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

Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and...

Date post: 07-Dec-2016
Category:
Upload: wouter
View: 214 times
Download: 0 times
Share this document with a friend
17
265 CHAPTER 13 Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology SUSAN S. SMYTH, ANPING DONG, JESSICA WHEELER, MANIKANDAN PANCHATCHARAM, and ANDREW J. MORRIS 13.1. INTRODUCTION Since the initial reports that lysophosphatidic acid (LPA) affected blood pres- sure in mammals (1, 2), a body of literature supports an important role for this bioactive lipid in regulating cardiovascular physiology and pathology. LPA exerts diverse effects on most blood and vascular cells. LPA circulates in plasma primarily bound to serum proteins and lipoprotein particles. However, whether circulating pools of LPA are the key determinant of biologic activity remain uncertain. In this chapter, the pathways for maintenance of plasma LPA are briefly reviewed; current understanding of blood and vascular cells responses to LPA are discussed; and evidence for a role in cardiovascular pathology, including a review of relevant methods, is presented. 13.2. CIRCULATING LPA LEVELS LPA is present in plasma at levels (100 nM) sufficient to elicit biologic effects on G protein-coupled receptors (3), although the contribution of plasma LPA to actions at cell surface receptors is not clear. The source for most extracellular, biologically active LPA is the secreted lysophospholipase D (lysoPLD) (4, 5) autotaxin, a member of the ectonucleotidase pyrophos- phate family encoded by Enpp2 and present in plasma at concentrations of 0.5–1 mg/L (approximately 5–10 μM) (6). Autotaxin generates LPA by 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 (LPA) Signaling and Cardiovascular Pathology

265

CHAPTER 13

Lysophosphatidic Acid (LPA) Signaling and Cardiovascular PathologySUSAN S. SMYTH, ANPING DONG, JESSICA WHEELER, MANIKANDAN PANCHATCHARAM, and ANDREW J. MORRIS

13.1. INTRODUCTION

Since the initial reports that lysophosphatidic acid (LPA) affected blood pres-sure in mammals (1, 2), a body of literature supports an important role for this bioactive lipid in regulating cardiovascular physiology and pathology. LPA exerts diverse effects on most blood and vascular cells. LPA circulates in plasma primarily bound to serum proteins and lipoprotein particles. However, whether circulating pools of LPA are the key determinant of biologic activity remain uncertain. In this chapter, the pathways for maintenance of plasma LPA are briefly reviewed; current understanding of blood and vascular cells responses to LPA are discussed; and evidence for a role in cardiovascular pathology, including a review of relevant methods, is presented.

13.2. CIRCULATING LPA LEVELS

LPA is present in plasma at levels (≥100 nM) sufficient to elicit biologic effects on G protein-coupled receptors (3), although the contribution of plasma LPA to actions at cell surface receptors is not clear. The source for most extracellular, biologically active LPA is the secreted lysophospholipase D (lysoPLD) (4, 5) autotaxin, a member of the ectonucleotidase pyrophos-phate family encoded by Enpp2 and present in plasma at concentrations of 0.5–1 mg/L (approximately 5–10 μM) (6). Autotaxin generates LPA by

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 (LPA) Signaling and Cardiovascular Pathology

266 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

hydrolysis of lysophosphatidylcholine (LPC), which is present in plasma at concentrations of approximately 100 μM. LPC is formed when lecithin-cholesterol acyltransferase (LCAT, also called phosphatidylcholine-sterol O-acyltransferase) converts free cholesterol into cholesteryl esters. However, LPA levels are normal in individuals lacking LCAT (7). Therefore, plasma LPA likely derives from LPC generated from phospholipase A (PLA) action on phosphatidylcholine (PC) present in lipoproteins, circulating microparti-cles, platelets, or other blood and vascular cells (7–9). In rodents, small-molecule autotaxin inhibitors and autotaxin antibodies lower LPA levels (10, 11), and mice genetically engineered to have reduced autotaxin expression also have lower LPA levels (12, 13). Plasma LPA may be generated in an autotaxin-independent manner by PLA2-mediated hydrolysis of phosphatidic acid (PA), although the PLA2 responsible has not been identified, and the contribution of this pathway to plasma LPA levels appears to be fairly small relative to that of autotaxin.

In addition to the requirement for autotaxin, blood platelets and platelet-derived microparticles have been implicated in plasma LPA generation. LPA levels are reduced in rats by antibody-mediated thrombocytopenia or treat-ment of mice with a platelet integrin inhibitor that also induces thrombocyto-penia (7, 14). Platelet-derived microparticles have also been cited as a source for the generation of LPA (9). Isolated, washed platelets produce relatively small amounts of LPA upon agonist stimulation (15, 16), and LPA production by isolated, activated platelets is prevented by autotaxin inhibitors (17). One explanation for these observations is that activated platelets, and potentially their microparticles, provide an LPC source for the generation of LPA by autotaxin. In keeping with this, Tigyi and colleagues recently purified a PLA1 from thrombin-activated platelets that increases the production of LPC and LPA in plasma (8). They identified the activity as acyl-protein thioesterase 1 (APT1, also called lysophospholipase A-I [LYPLA-I]) and proposed that sn-2 LPC generated by APT1 undergoes acyl migration to sn-1 LPC, which serves as a substrate for autotaxin (8).

The working model for platelet LPA production, in which PC is converted to LPC by APT1 and then to LPA by autotaxin, necessitates that autotaxin interacts with cell surfaces. How autotaxin accesses LPC generated during platelet activation and whether LPA is generated along the membrane bilayer or released freely or both are not known. The crystal structure of autotaxin reveals a hydrophobic substrate binding channel leading to the active site (18, 19). The channel is formed at the interface of the somatomedin B-like (SMB) and phosphodiesterase (PDE) domains of the protein and may provide a means to spatially restrict the generation and delivery of LPA. Autotaxin can localize to cell surfaces by binding to activated integrins, as has been observed with platelets (20), lymphocytes (21), and oligodendrocytes (22). The interac-tion of the SMB domain of autotaxin with activated integrins on platelets and other cells may alter autotaxin activity and enable localized production of cell-associated LPA.

Page 3: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

LPA SIGNALING IN BLOOD AND VASCULAR CELLS 267

13.2.1. Protocols for LPA Measurement in Plasma

Whole blood should be collected into an anticoagulant solution of EDTA (10 mM) and citrate-theophylline-adenoisne-dipyridamole (CTAD; 10% v/v, BD Biosciences, San Jose, CA). An autotaxin inhibitor can be added to prevent ex vivo LPA production during sample processing. We typically spin the blood at 12,000 g × 5 minutes and immediately freeze the resultant plasma. This protocol results in minimal platelet activation as judged by plasma platelet factor 4 levels. LPA circulates in plasma primarily bound to serum proteins and lipoprotein particles. The distribution of LPA can be monitored by size-exclusion fractionation of plasma. Typically, plasma is chromatographed on a Superdex 75 or similar column (Pharmacia, Kalamazoo, MI). LPA content in fractions is measured by high-performance liquid chromatography (HPLC)/electrospray ionization (ESI)/tandem mass spectrometry and lipoprotein frac-tions identified by immunoblot analysis or cholesterol content.

Studies in which autotaxin inhibitors or LPA is administered to animals indicate rapid metabolism of LPA occurs in circulation and whole blood, suggesting dynamic regulation by synthetic and metabolic/clearance path-ways (11, 23). At present, the mechanism(s) responsible for LPA metabolism and clearance in blood are not known, although a role for the lipid phosphate phosphatase 1 (LPP1) has been suggested (23). Unlike the rapidly metabo-lism observed in whole blood, plasma LPA is relatively stable. Therefore, measuring LPA accumulation in plasma incubated at 37°C for 60–120 minutes under conditions that support autotaxin activity is a simple and reliable method to monitor enzyme activity in plasma. Small-molecule inhibitors can be included in the assay to confirm the autotaxin-dependent nature of LPA production.

13.3. LPA SIGNALING IN BLOOD AND VASCULAR CELLS

Three receptor systems have been implicated in mediating LPA’s signaling effects on cells. These are a family of G protein-coupled receptors (24), peroxi-some proliferator-activating receptor γ (PPARγ) (25), and, more recently, the receptor for advanced glycation end products (RAGE) (26). At least six LPA G protein-coupled receptors, termed LPA1–6, may mediate responses to extracellular LPA. These LPA receptors couple to multiple heterotrimeric G proteins (Gi/o, Gq/11/14, Gs, and G12/13) and initiate various signal transduction pathways. In the vasculature, G13-mediated activation of Rho appears to play an especially prominent role. The contribution of LPA receptors to signaling in blood and vascular cells is described in more detail below. The nuclear receptor PPARγ, which is important in blood and vascular cell signaling, has been proposed as a receptor for LPA (25, 27). This pathway may be acti-vated by intracellular LPA generated by the glycerol-3-phosphate pathway

Page 4: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

268 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

because cells overexpressing glycerol-3-phosphate acyltransferase-1 have sixfold elevated levels of intracellular LPA and higher PPARγ activity, whereas the addition of extracellular LPA lacked effect on PPARγ (28). These findings suggest an alternate role in “transcellular” LPA signaling. Recently, RAGE has been implicated as a potential mediator of LPA signaling in the vascula-ture, and the absence of RAGE in mice resulted in a loss of vascular LPA signaling (26). The ability of LPA to bind with high affinity to soluble RAGE fragments suggests a direct role for the receptor in LPA signaling, although this remains to be firmly established.

13.4. REGULATION OF BLOOD PRESSURE

Exogenous administration of LPA alters blood pressure in animals. Intrave-nous injection of LPA transiently elevates arterial blood pressure in mice (29) and rats (2). Local application to porcine cerebral vessels elicits vasoconstric-tion (1). The receptors mediating this response are not known. In response to intravenously administered LPA, we have observed an increase in mean arte-rial pressure in mice lacking LPA1, LPA2, both LPA1 and LPA2 (29), LPA4, and PPARγ in smooth muscle cells (SMC) (unpublished observations), and others have reported that the elevated blood pressure occurs in Lpar3–/– mice.

13.4.1. Protocol for Arterial Pressure Measurements

Systolic blood pressure and heart rate can be measured noninvasively in con-scious mice by tail cuff analysis using methods that either detect cuff pressure (BP-2000, Visitech, Apex, NC) or volume pressure recordings (CODA, Kent Scientific, Torrington, CT). The mice are acclimated to the procedure by daily training for 1 week, and recordings are then made for five consecutive days. To measure acute changes in blood pressure in response to intravenous admin-istration of LPA, mice are anesthetized with isoflurane, placed on a mouse pad that has imbedded EKG electrodes and surface mounted semiconductor tem-perature sensor to distribute heat, and a pressure catheter (1.4 Fr Millar [Houston, TX] catheter) is introduced into the carotid artery. A data acquisi-tion and recording system (PowerLab with LabChart, ADInstruments, Colo-rado Springs, CO) is used to display readings in real time. When injected intravenously into either the jugular or the femoral vein, LPA elicits a tran-sient increase in mean arterial pressure that begins within ≈15 seconds and begins to return to baseline at ≈1 minute and is accompanied by a reflex bra-dycardia. The “gold standard” for blood pressure recordings in rodents is implantation of telemetric pressure transmitters, such as those supplied by Data Sciences International (St. Paul, MN) that record arterial pressure for prolonged periods of time in conscious, free-roaming animals. The drawbacks of implantable telemetric units include requirements for advanced skills in rodent survival surgery and expense of the monitoring equipment.

Page 5: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

VASCULAR PERMEABILITY 269

13.5. BLOOD VESSEL AND LYMPHATIC FORMATION

The most compelling evidence for the role of LPA in blood vessel formation comes from observations in mice with inherited absence of either autotaxin or LPA receptors. The lack of autotaxin results in embryonic lethality in mice due in part to improper vasculogenesis (12, 13). Vascular endothelial growth factor (VEGF) increases autotaxin expression in angiogenic endothelial cells in a phospholipase C γ (PLCγ)-dependent manner, and autotaxin may regulate endothelial tube remodeling (30). When mixed in Matrigel (BD Biosciences) and implanted subcutaneously, autotaxin elicits angiogenesis in mice (31). Although the mechanisms responsible are not fully understood, in cultured endothelial cells, LPA activates protein kinase D (PKD) and downregulates CD36, a receptor for the antiangiogenic protein thrombospondin (32). Attenu-ated expression of CD36 may blunt the inhibitory effects of thrombospondin and allow angiogenesis to proceed unchecked. LPA promotes endothelial migration largely through Rho-dependent processes that alter the actin cyto-skeleton and extracellular matrix composition (32). Genetic deficiency of the LPA4 receptor, which couples to G13 (33, 34), causes edema and hemorrhage in a subset of embryo and resultant lethality (35). In adult Lpar4-null mice, fewer endothelial-lined vessels form in Matrigel implants, impairments in lym-phatics are also present in Lpar4-null mice, and a role for LPA in lymphangio-genesis has been supported in in vitro systems (36). Thus, autotaxin and LPA via LPA4, and potentially other receptor systems, may regulate both vascular and lympathic vessel formation (35).

13.5.1. Protocol for Matrigel Angiogenesis Assay

Anesthetized mice are shaved to expose the skin on both flanks. Matrigel (200–250 μL or sterile, reduced-growth-factor Matrigel at 10 mg/mL) prepared with purified autotaxin (0.1–1 nM) (31) or basic fibroblast growth factor (bFGF; 250 ng/mL) and heparin (60 U/mL) are injected subcutaneously on the left center of the back and on the right center of the back using a 27 G needle and tuberculin syringe. Typically, the Matrigel plug on one side is used as the matched control for the other (e.g., catalytically inactive autotaxin or heparin only). The mice should be singly housed until fully recovered and the Matrigel plug has solidified. Vessel growth in the plug occurs within 7–14 days and can be visualized by intravenously injecting 200 μL of FITC-Dextran, (50 mg/mL, 2 × 106 m.w., Sigma, St. Louis, MO), either through a lateral tail vein or an exposed jugular vein. Mice are euthanized 10 minutes later. Vessels can also be visualized by immunohistochemical visualization of in the Matrigel implants using antibodies to endothelial and SMC-specific antigens.

13.6. VASCULAR PERMEABILITY

While studies in cultured endothelial cells have yielded variable results with regard to the effects of LPA on endothelial barrier function, in a bleomycin

Page 6: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

270 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

model of lung injury, mice lacking LPA1 display reduced fibrosis and vascular leak (37). In this injury model, bleomycin increases LPA levels in alveolar fluid. The protection from lung edema observed in the LPA1-deficient mice could reflect a pathologic role of LPA in promoting endothelial permeability. Some, but not all, studies of isolated endothelial cells support a model in which LPA disrupts endothelial barrier function (see References 38–41; reviewed in Ref-erence 42). In support of barrier destabilizing effects of LPA, topical applica-tion provokes cerebromicrovascular permeability in mice (43). Interestingly, in the bleomycin model, lack of LPA1 did not influence recruitment of inflam-matory cells into damaged lungs, which might be expected to be influenced by endothelial barrier function.

13.6.1. Vascular Permeability Protocol

Evans blue dye (200 μL of a 1% solution in phosphate-buffered saline [PBS]) is injected intravenously. Fifteen minutes later, mice are euthanized and per-fused with PBS containing heparin through the right ventricle at a constant rate to yield a pressure of 25 mmHg. The lungs are dissected, weighed, and the entire right lung scanned using an Odyssey Infrared Imaging System 2.1 (LI-COR Inc., Lincoln, NE) to detect Evans blue. As an alternative method to detect Evan blue, the lung can be immersed in 1 mL 4% formamide and Evans blue extracted by shaking in a water bath at 56°C for 24 hours and quantitated by absorbance at 600 nm. The values for Evans blue are normalized to lung weight. It is important to obtain measurements of both wet and dry lung weight. Vascular leak can be provoked with an inflammatory challenge, for example, bacterial LPS (2 mg/kg, E. coli 0111:B4; Sigma) administered intra-peritoneally 4–6 hours before Evans blue.

13.7. VASCULAR INFLAMMATION

LPA may promote inflammatory responses either indirectly through endothe-lial cells or by direct effects on leukocytes. In addition to altering endothelial barrier properties, LPA may “inflame” endothelial cells by upregulating the expression of endothelial chemoattractants (21, 44, 45), stimulating the secre-tion of the pentraxin-3, an acute phase reactant with both proinflammatory and prothrombotic features (46), and triggering exposure of endothelial adhe-sion receptors (47). The endothelial effects of LPA are mediated by both LPA1 and LPA3 and likely promote endothelial–white blood cell interactions in vivo. Autotaxin expression is upregulated in monocytic-like cells following LPS exposure (48), and localized autotaxin activity regulates lymphocyte traffick-ing (21). It is therefore reasonable to speculate that the generation of LPA during inflammation may have direct cellular consequences. Neutrophils (49, 50), eosinophils (51), and mononuclear phagocytes (48, 52) are responsive to LPA, and LPA triggers platelet–monocyte coaggregate formation in whole

Page 7: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

ATHEROTHROMBOSIS 271

blood (53). LPA also elicits the release of IL-6 from SMCs (54). In an air pouch model, LPA recruits both neutrophils and monocytes, triggers the release of IL-6 and KC, and enhances TNFα effects in an LPA1- and LPA3-dependent manner (55). Together, these results focus attention on LPA1 and LPA3 as potentially important mediators of vascular inflammation.

13.7.1. Inflammation Models

Two classic experimental models of acute inflammation are peritonitis and dermatitis. Peritonitis is induced by intraperitoneal injection of 1 mL of thio-glycollate (3% w/v). The mice are euthanized at 6 hours after the injection, at which time peritoneal fluid is collected to determine the number of leukocytes that transmigrated in response to the thioglycollate. Dermatitis is induced by topical application of croton oil (10 μL of 2% solution in 4:1 acetone : olive oil). After 6 hours, the mice are euthanized and the tissue removed and fixed in paraformaldehyde for subsequent histologic assessment, which includes leu-kocyte accumulation. To our knowledge, the effects of LPA in eliciting perito-nitis or dermatitis has not been examined. However, as discussed above, LPA does trigger neutrophil and monocyte migration into air pouches (55). In the air pouch model, 3 mL of sterile air is injected subcutaneously twice, 72 hours apart. Four days after the second air injection, 1 mL of PBS containing 0.1% bovine serum albumin and 1–6 μg of LPA (2.3–13.9 μM) is introduced into the pouch. The pouch is flushed with sterile buffer 6 hours later and leukocytes collected.

13.8. ATHEROTHROMBOSIS

Atherosclerosis is a chronic inflammatory condition with the hallmark features of endothelial dysfunction, lipid accumulation, vascular inflammation, and a fibroproliferative response of resident SMCs. The complications of atheroscle-rosis occur either as a consequence of tissue ischemia resulting from obstruc-tions in blood flow by the atheromatous plaque or when plaque ruptures or erodes and causes platelet-dependent thrombus formation, the proximate cause of most heart attacks and many strokes. LPA is present in human ath-eroma, particularly enriched in the lipid-rich core, and levels increase in exper-imental mouse models during progression of atherosclerosis (56, 57). Advanced lesions in Ldlr–/– mice, generated by a combination of diet and collar placement around the carotid artery, contain ≈20-fold higher levels of LPA than uninjured vessels, especially highly unsaturated long-chain acyl-LPA species (58). Several pathways may result in accumulation of LPA in atherosclerotic plaque. LPA is produced during mild oxidation of low-density lipoprotein (LDL) (56) and may also be generated from LPC, which markedly increases in atherosclerotic/injured arteries of Ldlr–/– mice (58). Within days to weeks of feeding male New Zealand white rabbits a diet with 1% cholesterol, serum LPC levels increase.

Page 8: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

272 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

The increase in LPC is accompanied by heightened generation of biologically active LPA in serum incubated ex vivo for 24 hours, presumably as a conse-quence of the lysoPLD actions of autotaxin (59).

LPA may serve a prothrombotic function based on its ability to elicit pro-coagulant phosphatidylserine exposure on erythrocytes (60), its platelet-activating effects (61), and its stimulation of tissue factor expression on SMCs (62). These observations have led to the speculation that LPA is an important prothrombotic component of atherosclerotic plaque (56, 63, 64). LPA’s effects on platelets are primarily through G13-dependent signaling, resulting in shape change, fibronectin matrix assembly, and potentiation of other platelet ago-nists (65–68). The platelet LPA response demonstrates an interesting pharma-cology, with alkyl glycerol phosphate (“1-alkyl-LPA”) being a more potent agonist than LPA (69), a pharmacologic profile shared by LPA5 (70). While human platelets and megakaryocytes contain mRNA for several LPA recep-tors, transcript levels for LPAR5 are among the highest for GPCRs in human platelets (71). RNAi targeting of LPAR5 in megakaryocyte lines prevents shape change in response to both LPA and to lipid extracts from atheroscle-rotic plaque (72). Taken together, these observations implicate LPA5 as the LPA-activating receptor on human platelets. Confirmation of LPA5 as the major platelet LPA receptor by use of genetic approaches in mice is ham-pered by the observation that LPA does not elicit a similar stimulatory effect on murine platelets (20), which, interestingly, lack expression of Lpar5 tran-scripts by RNA-seq analysis (73).

Phenotypic modulation of SMCs occurs in response to vascular injury and is a critical component in the development of intimal hyperplasia, a defining feature of atherosclerotic and restenotic lesions (74, 75). Phenotypic modula-tion involves the dedifferentiation, proliferation, and migration of normally quiescent SMC in the vessel wall and takes place when isolated SMCs are cultured in serum. As has been reviewed elsewhere in detail (76), LPA pro-motes all aspects of the phenotypic response in SMCs, including dedifferentia-tion (27, 54, 77–79), proliferation (80–82), and migration (83–87), and appears to be a key component of serum responsible for phenotypic modulation of SMCs (78, 88). LPA also triggers inflammatory and prothrombotic phenotypes in SMCs by upregulating expression of mediators such as IL-6 (54) and tissue factor (62). Thus, the effects of LPA on cultured SMCs would support a caus-ative role for the lipid in the development of intimal hyperplasia in the settings of atherosclerosis and restenosis. Such a role is supported by the discovery that exogenous application of LPA stimulates neointimal formation in both rat and mouse arteries (27, 77, 89). The process does not occur in the presence of an antagonist to PPARγ or in mice lacking PPARγ, although it occurs normally in mice deficient in Lpar1 and Lpar2 (89). In contrast, the application of genetic and pharmacologic tools in mice has suggested roles for LPA1 and LPA3 in vascular remodeling elicited by denudation injury (90) and flow-induced injury (29). The development of intimal hyperplasia likely results from effects of LPA on a combination of the processes described above, including recruitment of

Page 9: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

ATHEROTHROMBOSIS 273

inflammatory and progenitor cells and stimulation of resident SMCs. In par-ticular, LPA may regulate the ability of CXCL12/SDF to recruit smooth muscle progenitor cells to sites of injury (90). As similar events also occur in athero-sclerotic lesions, these results position LPA as a potential mediator of the development of atherosclerosis. Of particular interest from the standpoint of understanding its proatherosclerotic effects, LPA causes reverse transmigra-tion and neutral lipid uptake in monocyte cells (91). Thus, the accumulation of LPA in atherosclerotic lesions could limit macrophage egress during plaque progression. Indeed, treatment of mice with LPA promotes monocyte adhesion to the endothelium, stimulates perivascular macrophage accumulation, and heightens atherosclerotic plaque burden in Apoe–/– mice in an LPA1- and LPA3-dependent manner (44). Minimally oxidized LDL also triggers monocyte adhe-sion to the endothelium and requires the activity of autotaxin, suggesting that LPC in the minimally oxidized LDL may be converted to LPA along the surface of endothelial cells to mediate proatherosclerotic effects. Finally, a pharmacologic inhibitor of LPA1 and LPA3 reduces the development of ath-erosclerosis in Western diet-fed Apoe–/– mice (44).

13.8.1. Experimental Models for Studying the Development of Intimal Hyperplasia and Atherosclerosis

LPA can be infused in the carotid artery of rats or mice by clamping the origin of the vessel and inserting a PE10 catheter in the external common carotid artery of an anesthetized animal. After washing with buffer, the artery is filled with buffer containing LPA (2.5 μM) for 1 hour at which time the external carotid artery is ligated and the common carotid allowed to reperfuse with circulating blood. At various times after the injury, the common carotid can be isolated for histologic and biochemical analysis. To denude the endothelium from the femoral artery, the mouse is anesthetized with inhaled isoflurane and placed in supine position to expose the femoral vessels near the inguinal liga-ment and distal to the epigastric artery. The distal portion of the femoral artery is encircled with a 9-0 nylon suture; a vascular clamp is placed proximally at the level of the inguinal ligament; and a 0.010-in. (0.25-mm) diameter angio-plasty guidewire (Abbott Vascular, Santa Clara, CA) is introduced into the arterial lumen through an arteriotomy made just distal to the suture. After release of the clamp, the guidewire is advanced to the level of the aortic bifur-cation and immediately pulled back; this process is repeated two additional times to denude the endothelium. Experimental atherosclerosis is typically examined in mice on a proatherosclerotic background of either Apoe–/– or Ldlr–/–. The relative advantages of each of these strains has been previously discussed (92). Typically, mice are placed on a Western diet (D12079B, Research Diets, New Brunswick, NJ) for periods of up to 3 months. Atherosclerotic lesions are evaluated by en face staining of the aorta with Oil Red O or by histologic analysis of serial sections taken through the aorta at the level of the aortic valve or of sections from the brachiocephalic artery.

Page 10: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

274 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

13.9. FUTURE DIRECTIONS

From experimental observations made in isolated cells and animal models, a picture emerges in which the bioactive lipid LPA is poised as a key modulator of a variety of pathologic responses in blood and vascular cells. In particular, LPA may serve as a critical nexus between proinflammatory and prothrom-botic events in the vasculature. However, a causative role for LPA in the disease processes, including the development and complications of atheroscle-rosis, remains to be established. To date, the strongest evidence for a role for LPA comes from studies in which exogenous, superphysiologic concentrations of LPA have been provided to cells or administered to animals. Establishing a role for endogenous LPA generated in blood or vessels in response to injury or inflammation as a causative factor in disease progression will require the use of experimental therapeutics and genetic manipulations in animals. The recent advances in understanding LPA receptor biology and the generation of small-molecule drugs that inhibit LPA production or antagonize its signaling effects will undoubtedly translate into improved insight of its role in the car-diovasculature. Ultimately, findings in experimental animal models will require confirmation in humans to determine if pharmacologic targeting of LPA sig-naling improves clinical outcomes or if LPA signaling pathways in individuals predict complications and/or response to therapy.

ACKNOWLEDGMENTS

This research was supported by grants from the NIH (to S.S.S. and A.J.M), a VA Merit Award (S.S.S.), and a scientist development grant from the Ameri-can Heart Association (M.P.). This material is the result of work supported with the resources and use of the facilities at the Lexington VA Medical Center.

REFERENCES

1. Tigyi G, Hong L, Yakubu M, Parfenova H, Shibata M, Leffler CW. 1995. Lysophos-phatidic acid alters cerebrovascular reactivity in piglets. Am J Physiol 268:H2048–H2055.

2. Tokumura A, Fukuzawa K, Tsukatani H. 1978. Effects of synthetic and natural lysophosphatidic acids on the arterial blood pressure of different animal species. Lipids 13:572–574.

3. Morris AJ, Selim S, Salous A, Smyth SS. 2009. Blood relatives: dynamic regulation of bioactive lysophosphatidic acid and sphingosine-1-phosphate metabolism in the circulation. Trends Cardiovasc Med 19:135–140.

4. Nakanaga K, Hama K, Aoki J. 2010. Autotaxin—an LPA producing enzyme with diverse functions. J Biochem 148:13–24.

Page 11: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

REfERENCES 275

5. van Meeteren LA, Moolenaar WH. 2007. Regulation and biological activities of the autotaxin-LPA axis. Prog Lipid Res 46:145–160.

6. 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, Kuro-kawa M, Aoki J, Yatomi Y. 2008. Serum autotaxin measurement in haematological malignancies: a promising marker for follicular lymphoma. Br J Haematol 143:60–70.

7. 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.

8. Bolen AL, Naren AP, Yarlagadda S, Beranova-Giorgianni S, Chen L, Norman D, Baker DL, Rowland MM, Best MD, Sano T, Tsukahara T, Liliom K, Igarashi Y, Tigyi G. 2011. The phospholipase A1 activity of lysophospholipase A-I links platelet activation to LPA production during blood coagulation. J Lipid Res 52:958–970.

9. Fourcade O, Simon MF, Viode C, Rugani N, Leballe F, Ragab A, Fournie B, Sarda L, Chap H. 1995. Secretory phospholipase A2 generates the novel lipid mediator lysophosphatidic acid in membrane microvesicles shed from activated cells. Cell 80:919–927.

10. Gierse J, Thorarensen A, Beltey K, Bradshaw-Pierce E, Cortes-Burgos L, Hall T, Johnston A, Murphy M, Nemirovskiy O, Ogawa S, Pegg L, Pelc M, Prinsen M, Schnute M, Wendling J, Wene S, Weinberg R, Wittwer A, Zweifel B, Masferrer J. 2010. A novel autotaxin inhibitor reduces lysophosphatidic acid levels in plasma and the site of inflammation. J Pharmacol Exp Ther 334:310–317.

11. Albers HM, Dong A, van Meeteren LA, Egan DA, Sunkara M, van Tilburg EW, Schuurman K, van Tellingen O, Morris AJ, Smyth SS, Moolenaar WH, Ovaa H. 2010. Boronic acid-based inhibitor of autotaxin reveals rapid turnover of LPA in the circulation. Proc Natl Acad Sci U S A 107:7257–7262.

12. 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.

13. 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.

14. 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.

15. Eichholtz T, Jalink K, Fahrenfort I, Moolenaar WH. 1993. The bioactive phospho-lipid lysophosphatidic acid is released from activated platelets. Biochem J 291(Pt 3):677–680.

16. Gerrard JM, Robinson P. 1989. Identification of the molecular species of lysophos-phatidic acid produced when platelets are stimulated by thrombin. Biochim Biophys Acta 1001:282–285.

17. Fulkerson Z, Wu T, Sunkara M, Kooi CV, Morris AJ, Smyth SS. 2011. Binding of autotaxin to integrins localizes lysophosphatidic acid production to platelets and mammalian cells. J Biol Chem 286:34654–34663.

Page 12: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

276 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

18. Hausmann J, Kamtekar S, Christodoulou E, Day JE, Wu T, Fulkerson Z, Albers HM, van Meeteren LA, Houben AJ, van Zeijl L, Jansen S, Andries M, Hall T, Pegg LE, Benson TE, Kasiem M, Harlos K, Kooi CW, Smyth SS, Ovaa H, Bollen M, Morris AJ, Moolenaar WH, Perrakis A. 2011. Structural basis of substrate discrimi-nation and integrin binding by autotaxin. Nat Struct Mol Biol 18:198–204.

19. Nishimasu H, Okudaira S, Hama K, Mihara E, Dohmae N, Inoue A, Ishitani R, Takagi J, Aoki J, Nureki O. 2011. Crystal structure of autotaxin and insight into GPCR activation by lipid mediators. Nat Struct Mol Biol 18:205–212.

20. Pamuklar Z, Federico L, Liu S, Umezu-Goto M, Dong A, Panchatcharam M, Fulk-erson Z, Berdyshev E, Natarajan V, Fang X, van Meeteren LA, Moolenaar WH, Mills GB, Morris AJ, Smyth SS. 2009. Autotaxin/lysopholipase D and lysophospha-tidic acid regulate murine hemostasis and thrombosis. J Biol Chem 284:7385–7394.

21. Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. 2008. Autotaxin, an ectoenzyme that produces lysophosphatidic acid, promotes the entry of lympho-cytes into secondary lymphoid organs. Nat Immunol 9:415–423.

22. Fox MA, Alexander JK, Afshari FS, Colello RJ, Fuss B. 2004. Phosphodiesterase-I alpha/autotaxin controls cytoskeletal organization and FAK phosphorylation during myelination. Mol Cell Neurosci 27:140–150.

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

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

25. McIntyre TM, Pontsler AV, Silva AR, St Hilaire A, Xu Y, Hinshaw JC, Zimmerman GA, Hama K, Aoki J, Arai H, Prestwich GD. 2003. Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARgamma agonist. Proc Natl Acad Sci U S A 100:131–136.

26. Rai VTF, Chitayat S, Pei R, Lu Y, Li Q, Rosario R, Zhu Z, Qu W, Song F, Chazin W, Schmidt AM. 2011. Lysophosphatidic acid mediates vascular signaling via an immunoglobulin superfamily receptor. Arterioscler Thromb Vasc Biol 32:P188.

27. 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. 2004. Lysophos-phatidic acid induces neointima formation through PPARgamma activation. J Exp Med 199:763–774.

28. Stapleton CM, Mashek DG, Wang S, Nagle CA, Cline GW, Thuillier P, Leesnitzer LM, Li LO, Stimmel JB, Shulman GI, Coleman RA. 2011. Lysophosphatidic acid activates peroxisome proliferator activated receptor-gamma in CHO cells that over-express glycerol 3-phosphate acyltransferase-1. PLoS ONE 6:e18932.

29. Panchatcharam M, Miriyala S, Yang F, Rojas M, End C, Vallant C, Dong A, Lynch K, Chun J, Morris AJ, Smyth SS. 2008. Lysophosphatidic acid receptors 1 and 2 play roles in regulation of vascular injury responses but not blood pressure. Circ Res 103:662–670.

30. Sofat R, Hingorani AD, Smeeth L, Humphries SE, Talmud PJ, Cooper J, Shah T, Sandhu MS, Ricketts SL, Boekholdt SM, Wareham N, Khaw KT, Kumari M, Kivi-maki M, Marmot M, Asselbergs FW, van der Harst P, Dullaart RP, Navis G, van

Page 13: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

REfERENCES 277

Veldhuisen DJ, Van Gilst WH, Thompson JF, McCaskie P, Palmer LJ, Arca M, Quagliarini F, Gaudio C, Cambien F, Nicaud V, Poirer O, Gudnason V, Isaacs A, Witteman JC, van Duijn CM, Pencina M, Vasan RS, D’Agostino RB, Sr., Ordovas J, Li TY, Kakko S, Kauma H, Savolainen MJ, Kesaniemi YA, Sandhofer A, Paulwe-ber B, Sorli JV, Goto A, Yokoyama S, Okumura K, Horne BD, Packard C, Freeman D, Ford I, Sattar N, McCormack V, Lawlor DA, Ebrahim S, Smith GD, Kastelein JJ, Deanfield J, Casas JP. 2010. Separating the mechanism-based and off-target actions of cholesteryl ester transfer protein inhibitors with CETP gene polymor-phisms. Circulation 121:52–62.

31. Nam SW, Clair T, Kim YS, McMarlin A, Schiffmann E, Liotta LA, Stracke ML. 2001. Autotaxin (NPP-2), a metastasis-enhancing motogen, is an angiogenic factor. Cancer Res 61:6938–6944.

32. Ren B, Hale J, Srikanthan S, Silverstein RL. 2011. Lysophosphatidic acid suppresses endothelial cell CD36 expression and promotes angiogenesis via a PKD-1 depen-dent signaling pathway. Blood 117:6036–6045.

33. 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.

34. Yanagida K, Ishii S, Hamano F, Noguchi K, Shimizu T. 2007. LPA4/p2y9/GPR23 mediates rho-dependent morphological changes in a rat neuronal cell line. J Biol Chem 282:5814–5824.

35. 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.

36. Lin CH, Lu J, Lee H. 2011. Interleukin-1beta expression is required for lysophos-phatidic Acid-induced lymphangiogenesis in human umbilical vein endothelial cells. Int J Inflamm 2011:351010.

37. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, Polosukhin 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 vascular leak. Nat Med 14:45–54.

38. Neidlinger NA, Larkin SK, Bhagat A, Victorino GP, Kuypers FA. 2006. Hydrolysis of phosphatidylserine-exposing red blood cells by secretory phospholipase A2 generates lysophosphatidic acid and results in vascular dysfunction. J Biol Chem 281:775–781.

39. Alexander JS, Patton WF, Christman BW, Cuiper LL, Haselton FR. 1998. Platelet-derived lysophosphatidic acid decreases endothelial permeability in vitro. Am J Physiol 274:H115–H122.

40. English D, Kovala AT, Welch Z, Harvey KA, Siddiqui RA, Brindley DN, Garcia JG. 1999. Induction of endothelial cell chemotaxis by sphingosine 1-phosphate and stabilization of endothelial monolayer barrier function by lysophosphatidic acid, potential mediators of hematopoietic angiogenesis. J Hematother Stem Cell Res 8:627–634.

41. Minnear FL, Patil S, Bell D, Gainor JP, Morton CA. 2001. Platelet lipid(s) bound to albumin increases endothelial electrical resistance: mimicked by LPA. Am J Physiol Lung Cell Mol Physiol 281:L1337–L1344.

Page 14: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

278 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

42. Smyth SS, Cheng HY, Miriyala S, Panchatcharam M, Morris AJ. 2008. Roles of lysophosphatidic acid in cardiovascular physiology and disease. Biochim Biophys Acta 1781:563–570.

43. Sarker MH, Hu DE, Fraser PA. 2010. Regulation of cerebromicrovascular perme-ability by lysophosphatidic acid. Microcirculation 17:39–46.

44. Zhou Z, Subramanian P, Sevilmis G, Globke B, Soehnlein O, Karshovska E, Megens R, Heyll K, Chun J, Saulnier-Blache JS, Reinholz M, van Zandvoort M, Weber C, Schober A. 2011. Lipoprotein-derived lysophosphatidic acid promotes atheroscle-rosis by releasing CXCL1 from the endothelium. Cell Metab 13:592–600.

45. Lin CI, Chen CN, Chen JH, Lee H. 2006. Lysophospholipids increase IL-8 and MCP-1 expressions in human umbilical cord vein endothelial cells through an IL-1-dependent mechanism. J Cell Biochem 99:1216–1232.

46. Gustin C, Delaive E, Dieu M, Calay D, Raes M. 2008. Upregulation of pentraxin-3 in human endothelial cells after lysophosphatidic acid exposure. Arterioscler Thromb Vasc Biol 28:491–497.

47. Rizza C, Leitinger N, Yue J, Fischer DJ, Wang DA, Shih PT, Lee H, Tigyi G, Berliner JA. 1999. Lysophosphatidic acid as a regulator of endothelial/leukocyte interaction. Lab Invest 79:1227–1235.

48. Lee H, Liao JJ, Graeler M, Huang MC, Goetzl EJ. 2002. Lysophospholipid regula-tion of mononuclear phagocytes. Biochim Biophys Acta 1582:175–177.

49. Chettibi S, Lawrence AJ, Stevenson RD, Young JD. 1994. Effect of lysophosphatidic acid on motility, polarisation and metabolic burst of human neutrophils. FEMS Immunol Med Microbiol 8:271–281.

50. Itagaki K, Kannan KB, Hauser CJ. 2005. Lysophosphatidic acid triggers calcium entry through a non-store-operated pathway in human neutrophils. J Leukoc Biol 77:181–189.

51. Idzko M, Laut M, Panther E, Sorichter S, Durk T, Fluhr JW, Herouy Y, Mocken-haupt M, Myrtek D, Elsner P, Norgauer J. 2004. Lysophosphatidic acid induces chemotaxis, oxygen radical production, CD11b up-regulation, Ca2+ mobilization, and actin reorganization in human eosinophils via pertussis toxin-sensitive G pro-teins. J Immunol 172:4480–4485.

52. Fueller M, Wang DA, Tigyi G, Siess W. 2003. Activation of human monocytic cells by lysophosphatidic acid and sphingosine-1-phosphate. Cell Signal 15:367–375.

53. Haseruck N, Erl W, Pandey D, Tigyi G, Ohlmann P, Ravanat C, Gachet C, Siess W. 2004. The plaque lipid lysophosphatidic acid stimulates platelet activation and platelet-monocyte aggregate formation in whole blood: involvement of P2Y1 and P2Y12 receptors. Blood 103:2585–2592.

54. Hao F, Tan M, Wu DD, Xu X, Cui MZ. 2010. LPA induces IL-6 secretion from aortic smooth muscle cells via an LPA1-regulated, PKC-dependent, and p38alpha-mediated pathway. Am J Physiol Heart Circ Physiol 298:H974–H983.

55. Zhao C, Sardella A, Chun J, Poubelle PE, Fernandes MJ, Bourgoin SG. 2011. TNF-{alpha} promotes LPA1- and LPA3 -mediated recruitment of leukocytes in vivo through CXCR2 ligand chemokines. J Lipid Res 52:1307–1318.

56. Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, Corrinth C, Bittman R, Tigyi G, Aepfelbacher M. 1999. Lysophosphatidic acid mediates the rapid activation of platelets and endothelial cells by mildly oxidized low density lipoprotein and

Page 15: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

REfERENCES 279

accumulates in human atherosclerotic lesions. Proc Natl Acad Sci U S A 96:6931–6936.

57. Siess W. 2002. Athero- and thrombogenic actions of lysophosphatidic acid and sphingosine-1-phosphate. Biochim Biophys Acta 1582:204–215.

58. Bot M, Bot I, Lopez-Vales R, van de Lest CH, Saulnier-Blache JS, Helms JB, David S, van Berkel TJ, Biessen EA. 2010. Atherosclerotic lesion progression changes lysophosphatidic acid homeostasis to favor its accumulation. Am J Pathol 176:3073–3084.

59. Tokumura A, Kanaya Y, Kitahara M, Miyake M, Yoshioka Y, Fukuzawa K. 2002. Increased formation of lysophosphatidic acids by lysophospholipase D in serum of hypercholesterolemic rabbits. J Lipid Res 43:307–315.

60. Chung SM, Bae ON, Lim KM, Noh JY, Lee MY, Jung YS, Chung JH. 2007. Lyso-phosphatidic acid induces thrombogenic activity through phosphatidylserine expo-sure and procoagulant microvesicle generation in human erythrocytes. Arterioscler Thromb Vasc Biol 27:414–421.

61. Gerrard JM, Kindom SE, Peterson DA, White JG. 1979. Lysophosphatidic acids. II. Interaction of the effects of adenosine diphosphate and lysophosphatidic acids in dog, rabbit, and human platelets. Am J Pathol 97:531–547.

62. Cui MZ, Zhao G, Winokur AL, Laag E, Bydash JR, Penn MS, Chisolm GM, Xu X. 2003. Lysophosphatidic acid induction of tissue factor expression in aortic smooth muscle cells. Arterioscler Thromb Vasc Biol 23:224–230.

63. Essler M, Retzer M, Bauer M, Zangl KJ, Tigyi G, Siess W. 2000. Stimulation of platelets and endothelial cells by mildly oxidized LDL proceeds through activation of lysophosphatidic acid receptors and the Rho/Rho-kinase pathway. Inhibition by lovastatin. Ann N Y Acad Sci 905:282–286.

64. Maschberger P, Bauer M, Baumann-Siemons J, Zangl KJ, Negrescu EV, Reininger AJ, Siess W. 2000. Mildly oxidized low density lipoprotein rapidly stimulates via activation of the lysophosphatidic acid receptor Src family and Syk tyrosine kinases and Ca2+ influx in human platelets. J Biol Chem 275:19159–19166.

65. Gerrard JM, Kindom SE, Peterson DA, Peller J, Krantz KE, White JG. 1979. Lyso-phosphatidic acids. Influence on platelet aggregation and intracellular calcium flux. Am J Pathol 96:423–438.

66. Retzer M, Essler M. 2000. Lysophosphatidic acid-induced platelet shape change proceeds via Rho/Rho kinase-mediated myosin light-chain and moesin phosphory-lation. Cell Signal 12:645–648.

67. Olorundare OE, Peyruchaud O, Albrecht RM, Mosher DF. 2001. Assembly of a fibronectin matrix by adherent platelets stimulated by lysophosphatidic acid and other agonists. Blood 98:117–124.

68. Nilsson UK, Svensson SP, Grenegard M. 2002. Synergistic activation of human platelets by lysophosphatidic acid and adrenaline. Haematologica 87:730–739, dis-cussion 739.

69. Tokumura A, Yoshida J, Maruyama T, Fukuzawa K, Tsukatani H. 1987. Platelet aggregation induced by ether-linked phospholipids. 1. Inhibitory actions of bovine serum albumin and structural analogues of platelet activating factor. Thromb Res 46:51–63.

70. Williams JR, Khandoga AL, Goyal P, Fells JI, Perygin DH, Siess W, Parrill AL, Tigyi G, Fujiwara Y. 2009. Unique ligand selectivity of the GPR92/LPA5

Page 16: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

280 LPA SIGNALING AND CARDIOVASCULAR PATHOLOGY

lysophosphatidate receptor indicates role in human platelet activation. J Biol Chem 284:17304–17319.

71. Amisten S, Braun OO, Bengtsson A, Erlinge D. 2008. Gene expression profiling for the identification of G-protein coupled receptors in human platelets. Thromb Res 122:47–57.

72. Khandoga AL, Pandey D, Welsch U, Brandl R, Siess W. 2011. GPR92/LPA lyso-phosphatidate receptor mediates megakaryocytic cell shape change induced by human atherosclerotic plaques. Cardiovasc Res 90:157–164.

73. Rowley JW, Oler AJ, Tolley ND, Hunter BN, Low EN, Nix DA, Yost CC, Zimmer-man GA, Weyrich AS. 2011. Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood 118:e101–e111.

74. Damirin A, Tomura H, Komachi M, Liu JP, Mogi C, Tobo M, Wang JQ, Kimura T, Kuwabara A, Yamazaki Y, Ohta H, Im DS, Sato K, Okajima F. 2007. Role of lipoprotein-associated lysophospholipids in migratory activity of coronary artery smooth muscle cells. Am J Physiol Heart Circ Physiol 292:H2513–H2522.

75. Kaneyuki U, Ueda S, Yamagishi S, Kato S, Fujimura T, Shibata R, Hayashida A, Yoshimura J, Kojiro M, Oshima K, Okuda S. 2007. Pitavastatin inhibits lysophos-phatidic acid-induced proliferation and monocyte chemoattractant protein-1 expression in aortic smooth muscle cells by suppressing Rac-1-mediated reactive oxygen species generation. Vascul Pharmacol 46:286–292.

76. Morris AJ, Panchatcharam M, Cheng HY, Federico L, Fulkerson Z, Selim S, Miri-yala S, Escalante-Alcalde D, Smyth SS. 2009. Regulation of blood and vascular cell function by bioactive lysophospholipids. J Thromb Haemost 7 Suppl 1:38–43.

77. Yoshida K, Nishida W, Hayashi K, Ohkawa Y, Ogawa A, Aoki J, Arai H, Sobue K. 2003. Vascular remodeling induced by naturally occurring unsaturated lysophos-phatidic acid in vivo. Circulation 108:1746–1752.

78. Hayashi K, Takahashi M, Nishida W, Yoshida K, Ohkawa Y, Kitabatake A, Aoki J, Arai H, Sobue K. 2001. Phenotypic modulation of vascular smooth muscle cells induced by unsaturated lysophosphatidic acids. Circ Res 89:251–258.

79. Cui MZ, Laag E, Sun L, Tan M, Zhao G, Xu X. 2006. Lysophosphatidic acid induces early growth response gene 1 expression in vascular smooth muscle cells: CRE and SRE mediate the transcription. Arterioscler Thromb Vasc Biol 26:1029–1035.

80. Shano S, Moriyama R, Chun J, Fukushima N. 2008. Lysophosphatidic acid stimu-lates astrocyte proliferation through LPA(1). Neurochem Int 52:216–220.

81. Shano S, Hatanaka K, Ninose S, Moriyama R, Tsujiuchi T, Fukushima N. 2007. A lysophosphatidic acid receptor lacking the PDZ-binding domain is constitutively active and stimulates cell proliferation. Biochim Biophys Acta 1783:748–759.

82. Meyer zu Heringdorf D, Jakobs KH. 2007. Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism. Biochim Biophys Acta 1768:923–940.

83. Khurana S, Tomar A, George SP, Wang Y, Siddiqui MR, Guo H, Tigyi G, Mathew S. 2008. Autotaxin and lysophosphatidic acid stimulate intestinal cell motility by redistribution of the actin modifying protein villin to the developing lamellipodia. Exp Cell Res 314:530–542.

84. Lee MJ, Jeon ES, Lee JS, Cho M, Suh DS, Chang CL, Kim JH. 2008. Lysophospha-tidic acid in malignant ascites stimulates migration of human mesenchymal stem cells. J Cell Biochem 104:499–510.

Page 17: Lysophospholipid Receptors (Signaling and Biochemistry) || Lysophosphatidic Acid (LPA) Signaling and Cardiovascular Pathology

REfERENCES 281

85. Lee SY, Lee HY, Kim SD, Shim JW, Bae YS. 2007. Lysophosphatidylglycerol stimu-lates chemotactic migration and tube formation in human umbilical vein endothe-lial cells. Biochem Biophys Res Commun 363:490–494.

86. Hirakawa M, Karashima Y, Watanabe M, Kimura C, Ito Y, Oike M. 2007. Protein kinase A inhibits lysophosphatidic acid-induced migration of airway smooth muscle cells. J Pharmacol Exp Ther 321:1102–1108.

87. Schmitz U, Thommes K, Beier I, Vetter H. 2002. Lysophosphatidic acid stimulates p21-activated kinase in vascular smooth muscle cells. Biochem Biophys Res Commun 291:687–691.

88. Guo H, Makarova N, Cheng Y, Shuyu E, Ji RR, Zhang C, Farrar P, Tigyi G. 2008. The early- and late stages in phenotypic modulation of vascular smooth muscle cells: differential roles for lysophosphatidic acid. Biochim Biophys Acta 1781:571–581.

89. Cheng Y, Makarova N, Tsukahara R, Guo H, Shuyu E, Farrar P, Balazs L, Zhang C, Tigyi G. 2009. Lysophosphatidic acid-induced arterial wall remodeling: require-ment of PPARgamma but not LPA1 or LPA2 GPCR. Cell Signal 21:1874–1884.

90. Subramanian P, Karshovska E, Reinhard P, Megens RT, Zhou Z, Akhtar S, Schumann U, Li X, van Zandvoort M, Ludin C, Weber C, Schober A. 2010. Lysophosphatidic acid receptors LPA1 and LPA3 promote CXCL12-mediated smooth muscle pro-genitor cell recruitment in neointima formation. Circ Res 107:96–105.

91. Llodra J, Angeli V, Liu J, Trogan E, Fisher EA, Randolph GJ. 2004. Emigration of monocyte-derived cells from atherosclerotic lesions characterizes regressive, but not progressive, plaques. Proc Natl Acad Sci U S A 101:11779–11784.

92. Veniant MM, Withycombe S, Young SG. 2001. Lipoprotein size and atherosclerosis susceptibility in Apoe(–/–) and Ldlr(–/–) mice. Arterioscler Thromb Vasc Biol 21:1567–1570.


Recommended