Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Feb 1.
Published in final edited form as: J Immunol. 2014 Feb 1;192(3):851–857. doi: 10.4049/jimmunol.1302831

The autotaxin-LPA axis emerges as a novel regulator of lymphocyte homing and inflammation

Sara Knowlden 1, Steve N Georas 1,2
PMCID: PMC3905607  NIHMSID: NIHMS548099  PMID: 24443508

Abstract

Lyophosphatidic acid (LPA) is a pleiotropic lipid molecule with potent effects on cell growth and motility. Major progress has been made in recent years in deciphering the mechanisms of LPA generation and how it acts on target cells. Most research to-date has been conducted in other disciplines, but emerging data indicate that LPA has an important role to play in immunity. A key discovery was that autotaxin (ATX), an enzyme previously implicated in cancer cell motility, generates extracellular LPA from the precursor lysophosphatidylcholine (LPC). Steady-state ATX is expressed by only a few tissues including high-endothelial venules in lymph nodes, but inflammatory signals can up-regulate ATX expression in different tissues. Here we review current thinking about the ATX/LPA axis in lymphocyte homing, as well as in models of allergic airway inflammation and asthma. New insights into the role of LPA in regulating immune responses should be forthcoming in the near future.

Nomenclature and LPA generation

Lysophosphatidic acid is a member of the glycerophospholipid family, specifically a monoacylglycerophosphate (GP1005, according to Lipid Maps classification, http://www.lipidmaps.org). LPA has a three carbon glycerol backbone with one acyl side chain that can very in length and saturation (Figure 1). The prefix “lyso” indicates that only one fatty acid is attached to the phospholipid backbone, and distinguishes LPA from phosphatidic acid. The acyl group is usually located on the sn-1 carbon (i.e. 1-acyl-2-hydroxy-sn-glycero-3-phosphate). In serum, most LPA species are polyunsaturated (e.g. 18:2 and 20:4) or monounsaturated (e.g. 18:1) (1). Emerging data indicate that the acyl moiety affects the function of LPA, but this is a virtually unexplored area in the immune system. One exception is the observation that immature mouse bone marrow-derived dendritic cells (BM-DC) preferentially migrate in vitro to unsaturated (18:1 and 20:4) but not saturated (16:0 and 18:0) LPA (2), possibly reflecting affinity for the cell surface receptor LPA3 (see below).

Figure 1.

Figure 1

Two main sources of LPA generation are from phosphatidic acid (left) via the action of phospholipases, or by hydrolysis of the choline moiety of lysophosphatidylcholine (Lyso-PC), by autotaxin (also known as Lyso-PLD). Newly generated LPA can act on cell surface GPCR’s or intracellular receptors as indicated. Free LPA is rapidly degraded by lipid phosphate phosphatases into monoacyl glycerol.

LPA can be generated by multiple mechanisms, and a current challenge in the field is to relate different potential sources of LPA to biological activities in vivo. Extracellular LPA is thought to derive from at least two pathways. First LPA can be produced by the action of phospholipases on membrane microvesicles shed by activated platelets and erythrocytes (1, 3, 4). These pathways involve the direct hydrolysis of a fatty acid moiety from membrane-derived phosphatidic acid. Second, LPA can be generated from lysophoshpatidylcholine (LPC) by removal of the choline moiety by the enzyme lysophospholipase D (lyso-PLD). LPC is an intermediate in multiple lipid metabolic pathways and circulates in the blood stream at much higher concentration than LPA (5). These two pathways are not mutually exclusive, since certain phospholipases can produce LPC and other lysophospholipids from activated platelets that are then cleaved by lyso-PLD to produce LPA (4).

In 2002, the dominant lyso-PLD in serum was found to be an enzyme known as autotaxin (ATX) (6, 7). ATX had been discovered ten years earlier by Liotta and colleagues, who were studying autocrine factors that promoted cancer cell motility (8). Cloning of the autotaxin cDNA revealed domains similar to the ectonucleotide phosphodiesterase and pyrophosphatase (ENPP) family, and ATX was designated ENPP2 (9). However, the preferred substrates for ATX are not nucleotides but actually lysophospholipids including LPC (4), and it is becoming clear that many functions previously ascribed to ATX can be explained by its ability to generate LPA. This is especially true in the field of cancer biology, where the ATX/LPA axis is involved in the growth and metastasis of many different tumors and pathway antagonists are under active development (10).

LPA is unstable in plasma with a half-life of <5 minutes and technically challenging to measure (11, 12). Experiments using lipid phosphate phosphatase 1 (LPP1) hypomorphic mice revealed that this enzyme is a major determinant of LPA instability in vivo (11). Interestingly, there appear to be tissue-specific differences in LPP expression and activity suggesting that extracellular LPA may be more stable in some tissues than others (11). Relatively little is known about the stability of LPA in secondary lymphoid organs, although LPP expression is relatively high in the spleen.

LPA can be measured by colorimetric assays or mass spectrometry (MS), which has the advantage of detecting specific acyl side chains. A typical approach involves lipid extraction followed by high-pressure liquid chromatography (HPLC) and tandem MS in negative ion mode, monitoring for the glycerol phopshoryl moiety (m/z=152.9) and parent compound acyl groups. In order to estimate LPA concentrations, a closely related LPA species not normally present should be spiked into the original sample, extracted and processed identically, and used to generate a standard curve. However, the efficiency of extraction of different LPA species varies based on the sample and method used, and at present the true bioactive concentrations of LPA in plasma or biological fluids is not clear. Plasma concentrations are probably in the 100 nM range and are higher in women than men (5, 13, 14). In mice, similar variability in plasma LPA concentrations has been reported reflecting poorly-understood effects of age, gender and strain (11).

The function of circulating extracellular LPA is not known. Since the kD of cell surface LPA receptors is in the nM range, one possibility is that LPA receptors on circulating leukocytes or endothelial cells are continually engaged. Tonic engagement would lead to receptor desensitization, and is difficult to reconcile with the pro-inflammatory / barrier disruptive effects of LPA on endothelial cell subsets when studied in vitro (15). LPA likely circulates in microparticles or bound to albumin and other proteins, and it seems likely that this restricts its interaction with target receptors.

LPA binds to different cell surface receptors and intracellular targets

The discovery that LPA interacted with specific cell surface receptors was a major advance. A detailed discussion of LPA receptors is beyond the scope of this Brief Review, but this topic has been recently summarized (16). There are at least six G-protein coupled receptors (GPCR) that bind LPA and vary in their tissue distribution. LPA1 (Edg2), LPA2 (Edg4) and LPA3 (Edg7) belong to the Edg family, whereas LPA4-6 are more closely related to cell surface purinergic receptors (16, 17). Other Edg receptor family members bind sphingosine-1 phosphate (S1P), including S1P1 (Edg2). Signal transduction via LPA receptors leads to activation of mitogen-activated protein kinases, phosphoinositide-3 kinases (PI3K), and Rho kinases, which affect cell activation, survival and migration. This is a rapidly evolving field where new candidate receptors are being actively pursued, but the lack of antibodies directed against extracellular domains that detect surface expression or antagonize LPA binding is a major hindrance. The availability of LPA receptor deficient mice is starting to provide insights into the function of this molecule in vivo (18), although more research is needed in models of innate and adaptive immunity.

LPA can also bind to non-GPCR targets that are both extracellular (e.g. receptor for advanced glycosylation end products, RAGE) and intracellular (e.g. the nuclear receptor PPAR-γ and the cation channel TRPV1) (1921). Whereas most canonical LPA GPCR’s demonstrate preferential binding to longer chain length unsaturated species, PPARγ is only activated by unsaturated LPA species and is critically important for the effects of LPA on vascular remodeling (20). Very little is known about the role of these non-canonical LPA receptors in immune cells.

LPA receptors are expressed by T cells

LPA enhances the motility of human and mouse T cells in vitro, although generally not in a directed manner (2224). LPA also induces actin remodeling, uropod formation, and T cell polarization on ICAM-1 and CCL21 coated chambers (24, 25). The T cell LPA receptors involved in these responses is not known. LPA enhances the invasion of T lymphoma cells into tissue substrates, which in the case of Jurkat T cells depended on LPA2 (22, 23). LPA5 (GPR92) mRNA is abundant in the small intestine, especially in CD8+ intra-epithelial lymphocytes (26, 27), but the function of this receptor is currently unknown. More research using gene-targeted mice and LPA receptor antagonists should help clarify the role of LPA in T cell migration in the near future.

Autotaxin is expressed by HEV: role in T cell homing

In 2008, two reports revealed that ATX is constitutively expressed and released from lymphoid organ high endothelial venule (HEV) endothelial cells (EC’s) (24, 28). These and other studies challenged the notion that ATX was a trans-membrane protein, and suggested that secreted ATX could act at a distance from its site of production. One working model suggests that ATX is secreted into the lumen of the HEV where it binds to receptors on nearby cells, hydrolyzes the abundant LPC found in the plasma, and generates a locally restricted high concentration of LPA. Kanda et al. showed that ATX can bind to chemokine-activated human lymphocytes in a β1-integrin dependent manner, but binding to mouse lymphocytes interestingly appeared to be integrin-independent (24). Subsequently, Zhang et al. found that polarized T cells possess Mn+-activatable receptors for ATX that are localized at the leading edge (25). Since activated platelets can produce and bind ATX (4, 29), and also participate in lymphocyte trafficking at HEV (30, 31), an additional possibility is that platelets contribute to ATX-dependent LPA production in lymphoid organs.

The recent crystal structure of ATX provided insights into how this molecule may function in vivo (32, 33). In addition to phosphodiesterase domains, ATX contains N-terminal cysteine-rich somatomedin-B-like (SMB) domains and a C-terminal nuclease domain. Integrin binding by ATX appears to involve the SMB domains in an RGD-independent manner (32). A splice variant of ATX termed ATXα was recently shown to bind heparan sulfate proteogylcans (34). One unusual feature was the existence of a hydrophobic pocket and nearby channel that was large enough to accommodate a phospholipid with only a single acyl side chain. In additional to helping explain the preference of ATX for lysolipid substrates, these structural features indicate that ATX can act as a lipid carrier, and suggested a model in which integrin or substrate binding induces a conformational change, allowing ATX to release bound LPA to target receptors on nearby cells. A corollary of this model is that tethering of ATX will restrict newly generated bioactive LPA to an extremely precise location in vivo, such as HEV.

It is currently unknown if ATX-generated LPA acts on T cells, HEV EC, or both, and different models have been proposed. One model suggests that locally generated LPA provides a chemokinetic “boost” that facilitates T cell exit from the circulation and entry into lymph nodes along chemotactic gradients established by canonical lymph node associated cytokines. The receptors involved in LPA-dependent T cell emigration from HEV are not known. A second model suggests that the ATX/LPA axis regulates T cell homing indirectly by acting on HEV EC. These data are supported by the observations that: (i) HEV EC express LPA1 and LPA4, (ii) LPA induces changes in the EC cytoskeleton, and (iii) a potent small molecule inhibitor of ATX (HA130) did not affect lymphocyte accumulation at HEVs but did attenuate their extravasation (25, 28, 35). Furthermore, using novel imaging approaches Bai et al. demonstrated that after lymphocytes infiltrate the EC layer, migration across the HEV basal lamina required LPA (35). These data suggest that by inducing EC motility and shape change, LPA allows lymphocytes to detach from the HEV basal lamina and migrate into the lymph node parenchyma.

Manipulating the ATX/LPA axis with pathway antagonists

If the model proposed above is correct, then antagonists of the ATX/LPA axis should inhibit steady-state lymphocyte trafficking, whereas agonists should have the opposite effects. Experimental evidence supporting the ability of ATX/LPA antagonists to inhibit lymphocyte homing into lymphoid organs is starting to emerge, but we currently have no evidence that this affects generation of antigen-specific immunity. For example, incubating T cells with an inactive form of ATX (T120A), which probably acts as a dominant negative manner, attenuated cell homing to lymph nodes following adoptive transfer, as demonstrated by reduction of T cells in vicinity of HEVs 15 minutes later (24). Another small molecule inhibitor of ATX (HA130) specifically attenuated lymphocyte extravasation (discussed above) (25). However, an anti-ATX monoclonal antibody administered intraperitoneally that depleted plasma ATX did not inhibit lymphocyte trafficking to lymph nodes or the spleen, suggesting that ATX in the circulation is not as important as locally produced ATX at the HEV (28). BrP-LPA is a dual inhibitor of ATX and LPA receptors, and local administration of this compound diminished the trafficking of adoptively transferred lymphocytes into lymph nodes 30 minutes later (35). Ki16425 is an LPA receptor antagonist with selectivity for LPA1 and LPA3. When injected into the footpad of mice, Ki16425 did not significantly reduce T cell migration in vitro or lymphocyte trafficking to lymph nodes (35). These data are consistent with the possibility that T lymphocyte LPA2 is important for lymph node entry, but also support a role for LPA acting on endothelial cells in an LPA1/3-independent manner. Newer and more stable antagonists of ATX and LPA receptors are actively being developed, with the hope that these will demonstrate efficacy in cancer therapeutics (10). These compounds will also be useful tools to study the ATX/LPA axis in the initiation and regulation of adaptive immunity. However, clinical trials of ATX antagonists in cancer patients will need to be carefully monitored for untoward effects on the immune system, including immunosuppression. Is it possible to boost antigen-specific immunity by activating the ATX/LPA axis? Currently, there is little experimental evidence to support this possibility, but future studies investigating whether enhancing ATX activity or LPA production could boost generation of adaptive immunity seem worthwhile.

Regardless of mechanisms involved, the possibility that LPA promotes steady-state T recirculation through lymph nodes and secondary lymphoid organs is an exciting new direction, and complements the now well-recognized role for the related sphingolipid S1P in lymphocyte egress (36). Future studies in which LPA receptor expression can be regulated in a cell-type specific manner will be needed to determine the relative importance of LPA induced T cell motility, endothelial shape change, or both in regulating T cell homing. It will be interesting in future studies to determine if receptor desensitization influences the effects of LPA on T cell migration, similar to the effects of GRK2 on S1PR1 (37). Several other unanswered questions remain that should be readily addressable in the coming years. For example, what are the concentrations of LPA in afferent or efferent lymphatics? Does the ATX/LPA axis regulate T cell homing in a subset specific manner? What receptors are involved in this process? Do ATX/LPA regulate recirculation or LN entry of other immune cells besides T cells? Future research into these questions should enhance our understanding of how ATX and LPA regulate adaptive immunity in vivo.

LPA effects on dendritic cells and other cell types

In addition to lymphocytes, other immune cells express LPA receptors and are influenced by LPA including NK cells, mast cells, eosinophils, and B cells (summarized in Table 1) (3852). LPA induces chemotaxis of immature mouse BM-DC dendritic cells (DC), which involves pertussis-toxin sensitive receptors. Interestingly, this effect is lost in DC’s matured in the presence of lipopolysaccharide (LPS) (2, 53). LPS does not induce major changes in LPA receptor mRNA or protein expression in cell lysates (2, 53, 54), suggesting that G-protein coupling or LPA receptor surface expression are inhibited during DC maturation. Immature DC derived from LPA3 gene-targeted mice do not migrate to LPA in vitro, demonstrating a critical requirement for this receptor in LPA directed migration (2). In addition to inducing immature DC motility, LPA can influence DC function. LPA inhibited the production of IL-12 and TNF-α from LPS-stimulated DC in a pertussis toxin-insensitive manner (53). We recently reported that LPA inhibited LPS-dependent DC activation at least in part in an LPA2-dependent manner, and furthermore that LPA2-deficient DC were hyperactive after adoptive transfer in vivo (55). Consequently, the effects of LPA on DC are complex, and influenced by LPA species (discussed above), DC activation status, and receptors engaged. By inducing the mobility of both naive T cells and immature DC, LPA would seem to be well-suited to promote DC:T interactions in lymphoid organs and the initiation of adaptive immune responses.

Table 1.

Cell Type LPA receptors implicated Summary of findings LPA species (concentration) References

Natural Killer LPA1,2,3 Chemotaxis; mobilization of Ca2+ in activated cells; enhanced IFN-γ secretion; LPAR detected by flow cytometry 18:1 (1–10 μM) (38)
LPA2 Inhibited release of perforin and cytotoxic activity; enhances cAMP levels and activates PKA 14:0 (0.02–20 μM) (39)

Mast cells LPA1,2,3,4 (mRNA) Growth factor (human); induces proliferation and differentiation 18:1 (5 μM) (40)
NA Activates mast cells; causes tryptase release and vascular leakage in a mast-cell dependent manner in mice LPA gel (41)
LPA1,3 Histamine release, inhibited by DGPP 18:1 (10 μg/ml) (42)
LPA2 Production of MIP-1β, IL-8, MCP-1 in an IL-4 dependent manner 18:1 and 18:2 (1–50 μM) (43)
LPA5 (mRNA) MIP-1β release (human) 18:1 (1–5 μM) (44)

Neutrophils NA Infiltration of neutrophils in BALF of guinea pigs in Rho/ROCK dependent manner 18:1 (1–10 μg/ml) (45)
LPA1, 2 (mRNA and protein) Chemotaxis, greater in for neutrophils from pneumonia patients LPA (0.1–1 μM) (46)
NA Degranulation, PA production 18:1 (10–40 μM) (47)

Macrophage/Monocyte NA Survival factor for murine macrophages via PI3K 18:1 (7.7 μM) (48)
NA Upregulated IL-1β in mouse and human macrophages NA (49)
NA Monocyte migration to MCP-1 via iPLA2 at the leading edge NA (50)

B cells LPA2 (mRNA) B lymphoblast growth factor; MAPK activation; immunoglobulin production 18:1 (0.1–1 μM) (51)

Lymphatic endothelium LPA2 Proliferation and lymphangiogenesis via IL-8 18:1 (0.5–10 μM) (52)

Notes: NA = not available; BALF = bronchoalveolar lavage fluids; PA = phosphatidic acid

ATX is also upregulated during inflammation: lessons from asthma models

In addition to lymph node HEV, ATX is constitutively expressed in a few other tissues in adult mice and humans including adipocytes and the lung (24, 28, 5658). Interestingly, ATX was recently identified in mice in a screen for genes affecting lung development (59). Constitutive expression of ATX in the lung may help explain the fact that LPA is a normal component of epithelial lining fluids (60). The function of LPA in the lung at steady-state is not known, but it may play a role in maintaining homeostasis by promoting epithelial barrier integrity (61). Interestingly, both wild-type and lyso-PLD mutant ATX induce airway epithelial migration, suggesting that some of the effects of ATX in the airway are independent of LPA generation (62).

ATX expression and activity increase in different disease states, which suggests that the ATX/LPA axis may play a broader role in inflammation in general. For example, ATX is inducibly expressed on endothelial cells in the inflamed pancreas (28), and in lung epithelial cells following bleomycin exposure (63). Excitingly, inhibition or genetic deletion of ATX significantly attenuated lung fibrosis, supporting a crucial role for LPA in these models (63, 64). ATX expression is upregulated in synovial cells is from patients with rheumatoid arthritis, and recent studies have firmly implicated ATX-derived LPA in the pathogenesis of inflammatory arthritis (6568). The signals that regulate both steady-state and inducible ATX expression require further study. In monocytic THP1 cells, Li et al. reported that ATX mRNA and protein were induced by the toll-like receptor (TLR) 4 ligand lipopolysaccharide (69), but constitutive ATX expression in HEV was independent of the TLR adaptor Myd88 (28).

Growing evidence points to an important role for the ATX/LPA axis in mouse models of allergic airway inflammation and humans with asthma. Using HPLC-MS/MS, we found that LPA is constitutively detectable in bronchoalveolar lavage (BAL) fluids at baseline, but significantly increased 18 hours following segmental allergen challenge of allergic human subjects (from 483±77 to 1506±358 nM) (60). Assuming a ~100-fold dilution during BAL (70), we estimated that epithelial lining fluid LPA levels in the lung are in micromolar range. Interestingly, the accumulation of LPA species after allergen challenge was not uniform but enriched in poly-unsaturated species including 20:4 and 22:6 LPA (60). More recently, Park and Christman confirmed these findings and also showed that 22:5 LPA was also enriched in BAL fluids after allergen challenge (71). The function of polyunsaturated LPA in the lung is not entirely clear. Although LPA can promote eosinophil chemotaxis in vitro (72), we found that LPA levels did not correlate with eosinophil influx 18 hrs after challenge (60) arguing that LPA was not a dominant eosinophilic chemoattractant in this model. It remains to be seen whether LPA promotes the recruitment of leukocyte subsets or immature dendritic cells to the lung. Using a novel ATX antagonist in a triple allergen challenge mouse model of asthma, Park et al. showed that LPA generation in the allergic lung was dependent on ATX activity, and suggested based on immunohistochemical analysis and cell culture experiments that lung epithelial cells and IL-4 stimulated macrophages were potential sources of ATX (71). Furthermore, some of the inflammatory effects of triple allergen challenge were attenuated in LPA2-deficient mice (71).

Separate from its potential effects on cell migration, LPA has other functions and cellular targets relevant to the pathophysiology of asthma. For example, LPA can: (i) induce the differentiation and activation of mast cells (40, 43, 44) (see Table 1), (ii) promote airway epithelial chemokine/cytokine production (73, 74), and (iii) augment airway smooth muscle cell growth and contractility (7577). The ability of LPA to induce airway epithelial TSLP production is noteworthy (74), given the crucial role for this cytokine in initiation of Th2-dependent immunity. Although these studies suggest that LPA may be a novel pro-inflammatory molecule and contribute to initiation of allergic airway inflammation, other results paint a more nuanced picture. For example, we used mouse models of asthma involving both systemic immunization (intraperitoneal Ova + alum) and mucosal sensitization (inhaled Ova + low-dose LPS), and found that LPA2-deficient mice developed surprisingly more inflammation and AHR compared to their wild-type counterparts (55). Using reciprocal bone marrow chimeras, this mapped in part to an inhibitory effect of LPA2 on hematopoietic cells, and we concluded that LPA has a previously unsuspected inhibitory role on dendritic cell activation (55). These data are in keeping with other potentially inhibitory effects of LPA including: (i) suppression of DC cytokine production discussed above (53), (ii) protection from endotoxin-induced inflammation and neutrophilia (61, 78), (iii) induction of the decoy receptor IL-13Rα2 on epithelial cells (79), and (iv) stimulation of αvβ6 integrin-mediated activation of TGFβ (80). Taken together, these data point to both pro-inflammatory and potentially anti-inflammatory effects of LPA in asthma, depending on the timing and context of its production. Under conditions of acute inflammation such as following allergen challenge in allergic human subjects or mouse models of repetitive allergen exposure, LPA may act alone or in concert with other mediators to promote the recruitment or activation of inflammatory cell subsets. More chronically, LPA may play a regulatory role and it is tempting to speculate that over time repeated cycles of LPA generation might contribute to airway remodeling in chronic asthma.

Conclusions

New research will likely be soon forthcoming that will define with greater precision the role of the ATX/LPA axis in immune and inflammatory diseases. It seems apparent that ATX and LPA have previously underappreciated roles in steady-state lymphocyte homing, but we need to understand how effects on T cell migration translate into regulation of adaptive immunity. Newer generation and more stable ATX antagonists will likely prove useful tools in this regard in the near future. In addition to their clinical efficacy in cancer, these agents may have untoward effects on the immune system (e.g. immunosuppression), which will need to be carefully monitored. It will be interesting to determine if gain-of-function approaches that enhance ATX/LPA activity can be used to boost generation of immunity. More research is needed into the mechanisms and consequences of ATX upregulation in inflammatory diseases. It will be important to decipher whether the dominant action of LPA in these models is in the regulation of cell motility/migration, activation, or survival. Since both LPA and LPC are involved in numerous metabolic pathways, it is tempting to speculate that these novel lysolipid compounds are at the intersection of metabolism and immunity.

Acknowledgments

Funding Sources:

NIH R01 HL071933 and P30 ES001247 (SG); T32 AI007285 (SK)

References

  • 1.Sano T, Baker D, Virag T, Wada A, Yatomi Y, Kobayashi T, Igarashi Y, Tigyi G. Multiple mechanisms linked to platelet activation result in lysophosphatidic acid and sphingosine 1-phosphate generation in blood. J Biol Chem. 2002;277:21197–21206. doi: 10.1074/jbc.M201289200. [DOI] [PubMed] [Google Scholar]
  • 2.Chan LC, Peters W, Xu Y, Chun J, Farese RV, Jr, Cases S. LPA3 receptor mediates chemotaxis of immature murine dendritic cells to unsaturated lysophosphatidic acid (LPA) J Leukoc Biol. 2007;82:1193–1200. doi: 10.1189/jlb.0407221. [DOI] [PubMed] [Google Scholar]
  • 3.Fourcade O, Simon MF, Viode C, Rugani N, Leballe F, Ragab A, Fournie B, Sarda L, Chap H. Secretory phospholipase A2 generates the novel lipid mediator lysophosphatidic acid in membrane microvesicles shed from activated cells. Cell. 1995;80:919–927. doi: 10.1016/0092-8674(95)90295-3. [DOI] [PubMed] [Google Scholar]
  • 4.Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishimoto T, Mizuno K, Saku K, Taguchi R, Arai H. Serum lysophosphatidic acid is produced through diverse phospholipase pathways. J Biol Chem. 2002;277:48737–48744. doi: 10.1074/jbc.M206812200. [DOI] [PubMed] [Google Scholar]
  • 5.Nakamura K, Kishimoto T, Ohkawa R, Okubo S, Tozuka M, Yokota H, Ikeda H, Ohshima N, Mizuno K, Yatomi Y. Suppression of lysophosphatidic acid and lysophosphatidylcholine formation in the plasma in vitro: proposal of a plasma sample preparation method for laboratory testing of these lipids. Anal Biochem. 2007;367:20–27. doi: 10.1016/j.ab.2007.05.004. [DOI] [PubMed] [Google Scholar]
  • 6.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;158:227–233. doi: 10.1083/jcb.200204026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tokumura A, Majima E, Kariya Y, Tominaga K, Kogure K, Yasuda K, Fukuzawa K. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J Biol Chem. 2002;277:39436–39442. doi: 10.1074/jbc.M205623200. [DOI] [PubMed] [Google Scholar]
  • 8.Stracke ML, Krutzsch HC, Unsworth EJ, Arestad A, Cioce V, Schiffmann E, Liotta LA. Identification, purification, and partial sequence analysis of autotaxin, a novel motility-stimulating protein. J Biol Chem. 1992;267:2524–2529. [PubMed] [Google Scholar]
  • 9.Goding JW, Grobben B, Slegers H. Physiological and pathophysiological functions of the ecto-nucleotide pyrophosphatase/phosphodiesterase family. Biochim Biophys Acta. 2003;1638:1–19. doi: 10.1016/s0925-4439(03)00058-9. [DOI] [PubMed] [Google Scholar]
  • 10.Gotoh M, Fujiwara Y, Yue J, Liu J, Lee S, Fells J, Uchiyama A, Murakami-Murofushi K, Kennel S, Wall J, Patil R, Gupte R, Balazs L, Miller DD, Tigyi GJ. Controlling cancer through the autotaxin-lysophosphatidic acid receptor axis. Biochem Soc Trans. 2012;40:31–36. doi: 10.1042/BST20110608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tomsig JL, Snyder AH, Berdyshev EV, Skobeleva A, Mataya C, Natarajan V, Brindley DN, Lynch KR. Lipid phosphate phosphohydrolase type 1 (LPP1) degrades extracellular lysophosphatidic acid in vivo. Biochem J. 2009;419:611–618. doi: 10.1042/BJ20081888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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. Boronic acid-based inhibitor of autotaxin reveals rapid turnover of LPA in the circulation. Proc Natl Acad Sci U S A. 2010;107:7257–7262. doi: 10.1073/pnas.1001529107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hosogaya S, Yatomi Y, Nakamura K, Ohkawa R, Okubo S, Yokota H, Ohta M, Yamazaki H, Koike T, Ozaki Y. Measurement of plasma lysophosphatidic acid concentration in healthy subjects: strong correlation with lysophospholipase D activity. Annals of clinical biochemistry. 2008;45:364–368. doi: 10.1258/acb.2008.007242. [DOI] [PubMed] [Google Scholar]
  • 14.Block RC, Duff R, Lawrence P, Kakinami L, Brenna JT, Shearer GC, Meednu N, Mousa S, Friedman A, Harris WS, Larson M, Georas S. The effects of EPA, DHA, and aspirin ingestion on plasma lysophospholipids and autotaxin. Prostaglandins Leukot Essent Fatty Acids. 2010;82:87–95. doi: 10.1016/j.plefa.2009.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ren Y, Guo L, Tang X, Apparsundaram S, Kitson C, Deguzman J, Fuentes ME, Coyle L, Majmudar R, Allard J, Truitt T, Hamid R, Chen Y, Qian Y, Budd DC. Comparing the differential effects of LPA on the barrier function of human pulmonary endothelial cells. Microvasc Res. 2013;85:59–67. doi: 10.1016/j.mvr.2012.10.004. [DOI] [PubMed] [Google Scholar]
  • 16.Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, Chun J. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol. 2010;50:157–186. doi: 10.1146/annurev.pharmtox.010909.105753. [DOI] [PubMed] [Google Scholar]
  • 17.Davenport AP, Alexander SP, Sharman JL, Pawson AJ, Benson HE, Monaghan AE, Liew WC, Mpamhanga CP, Bonner TI, Neubig RR, Pin JP, Spedding M, Harmar AJ. International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands. Pharmacol Rev. 2013;65:967–986. doi: 10.1124/pr.112.007179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Choi JW, Lee CW, Chun J. Biological roles of lysophospholipid receptors revealed by genetic null mice: an update. Biochim Biophys Acta. 2008;1781:531–539. doi: 10.1016/j.bbalip.2008.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rai V, Toure F, Chitayat S, Pei R, Song F, Li Q, Zhang J, Rosario R, Ramasamy R, Chazin WJ, Schmidt AM. Lysophosphatidic acid targets vascular and oncogenic pathways via RAGE signaling. J Exp Med. 2012;209:2339–2350. doi: 10.1084/jem.20120873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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;199:763–774. doi: 10.1084/jem.20031619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nieto-Posadas A, Picazo-Juarez G, Llorente I, Jara-Oseguera A, Morales-Lazaro S, Escalante-Alcalde D, Islas LD, Rosenbaum T. Lysophosphatidic acid directly activates TRPV1 through a C-terminal binding site. Nat Chem Biol. 2012;8:78–85. doi: 10.1038/nchembio.712. [DOI] [PubMed] [Google Scholar]
  • 22.Zheng Y, Kong Y, Goetzl EJ. Lysophosphatidic acid receptor-selective effects on Jurkat T cell migration through a Matrigel model basement membrane. J Immunol. 2001;166:2317–2322. doi: 10.4049/jimmunol.166.4.2317. [DOI] [PubMed] [Google Scholar]
  • 23.Stam JC, Michiels F, van der Kammen RA, Moolenaar WH, Collard JG. Invasion of T-lymphoma cells: cooperation between Rho family GTPases and lysophospholipid receptor signaling. Embo J. 1998;17:4066–4074. doi: 10.1093/emboj/17.14.4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. Autotaxin, an ectoenzyme that produces lysophosphatidic acid, promotes the entry of lymphocytes into secondary lymphoid organs. Nat Immunol. 2008;9:415–423. doi: 10.1038/ni1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhang Y, Chen YC, Krummel MF, Rosen SD. Autotaxin through lysophosphatidic acid stimulates polarization, motility, and transendothelial migration of naive T cells. J Immunol. 2012;189:3914–3924. doi: 10.4049/jimmunol.1201604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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;281:23589–23597. doi: 10.1074/jbc.M603670200. [DOI] [PubMed] [Google Scholar]
  • 27.Kotarsky K, Boketoft A, Bristulf J, Nilsson NE, Norberg A, Hansson S, Owman C, Sillard R, Leeb-Lundberg LM, Olde B. Lysophosphatidic acid binds to and activates GPR92, a G protein-coupled receptor highly expressed in gastrointestinal lymphocytes. J Pharmacol Exp Ther. 2006;318:619–628. doi: 10.1124/jpet.105.098848. [DOI] [PubMed] [Google Scholar]
  • 28.Nakasaki T, Tanaka T, Okudaira S, Hirosawa M, Umemoto E, Otani K, Jin S, Bai Z, Hayasaka H, Fukui Y, Aozasa K, Fujita N, Tsuruo T, Ozono K, Aoki J, Miyasaka M. Involvement of the lysophosphatidic acid-generating enzyme autotaxin in lymphocyte-endothelial cell interactions. Am J Pathol. 2008;173:1566–1576. doi: 10.2353/ajpath.2008.071153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fulkerson Z, Wu T, Sunkara M, Kooi CV, Morris AJ, Smyth SS. Binding of autotaxin to integrins localizes lysophosphatidic acid production to platelets and mammalian cells. J Biol Chem. 2011;286:34654–34663. doi: 10.1074/jbc.M111.276725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Diacovo TG, Puri KD, Warnock RA, Springer TA, von Andrian UH. Platelet-mediated lymphocyte delivery to high endothelial venules. Science. 1996;273:252–255. doi: 10.1126/science.273.5272.252. [DOI] [PubMed] [Google Scholar]
  • 31.Herzog BH, Fu J, Wilson SJ, Hess PR, Sen A, McDaniel JM, Pan Y, Sheng M, Yago T, Silasi-Mansat R, McGee S, May F, Nieswandt B, Morris AJ, Lupu F, Coughlin SR, McEver RP, Chen H, Kahn ML, Xia L. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature. 2013 doi: 10.1038/nature12501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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. Structural basis of substrate discrimination and integrin binding by autotaxin. Nat Struct Mol Biol. 2011;18:198–204. doi: 10.1038/nsmb.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nishimasu H, Okudaira S, Hama K, Mihara E, Dohmae N, Inoue A, Ishitani R, Takagi J, Aoki J, Nureki O. Crystal structure of autotaxin and insight into GPCR activation by lipid mediators. Nat Struct Mol Biol. 2011;18:205–212. doi: 10.1038/nsmb.1998. [DOI] [PubMed] [Google Scholar]
  • 34.Houben AJ, van Wijk XM, van Meeteren LA, van Zeijl L, van de Westerlo EM, Hausmann J, Fish A, Perrakis A, van Kuppevelt TH, Moolenaar WH. The polybasic insertion in autotaxin alpha confers specific binding to heparin and cell surface heparan sulfate proteoglycans. J Biol Chem. 2013;288:510–519. doi: 10.1074/jbc.M112.358416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bai Z, Cai L, Umemoto E, Takeda A, Tohya K, Komai Y, Veeraveedu PT, Hata E, Sugiura Y, Kubo A, Suematsu M, Hayasaka H, Okudaira S, Aoki J, Tanaka T, Albers HM, Ovaa H, Miyasaka M. Constitutive lymphocyte transmigration across the basal lamina of high endothelial venules is regulated by the autotaxin/lysophosphatidic acid axis. J Immunol. 2013;190:2036–2048. doi: 10.4049/jimmunol.1202025. [DOI] [PubMed] [Google Scholar]
  • 36.Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol. 2005;23:127–159. doi: 10.1146/annurev.immunol.23.021704.115628. [DOI] [PubMed] [Google Scholar]
  • 37.Arnon TI, Xu Y, Lo C, Pham T, An J, Coughlin S, Dorn GW, Cyster JG. GRK2-dependent S1PR1 desensitization is required for lymphocytes to overcome their attraction to blood. Science. 2011;333:1898–1903. doi: 10.1126/science.1208248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jin Y, Knudsen E, Wang L, Maghazachi AA. Lysophosphatidic acid induces human natural killer cell chemotaxis and intracellular calcium mobilization. Eur J Immunol. 2003;33:2083–2089. doi: 10.1002/eji.200323711. [DOI] [PubMed] [Google Scholar]
  • 39.Lagadari M, Truta-Feles K, Lehmann K, Berod L, Ziemer M, Idzko M, Barz D, Kamradt T, Maghazachi AA, Norgauer J. Lysophosphatidic acid inhibits the cytotoxic activity of NK cells: involvement of Gs protein-mediated signaling. Int Immunol. 2009;21:667–677. doi: 10.1093/intimm/dxp035. [DOI] [PubMed] [Google Scholar]
  • 40.Bagga S, Price KS, Lin DA, Friend DS, Austen KF, Boyce JA. Lysophosphatidic acid accelerates the development of human mast cells. Blood. 2004;104:4080–4087. doi: 10.1182/blood-2004-03-1166. [DOI] [PubMed] [Google Scholar]
  • 41.Bot M, de Jager SC, MacAleese L, Lagraauw HM, van Berkel TJ, Quax PH, Kuiper J, Heeren RM, Biessen EA, Bot I. Lysophosphatidic acid triggers mast cell-driven atherosclerotic plaque destabilization by increasing vascular inflammation. J Lipid Res. 2013;54:1265–1274. doi: 10.1194/jlr.M032862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hashimoto T, Ohata H, Honda K. Lysophosphatidic acid (LPA) induces plasma exudation and histamine release in mice via LPA receptors. J Pharmacol Sci. 2006;100:82–87. doi: 10.1254/jphs.fpj05030x. [DOI] [PubMed] [Google Scholar]
  • 43.Lin DA, Boyce JA. IL-4 regulates MEK expression required for lysophosphatidic acid-mediated chemokine generation by human mast cells. J Immunol. 2005;175:5430–5438. doi: 10.4049/jimmunol.175.8.5430. [DOI] [PubMed] [Google Scholar]
  • 44.Lundequist A, Boyce JA. LPA5 is abundantly expressed by human mast cells and important for lysophosphatidic acid induced MIP-1beta release. PLoS ONE. 2012;6:e18192. doi: 10.1371/journal.pone.0018192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hashimoto T, Yamashita M, Ohata H, Momose K. Lysophosphatidic acid enhances in vivo infiltration and activation of guinea pig eosinophils and neutrophils via a Rho/Rho-associated protein kinase-mediated pathway. J Pharmacol Sci. 2003;91:8–14. doi: 10.1254/jphs.91.8. [DOI] [PubMed] [Google Scholar]
  • 46.Rahaman M, Costello RW, Belmonte KE, Gendy SS, Walsh MT. Neutrophil sphingosine 1-phosphate and lysophosphatidic acid receptors in pneumonia. Am J Respir Cell Mol Biol. 2006;34:233–241. doi: 10.1165/rcmb.2005-0126OC. [DOI] [PubMed] [Google Scholar]
  • 47.Tou JS, Gill JS. Lysophosphatidic acid increases phosphatidic acid formation, phospholipase D activity and degranulation by human neutrophils. Cell Signal. 2005;17:77–82. doi: 10.1016/j.cellsig.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 48.Koh JS, Lieberthal W, Heydrick S, Levine JS. Lysophosphatidic acid is a major serum noncytokine survival factor for murine macrophages which acts via the phosphatidylinositol 3-kinase signaling pathway. J Clin Invest. 1998;102:716–727. doi: 10.1172/JCI1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chang CL, Lin ME, Hsu HY, Yao CL, Hwang SM, Pan CY, Hsu CY, Lee H. Lysophosphatidic acid-induced interleukin-1 beta expression is mediated through Gi/Rho and the generation of reactive oxygen species in macrophages. Journal of biomedical science. 2008;15:357–363. doi: 10.1007/s11373-007-9223-x. [DOI] [PubMed] [Google Scholar]
  • 50.Mishra RS, Carnevale KA, Cathcart MK. iPLA2beta: front and center in human monocyte chemotaxis to MCP-1. J Exp Med. 2008;205:347–359. doi: 10.1084/jem.20071243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rosskopf D, Daelman W, Busch S, Schurks M, Hartung K, Kribben A, Michel MC, Siffert W. Growth factor-like action of lysophosphatidic acid on human B lymphoblasts. Am J Physiol. 1998;274:C1573–1582. doi: 10.1152/ajpcell.1998.274.6.C1573. [DOI] [PubMed] [Google Scholar]
  • 52.Mu H, Calderone TL, Davies MA, Prieto VG, Wang H, Mills GB, Bar-Eli M, Gershenwald JE. Lysophosphatidic acid induces lymphangiogenesis and IL-8 production in vitro in human lymphatic endothelial cells. Am J Pathol. 2012;180:2170–2181. doi: 10.1016/j.ajpath.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Panther E, Idzko M, Corinti S, Ferrari D, Herouy Y, Mockenhaupt M, Dichmann S, Gebicke-Haerter P, Di Virgilio F, Girolomoni G, Norgauer J. The influence of lysophosphatidic acid on the functions of human dendritic cells. J Immunol. 2002;169:4129–4135. doi: 10.4049/jimmunol.169.8.4129. [DOI] [PubMed] [Google Scholar]
  • 54.Chen R, Roman J, Guo J, West E, McDyer J, Williams MA, Georas SN. Lysophosphatidic acid modulates the activation of human monocyte-derived dendritic cells. Stem Cells Dev. 2006;15:797–804. doi: 10.1089/scd.2006.15.797. [DOI] [PubMed] [Google Scholar]
  • 55.Emo J, Meednu N, Chapman TJ, Rezaee F, Balys M, Randall T, Rangasamy T, Georas SN. Lpa2 Is a Negative Regulator of Both Dendritic Cell Activation and Murine Models of Allergic Lung Inflammation. J Immunol. 2012 doi: 10.4049/jimmunol.1102956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Valet P, Pages C, Jeanneton O, Daviaud D, Barbe P, Record M, Saulnier-Blache JS, Lafontan M. Alpha2-adrenergic receptor-mediated release of lysophosphatidic acid by adipocytes. A paracrine signal for preadipocyte growth. J Clin Invest. 1998;101:1431–1438. doi: 10.1172/JCI806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Giganti A, Rodriguez M, Fould B, Moulharat N, Coge F, Chomarat P, Galizzi JP, Valet P, Saulnier-Blache JS, Boutin JA, Ferry G. Murine and human autotaxin alpha, beta, and gamma isoforms: gene organization, tissue distribution, and biochemical characterization. J Biol Chem. 2008;283:7776–7789. doi: 10.1074/jbc.M708705200. [DOI] [PubMed] [Google Scholar]
  • 58.Dusaulcy R, Rancoule C, Gres S, Wanecq E, Colom A, Guigne C, van Meeteren LA, Moolenaar WH, Valet P, Saulnier-Blache JS. Adipose-specific disruption of autotaxin enhances nutritional fattening and reduces plasma lysophosphatidic acid. J Lipid Res. 2011;52:1247–1255. doi: 10.1194/jlr.M014985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ganguly K, Stoeger T, Wesselkamper SC, Reinhard C, Sartor MA, Medvedovic M, Tomlinson CR, Bolle I, Mason JM, Leikauf GD, Schulz H. Candidate genes controlling pulmonary function in mice: transcript profiling and predicted protein structure. Physiol Genomics. 2007;31:410–421. doi: 10.1152/physiolgenomics.00260.2006. [DOI] [PubMed] [Google Scholar]
  • 60.Georas SN, Berdyshev E, Hubbard W, Gorshkova IA, Usatyuk PV, Saatian B, Myers AC, Williams MA, Xiao HQ, Liu M, Natarajan V. Lysophosphatidic acid is detectable in human bronchoalveolar lavage fluids at baseline and increased after segmental allergen challenge. Clin Exp Allergy. 2007;37:311–322. doi: 10.1111/j.1365-2222.2006.02626.x. [DOI] [PubMed] [Google Scholar]
  • 61.He D, Su Y, Usatyuk PV, Spannhake EW, Kogut P, Solway J, Natarajan V, Zhao Y. Lysophosphatidic acid enhances pulmonary epithelial barrier integrity and protects endotoxin-induced epithelial barrier disruption and lung injury. J Biol Chem. 2009;284:24123–24132. doi: 10.1074/jbc.M109.007393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhao J, He D, Berdyshev E, Zhong M, Salgia R, Morris AJ, Smyth SS, Natarajan V, Zhao Y. Autotaxin induces lung epithelial cell migration through lysoPLD activity-dependent and -independent pathways. Biochem J. 2011;439:45–55. doi: 10.1042/BJ20110274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Oikonomou N, Mouratis MA, Tzouvelekis A, Kaffe E, Valavanis C, Vilaras G, Karameris A, Prestwich GD, Bouros D, Aidinis V. Pulmonary autotaxin expression contributes to the pathogenesis of pulmonary fibrosis. Am J Respir Cell Mol Biol. 2012;47:566–574. doi: 10.1165/rcmb.2012-0004OC. [DOI] [PubMed] [Google Scholar]
  • 64.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. The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat Med. 2008;14:45–54. doi: 10.1038/nm1685. [DOI] [PubMed] [Google Scholar]
  • 65.Aidinis V, Carninci P, Armaka M, Witke W, Harokopos V, Pavelka N, Koczan D, Argyropoulos C, Thwin MM, Moller S, Waki K, Gopalakrishnakone P, Ricciardi-Castagnoli P, Thiesen HJ, Hayashizaki Y, Kollias G. Cytoskeletal rearrangements in synovial fibroblasts as a novel pathophysiological determinant of modeled rheumatoid arthritis. PLoS Genet. 2005;1:e48. doi: 10.1371/journal.pgen.0010048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Nikitopoulou I, Oikonomou N, Karouzakis E, Sevastou I, Nikolaidou-Katsaridou N, Zhao Z, Mersinias V, Armaka M, Xu Y, Masu M, Mills GB, Gay S, Kollias G, Aidinis V. Autotaxin expression from synovial fibroblasts is essential for the pathogenesis of modeled arthritis. J Exp Med. 2012;209:925–933. doi: 10.1084/jem.20112012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nikitopoulou I, Kaffe E, Sevastou I, Sirioti I, Samiotaki M, Madan D, Prestwich GD, Aidinis V. A metabolically-stabilized phosphonate analog of lysophosphatidic Acid attenuates collagen-induced arthritis. PLoS One. 2013;8:e70941. doi: 10.1371/journal.pone.0070941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Orosa B, Garcia S, Martinez P, Gonzalez A, Gomez-Reino JJ, Conde C. Lysophosphatidic acid receptor inhibition as a new multipronged treatment for rheumatoid arthritis. Ann Rheum Dis. 2013 doi: 10.1136/annrheumdis-2012-202832. [DOI] [PubMed] [Google Scholar]
  • 69.Li S, Zhang J. Lipopolysaccharide induces autotaxin expression in human monocytic THP-1 cells. Biochem Biophys Res Commun. 2009;378:264–268. doi: 10.1016/j.bbrc.2008.11.047. [DOI] [PubMed] [Google Scholar]
  • 70.Rennard SI, Basset G, Lecossier D, O’Donnell KM, Pinkston P, Martin PG, Crystal RG. Estimation of volume of epithelial lining fluid recovered by lavage using urea as marker of dilution. J Appl Physiol. 1986;60:532–538. doi: 10.1152/jappl.1986.60.2.532. [DOI] [PubMed] [Google Scholar]
  • 71.Park GY, Lee YG, Berdyshev E, Nyenhuis S, Du J, Fu P, Gorshkova IA, Li Y, Chung S, Karpurapu M, Deng J, Ranjan R, Xiao L, Jaffe HA, Corbridge SJ, Kelly EA, Jarjour NN, Chun J, Prestwich GD, Kaffe E, Ninou I, Aidinis V, Morris AJ, Smyth SS, Ackerman SJ, Natarajan V, Christman JW. Autotaxin production of lysophosphatidic Acid mediates allergic asthmatic inflammation. Am J Respir Crit Care Med. 2013;188:928–940. doi: 10.1164/rccm.201306-1014OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Idzko M, Laut M, Panther E, Sorichter S, Durk T, Fluhr JW, Herouy Y, Mockenhaupt M, Myrtek D, Elsner P, Norgauer J. Lysophosphatidic acid induces chemotaxis, oxygen radical production, CD11b up-regulation, Ca2+ mobilization, and actin reorganization in human eosinophils via pertussis toxin-sensitive G proteins. J Immunol. 2004;172:4480–4485. doi: 10.4049/jimmunol.172.7.4480. [DOI] [PubMed] [Google Scholar]
  • 73.Barekzi E, Roman J, Hise K, Georas S, Steinke JW. Lysophosphatidic acid stimulates inflammatory cascade in airway epithelial cells. Prostaglandins Leukot Essent Fatty Acids. 2006;74:357–363. doi: 10.1016/j.plefa.2006.03.004. [DOI] [PubMed] [Google Scholar]
  • 74.Medoff BD, Landry AL, Wittbold KA, Sandall BP, Derby MC, Cao Z, Adams JC, Xavier RJ. CARMA3 mediates lysophosphatidic acid-stimulated cytokine secretion by bronchial epithelial cells. Am J Respir Cell Mol Biol. 2009;40:286–294. doi: 10.1165/rcmb.2008-0129OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Toews ML, Ustinova EE, Schultz HD. Lysophosphatidic acid enhances contractility of isolated airway smooth muscle. J Appl Physiol. 1997;83:1216–1222. doi: 10.1152/jappl.1997.83.4.1216. [DOI] [PubMed] [Google Scholar]
  • 76.Cerutis DR, Nogami M, Anderson JL, Churchill JD, Romberger DJ, Rennard SI, Toews ML. Lysophosphatidic acid and EGF stimulate mitogenesis in human airway smooth muscle cells. Am J Physiol. 1997;273:L10–15. doi: 10.1152/ajplung.1997.273.1.L10. [DOI] [PubMed] [Google Scholar]
  • 77.Hashimoto T, Nakano Y, Ohata H, Momose K. Lysophosphatidic acid enhances airway response to acetylcholine in guinea pigs. Life Sci. 2001;70:199–205. doi: 10.1016/s0024-3205(01)01382-0. [DOI] [PubMed] [Google Scholar]
  • 78.Fan H, Zingarelli B, Harris V, Tempel GE, Halushka PV, Cook JA. Lysophosphatidic acid inhibits bacterial endotoxin-induced pro-inflammatory response: potential anti-inflammatory signaling pathways. Mol Med. 2008;14:422–428. doi: 10.2119/2007-00106.Fan. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhao Y, He D, Zhao J, Wang L, Leff AR, Spannhake EW, Georas S, Natarajan V. Lysophosphatidic acid induces interleukin-13 (IL-13) receptor alpha2 expression and inhibits IL-13 signaling in primary human bronchial epithelial cells. J Biol Chem. 2007;282:10172–10179. doi: 10.1074/jbc.M611210200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Xu MY, Porte J, Knox AJ, Weinreb PH, Maher TM, Violette SM, McAnulty RJ, Sheppard D, Jenkins G. Lysophosphatidic acid induces alphavbeta6 integrin-mediated TGF-beta activation via the LPA2 receptor and the small G protein G alpha(q) Am J Pathol. 2009;174:1264–1279. doi: 10.2353/ajpath.2009.080160. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES