+ All Categories
Home > Documents > Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

Date post: 07-Dec-2016
Category:
Upload: wouter
View: 217 times
Download: 4 times
Share this document with a friend
19
661 CHAPTER 31 Lysophosphatidic Acid Receptors in Cancer ABIR MUKHERJEE, JINHUA WU, YONGLING GONG, and XIANJUN FANG 31.1. INTRODUCTION In the past decade, there have been considerable advances in our understand- ing of the sources and biological functions of lysophospholipids. The best characterized of these are lysophosphatidic acid (LPA) with a glycerol back- bone and the related sphingosine 1-phosphate (S1P) containing sphingosine (1, 2). These lysophospholipids are not only metabolites in biosynthesis of more complex lipids in eukaryotic and prokaryotic cells but also have emerged as pluripotent intercellular mediators to induce hormone- and growth factor- like responses in their target cells. LPA is a key intermediate in de novo lipid synthesis in all cells formed by acylation of glycerol 3-phosphate catalyzed by glycerophosphate acyltransfer- ase (3, 4). LPA is subsequently converted to phosphatidic acid (PA) by LPA acyltransferase and further to other phospholipids and triacylglycerols. These conversions occur intracellularly in the endoplasmic reticulum or mitochon- dria. The pathway is conserved in lower organisms in which extracellular LPA actions are not observed. There is no evidence for LPA release into the extra- cellular fluid or leaflets of plasma membranes. Thus, LPA produced through this route is not considered to be involved in extracellular LPA signaling. Another potential intracellular pathway for the synthesis of LPA is the phos- phorylation of monoacylglycerol by acylglycerol kinase (AGK) located at the mitochondria (5). Similarly, the contribution of AGK to the physiological levels of LPA is unknown due to the lack of information on potential LPA transporters responsible for secreting LPA from intracellular locations. Lysophospholipid Receptors: Signaling and Biochemistry, First Edition. Edited by Jerold Chun, Timothy Hla, Sara Spiegel, and Wouter Moolenaar. © 2013 John Wiley & Sons, Inc. Published 2013 by John Wiley & Sons, Inc.
Transcript
Page 1: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

661

CHAPTER 31

Lysophosphatidic Acid Receptors in CancerABIR MUKHERJEE, JINHUA WU, YONGLING GONG, and XIANJUN FANG

31.1. INTRODUCTION

In the past decade, there have been considerable advances in our understand-ing of the sources and biological functions of lysophospholipids. The best characterized of these are lysophosphatidic acid (LPA) with a glycerol back-bone and the related sphingosine 1-phosphate (S1P) containing sphingosine (1, 2). These lysophospholipids are not only metabolites in biosynthesis of more complex lipids in eukaryotic and prokaryotic cells but also have emerged as pluripotent intercellular mediators to induce hormone- and growth factor-like responses in their target cells.

LPA is a key intermediate in de novo lipid synthesis in all cells formed by acylation of glycerol 3-phosphate catalyzed by glycerophosphate acyltransfer-ase (3, 4). LPA is subsequently converted to phosphatidic acid (PA) by LPA acyltransferase and further to other phospholipids and triacylglycerols. These conversions occur intracellularly in the endoplasmic reticulum or mitochon-dria. The pathway is conserved in lower organisms in which extracellular LPA actions are not observed. There is no evidence for LPA release into the extra-cellular fluid or leaflets of plasma membranes. Thus, LPA produced through this route is not considered to be involved in extracellular LPA signaling. Another potential intracellular pathway for the synthesis of LPA is the phos-phorylation of monoacylglycerol by acylglycerol kinase (AGK) located at the mitochondria (5). Similarly, the contribution of AGK to the physiological levels of LPA is unknown due to the lack of information on potential LPA transporters responsible for secreting LPA from intracellular locations.

Lysophospholipid Receptors: Signaling and Biochemistry, First Edition. Edited by Jerold Chun, Timothy Hla, Sara Spiegel, and Wouter Moolenaar.© 2013 John Wiley & Sons, Inc. Published 2013 by John Wiley & Sons, Inc.

Page 2: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

662 LPA RECEPTORS IN CANCER

The molecular pathways for extracellular LPA production are better under-stood. The mechanism has been best characterized in activated platelets that are responsible for increased LPA levels in serum compared to the whole blood or plasma (6–8). LPA can be produced by activated platelets from newly generated, membrane-associated PA by the action of phospholipase D (PLD) followed by phospholipase A1 (PLA1) or phospholipase A2 (PLA2)-mediated deacylation (6, 7). However, the bulk of LPA arising from platelet activation results from the sequential cleavage of serum and membrane phospholipids to lysophospholipids by PLA1 and PLA2 secreted from platelets, followed by conversion to LPA by lysophospholipase D (lysoPLD) present in the plasma (6, 9, 10). The plasma lysoPLD is autotaxin (ATX) (11, 12), a member of the nucleotide pyrophosphatase and phosphodiesterase family of exo- and ecto-enzymes (13). Homozygous deletion of ATX leads to embryonic lethality at E9.5 due to impaired vessel formation in the yolk sac and embryo proper (14, 15), indicating that ATX is essential for embryonic vasculature. Blood LPA levels in ATX heterozygous mice are about half of those present in wild-type littermates, indicating that ATX is a major LPA-generating enzyme in the circulation (14, 15).

The LPA homeostasis is regulated mainly by the LPA synthesizing enzyme ATX and lipid phosphate phosphatases (LPPs) including LPP1/PAP2A, LPP3/PAP2B, and LPP2/PAP2C (16). LPPs are located at the cell membrane with the catalytic domain facing the extracellular media. The LPP enzymes dephos-phorylate LPA and other phospholipids at the external leaflet of the mem-brane (17, 18). Overexpression of LPPs has been shown to antagonize overall cellular responses to LPA and to reduce LPA concentrations in the medium (19, 20). In contrast, inhibition of LPP enzyme activity sensitizes platelets to LPA-induced responses as well as increases thrombin-induced LPA produc-tion (20). Together, these observations suggest that LPPs play a critical role in controlling levels of extracellular LPA or membrane accessible LPA. Tomsig et al. recently reported that LPP1 functions as a LPP that specifically catalyzes dephosphorylation of LPA (21).

31.2. G PROTEIN-COUPLED RECEPTORS (GPCRS) FOR LPA

Most biological actions of LPA are mediated through LPA activation of spe-cific GPCRs (22). At least six GPCRs have been identified to mediate a wide range of biological actions of LPA (23–29). The LPA1/Edg2, LPA2/Edg4, and LPA3/Edg7 receptors are members of the endothelial differentiation gene (Edg) family, sharing 50–57% homology in their amino acid sequences (23–26). These LPA receptors couple to Gi, Gq, and G12/13 that, in turn, activate diverse pathways including stimulation of phospholipase C (PLC) (30), inhibi-tion of adenylyl cyclase (30), and activation of mitogen-activated protein kinases (MAPKs) (31) and PI3K (32). Activation of these signaling events

Page 3: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

LPA1 IN CANCER 663

downstream of LPA receptors culminates in changes in cell morphology, growth, survival, migration, and tumor cell invasion (33).

In addition to the Edg LPA1–3 receptors, GPR23/P2Y9/LPA4 of the puriner-gic receptor family and the related GPR92/LPA5, and P2Y5/LPA6 have been identified as new LPA receptors (27–29). GPR87 was proposed to be the seventh LPA receptor (34), but further evidence is needed to validate its role as an additional LPA receptor. This new subgroup of non-Edg LPA receptors is structurally distant from the Edg LPA1–3 receptors (27–29). These novel LPA receptors couple to Gq, G12/13, and Gs, leading to activation of PLC, the small GTPase Rho, and adenylyl cyclase, respectively.

Only minor abnormalities such as craniofacial dysmorphism and defective sucking behavior were found in LPA1-deficient mice (35). However, further analysis of these Lpar1 knockout mice subjected to pathophysiological condi-tions has revealed that LPA1 is required for the initiation of neuropathic pain (36) and promotion of pulmonary and renal fibrosis (37, 38). Disruption of the Lpar2 gene results in no obvious abnormalities in mice (39). Targeted deletion of Lpar3 leads to identification of a specific function of this LPA receptor in female reproduction. LPA3-deficient female mice show a delayed implantation and defective embryo spacing, associated with reduced uterine expression of Cox-2 mRNA (40). Among the non-Edg LPA receptors, two independent groups have reported phenotypes of Lpar4 knockout mice (41, 42). In mice of C57BL/6 background, loss of LPA4 seems to cause partial embryonic lethality (41). However, in the C57BL/6 and 129/Sv mixed background, Lee et al. did not observe any apparent abnormalities in embryonic development (42). Instead, LPA4 null embryonic fibroblasts are more responsive to LPA-induced migration, pointing to the possibility that LPA4 plays a negative role in the regulation of cell motility (42). The observations in genetic models of LPA receptors suggest the diverse LPA receptors may act redundantly or antago-nistically to regulate cellular responses to LPA.

31.3. LPA1 IN CANCER

The potential role of LPA in the pathogenesis of cancers has received more and more attention in recent years. In addition to growth and survival-promoting effects, LPA is a potent motogen, driving random migration, che-motaxis, and tumor cell invasion (12, 33, 42, 43). Interestingly, the major LPA synthesizing enzyme ATX was originally identified from culture supernatants as a tumor cell motility-stimulating factor (11, 12). Stimulation of cell motility by LPA and ATX could contribute to the metastatic phenotype of cancer cells.

Although each of the Edg LPA receptors has been linked to promoting cell motility in certain cellular contexts, the LPA1 receptor is apparently the primary receptor subtype that plays a fundamental role in mediation of cell motility. Knockdown of the LPA1 receptor expression or pharmacological

Page 4: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

664 LPA RECEPTORS IN CANCER

inhibition of LPA1 with Ki16425 blocks the chemotactic response to LPA consistently in different lineages of mammalian cells (44). In nude mice, down-regulation of LPA1 expression or activity in breast cancer cell lines inhibited their metastasis to the bone (45). Conversely, topical expression of LPA1 alone in the nonresponsive cell lines such as Rh7777 (12), B103 (46), and SKBr-3 (45, 47) is sufficient to restore migratory response to LPA or to induce bone metastasis in vivo. The most convincing evidence to support a critical role for LPA1 in cell motility stemmed from the study by Hama et al., who showed mouse skin fibroblasts devoid of LPA1 failed to migrate toward LPA, while the cells lacking LPA2 showed a normal response similar to wild-type cells (44). Loss of LPA1 was associated with the deficiency in activation of Rac by LPA in these skin fibroblasts. The observation is consistent with the fact that LPA1 couples to Gi that links to activation of the downstream target Rac. Rac, along with Rho downstream of G12/13, mediate cell migration in a coordinate manner. Rac promotes lamellipodia protrusion and forward movement, whereas RhoA regulates actomyosin-driven cytoskeleton contraction and detachment of the rear of migrating cells (48).

Compared to other Edg LPA receptors, LPA1 is most widely expressed in tissues (49). Gene expression studies failed to show any consensus increase in LPA1 expression between normal and malignant cells (50–54). Instead, some expression or array analyses indicate modest decreases in LPA1 mRNA expres-sion in primary tumors (50, 52–55). However, most of these results are statisti-cally insignificant or lack appropriate normal controls for comparison.

One of the most exciting findings about regulation of LPA1 in cancer is that the metastatic tumor suppressor Nm23 inhibits LPA1 expression (56). Nm23 is the first identified metastasis suppressor gene (MSG) that, by definition, inhibits the process of metastasis but not growth of primary tumors (56). At least three biological functions of Nm23 may contribute to its metastasis-suppressive activity. These include the histidine kinase activity that phosphory-lates ATP-citrate lyase, aldolase C, and the kinase suppressor of ras, Nm23 interaction/inactivation of cellular and viral proteins that stimulate metastasis, and regulation of expression of metastatic proteins (56, 57). Manipulation of Nm23 expression in lung and breast cancer cells has revealed a number of genes that repressed by Nm23 including Wnt5B, uPA, MMPs, CTGF, c-Met, and recently, LPA1 (58–60). Among these targets repressed by Nm23, only LPA1 is capable of functionally overcoming the antimetastatic effects of Nm23 (57, 61). Topical expression of LPA2 only shows a minor effect (56), suggesting that suppression of LPA1 expression is an important and major downstream effector of the Nm23 metastasis suppressor.

LPA1 mRNA expression levels were analyzed in two published microarray cohorts of human breast carcinomas (62, 63). When the cohorts were separated into high and low Nm23-H1 expressing sets, LPA1 expression inversely cor-related with Nm23-H1 (61). In addition, an inverse correlation was also observed by immunohistochemical staining of LPA1 and Nm23-M1 in hepato-cellular carcinomas from wild-type and Nm23-M1 null mice (61). The next

Page 5: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

LPA2 IN CANCER 665

question is whether LPA1 expression is indeed upregulated in metastatic cancer compared to primary tumors. Such analysis is only available in ovarian cancer where LPA1 expression seems to be higher in metastatic tissues than in primary ovarian tumors (64).

The importance of LPA1 in the regulation of metastatic potential of cancer cells is further highlighted by the observation that LPA1 may lie downstream of fibroblast growth factor receptor 4 (FGFR4) (65). The germline polymor-phism of FGFR4 results in substitution of glycine by arginine at codon 388 (G388 to R388). The R388 FGFR4 does not increase tumor incidence. However, clinical studies demonstrate that cancer patients with the R388 polymorphism are more likely to develop metastatic diseases (66). This phenotype seems to correlate with higher levels of LPA1 receptor expression and migratory and invasive responses to LPA in tumors carrying R388 FGFR (65). In contrast, the G388 FGFR4 variant serves as a repressor of LPA1 receptor expression and LPA-dependent cellular effects (65).

Another interesting study suggests that LPA1 also downregulates the func-tion of the p53 tumor suppressor (67). Stimulation of lung carcinoma cell lines with LPA induces proteasomal degradation of p53 associated with inhibition of p53 nuclear accumulation and p53-dependent transcription. The effect of LPA on p53 could be also reproduced in hepatocellular carcinoma cells by overexpression of LPA receptors including LPA1 but not its inactive mutant form LPA1-R124A (67).

31.4. LPA2 IN CANCER

Different from LPA1, the LPA2 receptor does not seem to be a major mediator of cellular migratory response to LPA (68, 69). However, strong evidence indicates that expression of this receptor is most commonly increased in human malignancies. We previously examined mRNA expression of LPA1, LPA2, and LPA3 in normal ovarian epithelial cells, SV40 T antigen-immortalized ovarian epithelial cells, and ovarian cancer cell lines (70, 71). LPA2 and/or LPA3 mRNAs were highly expressed in ovarian cancer cell lines but were not present in normal ovarian epithelial cells (70, 71). The observation was later extended to the primary ovarian cancer samples directly obtained from patients. LPA2 is overexpressed in about 30% of ovarian cancers, whereas LPA3 is overexpressed in about 45% of ovarian cancers (70). Thus, a majority of ovarian cancers overexpress LPA2, LPA3, or both of them. These include early and late stages of ovarian cancer.

Many other groups have reported abnormal expression of the LPA2 recep-tor in various malignancies. For instance, overexpression of LPA2 mRNA was observed in differentiated thyroid cancer (72). In breast cancer, LPA2 overex-pression was observed in more than half (57%) of the most common invasive ductal carcinoma (73). Interestingly, LPA2 overexpression is more common in postmenopausal breast cancer patients (67%) than in premenopausal patients

Page 6: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

666 LPA RECEPTORS IN CANCER

(45%) (73). LPA2 was also commonly overexpressed in gastric cancer (74). The increase was more prevalent in the intestinal type (67%) than in the diffuse type of gastric cancer (32%). Of particular interest, LPA2 overexpres-sion in the diffuse-type gastric cancer correlated with a higher rate of lym-phatic and venous invasion, lymphatic metastasis, and advanced tumor stages (74). In colorectal cancer, LPA2 mRNA was found to be consistently increased, while that of LPA1 was lower compared to the matched normal intestinal mucosa (55). So, the LPA2/LPA1 ratio was dramatically augmented in all 26 cancer specimens examined. These studies of breast, gastric, and colorectal cancers also showed increases in immunohistochemical staining of LPA2 protein in carcinoma cells compared to the surrounding normal epithelial tissues (55, 73, 74).

These observations on increased LPA2 expression in malignancies did not establish a cause–effect relationship for this receptor in oncogenesis. Further, these observations did not answer how LPA2 is upregulated mechanistically in cancer cells. A number of recent studies, however, have provided compelling evidence that LPA2 is causally linked to tumorigenesis in animals. Ectopic expression of LPA2 in ovarian cancer cell lines enhances their tumorigenicity and aggressiveness in subcutaneous and intraperitoneal xenograft models in nude mice (75). Although LPA1 and LPA3 also showed partial effects, LPA2 was consistently the most effective in these assays. More recently, Liu et al reported that transgenic expression of LPA receptors driven by the MMTV promoter leads to development of mammary tumors including estrogen receptor-positive and -negative breast carcinomas (76). After repeated breed-ing cycles, the female mice developed late-onset invasive mammary carcino-mas, which was preceded by chronic mastitis, hyperplasia, and intraepithelial neoplasia, suggesting that LPA signaling underlies these preneoplastic pro-cesses culminating in mammary tumorigenesis (76). Once again, LPA2 trans-genic mice showed a more aggressive tumorigenic phenotype compared to the LPA1 or LPA3 transgenic animals. The LPA2 transgenic model in the ovary has been also reported (77). The directed expression of LPA2 did not seem to be sufficient to induce ovarian tumorigenesis but resulted in overexpression of the oncogenic factors vascular endothelial growth factor (VEGF) and urokinase plasminogen activator (uPA) in the transgenic mice (77).

The most direct evidence to implicate LPA2 in cancer stems from the recent studies by Yun’s group using the Lpar2 null mice (78, 79). Mice lacking LPA2 do not show any physiological abnormalities (39). However, compared to wild-type mice, the LPA2-deficient mice were more resistant to intestinal tumor formation induced by colitis or by ApcMin mutation (78, 79). The mechanism for the oncogenic action of LPA2 remains elusive. Most studies have been focused on the ability of LPA to stimulate expression of oncogenic protein factors including IL-6, VEGF, HIF1a, c-Myc, cyclin D1, Krüppel-like factor 5,and Cox-2 (80–84). LPA2 seems to be more effective than other LPA recep-tors in driving the transcriptional effects of LPA on these LPA target genes. Furthermore, LPA2 contains a postsynaptic density 95, disks large, and zonula

Page 7: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

DEREGULATION OF OTHER LPA RECEPTORS IN CANCER 667

occludens-1 (PDZ) binding motif at the carboxyl terminal end that enables interaction with multiple PDZ scaffold proteins (85, 86). Of particular interest is the Na+/H+ exchanger regulatory factor 2 (NHERF-2), which likely potenti-ates LPA2-dependent gene expression and other LPA-driven cellular pro-cesses (85–87). This is in sharp contrast to the LPA1 receptor where deletion of the PDZ domains enhances the receptor activity or makes it constitutively active (88). Several recent studies provide further evidence to support interac-tions of diverse PDZ scaffold proteins with the LPA2 PDZ binding domain, which regulate many aspects of LPA2 biological functions (87, 89, 90).

31.5. DEREGULATION OF OTHER LPA RECEPTORS IN CANCER

Different from LPA1 and LPA2, LPA3 is quite unique in that its expression is limited to a few normal tissues such as the uterus (40, 70). There is inconsistent information on its expression in cancer. For instance, it is overexpressed in ovarian cancer (70), prostate cancer (91), and superficial bladder cancer (53). However, the overexpression of LPA3 was not seen in many other types of cancer (Fig. 31.1). Recent studies in murine and human cancer cell lines suggest that LPA3 could be epigenetically silenced in tumor cells via hypermethylation of its promoter (92). There is also evidence that LPA3 inhibits LPA1-mediated tumor cell migration (93). Thus, a generalized role for LPA3 in cancer remains to be fully determined, although this receptor is capable of eliciting many cel-lular responses to LPA when exogenously overexpressed in mammalian cells.

Compared to the Edg LPA receptors, little is known about the biological roles in cancer of the novel subtype of LPA receptors LPA4–6. LPA4 mRNA is expressed in cells of both mesenchymal and epithelial origins (42). Expression profiling analyses have not led to any statistically meaningful conclusions about its expression in cancer (Fig. 31.1). Studies in culture, however, indicate that a majority of cancer cell lines lack significant expression of LPA4 mRNA when compared to the Edg LPA receptors (44, 94). This highlights the possibil-ity that LPA4 expression is generally repressed in malignant cells in contrast to the Edg LPA receptors. To determine whether LPA4 expression is sup-pressed in association with cell transformation, we transformed NIH 3T3 cells with activated Ras, Raf, or Mos oncogene. The oncogene-expressing clones showed transformed morphology and grew colonies in soft agar (data not shown). These Ras-, Raf-, or Mos-transformed clones expressed LPA4 mRNA at much reduced levels as revealed by Northern blotting (Fig. 31.2, left panel). The dramatic decrease in LPA4 in oncogene-transformed cells was readily reproduced by regular RT-PCR (Fig. 31.2, right panel). The transformation-linked downregulation of LPA4 was specific as expression of LPA1 or LPA2 was not compromised in Ras-, Raf-, or Mos-transformed cells.

GPR87, also known as GPR95 and recently proposed to be an additional LPA receptor (LPA7) (22, 34), is expressed at low levels in most tissues with the exception of the prostate, placenta, head, and neck (95). An interesting

Page 8: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

668 LPA RECEPTORS IN CANCER

Figure 31.1. Expression of LPA1–6 receptors in human malignancies and their corre-sponding normal tissues. Oncomine™ (Compendia Bioscience, Ann Arbor, MI) was used for analysis and visualization of microarray data from various published research articles. Expression levels of the LPA1–6 receptors in the indicated normal and cancer tissues were depicted as heat map of their log2 median-centered intensities. The numbers of normal and tumor samples examined in each category were indicated in brackets. (A) breast versus invasive ductal breast carcinoma (51), (B) ovary versus ovarian serous adenocarcinoma (52), (C) bladder versus superficial bladder cancer (53), and (D) colon versus colon carcinoma (54). (See color insert.)

A

B

C

D

Ovary (4) Ovarian Serous Adenocarcinoma (41)

Bladder (48) Superficial Bladder Cancer (28)

Colon (10) Colon Carcinoma (5)

Invasive Ductal Breast Carcinoma (33)Breast (9)

observation with GPR87 is its overexpression specifically in squamous cell carcinomas of the lung, cervix, skin, urinary bladder, testis, head, and neck (95). Earlier studies demonstrated that GPR87 possesses a prosurvival activity in cancer cells (96). It was later found that DNA damage and genotoxic stress induce GPR87 expression in a p53-dependent fashion (95, 97). This induction is likely mediated through a p53-responsive element present in the GPR87 promoter. Other p53 family members, p63 and p73, also promote GPR87 expression (95). Strikingly, knockdown of GPR87 expression sensitizes cancer cells to cell killing by genotoxic drugs, indicating that GPR87 mediates cell viability effect downstream of p53 (95, 97). Thus, targeting of GPR87 offers an opportunity for enhancing drug efficacy in cancer, especially in squamous cell carcinoma. In spite of all these interesting observations, no information is available on whether these biological functions of GPR87 rely on its putative ligand LPA.

Page 9: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

ATX IN CANCER 669

31.6. POTENTIAL MUTATIONS OF LPA RECEPTORS IN CANCER

Although not common, mutations of GPCRs are associated with cancer development. For example, activating mutations in the thyroid-stimulating hormone receptor are found in 30% of human thyroid adenomas and some carcinomas (98). It has been long speculated that certain LPA receptors may be activated or inactivated through genetic mutation in cancer. Recent finding that P2Y5/LPAR6 is mutated and responsible for hypotrichosis simplex (29) has further stimulated the interest in the possible mutation and dysfunction of certain LPA receptors in neoplastic tissues. Recently, Obo et al. reported missense mutations of Lpar1 in more than 40% rat hepatocellular carcinomas induced by N-nitrosodiethylamine (DEN) or by a choline-deficient l-amino acid-defined (CDAA) diet (99). A similar frequency of Lpar1 missense muta-tions was detected in lung adenocarcinoma in rats induced by N-nitrosobis(2-hydroxypropyl)amine (BHP) (100). The biological consequences of these mutations are not understood. Interestingly, various human cancer cell lines also harbor mutant forms of LPAR1 or LPAR2 (92, 101). It will be of interest to determine whether such mutations are indeed present in primary human cancers.

31.7. ATX IN CANCER

In addition to the altered expression of LPA receptors, there is also evidence for amplifying LPA signaling in cancer through overexpression of the major

Figure 31.2. Downregulation of LPA4 expression in NIH 3T3 cells transformed by oncogenes. NIH 3T3 cells were transformed with activated Ras, Raf, or Mos oncogene. LPA4 mRNA expression in oncogene-transformed cell lines (Raf transformed clone Raf-1; Ras transformed clones Ras-1 and Ras-2; and Mos transformed clone Mos-1), vector control cells (vector), and parental NIH 3T3 cells (parental) was analyzed by Northern blotting (left panel) and confirmed by RT-PCR (right panel).

NIH 3T3 NIH 3T3

Par

enta

l

Vec

tor

Raf

-1

Ras

-1

Mos

-1

LPA4

18S

LPA4

LPA1

LPA2

Northern blotting RT-PCRR

as-2

Par

enta

l

Vec

tor

Raf

-1

Ras

-1

Mos

-1

Page 10: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

670 LPA RECEPTORS IN CANCER

LPA-producing enzyme ATX. Serum ATX protein levels were elevated in patients with hematological malignancies including B-cell-derived follicle lym-phoma and Hodgkin’s lymphoma (102). The increases in ATX protein in these patients correlated with higher LPA levels in the plasma. It was not clear whether the increased ATX in these patients was made and secreted by tumor cells. Prostate cancer is a prototype model of stepwise progression of tumori-genesis. ATX was found to be absent in non-neoplastic prostate cells but elevated in a majority of high-grade intraepithelial neoplasia (103). There are also observations to support elevated expression of ATX in renal carcinoma (104), glioblastoma multiforme (94), poorly differentiated thyroid cancer (105), and non-small-cell lung carcinomas (106). Thus, ATX overexpression appears to be a common route to amplify LPA production and action in tumor microenvironments.

31.8. FUTURE PROSPECTS

Significant progresses have been made toward better understanding of the oncogenic actions of LPA. The field has been benefitted from knockout and transgenic animal models targeting LPA receptors or LPA production. Earlier studies of these genetic models lacking Lpar1, Lpar2, or even both of them did not provide direct evidence for LPA signaling in cancer (35, 39). More substantial studies of the susceptibility of these mice to carcinogenesis have clearly demonstrated that LPA signaling through its receptors, particularly the LPA2 receptor, contributes to oncogenic processes (78, 79). Overexpression of LPA receptors or ATX in transgenic models also supports the tumorigenic role of LPA (76). Therefore, LPA receptors and LPA metabolism are attractive novel anticancer targets. Several groups have developed a receptor subtype-specific antagonist of known LPA receptors with the most successful one, Ki16425, as a selective inhibitor for LPA1 and LPA3 (107). Due to the promi-nent role of LPA1 in the regulation of cell motility and tumor cell invasion, Ki16425 and other LPA1-specific blockers could be of potential application in the prevention of metastasis. Relatively speaking, limited progresses have been made in the identification and validation of LPA2-specific blockers. Among all LPA receptors, LPA2 has been found to be most consistently ele-vated in various human malignancies (55, 70–74). Therefore, effective and specific antagonists of LPA2 are highly desired. A number of potential inhibi-tors have been identified through high-throughput screenings (108). However, these compounds have not been thoroughly characterized and validated by independent groups as specific and potent antagonists for LPA2.

Further, alteration of LPA production or catabolism in the tumor microen-vironment may provide additional approaches to control LPA levels and LPA-associated tumor-promoting activities. A number of natural and synthetic compounds have been described to inhibit ATX activity and invasive pheno-

Page 11: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

REFERENCES 671

types of tumor cells (109, 110). More recently, calcium-independent phospho-lipase A2 (iPLA2), an enzyme that could be potentially involved in LPA production from membrane phospholipids (9), is required for tumorigenicity of ovarian and prostate cancer cell lines in xenograft mouse models (111–113). Intriguingly, iPLA2 activity in both host animals and injected tumor cells is needed for tumorigenicity of ovarian cancer cell lines (112). If this is mediated through regulation of LPA production, iPLA2 could offer an alternative way for the intervention of LPA action and tumor growth.

ACKNOWLEDGMENTS

The work was supported by the NIH/NCI grant 2R01CA102196 (X.F.), the Massey Cancer Center pilot project grant (X.F.), and the NIH grant P30 CA16059 to the Massey Cancer Center of Virginia Commonwealth University.

REFERENCES

1. Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, Chun J. 2010. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol 50:157–186.

2. Tani M, Ito M, Igarashi Y. 2007. Ceramide/sphingosine/sphingosine 1-phosphate metabolism on the cell surface and in the extracellular space. Cell Signal 19:229–237.

3. Pages C, Simon MF, Valet P, Saulnier-Blache JS. 2001. Lysophosphatidic acid syn-thesis and release. Prostaglandins Other Lipid Mediat 64:1–10.

4. Dircks L, Sul HS. 1999. Acyltransferases of de novo glycerophospholipid biosyn-thesis. Prog Lipid Res 38:461–479.

5. 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. J Cell Biol 169:801–811.

6. Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishimoto T, Mizuno K, Saku K, Taguchi R, Arai H. 2002. Serum lysophosphatidic acid is produced through diverse phospholipase pathways. J Biol Chem 277:48737–48744.

7. Aoki J. 2004. Mechanisms of lysophosphatidic acid production. Semin Cell Dev Biol 15:477–489.

8. Sano T, Baker D, Virag T, Wada A, Yatomi Y, Kobayashi T, Igarashi Y, Tigyi G. 2002. Multiple mechanisms linked to platelet activation result in lysophosphatidic acid and sphingosine 1-phosphate generation in blood. J Biol Chem 277:21197–21206.

9. Murakami M, Kudo I. 2002. Phospholipase A2. J Biochem 131:285–292. 10. Aoki J, Inoue A, Okudaira S. 2008. Two pathways for lysophosphatidic acid pro-

duction. Biochim Biophys Acta 1781:513–518.

Page 12: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

672 LPA RECEPTORS IN CANCER

11. Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure K, Yasuda K, Fukuzawa K. 2002. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J Biol Chem 277:39436–39442.

12. Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, Takio K, Yamori T, Mills GB, Inoue K, Aoki J, Arai H. 2002. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J Cell Biol 158:227–233.

13. Nam SW, Clair T, Campo CK, Lee HY, Liotta LA, Stracke ML. 2000. Autotaxin (ATX), a potent tumor motogen, augments invasive and metastatic potential of ras-transformed cells. Oncogene 19:241–247.

14. van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradere JP, Pettit TR, Wakelam MJ, Saulnier-Blache JS, Mummery CL, Moolenaar WH, Jonkers J. 2006. Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Mol Cell Biol 26:5015–5022.

15. Tanaka M, Okudaira S, Kishi Y, Ohkawa R, Iseki S, Ota M, Noji S, Yatomi Y, Aoki J, Arai H. 2006. Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophosphatidic acid. J Biol Chem 281:25822–25830.

16. Brindley DN. 2004. Lipid phosphate phosphatases and related proteins: signaling functions in development, cell division, and cancer. J Cell Biochem 92:900–912.

17. Waggoner DW, Xu J, Singh I, Jasinska R, Zhang QX, Brindley DN. 1999. Structural organization of mammalian lipid phosphate phosphatases: implications for signal transduction. Biochim Biophys Acta 1439:299–316.

18. Zhang QX, Pilquil CS, Dewald J, Berthiaume LG, Brindley DN. 2000. Identifica-tion of structurally important domains of lipid phosphate phosphatase-1: implica-tions for its sites of action. Biochem J 345(Pt 2):181–184.

19. Tanyi JL, Morris AJ, Wolf JK, Fang X, Hasegawa Y, Lapushin R, Auersperg N, Sigal YJ, Newman RA, Felix EA, Atkinson EN, Mills GB. 2003. The human lipid phosphate phosphatase-3 decreases the growth, survival, and tumorigenesis of ovarian cancer cells: validation of the lysophosphatidic acid signaling cascade as a target for therapy in ovarian cancer. Cancer Res 63:1073–1082.

20. Smyth SS, Sciorra VA, Sigal YJ, Pamuklar Z, Wang Z, Xu Y, Prestwich GD, Morris AJ. 2003. Lipid phosphate phosphatases regulate lysophosphatidic acid produc-tion and signaling in platelets: studies using chemical inhibitors of lipid phosphate phosphatase activity. J Biol Chem 278:43214–43223.

21. Tomsig JL, Snyder AH, Berdyshev EV, Skobeleva A, Mataya C, Natarajan V, Brindley DN, Lynch KR. 2009. Lipid phosphate phosphohydrolase type 1 (LPP1) degrades extracellular lysophosphatidic acid in vivo. Biochem J 419:611–618.

22. Chun J, Hla T, Lynch KR, Spiegel S, Moolenaar WH. 2010. International Union of Basic and Clinical Pharmacology. LXXVIII. Lysophospholipid receptor nomen-clature. Pharmacol Rev 62:579–587.

23. Hecht JH, Weiner JA, Post SR, Chun J. 1996. Ventricular zone gene-1 (vzg-1) encodes a lysophosphatidic acid receptor expressed in neurogenic regions of the developing cerebral cortex. J Cell Biol 135:1071–1083.

24. An S, Bleu T, Hallmark OG, Goetzl EJ. 1998. Characterization of a novel subtype of human G protein-coupled receptor for lysophosphatidic acid. J Biol Chem 273:7906–7910.

Page 13: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

REFERENCES 673

25. Bandoh K, Aoki J, Hosono H, Kobayashi S, Kobayashi T, Murakami-Murofushi K, Tsujimoto M, Arai H, Inoue K. 1999. Molecular cloning and characterization of a novel human G-protein-coupled receptor, EDG7, for lysophosphatidic acid. J Biol Chem 274:27776–27785.

26. Im DS, Heise CE, Harding MA, George SR, O’Dowd BF, Theodorescu D, Lynch KR. 2000. Molecular cloning and characterization of a lysophosphatidic acid receptor, Edg-7, expressed in prostate. Mol Pharmacol 57:753–759.

27. Noguchi K, Ishii S, Shimizu T. 2003. Identification of p2y9/GPR23 as a novel G protein-coupled receptor for lysophosphatidic acid, structurally distant from the Edg family. J Biol Chem 278:25600–25606.

28. 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. J Biol Chem 281:23589–23597.

29. Pasternack SM, von Kugelgen I, Aboud KA, Lee YA, Ruschendorf F, Voss K, Hillmer AM, Molderings GJ, Franz T, Ramirez A, Nurnberg P, Nothen MM, Betz RC. 2008. G protein-coupled receptor P2Y5 and its ligand LPA are involved in maintenance of human hair growth. Nat Genet 40:329–334.

30. van Corven EJ, Groenink A, Jalink K, Eichholtz T, Moolenaar WH. 1989. Lysophosphatidate-induced cell proliferation: identification and dissection of sig-naling pathways mediated by G proteins. Cell 59:45–54.

31. Howe LR, Marshall CJ. 1993. Lysophosphatidic acid stimulates mitogen-activated protein kinase activation via a G-protein-coupled pathway requiring p21ras and p74raf-1. J Biol Chem 268:20717–20720.

32. Stoyanov B, Volinia S, Hanck T, Rubio I, Loubtchenkov M, Malek D, Stoyanova S, Vanhaesebroeck B, Dhand R, Nurnberg B, Gierschik P, Seedorf K, Hsuan JJ, Waterfield MD, Wetzker R. 1995. Cloning and characterization of a G protein-activated human phosphoinositide-3 kinase. Science 269:690–693.

33. Mills GB, Moolenaar WH. 2003. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer 3:582–591.

34. Tabata K, Baba K, Shiraishi A, Ito M, Fujita N. 2007. The orphan GPCR GPR87 was deorphanized and shown to be a lysophosphatidic acid receptor. Biochem Biophys Res Commun 363(3):861–866.

35. Contos JJ, Fukushima N, Weiner JA, Kaushal D, Chun J. 2000. Requirement for the lpA1 lysophosphatidic acid receptor gene in normal suckling behavior. Proc Natl Acad Sci U S A 97:13384–13389.

36. Inoue M, Rashid MH, Fujita R, Contos JJ, Chun J, Ueda H. 2004. Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling. Nat Med 10:712–718.

37. Pradere JP, Klein J, Gres S, Guigne C, Neau E, Valet P, Calise D, Chun J, Bascands JL, Saulnier-Blache JS, Schanstra JP. 2007. LPA1 receptor activation promotes renal interstitial fibrosis. J Am Soc Nephrol 18:3110–3118.

38. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, Polo-sukhin V, Wain J, Karimi-Shah BA, Kim ND, Hart WK, Pardo A, Blackwell TS, Xu Y, Chun J, Luster AD. 2008. The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascu-lar leak. Nat Med 14:45–54.

Page 14: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

674 LPA RECEPTORS IN CANCER

39. Contos JJ, Ishii I, Fukushima N, Kingsbury MA, Ye X, Kawamura S, Brown JH, Chun J. 2002. Characterization of lpa(2) (Edg4) and lpa(1)/lpa(2) (Edg2/Edg4) lysophosphatidic acid receptor knockout mice: signaling deficits without obvious phenotypic abnormality attributable to lpa(2). Mol Cell Biol 22:6921–6929.

40. Ye X, Hama K, Contos JJ, Anliker B, Inoue A, Skinner MK, Suzuki H, Amano T, Kennedy G, Arai H, Aoki J, Chun J. 2005. LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature 435:104–108.

41. Sumida H, Noguchi K, Kihara Y, Abe M, Yanagida K, Hamano F, Sato S, Tamaki K, Morishita Y, Kano MR, Iwata C, Miyazono K, Sakimura K, Shimizu T, Ishii S. 2010. LPA4 regulates blood and lymphatic vessel formation during mouse embryo-genesis. Blood 116:5060–5070.

42. Lee Z, Cheng CT, Zhang H, Subler MA, Wu J, Mukherjee A, Windle JJ, Chen CK, Fang X. 2008. Role of LPA4/p2y9/GPR23 in negative regulation of cell motility. Mol Biol Cell 19:5435–5445.

43. Imamura F, Horai T, Mukai M, Shinkai K, Sawada M, Akedo H. 1993. Induction of in vitro tumor cell invasion of cellular monolayers by lysophosphatidic acid or phospholipase D. Biochem Biophys Res Commun 193(2):497–503.

44. Hama K, Aoki J, Fukaya M, Kishi Y, Sakai T, Suzuki R, Ohta H, Yamori T, Wata-nabe M, Chun J, Arai H. 2004. Lysophosphatidic acid and autotaxin stimulate cell motility of neoplastic and non-neoplastic cells through LPA1. J Biol Chem 279:17634–17639.

45. Boucharaba A, Serre CM, Guglielmi J, Bordet JC, Clezardin P, Peyruchaud O. 2006. The type 1 lysophosphatidic acid receptor is a target for therapy in bone metastases. Proc Natl Acad Sci U S A 103:9643–9648.

46. Van Leeuwen FN, Olivo C, Grivell S, Giepmans BN, Collard JG, Moolenaar WH. 2003. Rac activation by lysophosphatidic acid LPA1 receptors through the guanine nucleotide exchange factor Tiam1. J Biol Chem 278:400–406.

47. Boucharaba A, Serre CM, Gres S, Saulnier-Blache JS, Bordet JC, Guglielmi J, Clezardin P, Peyruchaud O. 2004. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest 114:1714–1725.

48. Nobes CD, Hall A. 1999. Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol 144:1235–1244.

49. Contos JJ, Ishii I, Chun J. 2000. Lysophosphatidic acid receptors. Mol Pharmacol 58:1188–1196.

50. Su LJ, Chang CW, Wu YC, Chen KC, Lin CJ, Liang SC, Lin CH, Whang-Peng J, Hsu SL, Chen CH, Huang CY. 2007. Selection of DDX5 as a novel internal control for Q-RT-PCR from microarray data using a block bootstrap re-sampling scheme. BMC Genomics 8:140.

51. Radvanyi L, Singh-Sandhu D, Gallichan S, Lovitt C, Pedyczak A, Mallo G, Gish K, Kwok K, Hanna W, Zubovits J, Armes J, Venter D, Hakimi J, Shortreed J, Donovan M, Parrington M, Dunn P, Oomen R, Tartaglia J, Berinstein NL. 2005. The gene associated with trichorhinophalangeal syndrome in humans is overex-pressed in breast cancer. Proc Natl Acad Sci U S A 102:11005–11010.

52. Hendrix ND, Wu R, Kuick R, Schwartz DR, Fearon ER, Cho KR. 2006. Fibroblast growth factor 9 has oncogenic activity and is a downstream target of Wnt signaling in ovarian endometrioid adenocarcinomas. Cancer Res 66:1354–1362.

Page 15: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

REFERENCES 675

53. Sanchez-Carbayo M, Socci ND, Lozano J, Saint F, Cordon-Cardo C. 2006. Defining molecular profiles of poor outcome in patients with invasive bladder cancer using oligonucleotide microarrays. J Clin Oncol 24:778–789.

54. Skrzypczak M, Goryca K, Rubel T, Paziewska A, Mikula M, Jarosz D, Pachlewski J, Oledzki J, Ostrowski J. 2010. Modeling oncogenic signaling in colon tumors by multidirectional analyses of microarray data directed for maximization of analyti-cal reliability. PLoS ONE 5:e13091.

55. Shida D, Watanabe T, Aoki J, Hama K, Kitayama J, Sonoda H, Kishi Y, Yamaguchi H, Sasaki S, Sako A, Konishi T, Arai H, Nagawa H. 2004. Aberrant expression of lysophosphatidic acid (LPA) receptors in human colorectal cancer. Lab Invest 84:1352–1362.

56. Marshall JC, Collins J, Marino N, Steeg P. 2010. The Nm23-H1 metastasis suppres-sor as a translational target. Eur J Cancer 46:1278–1282.

57. Lee JH, Marshall JC, Steeg PS, Horak CE. 2009. Altered gene and protein expres-sion by Nm23-H1 in metastasis suppression. Mol Cell Biochem 329:141–148.

58. Boissan M, De Wever O, Lizarraga F, Wendum D, Poincloux R, Chignard N, Desbois-Mouthon C, Dufour S, Nawrocki-Raby B, Birembaut P, Bracke M, Chavr-ier P, Gespach C, Lacombe ML. 2010. Implication of metastasis suppressor NM23-H1 in maintaining adherens junctions and limiting the invasive potential of human cancer cells. Cancer Res 70:7710–7722.

59. Oda Y, Naka T, Takeshita M, Iwamoto Y, Tsuneyoshi M. 2000. Comparison of histological changes and changes in nm23 and c-MET expression between primary and metastatic sites in osteosarcoma: a clinicopathologic and immunohistochemi-cal study. Hum Pathol 31:709–716.

60. Horak CE, Lee JH, Elkahloun AG, Boissan M, Dumont S, Maga TK, Arnaud-Dabernat S, Palmieri D, Stetler-Stevenson WG, Lacombe ML, Meltzer PS, Steeg PS. 2007. Nm23-H1 suppresses tumor cell motility by down-regulating the lyso-phosphatidic acid receptor EDG2. Cancer Res 67:7238–7246.

61. Horak CE, Mendoza A, Vega-Valle E, Albaugh M, Graff-Cherry C, McDermott WG, Hua E, Merino MJ, Steinberg SM, Khanna C, Steeg PS. 2007. Nm23-H1 sup-presses metastasis by inhibiting expression of the lysophosphatidic acid receptor EDG2. Cancer Res 67:11751–11759.

62. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D. 2000. Molecular portraits of human breast tumours. Nature 406:747–752.

63. van ’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH. 2002. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:530–536.

64. Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, Traficante N, Fereday S, Hung JA, Chiew YE, Haviv I, Gertig D, DeFazio A, Bowtell DD. 2008. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res 14:5198–5208.

Page 16: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

676 LPA RECEPTORS IN CANCER

65. Stadler CR, Knyazev P, Bange J, Ullrich A. 2006. FGFR4 GLY388 isotype sup-presses motility of MDA-MB-231 breast cancer cells by EDG-2 gene repression. Cell Signal 18:783–794.

66. Sugiyama N, Varjosalo M, Meller P, Lohi J, Chan KM, Zhou Z, Alitalo K, Taipale J, Keski-Oja J, Lehti K. 2010. FGF receptor-4 (FGFR4) polymorphism acts as an activity switch of a membrane type 1 matrix metalloproteinase-FGFR4 complex. Proc Natl Acad Sci U S A 107:15786–15791.

67. Murph MM, Hurst-Kennedy J, Newton V, Brindley DN, Radhakrishna H. 2007. Lysophosphatidic acid decreases the nuclear localization and cellular abundance of the p53 tumor suppressor in A549 lung carcinoma cells. Mol Cancer Res 5:1201–1211.

68. Komachi M, Tomura H, Malchinkhuu E, Tobo M, Mogi C, Yamada T, Kimura T, Kuwabara A, Ohta H, Im DS, Kurose H, Takeyoshi I, Sato K, Okajima F. 2009. LPA1 receptors mediate stimulation, whereas LPA2 receptors mediate inhibition, of migration of pancreatic cancer cells in response to lysophosphatidic acid and malignant ascites. Carcinogenesis 30:457–465.

69. Chen M, Towers LN, O’Connor KL. 2007. LPA2 (EDG4) mediates Rho-dependent chemotaxis with lower efficacy than LPA1 (EDG2) in breast carcinoma cells. Am J Physiol Cell Physiol 292:C1927–C1933.

70. Fang X, Schummer M, Mao M, Yu S, Tabassam FH, Swaby R, Hasegawa Y, Tanyi JL, LaPushin R, Eder A, Jaffe R, Erickson J, Mills GB. 2002. Lysophosphatidic acid is a bioactive mediator in ovarian cancer. Biochim Biophys Acta 1582:257–264.

71. Fang X, Gaudette D, Furui T, Mao M, Estrella V, Eder A, Pustilnik T, Sasagawa T, Lapushin R, Yu S, Jaffe RB, Wiener JR, Erickson JR, Mills GB. 2000. Lysophos-pholipid growth factors in the initiation, progression, metastases, and management of ovarian cancer. Ann N Y Acad Sci 905:188–208.

72. Schulte KM, Beyer A, Kohrer K, Oberhauser S, Roher HD. 2001. Lysophospha-tidic acid, a novel lipid growth factor for human thyroid cells: over-expression of the high-affinity receptor edg4 in differentiated thyroid cancer. Int J Cancer 92:249–256.

73. Kitayama J, Shida D, Sako A, Ishikawa M, Hama K, Aoki J, Arai H, Nagawa H. 2004. Over-expression of lysophosphatidic acid receptor-2 in human invasive ductal carcinoma. Breast Cancer Res 6:R640–R646.

74. Yamashita H, Kitayama J, Shida D, Ishikawa M, Hama K, Aoki J, Arai H, Nagawa H. 2006. Differential expression of lysophosphatidic acid receptor-2 in intestinal and diffuse type gastric cancer. J Surg Oncol 93:30–35.

75. Yu S, Murph MM, Lu Y, Liu S, Hall HS, Liu J, Stephens C, Fang X, Mills GB. 2008. Lysophosphatidic acid receptors determine tumorigenicity and aggressiveness of ovarian cancer cells. J Natl Cancer Inst 100:1630–1642.

76. Liu S, Umezu-Goto M, Murph M, Lu Y, Liu W, Zhang F, Yu S, Stephens LC, Cui X, Murrow G, Coombes K, Muller W, Hung MC, Perou CM, Lee AV, Fang X, Mills GB. 2009. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell 15:539–550.

77. Huang MC, Lee HY, Yeh CC, Kong Y, Zaloudek CJ, Goetzl EJ. 2004. Induction of protein growth factor systems in the ovaries of transgenic mice overexpressing

Page 17: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

REFERENCES 677

human type 2 lysophosphatidic acid G protein-coupled receptor (LPA2). Onco-gene 23:122–129.

78. Lin S, Wang D, Iyer S, Ghaleb AM, Shim H, Yang VW, Chun J, Yun CC. 2009. The absence of LPA2 attenuates tumor formation in an experimental model of colitis-associated cancer. Gastroenterology 136:1711–1720.

79. Lin S, Lee SJ, Shim H, Chun J, Yun CC. 2010. The absence of LPA receptor 2 reduces the tumorigenesis by ApcMin mutation in the intestine. Am J Physiol Gastrointest Liver Physiol 299:G1128–G1138.

80. Hu YL, Tee MK, Goetzl EJ, Auersperg N, Mills GB, Ferrara N, Jaffe RB. 2001. Lysophosphatidic acid induction of vascular endothelial growth factor expression in human ovarian cancer cells. J Natl Cancer Inst 93:762–768.

81. Zhang H, Bialkowska A, Rusovici R, Chanchevalap S, Shim H, Katz JP, Yang VW, Yun CC. 2007. Lysophosphatidic acid facilitates proliferation of colon cancer cells via induction of Krüppel-like factor 5. J Biol Chem 282:15541–15549.

82. Yang M, Zhong WW, Srivastava N, Slavin A, Yang J, Hoey T, An S. 2005. G protein-coupled lysophosphatidic acid receptors stimulate proliferation of colon cancer cells through the β-catenin pathway. Proc Natl Acad Sci U S A 102:6027–6032.

83. Oyesanya RA, Lee ZP, Wu J, Chen J, Song Y, Mukherjee A, Dent P, Kordula T, Zhou H, Fang X. 2008. Transcriptional and post-transcriptional mechanisms for lysophosphatidic acid-induced cyclooxygenase-2 expression in ovarian cancer cells. FASEB J 22:2639–2651.

84. Fang X, Yu S, Bast RC, Liu S, Xu HJ, Hu SX, LaPushin R, Claret FX, Aggarwal BB, Lu Y, Mills GB. 2004. Mechanisms for lysophosphatidic acid-induced cytokine production in ovarian cancer cells. J Biol Chem 279(10):9653–9661.

85. Lin FT, Lai YJ. 2008. Regulation of the LPA2 receptor signaling through the carboxyl-terminal tail-mediated protein-protein interactions. Biochim Biophys Acta 1781:558–562.

86. Oh YS, Jo NW, Choi JW, Kim HS, Seo SW, Kang KO, Hwang JI, Heo K, Kim SH, Kim YH, Kim IH, Kim JH, Banno Y, Ryu SH, Suh PG. 2004. NHERF2 specifically interacts with LPA2 receptor and defines the specificity and efficiency of receptor-mediated phospholipase C-beta3 activation. Mol Cell Biol 24:5069–5079.

87. Yun CC, Sun H, Wang D, Rusovici R, Castleberry A, Hall RA, Shim H. 2005. LPA2 receptor mediates mitogenic signals in human colon cancer cells. Am J Physiol Cell Physiol 289:C2–C11.

88. Shano S, Hatanaka K, Ninose S, Moriyama R, Tsujiuchi T, Fukushima N. 2008. A lysophosphatidic acid receptor lacking the PDZ-binding domain is constitutively active and stimulates cell proliferation. Biochim Biophys Acta 1783:748–759.

89. Choi JW, Lim S, Oh YS, Kim EK, Kim SH, Kim YH, Heo K, Kim J, Kim JK, Yang YR, Ryu SH, Suh PG. 2010. Subtype-specific role of phospholipase C-beta in bradykinin and LPA signaling through differential binding of different PDZ scaf-fold proteins. Cell Signal 22:1153–1161.

90. Lee SJ, Ritter SL, Zhang H, Shim H, Hall RA, Yun CC. 2010. MAGI-3 competes with NHERF-2 to negatively regulate LPA2 receptor signaling in colon cancer cells. Gastroenterology 140:924–934.

91. Zeng Y, Kakehi Y, Nouh MA, Tsunemori H, Sugimoto M, Wu XX. 2009. Gene expression profiles of lysophosphatidic acid-related molecules in the prostate: relevance to prostate cancer and benign hyperplasia. Prostate 69:283–292.

Page 18: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

678 LPA RECEPTORS IN CANCER

92. Tsujino M, Fujii M, Okabe K, Mori T, Fukushima N, Tsujiuchi T. 2010. Differential expressions and DNA methylation patterns of lysophosphatidic acid receptor genes in human colon cancer cells. Virchows Arch 457:669–676.

93. Hayashi M, Okabe K, Yamawaki Y, Teranishi M, Honoki K, Mori T, Fukushima N, Tsujiuchi T. 2011. Loss of lysophosphatidic acid receptor-3 enhances cell migra-tion in rat lung tumor cells. Biochem Biophys Res Commun 405:450–454.

94. Kishi Y, Okudaira S, Tanaka M, Hama K, Shida D, Kitayama J, Yamori T, Aoki J, Fujimaki T, Arai H. 2006. Autotaxin is overexpressed in glioblastoma multiforme and contributes to cell motility of glioblastoma by converting lysophosphatidyl-choline to lysophosphatidic acid. J Biol Chem 281:17492–17500.

95. Zhang Y, Scoumanne A, Chen X. 2010. G protein-coupled receptor 87: a promising opportunity for cancer drug discovery. Mol Cell Pharmacol 2:111–116.

96. Glatt S, Halbauer D, Heindl S, Wernitznig A, Kozina D, Su KC, Puri C, Garin-Chesa P, Sommergruber W. 2008. hGPR87 contributes to viability of human tumor cells. Int J Cancer 122:2008–2016.

97. Zhang Y, Qian Y, Lu W, Chen X. 2009. The G protein-coupled receptor 87 is neces-sary for p53-dependent cell survival in response to genotoxic stress. Cancer Res 69:6049–6056.

98. Polak M. 1999. Hyperfunctioning thyroid adenoma and activating mutations in the TSH receptor gene. Arch Med Res 30:510–513.

99. Obo Y, Yamada T, Furukawa M, Hotta M, Honoki K, Fukushima N, Tsujiuchi T. 2009. Frequent mutations of lysophosphatidic acid receptor-1 gene in rat liver tumors. Mutat Res 660:47–50.

100. Yamada T, Obo Y, Furukawa M, Hotta M, Yamasaki A, Honoki K, Fukushima N, Tsujiuchi T. 2009. Mutations of lysophosphatidic acid receptor-1 gene during pro-gression of lung tumors in rats. Biochem Biophys Res Commun 378:424–427.

101. Okabe K, Hayashi M, Fujii M, Honoki K, Mori T, Fukushima N, Tsujiuchi T. 2010. Mutations of lysophosphatidic acid receptor genes in human osteosarcoma cells. Pathobiology 77:278–282.

102. Masuda A, Nakamura K, Izutsu K, Igarashi K, Ohkawa R, Jona M, Higashi K, Yokota H, Okudaira S, Kishimoto T, Watanabe T, Koike Y, Ikeda H, Kozai Y, Kurokawa M, Aoki J, Yatomi Y. 2008. Serum autotaxin measurement in haemato-logical malignancies: a promising marker for follicular lymphoma. Br J Haematol 143:60–70.

103. Nouh MA, Wu XX, Okazoe H, Tsunemori H, Haba R, Abou-Zeid AM, Saleem MD, Inui M, Sugimoto M, Aoki J, Kakehi Y. 2009. Expression of autotaxin and acylglycerol kinase in prostate cancer: association with cancer development and progression. Cancer Sci 100:1631–1638.

104. Stassar MJ, Devitt G, Brosius M, Rinnab L, Prang J, Schradin T, Simon J, Petersen S, Kopp-Schneider A, Zoller M. 2001. Identification of human renal cell carcinoma associated genes by suppression subtractive hybridization. Br J Cancer 85:1372–1382.

105. Kehlen A, Englert N, Seifert A, Klonisch T, Dralle H, Langner J, Hoang-Vu C. 2004. Expression, regulation and function of autotaxin in thyroid carcinomas. Int J Cancer 109:833–838.

106. Yang Y, Mou L, Liu N, Tsao MS. 1999. Autotaxin expression in non-small-cell lung cancer. Am J Respir Cell Mol Biol 21:216–222.

Page 19: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid Receptors in Cancer

REFERENCES 679

107. Ohta H, Sato K, Murata N, Damirin A, Malchinkhuu E, Kon J, Kimura T, Tobo M, Yamazaki Y, Watanabe T, Yagi M, Sato M, Suzuki R, Murooka H, Sakai T, Nishitoba T, Im DS, Nochi H, Tamoto K, Tomura H, Okajima F. 2003. Ki16425, a subtype-selective antagonist for EDG-family lysophosphatidic acid receptors. Mol Pharmacol 64:994–1005.

108. Beck HP, Kohn T, Rubenstein S, Hedberg C, Schwandner R, Hasslinger K, Dai K, Li C, Liang L, Wesche H, Frank B, An S, Wickramasinghe D, Jaen J, Medina J, Hungate R, Shen W. 2008. Discovery of potent LPA2 (EDG4) antagonists as potential anticancer agents. Bioorg Med Chem Lett 18:1037–1041.

109. Parrill AL, Baker DL. 2008. Autotaxin inhibition: challenges and progress toward novel anti-cancer agents. Anticancer Agents Med Chem 8:917–923.

110. Xu X, Yang G, Zhang H, Prestwich GD. 2009. Evaluating dual activity LPA recep-tor pan-antagonist/autotaxin inhibitors as anti-cancer agents in vivo using engi-neered human tumors. Prostaglandins Other Lipid Mediat 89:140–146.

111. Song Y, Wilkins P, Hu W, Murthy KS, Chen J, Lee Z, Oyesanya R, Wu J, Barbour SE, Fang X. 2007. Inhibition of calcium-independent phospholipase A2 suppresses proliferation and tumorigenicity of ovarian carcinoma cells. Biochem J 406:427–436.

112. Li H, Zhao Z, Wei G, Yan L, Wang D, Zhang H, Sandusky GE, Turk J, Xu Y. 2010. Group VIA phospholipase A2 in both host and tumor cells is involved in ovarian cancer development. FASEB J 24:4103–4116.

113. Li H, Zhang H, Wei G, Cai Q, Yan L, Xu Y. 2010. Tumor cell group via phospho-lipase A is involved in prostate cancer development. Prostate 71:373–384.


Recommended