neck cancers. Among different pathological processes, the Not surprisingly for a platelet-produced role of LPA in ovarian cancer has been studied most growth factor, LPA have all the hallmarks of a extensively. In ovary cancer, LPA contribute to the wound healing agonist: they stimulate the prolifer- development, progression, and metastasis and their ation and migration of endothelial cells (Lee et al., concentration is increased in both plasma and as- 2000), smooth muscle cells (Siess, 2002) and migra- cites of ovary cancer patients, reaching 80 µM (in Vol. 55 Biological activity of LPA and cPA 235 comparison to the physiological 1–5 µM concentra- cancer with a high infiltration rate. In breast cancer, tion) (Fang et al., 2000). Assignment of serum lyso- ATX expression level strongly correlates with the in- phosphatidic acids can be a potential biomarker for vasiveness of cancer cells. In addition to stimulating ovarian cancer especially in light of the fact that proliferation and motility of tumor cells, autotaxin they are not produced by normal ovarian epithelial contributes to the progression of tumors by stabiliz- cells. Ovary cancer cells also produce LPA, thereby ing preformed blood vessels in their vicinity (Tana- maintaining an LPA-rich microenvironment. Elevat- ka et al., 2006). Much of the anti-apoptosis effect and ed LPA levels have been detected in 98% of ovary intracellular signaling attributed to ATX can be ex- cancer patients, including 90% of patients with stage plained by its ability to produce bioactive phospho- I disease, suggesting that LPA promote early events lipids, such as LPA. Whether the generation of an in ovary carcinoma dissemination. In ovarian can- LPA-rich tumor microenvironment can fully account cer cell lines, LPA show various activities including for the ability of autotoxin to promote metastasis re- the enhancement of cell adhesion/attachment, pro- mains to be established, especially in view of the fact duction of angiogenetic factors such as VEGF (Hu that ATX expression is strongly upregulated by the et al., 2001), IL-6 (Fang et al., 2004) and IL-8 (So et v-Jun oncogene (Black et al., 2004) and the cancer-as- al., 2004), enhancement of urokinase plasminogen sociated α6β4 integrin, apparently via the transcrip- activator (uPA) expression (Li et al., 2005), and pre- tion factor NFAT1, which binds to the ATX promo- vention of cell apoptosis (Kang et al., 2004). Ovarian tor at two distinct sites (Chen & O’Connor, 2005). cancers (as well as colorectal cancer) show markedly Nevertheless, the available evidence suggests that increased expression of LPA2 and LPA3 receptors the ATX–LPA axis is a promising target for pharma- (Wang et al., 2007). cological intervention. Furthermore, ATX is a drug- Lysophosphatidic acids support the progres- gable target because it is a soluble exo-enzyme that sion of breast and ovarian cancer metastasis to bone, is readily amenable to high-throughput screening. acting as inducers of IL-6 and IL-8. Although breast A lead towards developing ATX inhibitors has been cancer cell lines express LPA1, LPA2 and LPA3 re- provided by the discovery that this enzyme under- ceptors, most of the LPA activities on human breast goes end product inhibition, for example by LPA cancer MDA-BO2 cell proliferation and production (van Meeteren et al., 2005). Indeed, a limited number of proosteoclastic cytokines are LPA1-dependent. of ATX inhibitors that are LPA analogs have been re- Blocking the LPA1 receptor is a promising thera- ported to date (among them fatty alcohol phosphate peutic target in cancer, especially for metastasis to analogs, phosphatidic acid derivatives, and recently bone. Indeed, the LPA1 receptor antagonist Ki16425 β-hydroxy and β-keto phosphonate derivatives of (3-(4-[4-([1-(2-chlorophenyl)ethoxy]carbonyl amino)-3- LPA) (Cui et al., 2007). Considering the importance methyl-5-isoxazolyl]benzylsulfanyl) propanoic acid) of ATX as a determinant of blood LPA level, there blocked in vivo tumor cell proliferation and inhib- are also attempts to apply ATX activity in clinical ited the production of proosteoclastic cytokines by laboratory testing (Nakamura et al., 2008). tumor cells, whereas normal platelet functions were unaffected (Boucharaba et al., 2006). The role of LPA in the immune system

The LPA1 receptor could also be a target in prostate cancers’ therapy (Hao et al., 2007). LPA1, Lysophosphatidic acids are pro-inflammatory LPA2 and LPA3 have been detected in prostate can- factors that have been implicated in the development cer cells, however, only LPA1 was found to be re- of inflammation taking place in pathological processes sponsible for prostate cancer cell migration in vitro, such as asthma or allergy. In the normal individual, and the overexpression of LPA1 resulted in increased airway injury is properly repaired due to the release tumor growth. and actions of appropriate repair mediators, includ- There is a growing body of evidence that not ing LPA. The LPA-mediated effects on airway cells only LPA but also autotaxin is involved in tumor that may contribute to airway repair in a physiologi- growth. ATX was originally identified as a tumor cal setting include fibronectin release, fibroblast - pro cell motility factor (Umezu-Goto et al., 2002; Toku- liferation and contraction, and airway smooth muscle mura et al., 2002). The enzyme stimulates both pro- cell proliferation. LPA alone can stimulate prolifera- liferation and motility of cancer cells through LPA tion of human airway smooth muscle (HASM) cells production. In addition, overexpression of ATX is or they can synergize with epidermal growth factor frequently associated with malignant tumors such as (EGF) to further increase proliferation of HASM cells small cell lung cancer, renal cell cancer, hepatocel- (Toews et al., 2002). The chronic inflammation that for lular carcinoma, breast cancer, Hodgkin lymphoma, instance characterizes asthma leads to damage of the thyroid cancer, and glioblastoma (Mills & Moole- epithelium and epithelial cell shedding, thus exposing naar, 2003). The highest ATX expression is detected the underlying tissue and leading to the release of a in glioblastoma multiforme (GBM), a very malignant variety of inflammatory and repair mediators. In the 236 E. Gendaszewska-Darmach 2008 asthmatic airway, exaggerated or prolonged respons- signaling pathways. LPA cause the collapse of neu- es to LPA contribute to long-term airway remodeling. ron growth cone and tend to inhibit or reverse the Fibronectin secretion contributes to the thickening of morphological differentiation of many neuronal cell the lamina reticularis, and fibroblast proliferation and lines. In addition to cell intrinsic responses, systemic contraction can contribute to the subepithelial fibrosis. effects on larger populations of neuronal cells have Airway smooth muscle proliferation in synergy with also been observed. They induced changes both in EGF and other growth factors are a major stimulus neuroanatomy and in animal behavior. For example, for the thickening of the smooth muscle layer. Addi- LPA exposure can produce dramatic receptor-de- tionally, following proliferation, the cells respond by pendent changes in folding of developing cerebral upregulating cytokine production (VEGF, GM-CSF, cortex, along with increase in cell number (Kings- and TNF-α) leading to increased inflammation. bury et al., 2003). The direct stimulation of peripher-

LPA participate in the motility, polarization, al nociceptor endings by LPA through LPA1 recep- and metabolic burst of human neutrophils, induce tors also suggests its role in nociceptive processes. haptotactic migration of human monocytes (Zhang It has been reported recently that the activation of et al., 2006), and also enhance monocyte–endothelial the LPA1 receptor and its downstream Rho/Rho-ki- cell adhesion and monocyte chemotaxis toward en- nase pathway is required for the development of dothelial cells through the respective upregulation of neuropathic pain (Inoue et al., 2004). The increased IL-8 and MCP-1 (monocyte chemoattractant protein- expressions of the γ isoform of protein kinase C

1) expressions in endothelial cells, thus promoting in- (PKCγ) in the spinal dorsal horn and the α2 δ-1 sub- flammation processes. Expression of LPA-induced in- unit of voltage-gated calcium channels (Caα2 δ-1) in flammatory response genes is mediated by LPA1 and the dorsal root ganglion are two important mark- LPA3, which suggests the utilization of LPA1 or LPA3 ers of neuropathic pain. The intrathecal injection of as drug targets to treat severe inflammation (Lin et LPA caused an increase of PKCγ expression in the al., 2007) spinal dorsal horn. An up-regulation of PKCγ was also observed after partial sciatic nerve injury. Simi- LPA and neuronal cells larly, LPA- and nerve injury-induced up-regulations

were observed for the Cav α2 δ-1 subunit in spinal −/− LPA receptors have multiple activities in ner- dorsal horn. The use of lpa1 mice has also demon- vous system cells achieved via activation of various strated that endogenous LPA plays a role in induc-

Y O O O O R C O X P sn-1 LPA P O O O Z

Z=CH2, CHOH, CHBr, CHF, CF2 migration Methylenephosphonates O O O O R P P O O F C O F F O O sn-2 LPA Y O P Cyclic fluoromethylenephosphonates O O O O CH2F CH3 X=OH (LPA), F, OCH3, OC2H4OH P P

Y= OH (LPA), F, CHF2, CH2CHF2, OCH3, OC2H4OH O O O O

Methylphosphonates S S O O P P O O O

Phosphorothioates and cyclic phosphonothioates

O O Figure 4. Chemically syn- S thesized LPA analogs re-

N CF3 sistant to enzymatic deg- H radation. Trifluoromethylsulfonamides Vol. 55 Biological activity of LPA and cPA 237 ing underlying mechanisms such as demyelination, specific ATX isoforms will be necessary to elucidate decrease in protein and gene expressions of myelin whether autotaxin is a valuable therapeutic target. basic protein (MBP) and peripheral myelin protein 22 kDa (PMP22 ), which are reduced in the demye- linated status. Up-regulation of PKCγ and of Cav α2 CONCLUDING REMARKS δ-1 in mice giving partial sciatic nerve ligation was also observed (Ueda, 2006). The demyelination and At the beginning of the 2000s Hla et al. (2001)

Cav α2 δ-1 up-regulation are thought to underlie the concluded that lysophospholipid receptor biology sensitization of Aδ/β nociceptive transmission pos- had generated insights into fundamental cellular sibly through ephapsis, ectopic discharge, and col- mechanisms and might provide therapeutic targets lateral sprouting, leading to allodynia and hyperal- for drug development. However, further findings gesia. PKCγ up-regulation in lamina II of the dorsal and discoveries have shown that this area of cel- spinal cord, on the other hand, may be involved in lular biology is more surprising than one could ex- the process of central sensitization, which is thought pect. Research of the last decade has demonstrated to be a common cause of neuropathic allodynia and that lysophosphatidic acids, their receptors as well hyperalgesia. Altogether, these findings suggest that as enzymes of LPA metabolism are novel targets for novel analgesics for the treatment of the initial phas- therapeutic intervention in many pathophysiological es of neuropathic pain may consist of antagonists or processes. Targeted deletion of LPA receptors, par- inhibitors of any of these identified components of ticularly in conjunction with the use of receptor ago- the LPA signaling pathway. nists and/or antagonists, has revealed roles of indi- vidual receptors in many disease models, e.g. loss of

Chemically synthesized LPA analogs LPA1 signaling has been reported to block the initia- tion of nerve injury-induced pain and alter the bal- The recent progress in elucidation of LPA- ance of adipocytes and their precursors in adipocyte dependent signaling and LPA metabolism has been tissue. The effects of a loss of 3 LPA have indicated possible due to the use of their analogs in which the importance of LPA signaling in fertility. labile groups have been chemically stabilized. For Extremely important for LPA signaling is au- example, the phosphate group can be replaced totaxin expression, activity and regulation. There- with charged or neutral phosphoromimetics includ- fore, ATX is also an attractive pharmacological target ing methylene (–CH2–) phosphonates, α-X methyl- since blockage of LPA production via this enzyme ene phosphonates (where X is –CHF–, –CHBr– or inhibition could be a useful anticancer chemotherapy. –CHOH–), phosphorothioates or phosphonothioates. While much has recently been learned about ATX ac- These modifications make such analogs resistant tivity and LPA function, many problems remain to be against lysophospholipid phosphatases (LPP). The solved. How many tissue-specific isoforms of ATX do sn-1 or sn-2 hydroxy groups can be substituted with exist? What are the differences between them? Which fluorine or methoxy groups. Such LPA analogs are pool of LPC or other substrates is preferentially de- unable to undergo acyl migration, effectively “freez- graded by autotaxin? How is the enzyme activity reg- ing” the acyl chain in the sn-1 or sn-2 position, re- ulated? Are there any membrane cPA receptors as is spectively. Additional stabilization of LPA analogs the case for LPA receptors? Which molecular factors can be achieved by replacement of the sn-1 O-acyl switch ATX activity from LPA synthesis to cPA pro- group with an O-alkyl ether (Fig. 4). Chemically duction? Hopefully, answers to these questions will synthesized LPA analogs are not only more resistant emerge in the near future. to the action of the LPP or LPAAT enzymes. Some of them have been reported to be potent and selec- Acknowledgements tive agonists of LPA receptors, especially the LPA3 receptor or PPARγ. However, taking into account This work was supported by a grant (PBZ- the multitude of LPA receptors, it seems more rea- MNiSW-07/I/2007) from the Ministry of Science and sonable to search for specific inhibitors of ATX than Higher Education. of individual LPA receptors. Unfortunately, among I thank Professor Maria Koziolkiewicz for her the LPA analogs synthesized so far, only a few com- valuable consultations and help in preparation of pounds have shown an inhibitory effect towards this this manuscript. enzyme (Cui et al., 2007). Therefore, a recent report describing phosphorothioate inhibitors of NPP1, an enzyme related to autotoxin, may be useful for fur- REFERENCES ther search of ATX inhibitors (Wójcik et al., 2007). Synthesis and optimization of small-molecule in- An S, Bleu T, Hallmark OG, Goetzl EJ (1998) Characteriza- hibitors of ATX as well as characteristics of tissue- tion of a novel subtype of human G protein-coupled 238 E. Gendaszewska-Darmach 2008

receptor for lysophosphatidic acid. J Biol Chem 273: evaluation of phosphonate derivatives as autotaxin 7906–7910. (ATX) inhibitors. Bioorg Med Chem Lett 17: 1634–1640. Aoki J (2004) Mechanisms of lysophosphatidic acid pro- Dever G, Wainwright CL, Kennedy S, Spickett CM (2006) duction. Semin Cell Dev Biol 15: 477–489. Fatty acid and phospholipid chlorohydrins cause cell Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishi- stress and endothelial adhesion. Acta Biochim Polon 53: moto T, Mizuno K, Saku K, Taguchi R, Arai H (2002) 761–768. Serum lysophosphatidic acid is produced through di- Eichholtz T, Jalink K, Fahrenfort I, Moolenaar WH (1993) verse phospholipase pathways. J Biol Chem 277: 48737– The bioactive phospholipid lysophosphatidic acid is re- 48744. leased from activated platelets. Biochem J 291: 677–680. Baker DL, Desiderio DM, Miller DD, Tolley B, Tigyi GJ English D, Cui Y, Siddiqui R, Patterson C, Natarajan V, (2001) Direct quantitative analysis of lysophosphatidic Brindley DN, Garcia JG (1999) Induction of endothe- acid molecular species by stable isotope dilution elec- lial monolayer permeability by phosphatidate. J Cell trospray ionization liquid chromatography — mass Biochem 75: 105–117. spectrometry. Anal Biochem 292: 287–295. Fang X, Gaudette D, Furui T, Mao M, Estrella V, Eder A, Baker DL, Fujiwara Y, Pigg KR, Tsukahara R, Kobayashi Pustilnik T, Sasagawa A, Lapushin R, Yu S, Jaffe RB, S, Murofushi H, Uchiyama A, Murakami-Murofushi Wiener JR, Erickson JR, Mills GB (2000) Lysophospho- K, Koh E, Bandle RW, Byun H-S, Bittman R, Fan D, lipid growth factors in the initiation, progression, me- Murph M, Mills GB, Tigyi G (2006) Carba analogs of tastases, and management of ovarian cancer. Ann N Y cyclic phosphatidic acid are selective inhibitors of au- Acad Sci 905: 188–208. totaxin and cancer cell invasion and metastasis. J Biol Fang X, Yu S, Bast RC, Liu S, Xu HJ, Hu SX, LaPushin R, Chem 281: 22786–22793. Claret FX, Aggarwal BB, Lu Y, Mills GB (2004) Mecha- Balazs L, Okolicany J, Ferrebee M, Tolley B, Tigyi G (2001) nisms for lysophosphatidic acid-induced cytokine pro- Topical application of the phospholipid growth factor duction in ovarian cancer cells. J Biol Chem 279: 9653– lysophosphatidic acid promotes wound healing in vivo. 9661. Am J Physiol 280: R466–R472. Fourcade O, Simon MF, Viode C, Rugani N, Leballe F, Bandoh K, Aoki J, Hosono H, Kobayashi S, Kobayashi T, Ragab A, Fournie B, Sarda L, Chap H (1995) Secretory Murakami-Murofushi K, Tsujimoto M, Arai H, Inoue phospholipase A2 generates the novel lipid mediator K (1999) Molecular cloning and characterization of a lysophosphatidic acid in membrane microvesicles shed novel human G-protein-coupled receptor, EDG7, for from activated cells. Cell 80: 919–927. lysophosphatidic acid. J Biol Chem 274: 27776–27785. Gardell SE, Dubin AE, Chun J (2006) Emerging medicinal Bauer M, Maschberger P, Quek L, Briddon SJ, Dash D, roles for lysophospholipid signaling. Trends Mol Med Weiss M, Watson SP, Siess W (2001) Genetic and phar- 12: 65–75. macological analyses of involvement of Src-family, Hao F, Tan M, Xu X, Han J, Miller DD, Tigyi G, Cui MZ Syk and Btk tyrosine kinases in platelet shape change: (2007) Lysophosphatidic acid induces prostate cancer

Src-kinases mediate integrin αIbβ3 inside-out signalling PC3 cell migration via activation of LPA1, p42 and during shape change. Thromb Haemost 85: 331–340. p38α. Biochim Biophys Acta 1771: 883–892. Black EJ, Clair T, Delrow J, Neiman P, Gillespie DA (2004) Hecht JH, Weiner JA, Post SR, Chun J (1996) Ventricular Microarray analysis identifies autotaxin, a tumour cell zone gene-1 (vzg-1) encodes a lysophosphatidic acid re- motility and angiogenic factor with lysophospholipase ceptor expressed in neurogenic regions of the develop- D activity, as a specific target of cell transformation by ing cerebral cortex. J Cell Biol 135: 1071–1083. v-Jun. Oncogene 23: 2357–2366. Hla T, Lee MJ, Ancellin N, Paik JH, Kluk MJ (2001) Ly- Boucharaba A, Serre CM, Guglielmi J, Bordet JC, Clézard- sophospholipids — receptor revelations. Science 294: in P, Peyruchaud O (2006) The type 1 lysophosphatidic 1875–1878. acid receptor is a target for therapy in bone metastases. Hu YL, Tee MK, Goetzl EJ, Auersperg N, Mills GB, Fer- Proc Natl Acad Sci USA 103: 9643–9648. rara N, Jaffe RB (2001) Lysophosphatidic acid - induc Clair T, Aoki J, Koh E, Bandle RW, Nam SW, Ptaszynska tion of vascular endothelial growth factor expression MM, Mills GB, Schiffmann E, Liotta LA, Stracke ML in human ovarian cancer cells. J Natl Cancer Inst 93: (2003). Autotaxin hydrolyzes sphingosylphosphoryl- 762–768. choline to produce the regulator of migration, sphingo- Inoue M, Rashid MH, Fujita R, Contos JJ, Chun J, Ueda H. sine-1–phosphate. Cancer Res 63: 5446–5453. (2004) Initiation of neuropathic pain requires lysophos- Cerutis DR, Dreyer AC, Vierra MJ, King JP, Wagner DJ, phatidic acid receptor signaling. Nat Med 10: 712–718. Fimple JL, Cordini F, McVaney TP, Parrish LC, Wilw- Ishihara R, Tatsuta M, Iishi H, Baba M, Uedo N, Higash- erding TM, Mattson JS (2007) Lysophosphatidic acid ino K, Mukai M, Ishiguro S, Kobayashi S, Murakami- modulates the healing responses of human periodontal Murofushi (2004) Attenuation by cyclic phosphatidic ligament fibroblasts and enhances the actions of plate- acid of peritoneal metastasis of azoxymethane-induced let-derived growth factor. J Periodontol 78: 1136–1145. intestinal cancers in Wistar rats. Int J Cancer 110: 188– Chen M, O’Connor KL (2005) Integrin α6β4 promotes ex- 193. pression of autotaxin/ENPP2 autocrine motility factor Jansen S, Stefan C, Creemers JWM, Waelkens E, van Eynde in breast carcinoma cells. Oncogene 24: 5125–5130. A, Stalmans W, Bollen M (2005) Proteolytic maturation Chen X, Yang XY, Wang ND, Ding C, Yang YJ, You ZJ, Su and activation of autotaxin (NPP2), a secreted metas- Q, Chen JH (2003) Serum lysophosphatidic acid con- tasis-enhancing lysophospholipase D. J Cell Sci 118: centrations measured by dot immunogold filtration as- 3081–3089. say in patients with acute myocardial infarction. Scand Jansen S, Callewaert N, Dewerte I, Andries M, Ceulemans J Clin Lab Invest 63: 497–503. H, Bollen M (2007) An essential oligomannosidic gly- Contos JJ, Ishii I, Chun J (2000) Lysophosphatidic acid re- can chain in the catalytic domain of autotaxin, a secret- ceptors. Mol Pharmacol 58: 1188–1196. ed lysophospholipase-D. J Biol Chem 282: 11084–11091. Cui P, Tomsig JL, McCalmont WF, Lee S, Becker CJ, Lynch Kang YC, Kim KM, Lee, Namkoong S, Lee SJ, Han JA, KR, Macdonald TL (2007) Synthesis and biological Jeoung D, Ha KS, Kwon YG, Kim M (2004) Serum bio- active lysophospholipids prevent TRAIL-induced apop- Vol. 55 Biological activity of LPA and cPA 239

tosis via PI3K/Akt-dependent cFLIP expression and Mukai M, Nakamura H, Tatsuta M, Iwasaki T, Togawa A, Bad phosphorylation. Cell Death Differ 11: 1287–1298. Imamura F, Akedo H (2000) Hepatoma cell migration Kingsbury MA, Rehen SK, Contos JJ, Higgins CM, Chun through a mesothelial cell monolayer is inhibited by J (2003) Non-proliferative effects of lysophosphatidic cyclic AMP-elevating agents via a Rho-dependent path- acid enhance cortical growth and folding. Nat Neurosci way. FEBS Lett 484: 69–73. 6: 1292–1299. Murakami-Murofushi K, Shioda M, Kaji K, Yoshida S, Kishi Y, Okudaira S, Tanaka M, Hama K, Shida D, Ki- Murofushi H (1992) Inhibition of eukaryotic DNA tayama J, Yamori T, Aoki J, Fujimaki T, Arai H (2006) polymerase alpha with a novel lysophosphatidic acid Autotaxin is overexpressed in glioblastoma multiforme (PHYLPA) isolated from myxoamoebae of Physarum and contributes to cell motility of glioblastoma by con- polycephalum. J Biol Chem. 267: 21512–21517. verting lysophosphatidylcholine to lysophosphatidic Murakami-Murofushi K, Kaji K, Kano K, Fukuda M, Shio- acid. J Biol Chem 281: 17492–17500. da M, Murofushi H (1993) Inhibition of cell prolifera- Kobayashi T, Tanaka-Ishii R, Taguchi R, Ikezawa H, Mu- tion by a unique lysophosphatidic acid, PHYLPA, iso- rakami-Murofushi K (1999) Existence of a bioactive li- lated from Physarum polycephalum: signaling events of pid, cyclic phosphatidic acid, bound to human serum antiproliferative action by PHYLPA. Cell Struct Funct albumin. Life Sci 65: 2185–2191. 18: 363–370. Kotarsky K, Boketoft A, Bristulf J, Nilsson NE, Norberg A, Nakamura K, Igarashi K, Ide K, Ohkawa R, Okubo S, Hansson S, Owman C, Sillard R, Leeb-Lundberg LMF Yokota H, Masuda A, Oshima N, Takeuchi T, Nan- Olde B (2006) Lysophosphatidic acid binds to and ac- gaku M, Okudaira S, Arai H, Ikeda H, Aoki J, Yatomi tivates GPR92, a G protein-coupled receptor highly Y (2008) Validation of an autotaxin enzyme immu- expressed in gastrointestinal lymphocytes. J Pharmacol noassay in human serum samples and its application Exp Ther 318: 619–628. to hypoalbuminemia differentiation. Clin Chim Acta Lee CH, Olson P, Evans RM (2003a) Minireview: lipid me- 388: 51–58. tabolism, metabolic diseases, and peroxisome prolifera- Noguchi K, Ishii S, Shimizu T (2003) Identification of p2y9/ tor-activated receptors. Endocrinology 144: 2201–2207. GPR23 as a novel G protein-coupled receptor for lyso- Lee CW, Rivera R, Gardell S, Dubin AE, Chun J (2006a) phosphatidic acid, structurally distant from the Edg

GPR92 as a new G12/13 and Gq coupled lysophosphatid- family. J Biol Chem 278: 25600–25606. ic acid receptor that increases cAMP: LPA5. J Biol Chem Pilquil C, Dewald J, Cherney A, Gorshkova I, Tigyi G, 281: 23589–23597. English D, Natarajan V, Brindley DN (2006) Lipid Lee DJ, Ho C-H, Grinnell F (2003b) LPA-stimulated fibrob- phosphate -1 regulates lysophosphatidate- last contraction of floating collagen matrices does not induced fibroblast migration by controlling phospholi-

require Rho kinase activity or retraction of fibroblast pase D2-dependent phosphatidate generation. J Biol extensions. Exp Cell Res 289: 86–94. Chem 281: 38418–38429. Lee H, Geotzl EJ, An S (2000) Lysophosphatidic acid and Pyne S, Kong K-C, Darroch PI (2004) Lysophosphatidic sphingosine 1-phosphate stimulate endothelial cell acid and sphingosine 1-phosphate biology: the role of wound healing. Am J Physiol 47: C612–C617. lipid phosphate phosphatases. Semin Cell Dev Biol 15: Lee J, Park SY, Lee EK, Park CG, Chung HC, Rha SY, 491–501. Kim YK, Bae GU, Kim BK, Han JW, Lee HY (2006b) Retzer M, Essler M (2000) Lysophosphatidic acid-induced Activation of hypoxia-inducible factor-1{alpha} is nec- platelet shape change proceeds via Rho/Rho kinase-me- essary for lysophosphatidic acid-induced vascular en- diated myosin light-chain and moesin phosphorylation. dothelial growth factor expression. Clin Cancer Res 12: Cell Signal 12: 645–648. 6351–6358. Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, Tigyi Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, G, Siess W (2003) Subtype-selective antagonists of lys- Willson TM, Kliewer SA (1995) An antidiabetic thia- ophosphatidic acid receptors inhibit platelet activation zolidinedione is a high affinity ligand for peroxisome triggered by the lipid core of atherosclerotic plaques. proliferator-activated receptor gamma (PPAR gamma). Circulation 108: 741–747. J Biol Chem 270: 12953–12956. Sano T, Baker D, Virag T, Wada A, Yatomi Y, Kobayashi Li H, Ye X, Mahanivong C, Bian D, Chun J, Huang S T, Igarashi Y, Tigyi G (2002) Multiple mechanisms (2005) Signaling mechanisms responsible for lysophos- linked to platelet activation result in lysophosphatidic phatidic acid-induced urokinase plasminogen activa- acid and sphingosine 1-phosphate generation in blood. tor expression in ovarian cancer cells. J Biol Chem 280: J Biol Chem 277: 21197–21206. 10564–10571. Siess W (2002) Athero- and thrombogenic actions of lyso- Lin CI, Chen CN, Lin PW, Chang KJ, Hsieh FJ, Lee H phosphatidic acid and sphingosine-1-phosphate. Bio- (2007) Lysophosphatidic acid regulates inflammation- chim Biophys Acta 1582: 204–215.

related genes in human endothelial cells through LPA1 Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, Cor- and LPA3. Biochem Biophys Res Commun 363: 1001–1008. rinth C, Bittman R, Tigyi G, Aepfelbacher M (1999) McIntyre TM, Pontsler AV, Silva AR, Hilaire AS, Xu Y, Lysophosphatidic acid mediates the rapid activation of Hinshaw JC, Zimmerman GA, Hama K, Aoki J, Arai platelets and endothelial cells by mildly oxidized low H, Prestwich GD (2003) Identification of an intracel- density lipoprotein and accumulates in human athero- lular receptor for lysophosphatidic acid (LPA): LPA is sclerotic lesions. Proc Natl Acad Sci USA 96: 6931–6936. a transcellular PPARγ agonist. Proc Natl Acad Sci USA Simon MF, Rey A, Castan-Laurel I, Gres S, Sibrac D, 100: 131–136. Valet P, Saulnier-Blache JS (2002) Expression of ectoli- Meyer zu Heringdorf D, Jacobs KH (2007) Lysophospholi- pid phosphate phosphohydrolases in 3T3F442A preadi- pid receptors: signalling, pharmacology and regulation pocytes and adipocytes. Involvement in the control by lysophospholipid metabolism. Biochim Biophys Acta of lysophosphatidic acid production. J Biol Chem 277: 1768: 923–940. 23131–23136. Mills GB, Moolenaar WH (2003) The emerging role of lyso- Simon MF, Daviaud D, Pradere JP, Gres S, Guigne C, Wa- phosphatidic acid in cancer. Nat Rev Cancer 3: 582–591. bitsch M, Chun J, Valet P, Saulnier-Blache JS (2005) Lysophosphatidic acid inhibits adipocyte differen- 240 E. Gendaszewska-Darmach 2008

tiation via lysophosphatidic acid 1 receptor-dependent Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, down-regulation of peroxisome proliferator-activated Takio K, Yamori T, Mills GB, Inoue K, Aoki J, Arai receptor gamma2. J Biol Chem 280: 14656–14662. H (2002) Autotaxin has lysophospholipase D activity So J, Navari J, Wang FQ, Fishman DA (2004) Lysophos- leading to tumor cell growth and motility by lysophos- phatidic acid enhances epithelial ovarian carcinoma in- phatidic acid production. J Cell Biol 158: 227–233. vasion through the increased expression of interleukin- van Corven EJ, Groenink A, Jalink K, Eichholtz T, Moo- 8. Gynecol Oncol 95: 314–322. lenaar WH (1989) Lysophosphatidate-induced cell Stefan C, Jansen S, Bollen M (2005) NPP-type ectophos- proliferation: identification and dissection of signaling phodiesterases: unity in diversity. Trends Biochem Sci pathways mediated by G proteins. Cell 59: 45–54. 30: 542–550. van Meeteren LA, Ruurs P, Christodoulou E, Goding JW, Sturm A, Dignass AU (2002) Modulation of gastrointesti- Takakusa H, Kikuchi K, Perrakis A, Nagano T, Moo- nal wound repair and inflammation by phospholipids. lenaar W (2005) Inhibition of autotaxin by lysophos- Biochim Biophys Acta 1582: 282–288. phatidic acid and sphingosine 1-phosphate. J Biol Chem Sugiura T, Nakane S, Kishimoto S, Waku K, Yoshioka Y, 280: 21155–21161. Tokumura A (2002) Lysophosphatidic acid, a growth van Meeteren LA, Moolenaar WH (2007) Regulation and factor-like lipid, in the saliva. J Lipid Res 43: 2049–2055. biological activities of the autotaxin-LPA axis. Prog Li- Tigyi G, Parrill AL (2003) Molecular mechanisms of lyso- pid Res 46: 145–160. phosphatidic acid action. Prog Lipid Res 42: 498–526. Wang P, Wu X, Chen W, Liu J, Wang X (2007) The lyso- Tanaka M, Okudaira S, Kishi Y, Ohkawa R, Iseki S, Ota phosphatidic acid (LPA) receptors their expression and M, Noji S, Yatomi Y, Aoki J, Arai H (2006) Autotaxin significance in epithelial ovarian neoplasms. Gynecol stabilizes blood vessels and is required for embryonic Oncol 104: 714–720. vasculature by producing lysophosphatidic acid. J Biol Watterson KR, Lanning DA, Diegelmann RF, Spiegel S Chem 281: 25822–25830. (2007) Regulation of fibroblast functions by lysophos- Toews ML, Ediger TL, Romberger DJ, Rennard SI (2002) pholipid mediators: potential roles in wound healing. Lysophosphatidic acid in airway function and disease. Wound Repair Regen 15: 607–616. Biochim Biophys Acta 1582: 240–250. Wójcik M, Cieślak M, Stec WJ, Goding JW, Koziolkiewicz Tokumura A, Fukuzawa K, Tsukatani H (1978) Effects of M (2007) Nucleotide pyrophosphatase/phosphodi- synthetic and natural lysophosphatidic acids on the ar- 1 is responsible for degradation of antisense terial blood pressure of different animal species. Lipids phosphorothioate oligonucleotides. Oligonucleotides 17: 13: 572–574. 134–145. Tokumura A, Harada K, Fukuzawa K, Tsukanani H (1986) Wu WT, Chen C-N, Lin CI, Chen JH, Lee H (2005) Lyso- Involvement of lysophospholipase D in the production phospholipids enhance matrix metalloproteinase-2 ex- of lysophosphatidic acid in rat plasma. Biochim Biophys pression in human endothelial cells. Endocrinology 146: Acta 875: 31–38. 3387–3400. Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure Yoshida K, Nishida W, Hayashi K, Ohkawa Y, Ogawa A, K, Yasuda K, Fukuzawa K (2002) Identification of - hu Aoki J, Arai H, Sobue K (2003) Vascular remodeling man plasma lysophospholipase D, a lysophosphatidic induced by naturally occurring unsaturated lysophos- acid-producing enzyme, as autotaxin, a multifunctional phatidic acid in vivo. Circulation 108: 1746–1752. phosphodiesterase. J Biol Chem 277: 39436–39442. Zhang C, Baker DL, Yasuda S, Makarova N, Balans L, Tsuda S, Okudaira S, Moriya-Ito K, Shimamoto C, Tanaka Johnson LR, Marathe GK, McIntyre TM, Xu Y, Prest- M, Aoki J, Arai H, Murakami-Murofushi K, Kobayashi wich GD, Byun H-S, Bittman R, Tigyi G (2004)- Lyso T (2006) Cyclic phosphatidic acid is produced by auto- phosphatidic acid induces neointima formation through taxin in blood. J Biol Chem 281: 26081–26088. PPARγ activation. J Exp Med 199: 763–774. Uchiyama A, Mukai M, Fujiwara Y, Kobayashi S, Kawai Zhang Z, Liu Z, Meier KE (2006) Lysophosphatidic acid as N, Murofushi H, Inoue M, Enoki S, Tanaka Y, Niki T, a mediator for proinflammatory agonists in a human Kobayashi T, Tigyi G, Murakami-Murofushi K (2007) corneal epithelial cell line. Am J Physiol Cell Physiol 291: Inhibition of transcellular tumor cell migration and C1089–C1098. metastasis by novel carba-derivatives of cyclic phos- phatidic acid. Biochim Biophys Acta 1771: 103–112. Ueda H (2006) Molecular mechanisms of neuropathic pain- phenotypic switch and initiation mechanisms. Pharma- col Ther 109: 57–77.