Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Mar 1.
Published in final edited form as: Biochim Biophys Acta. 2008 May 3;1781(9):558–562. doi: 10.1016/j.bbalip.2008.04.013

Regulation of the LPA2 Receptor Signaling through the Carboxyl-Terminal Tail-Mediated Protein-Protein Interactions

Fang-Tsyr Lin 1,*, Yun-Ju Lai 1
PMCID: PMC2574565  NIHMSID: NIHMS59191  PMID: 18501721

Abstract

While it is well known that lysophosphatidic acid (LPA) mediates diverse physiological and pathophysiological responses through the activation of G protein-coupled LPA receptors, the specificity and molecular mechanisms by which different LPA receptors mediate these biological responses remain largely unknown. Recent identification of several PDZ proteins and zinc finger proteins that interact with the carboxyl-terminal tail of the LPA2 receptor provides a considerable progress towards the understanding of the mechanisms how the LPA2 receptor specifically mediates LPA signaling pathways. These findings have led to the proposal that there are at least two distinct protein interaction motifs present in the carboxyl terminus of the LPA2 receptor. Together, these data provide a new concept that the efficiency and specificity of the LPA2 receptor-mediated signal transduction can be achieved through the cross-regulation between the classical G protein-activated signaling cascades and the interacting partner-mediated signaling pathways.

Keywords: LPA, LPA2 receptor, PDZ, LIM, zinc finger, protein-protein interactions

1. Introduction

LPA is a growth factor-like phospholipid, which mediates diverse biological responses such as cell proliferation, differentiation, migration, survival, angiogenesis, inflammation and platelet aggregation [1-5]. In the past decade, the identification of at least seven G protein-coupled LPA receptors, including LPA1/EDG2, LPA2/EDG4 and LPA3/EDG7 of the Endothelial Differentiation Gene (EDG) family and the structurally distinct LPA4/P2Y9/GPR23, LPA5/GPR92, GPR87, and P2Y5 of the P2Y purinoreceptor family has provided a big step toward the understanding of LPA actions [6-12]. These LPA-bound membrane receptors couple to Gs, Gi/o, Gq and/or G12/13 proteins to activate various signaling pathways. In addition, LPA can bind to the nuclear peroxisome proliferator-activated receptor γ (PPARγ) to regulate vascular remodeling [13, 14]. Along with the rapid development of this field, more efforts have been made to determine the specificity and mechanisms by which different LPA receptors mediate LPA actions. Among all of the LPA receptors, the LPA2 receptor is unique in the carboxyl-terminal tail, in which it contains two distinct protein-protein interaction domains to bind several PDZ proteins and zinc finger proteins, respectively [15-21]. These protein-protein interactions confer the specificity, efficiency and diversity by which the LPA2 receptor mediates LPA-evoked signal transduction, which will be the focus of this review.

2. Functional roles of the LPA2 receptor

In response to LPA stimulation, the LPA2 receptor couples to Gi/o, Gq and G12/13 proteins to mediate cellular responses such as DNA synthesis, MAP kinase activation, AKT activation, inhibition of adenylyl cyclase, increase of inositol phosphates and intracellular calcium concentration, cell rounding, cell adhesion and cell migration [8]. These signaling events are partially impaired in mouse embryonic fibroblasts (MEFs) derived from the LPA2−/− mice, and are dramatically reduced or completely abolished in MEFs derived from the LPA1−/−, LPA2−/− mice, suggesting a functional redundancy of LPA1 and LPA2 receptors in some of the signaling pathways. However, disruption of the LPA2 receptor gene does not result in any developmental abnormality [22].

Although the LPA2 receptor is not required for growth and development, increasing evidence has implicated LPA and the LPA2 receptor as the key mediators in cancer progression. It has been reported that the expression of the LPA2 receptor is up-regulated in ovarian cancers, invasive ductal carcinoma tissue of breast cancers, colorectal cancers and thyroid cancers [23-26]. Recently autotaxin/lysophospholipase D (ATX/lysoPLD) has been known to mediate tumor invasion, neovascularization and metastasis through the production of LPA in the cellular microenvironment [5, 27]. And the platelet-derived LPA has been shown to act as a tumor cell mitogen and promoting factor of osteolysis during bone metastasis [28]. Although several actions of LPA can contribute to tumorigenesis, including cell migration, cell proliferation, cell survival and transcriptional activation of genes involved in these signaling events [5], it remains unclear whether the LPA2 receptor meditates these actions through specific signaling pathways.

3. Regulation of LPA2 receptor functions through the carboxyl-terminal tail-mediated protein-protein interactions

The classical paradigm of G protein-coupled receptor activation involves the coupling of ligand-bound receptors to heterotrimeric G proteins, which sequentially activates the downstream effector molecules and induces cellular responses. However, emerging evidence has revealed that other mechanisms can cross-regulate GPCR signaling pathways. For example, a number of GPCRs can interact with cellular proteins through the intracellular loops or carboxyl-terminal tail. These interacting partners participate in the regulation of receptor trafficking, dimerization, effector coupling, membrane expression and subcellular localization, etc., thereby determining the signal specificity and efficiency [29, 30].

The LPA1, LPA2 and LPA3 receptors share a high homology in the sequences except for the carboxyl-terminal tail, suggesting that this region may specifically regulate the unique functions of each receptor. The structure of LPA2-CT (a.a. 296-351) contains a di-leucine motif and several putative palmitoylated cysteine residues in the proximal region, and a class I PDZ-binding motif at the distal end. It also contains several serine and threonine residues presumably can be phosphorylated by G protein-coupled receptor kinases (GRKs), which are important for β-arrestin binding and receptor internalization. The last four amino acids, DSTL, known as a class I PDZ-binding motif, mediates the interactions with NHERF2 (Na+/H+ exchanger regulatory factor 2), PDZ-RhoGEF, LARG (Leukemia-associated RhoGEF) and MAGI-3 (membrane-associated guanylate kinase with an inverted domain structure-3) [16-18, 20, 21], whereas the proximal region is responsible for the association with several zinc finger proteins, including the LIM domain-containing TRIP6 (thyroid hormone receptor-interacting protein 6) and the proapoptotic Siva-1 protein [15, 19] (Table 1). Except for PDZ-RhoGEF and LARG that can bind to both LPA1 and LPA2 receptors [18], other proteins selectively interact with the LPA2 receptor but not other LPA receptors, suggesting that these interacting partners may play specific roles in regulating LPA2 receptor functions.

Table I.

Proteins interacting with the carboxyl-terminal tail of the LPA2 receptor

Binding motif Interacting protein Functional significance References
PDZ-binding motif
(distal end)
NHERF2 Activation of PLC-β3 signaling pathways [17]
Activation of AKT and ERKs [21]
Linking LPA2 with CFTR to inhibit CFTR activity [20]
PDZ-RhoGEF
LARG
RhoA activation [18]
MAGI-3 Activation of RhoA and ERKs [16]
Zinc finger-binding
motif
(proximal region)
TRIP6 Regulation of cell migration through
c-Src/PTPL1-dependent phosphorylation
and dephosphorylation
[15, 64, 66]
ERK activation
Siva-1 Interactions mediate down-regulation of
Siva-1 to promote cell survival
[19]

3.1. PDZ-mediated protein-protein interactions

PDZ domains, derived from the initials of three proteins containing such domains including PSD-95, the Drosophila discs-large tumor suppressor protein DlgA and the tight junction protein ZO-1, are composed of ∼90 amino acids that function as the protein interaction modules, which bind to the specific short peptide motif found in the carboxyl-terminus or internal region of a variety of target proteins [31-35]. Most of the PDZ domains are involved in the organization of multi-protein complexes, which allows the stable localization of interacting proteins to achieve the efficiency and fidelity of the signal transduction. The PDZ-binding motifs can be classified into three groups, which are composed of the four amino acids, X-S/T-X-L/V (class I), X-ϕ-X-ϕ (class II) and X-D-X-V (class III), respectively (ϕ, hydrophobic amino acid; X, unspecified amino acid) [36]. These PDZ-binding motifs can be found in the carboxyl-terminus of several LPA receptors, such as LPA1 (HSVV), LPA2 (DSTL) and LPA5 (DSAL).

Interestingly, the first reported LPA2 cDNA clone was derived from an ovarian tumor library, which lacks the last four amino acids (DSTL), but is 31 amino acids longer than the reported human cDNA sequences [8]. This can be explained by one guanine base deletion at the 3' end of the coding sequences, which leads to a frame-shift mutation in the carboxyl-terminal region of the protein. However, whether these mutations lead to any de-regulation of LPA2 in tumor development is not known.

3.1.1. NHERF2 regulates LPA-mediated phospholipase C-β signaling pathway and the activation of ERKs and AKT

The Na+/H+ exchanger regulatory factors NHERF1 (also known as EBP50) and NHERF2 (also known as E3KARP) possess two tandem PDZ domains and a carboxyl-terminal domain that binds to the actin-binding proteins ezrin, radixin, moesin and merlin [37, 38]. The NHERF family members function as scaffold proteins by associating with a number of transporter, ion channels, transcription factors, different signaling molecules and growth factor receptors [38-40]. Emerging data also show that the two PDZ domains of NHERF1 and NHERF2 can interact with the carboxyl-terminal PDZ-binding motifs of a number of G protein-coupled receptors, and regulate receptor trafficking, localization and/or other functions [41]. However, NHERF1 and NHERF2 seem to have a distinct peptide-binding specificity. For example, PDZ1 of NHERF1 binds to the β2-adrenergic receptor (DSLL), P2Y1 purinergic receptor (DTSL), and cystic fibrosis transmembrane regulator (CFTR)(DTRL) [42, 43]. But NHERF1 does not bind to the LPA1 receptor (HSVV) or LPA2 receptor (DSTL) [17]. Likewise, PDZ1 of NHERF2 binds to the β2-adrernergic receptor (DSLL) [42]; however, PDZ2 of NHERF2 specifically binds to the LPA2 receptor (DSTL) but not LPA1 receptor (HSVV) [17]. These observations suggest that although the last four amino acid residues of the target receptors are required for PDZ domain recognition, other adjacent residues are also involved in determining the specificity and/or affinity of the interaction.

Through the interaction with the LPA2 receptor, NHERF2 potentiates LPA-induced ERK activation and COX-2 induction [17]. This effect can be partly attributed to the specific coupling of the LPA2 receptor to phospholipase C-β3 (PLC-β3) by NHERF2, which regulates inositol phosphoate production, calcium mobilization and protein kinase C activation. Intriguingly, both PLC-β3 and the LPA2 receptor bind to the same PDZ domain of NHERF2. Since NHERF proteins can form oligomers through PDZ domains [44, 45], NHERF2 must be self-associated to form a multi-protein complex containing the LPA2 receptor and PLC-β3. In addition, it has been reported that LPA-induced activation of ERKs and AKT is attenuated by the inhibition of NHERF2 expression in Caco-2 colon cancer cells that predominantly express the LPA2 receptor but not LPA1 or LPA3 receptor [21].

3.1.2. NHERF2 links the LPA2 receptor with CFTR

Cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-regulated chloride channel, which plays a critical role in cholera toxin-induced secretory diarrhea [46]. Through the PDZ domain-mediated interactions, NHERF2 bridges the LPA2 receptor and CFTR, and facilitates the macromolecular complex formation at the apical plasma membrane of intestinal epithelial cells [20]. When activated by LPA, the LPA2 receptor couples to Gi proteins to reduce cAMP levels, thereby inhibiting PKA-mediated activation of CFTR and cholera toxin-induced secretory diarrhea [20]. These studies demonstrate that the triple complex formation among the LPA2 receptor, NHERF2 and CFTR plays a key role in LPA-mediated inhibition of CFTR activity.

3.1.3. PDZ domain-containing Rho GEFs and MAGI-3 regulate LPA2 functions in RhoA and ERK activation

In line with the discovery of NHERF2 as an LPA2 receptor-interacting protein, two reports have demonstrated that the PDZ-binding motif of LPA2 receptor can also interact with the PDZ domains of two RhoA-specific guanine nucleotide exchange factors (GEFs) and the MAGI family members [16, 18]. The activated LPA receptors couple to G12/13 proteins, which can activate RhoA signaling through the recruitment of the RGS domain-containing Rho GEFs, including PDZ-RhoGEF, LARG and p115-RhoGEF [47-50]. Among these Rho GEFs, the PDZ domain-containing PDZ-RhoGEF and LARG are capable of binding to the LPA1 and LPA2 receptors, and regulate LPA-induced RhoA activation [18]. These findings suggest that the PDZ-mediated interaction may facilitate the recruitment of PDZ domain-containing RhoGEFs to the proximity of G12/13 proteins, thereby enhancing their ability to catalyze the guanine nucleotide exchange of RhoA.

Using a PDZ proteomic array to screen the proteins that interact with the LPA2 receptor, several PDZ domain-containing proteins have been identified, which include NHERF1 (PDZ1), NHERF2 (PDZ2), neurabin, MAGI-2 (PDZ5) and MAGI-3 (PDZ5) [16]. However, PDZ-RhoGEF and LARG were not found by this assay, suggesting a differential in vitro binding affinity among these PDZ-mediated interactions. MAGI proteins contain multiple PDZ domains, WW domains and a guanylate kinase-like domain, allowing them to function as scaffold proteins to enable multi-protein complex formation [51]. These proteins are present at the areas of cell-cell contact such as the synaptic junctions in neurons and the tight junctions in epithelial cells [51]. MAGI-3 can interact with PTEN, receptor tyrosine phosphatase-β, transforming growth factor-α (TGF-α), frizzled receptor and the β1-adrenergic receptor [52-56]. It has been reported that MAGI-3 can interact with PTEN to negatively modulate AKT activity, and bind to the β1-adrenergic receptor to impair Gi-mediated AKT activation [52, 55]. However, overexpression of MAGI-3 or inhibition of MAGI-3 expression does not significantly affect LPA-induced AKT activation [16]. Instead, the interaction of MAGI-3 with the LPA2 receptor regulates LPA-induced activation of RhoA and ERKs [16]. Overexpression of MAGI-3 enhances LPA-induced RhoA activation but has little effect on ERK activation, whereas knockdown of MAGI-3 inhibits LPA-induced RhoA and ERK activation. Together, these data demonstrate that the LPA2 receptor functions can be differentially regulated through PDZ-mediated protein-protein interactions in different cellular contexts.

3.2. Zinc finger-mediated protein-protein interactions

Using the carboxyl-terminal tail of the LPA2 receptor as bait in a yeast two-hybrid screen, two zinc finger-containing molecules have been identified, including TRIP6 and Siva-1 [15, 19]. The structure of TRIP6 contains an N-terminal proline-rich domain, three carboxyl LIM domains and a PDZ-binding motif at the carboxyl-terminal end. The LIM domains (named by the initials of Lin-11, Isl-1, and Mec-3) contain two zinc finger motifs, which are critical for protein-protein interactions [57]. TRIP6 binds to the LPA2 receptor mainly through LIM2-3 domains; however, LIM1 can facilitate this interaction [15]. The structure of Siva-1 protein contains an internal death domain homology region (DDHR) and two carboxyl zinc finger motifs; however, lacking histidine residues [58]. The association with the LPA2 receptor is mediated by the carboxyl-terminal zinc finger motif of Siva-1, which resembles the second half of TRIP6-LIM3 domain [19]. Indeed, the domain mapping shows that these two proteins bind to the same motif located in the proximal region of the carboxyl-terminal tail of the LPA2 receptor (F.-T. Lin and G. Tigyi, unpublished). However, there are some differences in their interactions. TRIP6 associates with the membrane-bound LPA2 receptor in an LPA-dependent manner, whereas Siva-1 binds to the LPA2 receptor on the membrane or in the cytosol in the absence or presence of LPA [15, 19]. The interaction of TRIP6 with the LPA2 receptor is transient since LPA treatment for 15-20 min induces the translocation of TRIP6 to focal adhesion plaques where the LPA2 receptor is not present. In contrast, Siva-1 seems to preferentially bind to the internalized LPA2 receptor.

3.2.1. The LIM domain-containing TRIP6 regulates LPA2 receptor-mediated cell migration

TRIP6, also known as ZRP-1 (Zyxin-related Protein 1), is a focal adhesion molecule known to regulate cell motility and transcriptional control [15, 59-62]. Together with LPP, zyxin, and more distantly with ajuba and LIMD1, these proteins form the zyxin family members [59]. Through the LIM domain-mediated protein-protein interactions, these molecules function as scaffold or adaptor proteins for the assembly of multi-protein complexes involved in actin rearrangement, cell adhesion and motility [63]. LPA stimulation promotes the recruitment of TRIP6 to the activated LPA2 receptor, and induces the association of TRIP6 with the components of focal complexes, including paxillin, p130cas, FAK and c-Src [15]. Subsequently, TRIP6 is phosphorylated by c-Src at Tyr-55, a unique site only present in TRIP6 but not other zyxin family members [64]. This phosphorylation transforms the pY55-Q-A-P motif of TRIP6 into a docking site for Crk SH2 domain, thereby enhancing LPA-induced morphological changes and cell migration. In addition, knockdown of TRIP6 attenuates LPA-induced, c-Src-mediated ERK activation. In contrast to c-Src, the PTPL1/FAP-1 (Fas-associated phosphatasae-1) tyrosine phosphatase binds to the carboxyl-terminal PDZ-binding motif and LIM3 of TRIP6 [65]. Dephosphorylation of TRIP6 by PTPL1/FAP-1 serves as a molecular switch by inhibiting TRIP6 binding to Crk, thereby negatively regulating TRIP6 function in LPA-induced adhesion turnover and cell migration [66]. Given the high sequence homology among the LIM domains of TRIP6, LPP and zyxin, LPP and zyxin are also found to bind to the LPA2 receptor in an LPA-dependent manner.

3.2.2 The interaction of the LPA2 receptor with Siva-1 promotes LPA-mediated down-regulation of Siva-1

LPA is known as a promoting factor for cell survival [67]. Several pro-survival signaling pathways have been defined for this protective effect, including the activation of AKT/PKB, ERKs and NF-κB [68, 69]. Among different LPA receptors, the LPA1 and LPA2 receptors have been shown to mediate these effects [70-72]. In contrast to the LPA2 receptor, Siva-1 functions as a proapoptotic protein, which is transcriptionally activated by p53 and E2F1 during DNA damage response [73]. Although the mechanism how Siva-1 regulates apoptosis is not fully understood, it has been reported hat Siva-1 promotes apoptosis through a caspase-3-dependent mitochondrial pathway [74], and can function as a negative regulator of NF-κB in T cell receptor-mediated activation-induced cell death [75]. In addition, Siva-1 binds to Bcl-xL and inhibits Bcl-xL-mediated protection against UV radiation-induced apoptosis [76].

Several G protein-coupled receptors have been shown to undergo ligand-stimulated ubiquitination and degradation through proteasomal or lysosomal pathways [77]. It has been noted that prolonged LPA stimulation not only down-regulates the LPA2 receptor but also concomitantly targets the LPA2 receptor-interacting Siva-1 for ubiquitination and proteasomal degradation [19]. As a result, the proapoptotic activity of Siva-1 during DNA damage response is attenuated by LPA. Thus, in addition to activating AKT/PKB and ERK pathways to promote cell survival, LPA can directly inhibit the proapoptotic function of Siva-1 through the specific binding of Siva-1 to the LPA2 receptor.

4. Concluding remarks

As different LPA receptors may couple to the same G proteins to activate the downstream cascades, it appears that the LPA2 receptor signaling can be achieved rapidly and specifically through these G protein-independent protein-protein interactions. It is of interest to note that these LPA2 receptor-interacting PDZ proteins and LIM-containing proteins possess similar functions in the association with actin, and serve as scaffold or adaptor proteins for the assembly of multi-protein complexes. In particular, TRIP6 and the structurally related LPP contain a class I PDZ-binding motif at their carboxyl-termini, allowing them to interact with PDZ proteins. The PDZ-containing NHERF2, PDZ-RhoGEF, LARG and MAGI-3 mediate LPA-induced activation of RhoA, ERKs and/or AKT, and the LIM-containing TRIP6 regulates LPA-induced ERK activation, adhesion turnover and cell migration in a c-Src-dependent manner. Thus, it is likely that the LPA2 receptor, PDZ proteins and LIM-containing proteins form a supramolecular complex in the microdomain of plasma membrane, and coordinately regulate LPA2 receptor functions in actin remodeling, cell adhesion, migration and/or cell survival. However, several questions remain to be answered, such as the physiological relevance of these protein-protein interactions and the downstream signaling events initiated by these interacting proteins. As the number of the identified LPA receptors is increasing, the LPA signaling pathways are more complex than were previously imagined. To specifically target one particular LPA receptor subtype for therapeutic intervention, it is prerequisite to understand how different interacting proteins regulate the specific signal transduction of each LPA receptor. It is also anticipated that more interacting partners of a particular subtype of LPA receptor will be identified in the near future.

Acknowledgments

We would like to acknowledge the multiple investigators who made important contributions to the studies of LPA2 mentioned in this article. This work was supported by the National Institute of Health Grant CA100848 (to F.-T. L).

References

  • 1.Contos JJ, Ishii I, Chun J. Lysophosphatidic acid receptors. Mol Pharmacol. 2000;58:1188–96. doi: 10.1124/mol.58.6.1188. [DOI] [PubMed] [Google Scholar]
  • 2.Moolenaar WH, van Meeteren LA, Giepmans BN. The ins and outs of lysophosphatidic acid signaling. Bioessays. 2004;26:870–81. doi: 10.1002/bies.20081. [DOI] [PubMed] [Google Scholar]
  • 3.Moolenaar WH. Bioactive lysophospholipids and their G protein-coupled receptors. Exp Cell Res. 1999;253:230–8. doi: 10.1006/excr.1999.4702. [DOI] [PubMed] [Google Scholar]
  • 4.Moolenaar WH. LPA: a novel lipid mediator with diverse biological actions. Trends Cell Biol. 1994;4:213–9. doi: 10.1016/0962-8924(94)90144-9. [DOI] [PubMed] [Google Scholar]
  • 5.Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer. 2003;3:582–91. doi: 10.1038/nrc1143. [DOI] [PubMed] [Google Scholar]
  • 6.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. G protein-coupled receptor P2Y5 and its ligand LPA are involved in maintenance of human hair growth. Nat Genet. 2008;40:329–34. doi: 10.1038/ng.84. [DOI] [PubMed] [Google Scholar]
  • 7.Hecht JH, Weiner JA, Post SR, Chun J. Ventricular zone gene-1 (vzg-1) encodes a lysophosphatidic acid receptor expressed in neurogenic regions of the developing cerebral cortex. J Cell Biol. 1996;135:1071–83. doi: 10.1083/jcb.135.4.1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.An S, Bleu T, Hallmark OG, Goetzl EJ. Characterization of a novel subtype of human G protein-coupled receptor for lysophosphatidic acid. J Biol Chem. 1998;273:7906–10. doi: 10.1074/jbc.273.14.7906. [DOI] [PubMed] [Google Scholar]
  • 9.Aoki J, Bandoh K, Inoue K. A novel human G-protein-coupled receptor, EDG7, for lysophosphatidic acid with unsaturated fatty-acid moiety. Ann N Y Acad Sci. 2000;905:263–6. doi: 10.1111/j.1749-6632.2000.tb06556.x. [DOI] [PubMed] [Google Scholar]
  • 10.Noguchi K, Ishii S, Shimizu T. Identification of p2y9/GPR23 as a novel G protein-coupled receptor for lysophosphatidic acid, structurally distant from the Edg family. J Biol Chem. 2003;278:25600–6. doi: 10.1074/jbc.M302648200. [DOI] [PubMed] [Google Scholar]
  • 11.Lee CW, Rivera R, Gardell S, Dubin AE, Chun J. GPR92 as a new G12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. J Biol Chem. 2006;281:23589–97. doi: 10.1074/jbc.M603670200. [DOI] [PubMed] [Google Scholar]
  • 12.Tabata K, Baba K, Shiraishi A, Ito M, Fujita N. The orphan GPCR GPR87 was deorphanized and shown to be a lysophosphatidic acid receptor. Biochem Biophys Res Commun. 2007;363:861–6. doi: 10.1016/j.bbrc.2007.09.063. [DOI] [PubMed] [Google Scholar]
  • 13.Zhang C, Baker DL, Yasuda S, Makarova N, Balazs L, Johnson LR, Marathe GK, McIntyre TM, Xu Y, Prestwich GD, Byun HS, Bittman R, Tigyi G. Lysophosphatidic acid induces neointima formation through PPARgamma activation. J Exp Med. 2004;199:763–74. doi: 10.1084/jem.20031619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.McIntyre TM, Pontsler AV, Silva AR, Hilaire A, Xu Y, Hinshaw JC, Zimmerman GA, Hama K, Aoki J, Arai H, Prestwich GD. Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARgamma agonist. Proc Natl Acad Sci U S A. 2003;100:131–6. doi: 10.1073/pnas.0135855100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Xu J, Lai YJ, Lin WC, Lin FT. TRIP6 enhances lysophosphatidic acid-induced cell migration by interacting with the lysophosphatidic acid 2 receptor. J Biol Chem. 2004;279:10459–68. doi: 10.1074/jbc.M311891200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhang H, Wang D, Sun H, Hall RA, Yun CC. MAGI-3 regulates LPA-induced activation of Erk and RhoA. Cell Signal. 2007;19:261–8. doi: 10.1016/j.cellsig.2006.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.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. NHERF2 specifically interacts with LPA2 receptor and defines the specificity and efficiency of receptor-mediated phospholipase C-beta3 activation. Mol Cell Biol. 2004;24:5069–79. doi: 10.1128/MCB.24.11.5069-5079.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yamada T, Ohoka Y, Kogo M, Inagaki S. Physical and functional interactions of the lysophosphatidic acid receptors with PDZ domain-containing Rho guanine nucleotide exchange factors (RhoGEFs) J Biol Chem. 2005;280:19358–63. doi: 10.1074/jbc.M414561200. [DOI] [PubMed] [Google Scholar]
  • 19.Lin FT, Lai YJ, Makarova N, Tigyi G, Lin WC. The Lysophosphatidic Acid 2 Receptor Mediates Down-regulation of Siva-1 to Promote Cell Survival. J Biol Chem. 2007;282:37759–69. doi: 10.1074/jbc.M705025200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Li C, Dandridge KS, Di A, Marrs KL, Harris EL, Roy K, Jackson JS, Makarova NV, Fujiwara Y, Farrar PL, Nelson DJ, Tigyi GJ, Naren AP. Lysophosphatidic acid inhibits cholera toxin-induced secretory diarrhea through CFTR-dependent protein interactions. J Exp Med. 2005;202:975–86. doi: 10.1084/jem.20050421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yun CC, Sun H, Wang D, Rusovici R, Castleberry A, Hall RA, Shim H. LPA2 receptor mediates mitogenic signals in human colon cancer cells. Am J Physiol Cell Physiol. 2005;289:C2–11. doi: 10.1152/ajpcell.00610.2004. [DOI] [PubMed] [Google Scholar]
  • 22.Contos JJ, Ishii I, Fukushima N, Kingsbury MA, Ye X, Kawamura S, Brown JH, Chun J. 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. 2002;22:6921–9. doi: 10.1128/MCB.22.19.6921-6929.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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. Aberrant expression of lysophosphatidic acid (LPA) receptors in human colorectal cancer. Lab Invest. 2004;84:1352–62. doi: 10.1038/labinvest.3700146. [DOI] [PubMed] [Google Scholar]
  • 24.Kitayama J, Shida D, Sako A, Ishikawa M, Hama K, Aoki J, Arai H, Nagawa H. Over-expression of lysophosphatidic acid receptor-2 in human invasive ductal carcinoma. Breast Cancer Res. 2004;6:R640–6. doi: 10.1186/bcr935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.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. Lysophospholipid growth factors in the initiation, progression, metastases, and management of ovarian cancer. Ann N Y Acad Sci. 2000;905:188–208. doi: 10.1111/j.1749-6632.2000.tb06550.x. [DOI] [PubMed] [Google Scholar]
  • 26.Schulte KM, Beyer A, Kohrer K, Oberhauser S, Roher HD. Lysophosphatidic 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. 2001;92:249–56. doi: 10.1002/1097-0215(200102)9999:9999<::aid-ijc1166>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
  • 27.Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, Takio K, Yamori T, Mills GB, Inoue K, Aoki J, Arai H. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J Cell Biol. 2002;158:227–33. doi: 10.1083/jcb.200204026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boucharaba A, Serre CM, Gres S, Saulnier-Blache JS, Bordet JC, Guglielmi J, Clezardin P, Peyruchaud O. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest. 2004;114:1714–25. doi: 10.1172/JCI22123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hur EM, Kim KT. G protein-coupled receptor signalling and cross-talk: achieving rapidity and specificity. Cell Signal. 2002;14:397–405. doi: 10.1016/s0898-6568(01)00258-3. [DOI] [PubMed] [Google Scholar]
  • 30.Maudsley S, Martin B, Luttrell LM. The origins of diversity and specificity in g protein-coupled receptor signaling. J Pharmacol Exp Ther. 2005;314:485–94. doi: 10.1124/jpet.105.083121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cho KO, Hunt CA, Kennedy MB. The rat brain postsynaptic density fraction contains a homolog of the Drosophila discs-large tumor suppressor protein. Neuron. 1992;9:929–42. doi: 10.1016/0896-6273(92)90245-9. [DOI] [PubMed] [Google Scholar]
  • 32.Kim E, Niethammer M, Rothschild A, Jan YN, Sheng M. Clustering of Shaker-type K+ channels by interaction with a family of membrane-associated guanylate kinases. Nature. 1995;378:85–8. doi: 10.1038/378085a0. [DOI] [PubMed] [Google Scholar]
  • 33.Fanning AS, Anderson JM. Protein-protein interactions: PDZ domain networks. Curr Biol. 1996;6:1385–8. doi: 10.1016/s0960-9822(96)00737-3. [DOI] [PubMed] [Google Scholar]
  • 34.Kornau HC, Schenker LT, Kennedy MB, Seeburg PH. Domain interaction between NMDA receptor subunits and the postsynaptic density protein PSD-95. Science. 1995;269:1737–40. doi: 10.1126/science.7569905. [DOI] [PubMed] [Google Scholar]
  • 35.Sato T, Irie S, Kitada S, Reed JC. FAP-1: a protein tyrosine phosphatase that associates with Fas. Science. 1995;268:411–5. doi: 10.1126/science.7536343. [DOI] [PubMed] [Google Scholar]
  • 36.Sheng M, Sala C. PDZ domains and the organization of supramolecular complexes. Annu Rev Neurosci. 2001;24:1–29. doi: 10.1146/annurev.neuro.24.1.1. [DOI] [PubMed] [Google Scholar]
  • 37.Weinman EJ, Shenolikar S. Regulation of the renal brush border membrane Na(+)-H+ exchanger. Annu Rev Physiol. 1993;55:289–304. doi: 10.1146/annurev.ph.55.030193.001445. [DOI] [PubMed] [Google Scholar]
  • 38.Shenolikar S, Weinman EJ. NHERF: targeting and trafficking membrane proteins. Am J Physiol Renal Physiol. 2001;280:F389–95. doi: 10.1152/ajprenal.2001.280.3.F389. [DOI] [PubMed] [Google Scholar]
  • 39.Weinman EJ, Shenolikar S. The Na-H exchanger regulatory factor. Exp Nephrol. 1997;5:449–52. [PubMed] [Google Scholar]
  • 40.Kleizen B, Braakman I, de Jonge HR. Regulated trafficking of the CFTR chloride channel. Eur J Cell Biol. 2000;79:544–56. doi: 10.1078/0171-9335-00078. [DOI] [PubMed] [Google Scholar]
  • 41.Weinman EJ, Hall RA, Friedman PA, Liu-Chen LY, Shenolikar S. The association of NHERF adaptor proteins with g protein-coupled receptors and receptor tyrosine kinases. Annu Rev Physiol. 2006;68:491–505. doi: 10.1146/annurev.physiol.68.040104.131050. [DOI] [PubMed] [Google Scholar]
  • 42.Hall RA, Ostedgaard LS, Premont RT, Blitzer JT, Rahman N, Welsh MJ, Lefkowitz RJ. A C-terminal motif found in the beta2-adrenergic receptor, P2Y1 receptor and cystic fibrosis transmembrane conductance regulator determines binding to the Na+/H+ exchanger regulatory factor family of PDZ proteins. Proc Natl Acad Sci U S A. 1998;95:8496–501. doi: 10.1073/pnas.95.15.8496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hall RA, Premont RT, Chow CW, Blitzer JT, Pitcher JA, Claing A, Stoffel RH, Barak LS, Shenolikar S, Weinman EJ, Grinstein S, Lefkowitz RJ. The beta2-adrenergic receptor interacts with the Na+/H+-exchanger regulatory factor to control Na+/H+ exchange. Nature. 1998;392:626–30. doi: 10.1038/33458. [DOI] [PubMed] [Google Scholar]
  • 44.Shenolikar S, Minkoff CM, Steplock DA, Evangelista C, Liu M, Weinman EJ. N-terminal PDZ domain is required for NHERF dimerization. FEBS Lett. 2001;489:233–6. doi: 10.1016/s0014-5793(01)02109-3. [DOI] [PubMed] [Google Scholar]
  • 45.Lau AG, Hall RA. Oligomerization of NHERF-1 and NHERF-2 PDZ domains: differential regulation by association with receptor carboxyl-termini and by phosphorylation. Biochemistry. 2001;40:8572–80. doi: 10.1021/bi0103516. [DOI] [PubMed] [Google Scholar]
  • 46.Guggino SE. Gates of Janus: cystic fibrosis and diarrhea. Trends Microbiol. 1994;2:91–4. doi: 10.1016/0966-842x(94)90541-x. [DOI] [PubMed] [Google Scholar]
  • 47.Vazquez-Prado J, Basile J, Gutkind JS. Modular architecture and novel protein-protein interactions regulating the RGS-containing Rho guanine nucleotide exchange factors. Methods Enzymol. 2004;390:259–85. doi: 10.1016/S0076-6879(04)90017-1. [DOI] [PubMed] [Google Scholar]
  • 48.Bian D, Mahanivong C, Yu J, Frisch SM, Pan ZK, Ye RD, Huang S. The G12/13-RhoA signaling pathway contributes to efficient lysophosphatidic acid-stimulated cell migration. Oncogene. 2006;25:2234–44. doi: 10.1038/sj.onc.1209261. [DOI] [PubMed] [Google Scholar]
  • 49.Dhanasekaran DN. Transducing the signals: a G protein takes a new identity. Sci STKE. 2006;2006:pe31. doi: 10.1126/stke.3472006pe31. [DOI] [PubMed] [Google Scholar]
  • 50.Gohla A, Harhammer R, Schultz G. The G-protein G13 but not G12 mediates signaling from lysophosphatidic acid receptor via epidermal growth factor receptor to Rho. J Biol Chem. 1998;273:4653–9. doi: 10.1074/jbc.273.8.4653. [DOI] [PubMed] [Google Scholar]
  • 51.Dobrosotskaya I, Guy RK, James GL. MAGI-1, a membrane-associated guanylate kinase with a unique arrangement of protein-protein interaction domains. J Biol Chem. 1997;272:31589–97. doi: 10.1074/jbc.272.50.31589. [DOI] [PubMed] [Google Scholar]
  • 52.Wu Y, Dowbenko D, Spencer S, Laura R, Lee J, Gu Q, Lasky LA. Interaction of the tumor suppressor PTEN/MMAC with a PDZ domain of MAGI3, a novel membrane-associated guanylate kinase. J Biol Chem. 2000;275:21477–85. doi: 10.1074/jbc.M909741199. [DOI] [PubMed] [Google Scholar]
  • 53.Adamsky K, Arnold K, Sabanay H, Peles E. Junctional protein MAGI-3 interacts with receptor tyrosine phosphatase beta (RPTP beta) and tyrosinephosphorylated proteins. J Cell Sci. 2003;116:1279–89. doi: 10.1242/jcs.00302. [DOI] [PubMed] [Google Scholar]
  • 54.Franklin JL, Yoshiura K, Dempsey PJ, Bogatcheva G, Jeyakumar L, Meise KS, Pearsall RS, Threadgill D, Coffey RJ. Identification of MAGI-3 as a transforming growth factor-alpha tail binding protein. Exp Cell Res. 2005;303:457–70. doi: 10.1016/j.yexcr.2004.10.007. [DOI] [PubMed] [Google Scholar]
  • 55.He J, Bellini M, Inuzuka H, Xu J, Xiong Y, Yang X, Castleberry AM, Hall RA. Proteomic analysis of beta1-adrenergic receptor interactions with PDZ scaffold proteins. J Biol Chem. 2006;281:2820–7. doi: 10.1074/jbc.M509503200. [DOI] [PubMed] [Google Scholar]
  • 56.Yao R, Natsume Y, Noda T. MAGI-3 is involved in the regulation of the JNK signaling pathway as a scaffold protein for frizzled and Ltap. Oncogene. 2004;23:6023–30. doi: 10.1038/sj.onc.1207817. [DOI] [PubMed] [Google Scholar]
  • 57.Bach I. The LIM domain: regulation by association. Mech Dev. 2000;91:5–17. doi: 10.1016/s0925-4773(99)00314-7. [DOI] [PubMed] [Google Scholar]
  • 58.Prasad KV, Ao Z, Yoon Y, Wu MX, Rizk M, Jacquot S, Schlossman SF. CD27, a member of the tumor necrosis factor receptor family, induces apoptosis and binds to Siva, a proapoptotic protein. Proc Natl Acad Sci U S A. 1997;94:6346–51. doi: 10.1073/pnas.94.12.6346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Y, Gilmore TD. Zyxin and paxillin proteins: focal adhesion plaque LIM domain proteins go nuclear. Biochim Biophys Acta. 2003;1593:115–20. doi: 10.1016/s0167-4889(02)00349-x. [DOI] [PubMed] [Google Scholar]
  • 60.Zhao MK, Wang Y, Murphy K, Yi J, Beckerle MC, Gilmore TD. LIM domain-containing protein trip6 can act as a coactivator for the v- Rel transcription factor. Gene Expr. 1999;8:207–17. [PMC free article] [PubMed] [Google Scholar]
  • 61.Kassel O, Schneider S, Heilbock C, Litfin M, Gottlicher M, Herrlich P. A nuclear isoform of the focal adhesion LIM-domain protein Trip6 integrates activating and repressing signals at AP-1- and NF-kappaB-regulated promoters. Genes Dev. 2004;18:2518–28. doi: 10.1101/gad.322404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yi J, Kloeker S, Jensen CC, Bockholt S, Honda H, Hirai H, Beckerle MC. Members of the Zyxin family of LIM proteins interact with members of the p130Cas family of signal transducers. J Biol Chem. 2002;277:9580–9. doi: 10.1074/jbc.M106922200. [DOI] [PubMed] [Google Scholar]
  • 63.Wang Y, Gilmore TD. LIM domain protein Trip6 has a conserved nuclear export signal, nuclear targeting sequences, and multiple transactivation domains. Biochim Biophys Acta. 2001;1538:260–72. doi: 10.1016/s0167-4889(01)00077-5. [DOI] [PubMed] [Google Scholar]
  • 64.Lai YJ, Chen CS, Lin WC, Lin FT. c-Src-mediated phosphorylation of TRIP6 regulates its function in lysophosphatidic acid-induced cell migration. Mol Cell Biol. 2005;25:5859–68. doi: 10.1128/MCB.25.14.5859-5868.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Murthy KK, Clark K, Fortin Y, Shen SH, Banville D. ZRP-1, a zyxin-related protein, interacts with the second PDZ domain of the cytosolic protein tyrosine phosphatase hPTP1E. J Biol Chem. 1999;274:20679–87. doi: 10.1074/jbc.274.29.20679. [DOI] [PubMed] [Google Scholar]
  • 66.Lai YJ, Lin WC, Lin FT. PTPL1/FAP-1 Negatively Regulates TRIP6 Function in Lysophosphatidic Acid-induced Cell Migration. J Biol Chem. 2007;282:24381–7. doi: 10.1074/jbc.M701499200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ye X, Ishii I, Kingsbury MA, Chun J. Lysophosphatidic acid as a novel cell survival/apoptotic factor. Biochim Biophys Acta. 2002;1585:108–13. doi: 10.1016/s1388-1981(02)00330-x. [DOI] [PubMed] [Google Scholar]
  • 68.Radeff-Huang J, Seasholtz TM, Matteo RG, Brown JH. G protein mediated signaling pathways in lysophospholipid induced cell proliferation and survival. J Cell Biochem. 2004;92:949–66. doi: 10.1002/jcb.20094. [DOI] [PubMed] [Google Scholar]
  • 69.Goetzl EJ, Lee H, Dolezalova H, Kalli KR, Conover CA, Hu YL, Azuma T, Stossel TP, Karliner JS, Jaffe RB. Mechanisms of lysolipid phosphate effects on cellular survival and proliferation. Ann N Y Acad Sci. 2000;905:177–87. doi: 10.1111/j.1749-6632.2000.tb06549.x. [DOI] [PubMed] [Google Scholar]
  • 70.Hu X, Haney N, Kropp D, Kabore AF, Johnston JB, Gibson SB. Lysophosphatidic acid (LPA) protects primary chronic lymphocytic leukemia cells from apoptosis through LPA receptor activation of the anti-apoptotic protein AKT/PKB. J Biol Chem. 2005;280:9498–508. doi: 10.1074/jbc.M410455200. [DOI] [PubMed] [Google Scholar]
  • 71.Virag T, Elrod DB, Liliom K, Sardar VM, Parrill AL, Yokoyama K, Durgam G, Deng W, Miller DD, Tigyi G. Fatty alcohol phosphates are subtype-selective agonists and antagonists of lysophosphatidic acid receptors. Mol Pharmacol. 2003;63:1032–42. doi: 10.1124/mol.63.5.1032. [DOI] [PubMed] [Google Scholar]
  • 72.Deng W, Shuyu E, Tsukahara R, Valentine WJ, Durgam G, Gududuru V, Balazs L, Manickam V, Arsura M, VanMiddlesworth L, Johnson LR, Parrill AL, Miller DD, Tigyi G. The lysophosphatidic acid type 2 receptor is required for protection against radiation-induced intestinal injury. Gastroenterology. 2007;132:1834–51. doi: 10.1053/j.gastro.2007.03.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fortin A, MacLaurin JG, Arbour N, Cregan SP, Kushwaha N, Callaghan SM, Park DS, Albert PR, Slack RS. The proapoptotic gene SIVA is a direct transcriptional target for the tumor suppressors p53 and E2F1. J Biol Chem. 2004;279:28706–14. doi: 10.1074/jbc.M400376200. [DOI] [PubMed] [Google Scholar]
  • 74.Py B, Slomianny C, Auberger P, Petit PX, Benichou S. Siva-1 and an alternative splice form lacking the death domain, Siva-2, similarly induce apoptosis in T lymphocytes via a caspase-dependent mitochondrial pathway. J Immunol. 2004;172:4008–17. doi: 10.4049/jimmunol.172.7.4008. [DOI] [PubMed] [Google Scholar]
  • 75.Gudi R, Barkinge J, Hawkins S, Chu F, Manicassamy S, Sun Z, Duke-Cohan JS, Prasad KV. Siva-1 negatively regulates NF-kappaB activity: effect on T-cell receptor-mediated activation-induced cell death (AICD) Oncogene. 2006;25:3458–62. doi: 10.1038/sj.onc.1209381. [DOI] [PubMed] [Google Scholar]
  • 76.Xue L, Chu F, Cheng Y, Sun X, Borthakur A, Ramarao M, Pandey P, Wu M, Schlossman SF, Prasad KV. Siva-1 binds to and inhibits BCL-X(L)-mediated protection against UV radiation-induced apoptosis. Proc Natl Acad Sci U S A. 2002;99:6925–30. doi: 10.1073/pnas.102182299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wojcikiewicz RJ. Regulated ubiquitination of proteins in GPCR-initiated signaling pathways. Trends Pharmacol Sci. 2004;25:35–41. doi: 10.1016/j.tips.2003.11.008. [DOI] [PubMed] [Google Scholar]

RESOURCES