Journal List > Int J Stem Cells > v.13(2) > 1158835

Lee, Kim, and Kim: The Role of Lysophosphatidic Acid in Adult Stem Cells

Abstract

Stem cells are undifferentiated multipotent precursor cells that are capable both of perpetuating themselves as stem cells (self-renewal) and of undergoing differentiation into one or more specialized types of cells. And these stem cells have been reported to reside within distinct anatomic locations termed “niches”. The long-term goals of stem cell biology range from an understanding of cell-lineage determination and tissue organization to cellular therapeutics for degenerative diseases. Stem cells maintain tissue function throughout an organism’s lifespan by replacing differentiated cells. To perform this function, stem cells provide a unique combination of multilineage developmental potential and the capacity to undergo self-renewing divisions. The loss of self-renewal capacity in stem cells underlies certain degenerative diseases and the aging process. This self-renewal regulation must balance the regenerative needs of tissues that persist throughout life. Recent evidence suggests lysophosphatidic acid (LPA) signaling pathway plays an important role in the regulation of a variety of stem cells. In this review, we summarize the evidence linking between LPA and stem cell regulation. The LPA-induced signaling pathway regulates the proliferation and survival of stem cells and progenitors, and thus are likely to play a role in the maintenance of stem cell population in the body. This lipid mediator regulatory system can be a novel potential therapeutics for stem cell maintenance.

Introduction

LPA is a phospholipid that induces a variety of cellular responses in most cell types, including intracellular calcium mobilization, stress fiber formation, cell rounding, neurite retraction, proliferation, cell survival, migration, and differentiation (1-4). The LPA-induced cellular responses occur through activation of their G-coupled LPA receptors, and LPA activates these receptors through heterotrimeric Gα proteins (5, 6). Up to date, six G-coupled LPA receptors (LPA1-6) have been identified, and they have a broad tissue distribution (7). LPA1-3 receptors have been shown to mediate their cellular effects through mechanisms involving phospholipase C activation and calcium mobilization (3, 8, 9). Whereas, LPA1 and LPA2 receptors can mediate LPA-induced Rho activation required for morphological effects (10-15). LPA canbe produced from lysophosphatidylcholine (LPC) by removal of the choline moiety by the lysophospholipase D (lyso-PLD) (16). LPC is an intermediate in multiple lipid metabolic pathways. These results suggest the distinct signaling mechanisms of LPA receptors are responsible for the diverse function of LPA in different cell types (Fig. 1).

Role of the LPA in Embryonic Development

During the development, LPA is involved in various biological processes, including brain development (17-19). LPA mediates numerous aspects of progenitor behavior, including proliferation and cell cycle-associated morphological changes in the embryonic cerebral cortex (20, 21). The LPA1 receptor is abundantly expressed in progenitor cells of the embryonic cerebral cortex (21, 22). LPA1 receptor knockout (KO) mice were approximately 50% neonatal lethality and result in craniofacial dysmorphism due to defective suckling behavior, and generation of a small fraction of pups with a frontal hematoma (23). However, LPA2 receptor KO mice displayed no obvious phenotypic abnormalities. LPA1/2 receptors double knockout (DKO) mice displayed no additional phenotypic abnormalities relative to LPA1 receptor KO mice except for an increased incidence of perinatal frontal hematoma (17). Furthermore, LPA-induced responses, including phospholipase C activation, Ca2+mobilization, adenylyl cyclase activation, proliferation, JNK activation, AKT activation, and stress fiber formation were absent or severely reduced from LPA1/2 receptors DKO mouse embryonic fibroblast. Thus, these results supported a role for LPA signaling through the LPA1 receptor in nervous system development. LPA3 receptor-deficient female mice showed delayed embryo implantation, altered embryo spacing, and reduced litter size, resulting in the delayedembryonic development and hypertrophic placentas and embryonic death (24). This was attributed to a down-regulationof cyclooxygenase 2 which led to reduced levels of prostaglandins E2 and I2, which are essential players in implantation (17). The LPA4 receptor was shown to mediate the LPA-induced suppression of cell migration in vitro (25). LPA4 receptorKO embryos died during embryonic development and showed several abnormalities in the blood and lymphatic vascular system (26). LPA4 receptor deletion caused a potentiation of AKT and Rac activation, implying that the LPA4 receptor negatively regulates the PI3K pathway, which is in contrast to activation of this pathway by other LPA receptors (27).

LPA in the Regulation of Pluripotent Stem Cells

Embryonic stem cells are derived from the blastocyst stage of early mammalian embryos, are distinguished by their ability to differentiate into any embryonic cell type and by their ability to self-renew. The totipotent cells are the fertilized eggs of mammals and able to generate new individuals (28). Embryonic stem cells are pluripotent, having the ability to generate all body and extra-embryonic tissues. Also, embryonic stem cells have a normal karyotype, maintaininghigh telomerase activity, and exhibit remarkable long-term proliferative potential (29).
In the mouse embryonic stem cells, the LPA5 receptor has been identified (30, 31), and while the physiological relevance of LPA in mouse embryonic stem cells has not been established, LPA is known to stimulate the phosphorylation of ERK and JNK and result in the c-Fos induction (32). In the human embryonic stem cells, LPA1-3 receptors have been identified (33, 34), and LPA maintains undifferentiated human embryonic stem cell lines in the presence of mouse embryonic fibroblasts (33). These human embryonic stem cells retain functional gap junctions (35).
Induced pluripotent stem cells are pluripotent stem cells that can be reprogramed directly from somatic cells by introducing four specific genes (c-Myc, Oct3/4, Sox2, and Klf4) (36). Recent studies demonstrate that LPA modulates the Hippo pathway in both human embryonic stem cells and human-induced pluripotent stem cells by activating YAP/TAZ (37, 38). These data suggest that differential LPA receptors affect pluripotent stem cell maintenance.

LPA in the Regulation of Neural Stem Cells

Neural stem cells are self-renewing, multipotent cells that firstly generate the radial glial progenitor cells that generate the neurons and glia of the nervous system of all animals during embryonic development (39). These neural stem cells are located in the subventricular zone and the spinal cord (40). These stem cells can give rise to either neural or neuronal progenitor cells and are involved in the neurogenesis of the central nervous system.
A recent study has been reported that neural stem cells migrate to the sites of injury for the repair of damaged tissue (41). LPA signaling influences several developmental processes within the nervous system (18). LPA1-3 receptors are expressed in neural stem cells (42). LPA is found in the embryonic brain, neural tube, choroid plexus, meninges, blood vessels, spinal cord and cerebrospinal fluid (5), and regulates morphological rearrangements, proliferation, and differentiation of neural stem cells (21, 23, 43, 44). Neural stem cells can be maintained in culture as neurospheres by the presence of basic fibroblast growth factor and epidermal growth factor (45, 46). LPA inhibited the basic fibroblast growth factor-induced growth of neurospheres from cortical neural stem cells (47). In contrast, LPA has been shown to induce neurosphere formation from mouse forebrain neural stem cells (42). In rat cortical neural stem and progenitor cells, while LPA stimulates neuronal differentiation and migration, low concentrations of LPA induce proliferation (48). Besides, it has been reported that LPA does not induce proliferation but affects morphological rearrangements in rat hippocampal neural stem and progenitor cells (49-51).
LPA stimulates neuronal differentiation of cortical neuroblasts, neural progenitors, and early cortical neurons via the LPA1 receptor (44, 47). LPA is an essential factor for cortical neurogenesis (52) that induces the depolarization of mouse cortical neuroblasts and activates the electrical responses in neuroblasts via GABA signaling (53). Oligodendrocyte progenitors share properties with both stem cells and progenitor cells and give rise to oligodendrocytes which is responsible for neuron myelination within the central nervous system (54, 55). LPA1 receptor on oligodendrocyte progenitors has only been examined in rodents and inducesthe retraction of processes of oligodendrocyte progenitors (56, 57). Further, LPA was shown to induce cell proliferation in cultured astrocytes, which express Lpa1, Lpa2, and Lpa3 genes (58, 59). Also, LPA stimulates astrocyte proliferation through the LPA1 receptor (60). LPA1 receptor gene expression may be induced by LPA stimulation in astrocytes in vivo. Together, these studies suggest differential roles of LPA in the biology of neural stem cells and their progenitors (Fig. 2).

LPA in the Regulation of Hematopoietic Stem Cells

Hematopoietic stem cells mainly reside in the microenvironments of the bone marrow, where they pass through multiple developmental steps to produce mature blood cells (61, 62). Hematopoietic stem cells give rise to different types of blood cells, in lines called myeloid and lymphoid (62). Myeloid cells include monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, and megakaryocytes to platelets. Lymphoid cells include T cells, B cells, natural killer cells, and innate lymphoid cells.
LPA induced migration of c-Kit+ cell and enhanced the chemotactic migratory response of the primitive hematopoietic stem cells to stromal-derived factor 1 through a mechanism involving the activation of the Rac, Rho, and Cdc42 proteins (63). LPA decreased the adhesion of the myeloid progenitor cell line TF1 through a Rho-dependent pathway (64). LPA facilitates the migration of CD34+ hematopoietic progenitor cells (65) and triggers an invasion of hematopoietic stem cells to the stromal cell layer (66). Furthermore, LPA participates in EPO-dependent erythropoiesis by activating the LPA3 receptor (67). Also, in vitro stimulation of CD34+ human hematopoietic progenitors by LPA induced myeloid differentiation but did not affect lymphoid differentiation (68).
LPA receptors were expressed at significantly higher levels on common myeloid progenitors than common lymphoid progenitors suggesting that LPA acts on lineage specification (68). Especially, LPA1 and LPA2 receptors are expressed in LinSca1+c-Kit+ hematopoietic stem and progenitor cells (63). In contrast, the less primitive cKit cells expressed the LPA2 receptor, but not the LPA1 receptor. Also, the LPA1 receptor has important roles in the regulation of migration in hematopoietic stem cells (65). The pharmacological and genetic blockage of the LPA1 receptor inhibited hematopoietic differentiation of mouse embryonic stem cells and impaired the formation of hemangioblasts (69). In K562 human erythroleukemia cells, knockdown of LPA2 receptor enhanced erythropoiesis, whereas knockdown of LPA3 receptor inhibited RBC differentiation (70). The pharmacological activation of LPA receptors can be novel strategies for augmenting or inhibiting erythropoiesis and/or hematopoiesis.

LPA in the Regulation of Mesenchymal Stem Cells

Mesenchymal stem cells are multipotent stromal cells that can differentiate into a variety of cell types, including osteoblasts, chondrocytes, myocytes, and adipocytes (71, 72). These mesenchymal stem cells play an important role in hematopoietic stem cell differentiation into mature blood cells (73-75). Moreover, bone marrow stromal cells can differentiate into adipose, tendon, cartilage, and bone (76, 77).
LPA1-3 receptors have been identified in bone marrow mesenchymal stem cells, and LPA influenced the survival of mesenchymal stem cells (40, 78). LPA increased mesenchymal stem cells survival through ERK1/2 and PI3K/AKT signaling pathway and inhibitshypoxia/serum deprivation-induced apoptosis in mesenchymal stem cells. LPA also inhibited caspase12 pathways via inhibition of p38 signaling in mesenchymal stem cells, and it is involved in LPA1 and LPA3 receptors-linked ERK1/2 pathway (79). This study suggests LPA as a new anti-apoptotic target of mesenchymal stem cells for the therapeuticpotential of cardiac repair. Further, LPA stimulated the migration of bone marrow stromal cells via a Rho-dependent mechanism (80, 81). LPA stimulated VEGF secretion in mesenchymal stem cells but not in cardiomyocytes or cardiac fibroblasts via LPA1 and LPA3 receptors (82). Also, LPA promoted osteogenic differentiation in human mesenchymal stem cells via LPA1 and LPA4 receptors (83). These results suggest a potential role of LPA in the regulation of self-renewal and differentiation of mesenchymal stem cells.

LPA in the Regulation of Hepatic Stem Cells

Oval cells, also known as hepatic stem cells, are reportedly involved in the regeneration of the liver following injury (84), whereas relatively little is known about their response to LPA. During chronic liver injury, oval stem cell proliferation is associated with the up-regulation of the expression of the LPA1-3 receptors (85). Taken together, these results suggest that LPA plays critical roles during liver regeneration.

LPA in the Regulation of Cancer Stem Cells

Cancer stem cells were first identified by Bonnet and Dick in acute myeloid leukemia in 1997 (86). Cancer stem cells are cancer cells that have self-renewal capacity and show tumorigenic potential (87-89). These cancer stem cells are hypothesized to persist in tumors as a distinct population and cause relapse and metastasis (90). Cancer stem cells have implications for cancer therapy, including for disease identification, selective drug targets, prevention of metastasis. Thus, the development of targeted therapies can improve the survival and quality of life of cancer patients, especially for patients with metastatic disease.
Elevated LPA levels have been detected in the ascites of 98% of ovarian cancer patients (27). LPA treatment stimulates the expression of OCT4, SOX2, and ALDH1 genes which are cancer stem cell-associated, and drug transporters in ovarian cancer stem cells (91). In ovarian cancer stem cells, LPA was found to upregulate the expression of hypoxia-inducible factor-1-alpha, which plays a central role in tumor angiogenesis (92), whereas this was inhibited by the knockdown of LPA2 or LPA3 receptors (93). These results show that LPA is implicated in tumor angiogenesis.
Moreover, LPA promoted cancer stem cell-like characteristics, such as sphere-forming ability, resistance to anti-cancer drugs, and tumorigenic potential in xenograft transplantation (91). Knockdown or pharmacological inhibition of the LPA1 receptor reduced the LPA-stimulated proliferation and acquisition of cancer stem cell-like properties in ovarian cancer cells (88). These results suggest that LPA plays a key role in the self-renewal, therapeutic resistance, and metastasis of ovarian cancer stem cells.

LPA in the Regulation of Adult Somatic Cells

LPA is also implicated in vasculardevelopment and endothelial cell development, such as vasculogenesis, angiogenesis, and vascular maturation during the development. The first study that linked LPA to vascular development was that of the Autotaxin KO mice during the development (94). Autotaxin, a member of the ectonucleotide pyrophosphate and phosphatase family, primarily catalyzes the hydrolysis of lysophosphatidylcholine, resulting in LPA production (7, 95). Autotaxin-deficient mice die at E9.5 with profound vascular defects in the yolk sac and embryo, and neural tube malformations. LPA1/2 receptors DKO mice also showed blood within the lateral ventricles during the development. LPA was observed to facilitate the closure of wounded endothelial cells in vitro. This was due to a stimulation of both endothelial cell migration and proliferation by LPA treatment (96). Also, LPA modulates circulating monocyte migration via LPA-stimulated endothelial cells (97).
LPA has also been implicated in the regulation of pathophysiologic vascular responses in the endothelial and vascular smooth muscle cells. LPA was found to signal through Gαq to promote the growth and migration of vascular smooth muscle cells (98). Mice deficient in LPA1 and LPA2 receptors were protected from intimal hyperplasia (99) that induce migration of smooth muscle cells. LPA promoted human platelet development by inducing platelet shape change and calcium mobilization (27). LPA also induced the aggregation of platelets, which is essential in thrombosis (100). LPA is involved in vascular injury by causing endothelial cell death and vascular degeneration (101, 102). Loss of confluence and vascular integrity of endothelial cells was observed in vivo when exposed to LPA. Collectively, recent studies indicate that LPA signaling plays a role in the development of the vasculature and endothelial cells.
Liver sinusoidal endothelial cells constitute the innermost layer of hepatic blood vessels (103), and thus, directly encounter circulating erythrocytes and immune cells and provide a physical barrier for underlying hepatocytes (104). LSECs contribute to the maintenance of liver homeostasis and participate in metabolite transportation, immune regulation, and the development or resolution of pathological conditions, such asinflammation, cirrhosis, hepatocellular carcinoma, and liver regeneration (105, 106). LPA1 and LPA3 receptors are expressed in mouse liver sinusoidal endothelial cells (107). LPA has potent effects on cell migration (96) and membrane permeability in liver sinusoidal endothelial cell membranes (108).
In the immune cells, LPA enhances the motility of human and mouse T cells in vitro (109-111), induces human natural killer cell chemotaxis (112), accelerates the development of human mast cells (113), influences Neutrophils (114-116), Macrophages (117), and B cells (118). Also, LPA induces chemotaxis of immature mouse bone marrow-derived Dendritic cells (119).
These results suggest that LPA plays a key role in lymphocyte homing and inflammation.

Conclusions

Remarkable progress has been made in the last decade in understanding the role of LPA in the regulation of stem cells. This has been driven largely by the discovery of G protein-coupled receptors for LPA and also by the characterization of many of the enzymes involved in lysophospholipid metabolism. LPA controlsthe reproductive, gastrointestinal, vascular, nervous, and immune systems (120), and it plays an important role in the regulation of stem cells and their progenitors (121). LPA also appears to regulate the proliferation, survival, mobilization, migration, and differentiation of the stem cells. Thus, it is likely that LPA plays a role in the maintenance of stem cell populations in the body. Collectively, the modulation of LPA-mediated signaling pathways may provide opportunities for future therapeutics by regulating the self-renewal and differentiation of stem cells.

Acknowledgments

This work was supported by a 2-year research grant from Pusan National University (D.L.).

Notes

Potential Conflict of Interest

The authors have no conflicting financial interest.

References

1. Valdés-Rives SA, González-Arenas A. 2017; Autotaxin-lysophosphatidic acid: from inflammation to cancer development. Mediators Inflamm. 2017:9173090. DOI: 10.1155/2017/9173090. PMID: 29430083. PMCID: PMC5753009.
crossref
2. Chun J. 1999; Lysophospholipid receptors: implications for neural signaling. Crit Rev Neurobiol. 13:151–168. DOI: 10.1615/CritRevNeurobiol.v13.i2.20. PMID: 10512488.
crossref
3. Fukushima N, Ishii I, Contos JJ, Weiner JA, Chun J. 2001; Lysophospholipid receptors. Annu Rev Pharmacol Toxicol. 41:507–534. DOI: 10.1146/annurev.pharmtox.41.1.507. PMID: 11264467.
crossref
4. Moolenaar WH, Kranenburg O, Postma FR, Zondag GC. 1997; Lysophosphatidic acid: G-protein signalling and cellular responses. Curr Opin Cell Biol. 9:168–173. DOI: 10.1016/S0955-0674(97)80059-2. PMID: 9069262.
crossref
5. Yung YC, Stoddard NC, Chun J. 2014; LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res. 55:1192–1214. DOI: 10.1194/jlr.R046458. PMID: 24643338. PMCID: PMC4076099.
crossref
6. Leblanc R, Peyruchaud O. 2015; New insights into the autotaxin/LPA axis in cancer development and metastasis. Exp Cell Res. 333:183–189. DOI: 10.1016/j.yexcr.2014.11.010. PMID: 25460336.
crossref
7. van Meeteren LA, Moolenaar WH. 2007; Regulation and biological activities of the autotaxin-LPA axis. Prog Lipid Res. 46:145–160. DOI: 10.1016/j.plipres.2007.02.001. PMID: 17459484.
crossref
8. Lynch KR. 2002; Lysophospholipid receptor nomenclature. Biochim Biophys Acta. 1582:70–71. DOI: 10.1016/S1388-1981(02)00138-5. PMID: 12069811.
crossref
9. Contos JJ, Ishii I, Chun J. 2000; Lysophosphatidic acid receptors. Mol Pharmacol. 58:1188–1196. DOI: 10.1124/mol.58.6.1188. PMID: 11093753.
crossref
10. An S, Bleu T, Hallmark OG, Goetzl EJ. 1998; Characterization of a novel subtype of human G protein-coupled receptor for lysophosphatidic acid. J Biol Chem. 273:7906–7910. DOI: 10.1074/jbc.273.14.7906. PMID: 9525886.
crossref
11. An S, Bleu T, Zheng Y, Goetzl EJ. 1998; Recombinant human G protein-coupled lysophosphatidic acid receptors mediate intracellular calcium mobilization. Mol Pharmacol. 54:881–888. DOI: 10.1124/mol.54.5.881. PMID: 9804623.
crossref
12. Bandoh K, Aoki J, Hosono H, Kobayashi S, Kobayashi T, Murakami-Murofushi K, Tsujimoto M, Arai H, Inoue K. 1999; Molecular cloning and characterization of a novel human G-protein-coupled receptor, EDG7, for lysophosphatidic acid. J Biol Chem. 274:27776–27785. DOI: 10.1074/jbc.274.39.27776. PMID: 10488122.
crossref
13. Fukushima N, Kimura Y, Chun J. 1998; A single receptor encoded by vzg-1/lpA1/edg-2 couples to G proteins and mediates multiple cellular responses to lysophosphatidic acid. Proc Natl Acad Sci U S A. 95:6151–6156. DOI: 10.1073/pnas.95.11.6151. PMID: 9600933. PMCID: PMC27607.
crossref
14. Im DS, Heise CE, Harding MA, George SR, O'Dowd BF, Theodorescu D, Lynch KR. 2000; Molecular cloning and characterization of a lysophosphatidic acid receptor, Edg-7, expressed in prostate. Mol Pharmacol. 57:753–759. DOI: 10.1124/mol.57.4.753. PMID: 10727522.
crossref
15. Ishii I, Contos JJ, Fukushima N, Chun J. 2000; Functional comparisons of the lysophosphatidic acid receptors, LP(A1)/VZG-1/EDG-2, LP(A2)/EDG-4, and LP(A3)/EDG-7 in neuronal cell lines using a retrovirus expression system. Mol Pharmacol. 58:895–902. DOI: 10.1124/mol.58.5.895. PMID: 11040035.
crossref
16. Nakamura K, Kishimoto T, Ohkawa R, Okubo S, Tozuka M, Yokota H, Ikeda H, Ohshima N, Mizuno K, Yatomi Y. 2007; 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. 367:20–27. DOI: 10.1016/j.ab.2007.05.004. PMID: 17568554.
crossref
17. Contos JJ, Ishii I, Fukushima N, Kingsbury MA, Ye X, Kawamura S, Brown JH, Chun J. 2002; Characterization of lpa2 (Edg4) and lpa1/lpa2 (Edg2/Edg4) lysophosphatidic acid receptor knockout mice: signaling deficits without obvious phenotypic abnormality attributable to lpa2. Mol Cell Biol. 22:6921–6929. DOI: 10.1128/MCB.22.19.6921-6929.2002. PMID: 12215548. PMCID: PMC134025.
crossref
18. Yung YC, Stoddard NC, Mirendil H, Chun J. 2015; Lysophosphatidic acid signaling in the nervous system. Neuron. 85:669–682. DOI: 10.1016/j.neuron.2015.01.009. PMID: 25695267. PMCID: PMC4400838.
crossref
19. Sheng X, Yung YC, Chen A, Chun J. 2015; Lysophosphatidic acid signalling in development. Development. 142:1390–1395. DOI: 10.1242/dev.121723. PMID: 25852197. PMCID: PMC4392601.
crossref
20. Fukushima N, Weiner JA, Kaushal D, Contos JJ, Rehen SK, Kingsbury MA, Kim KY, Chun J. 2002; Lysophosphatidic acid influences the morphology and motility of young, postmitotic cortical neurons. Mol Cell Neurosci. 20:271–282. DOI: 10.1006/mcne.2002.1123. PMID: 12093159.
crossref
21. Hecht JH, Weiner JA, Post SR, Chun J. 1996; Ventricular zone gene-1 (vzg-1) encodes a lysophosphatidic acid receptor expressed in neurogenic regions of the developing cerebral cortex. J Cell Biol. 135:1071–1083. DOI: 10.1083/jcb.135.4.1071. PMID: 8922387. PMCID: PMC2133395.
crossref
22. Weiner JA, Chun J. 1999; Schwann cell survival mediated by the signaling phospholipid lysophosphatidic acid. Proc Natl Acad Sci U S A. 96:5233–5238. DOI: 10.1073/pnas.96.9.5233. PMID: 10220449. PMCID: PMC21847.
crossref
23. Contos JJ, Fukushima N, Weiner JA, Kaushal D, Chun J. 2000; Requirement for the lpA1 lysophosphatidic acid receptor gene in normal suckling behavior. Proc Natl Acad Sci U S A. 97:13384–13389. DOI: 10.1073/pnas.97.24.13384. PMID: 11087877. PMCID: PMC27233.
crossref
24. Ye X, Hama K, Contos JJ, Anliker B, Inoue A, Skinner MK, Suzuki H, Amano T, Kennedy G, Arai H, Aoki J, Chun J. 2005; LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature. 435:104–108. DOI: 10.1038/nature03505. PMID: 15875025. PMCID: PMC1369590.
crossref
25. Lee Z, Cheng CT, Zhang H, Subler MA, Wu J, Mukherjee A, Windle JJ, Chen CK, Fang X. 2008; Role of LPA4/p2y9/GPR23 in negative regulation of cell motility. Mol Biol Cell. 19:5435–5445. DOI: 10.1091/mbc.e08-03-0316. PMID: 18843048. PMCID: PMC2592656.
26. 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 embryogenesis. Blood. 116:5060–5070. DOI: 10.1182/blood-2010-03-272443. PMID: 20713964.
crossref
27. Teo ST, Yung YC, Herr DR, Chun J. 2009; Lysophosphatidic acid in vascular development and disease. IUBMB Life. 61:791–799. DOI: 10.1002/iub.220. PMID: 19621353. PMCID: PMC4307796.
crossref
28. Verfaillie CM, Pera MF, Lansdorp PM. 2002; Stem cells: hype and reality. Hematology Am Soc Hematol Educ Program. 369–391. DOI: 10.1182/asheducation-2002.1.369. PMID: 12446433.
crossref
29. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. 1998; Embryonic stem cell lines derived from human blastocysts. Science. 282:1145–1147. DOI: 10.1126/science.282.5391.1145. PMID: 9804556.
crossref
30. Lee CW, Rivera R, Gardell S, Dubin AE, Chun J. 2006; GPR92 as a new G12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. J Biol Chem. 281:23589–23597. DOI: 10.1074/jbc.M603670200. PMID: 16774927.
crossref
31. Kleger A, Busch T, Liebau S, Prelle K, Paschke S, Beil M, Rolletschek A, Wobus A, Wolf E, Adler G, Seufferlein T. 2007; The bioactive lipid sphingosylphosphorylcholine induces differentiation of mouse embryonic stem cells and human promyelocytic leukaemia cells. Cell Signal. 19:367–377. DOI: 10.1016/j.cellsig.2006.07.015. PMID: 16978842.
crossref
32. Schuck S, Soloaga A, Schratt G, Arthur JS, Nordheim A. 2003; The kinase MSK1 is required for induction of c-fos by lysophosphatidic acid in mouse embryonic stem cells. BMC Mol Biol. 4:6. DOI: 10.1186/1471-2199-4-6. PMID: 12769834. PMCID: PMC161794.
33. Pébay A, Wong RC, Pitson SM, Wolvetang EJ, Peh GS, Filipczyk A, Koh KL, Tellis I, Nguyen LT, Pera MF. 2005; Essential roles of sphingosine-1-phosphate and platelet-derived growth factor in the maintenance of human embryonic stem cells. Stem Cells. 23:1541–1548. DOI: 10.1634/stemcells.2004-0338. PMID: 16081668.
crossref
34. Inniss K, Moore H. 2006; Mediation of apoptosis and proliferation of human embryonic stem cells by sphingosine-1-phosphate. Stem Cells Dev. 15:789–796. DOI: 10.1089/scd.2006.15.789. PMID: 17253942.
crossref
35. Wong RC, Dottori M, Koh KL, Nguyen LT, Pera MF, Pébay A. 2006; Gap junctions modulate apoptosis and colony growth of human embryonic stem cells maintained in a serum-free system. Biochem Biophys Res Commun. 344:181–188. DOI: 10.1016/j.bbrc.2006.03.127. PMID: 16616002.
crossref
36. Takahashi K, Yamanaka S. 2006; Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. DOI: 10.1016/j.cell.2006.07.024. PMID: 16904174.
crossref
37. Hsiao C, Lampe M, Nillasithanukroh S, Han W, Lian X, Palecek SP. 2016; Human pluripotent stem cell culture density modulates YAP signaling. Biotechnol J. 11:662–675. DOI: 10.1002/biot.201500374. PMID: 26766309. PMCID: PMC4850094.
crossref
38. Qin H, Hejna M, Liu Y, Percharde M, Wossidlo M, Blouin L, Durruthy-Durruthy J, Wong P, Qi Z, Yu J, Qi LS, Sebastiano V, Song JS, Ramalho-Santos M. 2016; YAP Induces Human Naive Pluripotency. Cell Rep. 14:2301–2312. DOI: 10.1016/j.celrep.2016.02.036. PMID: 26947063. PMCID: PMC4807727.
crossref
39. Beattie R, Hippenmeyer S. 2017; Mechanisms of radial glia progenitor cell lineage progression. FEBS Lett. 591:3993–4008. DOI: 10.1002/1873-3468.12906. PMID: 29121403. PMCID: PMC5765500.
crossref
40. Pébay A, Bonder CS, Pitson SM. 2007; Stem cell regulation by lysophospholipids. Prostaglandins Other Lipid Mediat. 84:83–97. DOI: 10.1016/j.prostaglandins.2007.08.004. PMID: 17991611.
crossref
41. Picard-Riera N, Nait-Oumesmar B, Baron-Van Evercooren A. 2004; Endogenous adult neural stem cells: limits and potential to repair the injured central nervous system. J Neurosci Res. 76:223–231. DOI: 10.1002/jnr.20040. PMID: 15048920.
crossref
42. Svetlov SI, Ignatova TN, Wang KK, Hayes RL, English D, Kukekov VG. 2004; Lysophosphatidic acid induces clonal generation of mouse neurospheres via proliferation of Sca-1- and AC133-positive neural progenitors. Stem Cells Dev. 13:685–693. DOI: 10.1089/scd.2004.13.685. PMID: 15684836.
crossref
43. Fukushima N, Weiner JA, Chun J. 2000; Lysophosphatidic acid (LPA) is a novel extracellular regulator of cortical neuroblast morphology. Dev Biol. 228:6–18. DOI: 10.1006/dbio.2000.9930. PMID: 11087622.
crossref
44. Kingsbury MA, Rehen SK, Contos JJ, Higgins CM, Chun J. 2003; Non-proliferative effects of lysophosphatidic acid enhance cortical growth and folding. Nat Neurosci. 6:1292–1299. DOI: 10.1038/nn1157. PMID: 14625558.
crossref
45. Reynolds BA, Weiss S. 1992; Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 255:1707–1710. DOI: 10.1126/science.1553558. PMID: 1553558.
crossref
46. Reynolds BA, Rietze RL. 2005; Neural stem cells and neurospheres--re-evaluating the relationship. Nat Methods. 2:333–336. DOI: 10.1038/nmeth758. PMID: 15846359.
crossref
47. Fukushima N, Shano S, Moriyama R, Chun J. 2007; Lysophosphatidic acid stimulates neuronal differentiation of cortical neuroblasts through the LPA1-G(i/o) pathway. Neurochem Int. 50:302–307. DOI: 10.1016/j.neuint.2006.09.008. PMID: 17056154.
crossref
48. Cui HL, Qiao JT. 2006; Promotive action of lysophosphatidic acid on proliferation of rat embryonic neural stem cells and their differentiation to cholinergic neurons in vitro. Sheng Li Xue Bao. 58:547–555. PMID: 17173189.
49. Harada J, Foley M, Moskowitz MA, Waeber C. 2004; Sphingosine-1-phosphate induces proliferation and morphological changes of neural progenitor cells. J Neurochem. 88:1026–1039. DOI: 10.1046/j.1471-4159.2003.02219.x. PMID: 14756825.
crossref
50. Yuan XB, Jin M, Xu X, Song YQ, Wu CP, Poo MM, Duan S. 2003; Signalling and crosstalk of Rho GTPases in mediating axon guidance. Nat Cell Biol. 5:38–45. DOI: 10.1038/ncb895. PMID: 12510192.
crossref
51. Campbell DS, Holt CE. 2001; Chemotropic responses of retinal growth cones mediated by rapid local protein synthesis and degradation. Neuron. 32:1013–1026. DOI: 10.1016/S0896-6273(01)00551-7. PMID: 11754834.
crossref
52. Kingsbury MA, Rehen SK, Ye X, Chun J. 2004; Genetics and cell biology of lysophosphatidic acid receptor-mediated signaling during cortical neurogenesis. J Cell Biochem. 92:1004–1012. DOI: 10.1002/jcb.20061. PMID: 15258921.
crossref
53. Dubin AE, Bahnson T, Weiner JA, Fukushima N, Chun J. 1999; Lysophosphatidic acid stimulates neurotransmitter-like conductance changes that precede GABA and L-glutamate in early, presumptive cortical neuroblasts. J Neurosci. 19:1371–1381. DOI: 10.1523/JNEUROSCI.19-04-01371.1999. PMID: 9952414. PMCID: PMC6786022.
crossref
54. Zhao C, Fancy SP, Magy L, Urwin JE, Franklin RJ. 2005; Stem cells, progenitors and myelin repair. J Anat. 207:251–258. DOI: 10.1111/j.1469-7580.2005.00456.x. PMID: 16185249. PMCID: PMC1571531.
crossref
55. Weiner JA, Hecht JH, Chun J. 1998; Lysophosphatidic acid receptor gene vzg-1/lpA1/edg-2 is expressed by mature oligodendrocytes during myelination in the postnatal murine brain. J Comp Neurol. 398:587–598. DOI: 10.1002/(SICI)1096-9861(19980907)398:4<587::AID-CNE10>3.0.CO;2-5.
crossref
56. Dawson J, Hotchin N, Lax S, Rumsby M. 2003; Lysophosphatidic acid induces process retraction in CG-4 line oligodendrocytes and oligodendrocyte precursor cells but not in differentiated oligodendrocytes. J Neurochem. 87:947–957. DOI: 10.1046/j.1471-4159.2003.02056.x. PMID: 14622125.
crossref
57. Jaillard C, Harrison S, Stankoff B, Aigrot MS, Calver AR, Duddy G, Walsh FS, Pangalos MN, Arimura N, Kaibuchi K, Zalc B, Lubetzki C. 2005; Edg8/S1P5: an oligodendroglial receptor with dual function on process retraction and cell survival. J Neurosci. 25:1459–1469. DOI: 10.1523/JNEUROSCI.4645-04.2005. PMID: 15703400. PMCID: PMC6726002.
crossref
58. Rao TS, Lariosa-Willingham KD, Lin FF, Palfreyman EL, Yu N, Chun J, Webb M. 2003; Pharmacological characterization of lysophospholipid receptor signal transduction pathways in rat cerebrocortical astrocytes. Brain Res. 990:182–194. DOI: 10.1016/S0006-8993(03)03527-3. PMID: 14568343.
crossref
59. Sorensen SD, Nicole O, Peavy RD, Montoya LM, Lee CJ, Murphy TJ, Traynelis SF, Hepler JR. 2003; Common signaling pathways link activation of murine PAR-1, LPA, and S1P receptors to proliferation of astrocytes. Mol Pharmacol. 64:1199–1209. DOI: 10.1124/mol.64.5.1199. PMID: 14573770.
crossref
60. Shano S, Moriyama R, Chun J, Fukushima N. 2008; Lysophosphatidic acid stimulates astrocyte proliferation through LPA1. Neurochem Int. 52:216–220. DOI: 10.1016/j.neuint.2007.07.004. PMID: 17692995. PMCID: PMC3265988.
61. Adams GB, Scadden DT. 2006; The hematopoietic stem cell in its place. Nat Immunol. 7:333–337. DOI: 10.1038/ni1331. PMID: 16550195.
crossref
62. Scadden DT. 2006; The stem-cell niche as an entity of action. Nature. 441:1075–1079. DOI: 10.1038/nature04957. PMID: 16810242.
crossref
63. Whetton AD, Lu Y, Pierce A, Carney L, Spooncer E. 2003; Lysophospholipids synergistically promote primitive hematopoietic cell chemotaxis via a mechanism involving Vav 1. Blood. 102:2798–2802. DOI: 10.1182/blood-2002-12-3635. PMID: 12829605.
crossref
64. Lai JM, Lu CY, Yang-Yen HF, Chang ZF. 2001; Lysophosphatidic acid promotes phorbol-ester-induced apoptosis in TF-1 cells by interfering with adhesion. Biochem J. 359:227–233. DOI: 10.1042/bj3590227. PMID: 11563987. PMCID: PMC1222139.
crossref
65. Ortlepp C, Steudel C, Heiderich C, Koch S, Jacobi A, Ryser M, Brenner S, Bornhäuser M, Brors B, Hofmann WK, Ehninger G, Thiede C. 2013; Autotaxin is expressed in FLT3-ITD positive acute myeloid leukemia and hematopoietic stem cells and promotes cell migration and proliferation. Exp Hematol. 41:444–461.e4. DOI: 10.1016/j.exphem.2013.01.007. PMID: 23377000.
crossref
66. Yanai N, Matsui N, Furusawa T, Okubo T, Obinata M. 2000; Sphingosine-1-phosphate and lysophosphatidic acid trigger invasion of primitive hematopoietic cells into stromal cell layers. Blood. 96:139–144. DOI: 10.1182/blood.V96.1.139. PMID: 10891442.
crossref
67. Chiang CL, Chen SS, Lee SJ, Tsao KC, Chu PL, Wen CH, Hwang SM, Yao CL, Lee H. 2011; Lysophosphatidic acid induces erythropoiesis through activating lysophosphatidic acid receptor 3. Stem Cells. 29:1763–1773. DOI: 10.1002/stem.733. PMID: 21915944.
crossref
68. Evseenko D, Latour B, Richardson W, Corselli M, Sahaghian A, Cardinal S, Zhu Y, Chan R, Dunn B, Crooks GM. 2013; Lysophosphatidic acid mediates myeloid differentiation within the human bone marrow microenvironment. PLoS One. 8:e63718. DOI: 10.1371/journal.pone.0063718. PMID: 23696850. PMCID: PMC3655943.
crossref
69. Li H, Yue R, Wei B, Gao G, Du J, Pei G. 2014; Lysophosphatidic acid acts as a nutrient-derived developmental cue to regulate early hematopoiesis. EMBO J. 33:1383–1396. DOI: 10.15252/embj.201387594. PMID: 24829209. PMCID: PMC4194126.
crossref
70. Lin KH, Ho YH, Chiang JC, Li MW, Lin SH, Chen WM, Chiang CL, Lin YN, Yang YJ, Chen CN, Lu J, Huang CJ, Tigyi G, Yao CL, Lee H. 2016; Pharmacological activation of lysophosphatidic acid receptors regulates erythropoiesis. Sci Rep. 6:27050. DOI: 10.1038/srep27050. PMID: 27244685. PMCID: PMC4886675.
crossref
71. Ankrum JA, Ong JF, Karp JM. 2014; Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 32:252–260. DOI: 10.1038/nbt.2816. PMID: 24561556. PMCID: PMC4320647.
crossref
72. Mahla RS. 2016; Stem cells applications in regenerative medicine and disease therapeutics. Int J Cell Biol. 2016:6940283. DOI: 10.1155/2016/6940283. PMID: 27516776. PMCID: PMC4969512.
crossref
73. Schofield R. 1978; The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 4:7–25.
74. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. 2003; Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 425:841–846. DOI: 10.1038/nature02040. PMID: 14574413.
crossref
75. Li L, Xie T. 2005; Stem cell niche: structure and function. Annu Rev Cell Dev Biol. 21:605–631. DOI: 10.1146/annurev.cellbio.21.012704.131525. PMID: 16212509.
crossref
76. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. 1999; Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. DOI: 10.1126/science.284.5411.143. PMID: 10102814.
crossref
77. Passier R, Mummery C. 2003; Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc Res. 58:324–335. DOI: 10.1016/S0008-6363(02)00770-8. PMID: 12757867.
crossref
78. Chen J, Baydoun AR, Xu R, Deng L, Liu X, Zhu W, Shi L, Cong X, Hu S, Chen X. 2008; Lysophosphatidic acid protects mesenchymal stem cells against hypoxia and serum deprivation-induced apoptosis. Stem Cells. 26:135–145. DOI: 10.1634/stemcells.2007-0098. PMID: 17932426.
crossref
79. Li Z, Wei H, Liu X, Hu S, Cong X, Chen X. 2010; LPA rescues ER stress-associated apoptosis in hypoxia and serum deprivation-stimulated mesenchymal stem cells. J Cell Biochem. 111:811–820. DOI: 10.1002/jcb.22731. PMID: 20533299.
crossref
80. Jaganathan BG, Ruester B, Dressel L, Stein S, Grez M, Seifried E, Henschler R. 2007; Rho inhibition induces migration of mesenchymal stromal cells. Stem Cells. 25:1966–1974. DOI: 10.1634/stemcells.2007-0167. PMID: 17510214.
crossref
81. Annabi B, Thibeault S, Lee YT, Bousquet-Gagnon N, Eliopoulos N, Barrette S, Galipeau J, Béliveau R. 2003; Matrix metalloproteinase regulation of sphingosine-1-phosphate-induced angiogenic properties of bone marrow stromal cells. Exp Hematol. 31:640–649. DOI: 10.1016/S0301-472X(03)00090-0. PMID: 12842709.
crossref
82. Wei H, Wang F, Wang X, Yang J, Li Z, Cong X, Chen X. 2013; Lysophosphatidic acid promotes secretion of VEGF by increasing expression of 150-kD Oxygen-regulated protein (ORP150) in mesenchymal stem cells. Biochim Biophys Acta. 1831:1426–1434. DOI: 10.1016/j.bbalip.2013.05.003. PMID: 23707263.
crossref
83. Liu YB, Kharode Y, Bodine PV, Yaworsky PJ, Robinson JA, Billiard J. 2010; LPA induces osteoblast differentiation through interplay of two receptors: LPA1 and LPA4. J Cell Biochem. 109:794–800. DOI: 10.1002/jcb.22471. PMID: 20069565.
crossref
84. Lee KP, Lee JH, Kim TS, Kim TH, Park HD, Byun JS, Kim MC, Jeong WI, Calvisi DF, Kim JM, Lim DS. 2010; The Hippo-Salvador pathway restrains hepatic oval cell proliferation, liver size, and liver tumorigenesis. Proc Natl Acad Sci U S A. 107:8248–8253. DOI: 10.1073/pnas.0912203107. PMID: 20404163. PMCID: PMC2889558.
crossref
85. Sautin YY, Jorgensen M, Petersen BE, Saulnier-Blache JS, Crawford JM, Svetlov SI. 2002; Hepatic oval (stem) cell expression of endothelial differentiation gene receptors for lysophosphatidic acid in mouse chronic liver injury. J Hematother Stem Cell Res. 11:643–649. DOI: 10.1089/15258160260194785. PMID: 12201952.
crossref
86. Bonnet D, Dick JE. 1997; Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 3:730–737. DOI: 10.1038/nm0797-730. PMID: 9212098.
crossref
87. Visvader JE, Lindeman GJ. 2008; Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 8:755–768. DOI: 10.1038/nrc2499. PMID: 18784658.
crossref
88. Lee D, Suh DS, Lee SC, Tigyi GJ, Kim JH. 2018; Role of autotaxin in cancer stem cells. Cancer Metastasis Rev. 37:509–518. DOI: 10.1007/s10555-018-9745-x. PMID: 29926310. PMCID: PMC6309296.
crossref
89. Sreepadmanabh M, Toley BJ. 2018; Investigations into the cancer stem cell niche using in-vitro 3-D tumor models and microfluidics. Biotechnol Adv. 36:1094–1110. DOI: 10.1016/j.biotechadv.2018.03.009. PMID: 29559382.
crossref
90. Reya T, Morrison SJ, Clarke MF, Weissman IL. 2001; Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. DOI: 10.1038/35102167. PMID: 11689955.
crossref
91. Seo EJ, Kwon YW, Jang IH, Kim DK, Lee SI, Choi EJ, Kim KH, Suh DS, Lee JH, Choi KU, Lee JW, Mok HJ, Kim KP, Matsumoto H, Aoki J, Kim JH. 2016; Autotaxin regulates maintenance of ovarian cancer stem cells through lysophosphatidic acid-mediated autocrine mechanism. Stem Cells. 34:551–564. DOI: 10.1002/stem.2279. PMID: 26800320.
crossref
92. Kim KS, Sengupta S, Berk M, Kwak YG, Escobar PF, Belinson J, Mok SC, Xu Y. 2006; Hypoxia enhances lysophosphatidic acid responsiveness in ovarian cancer cells and lysophosphatidic acid induces ovarian tumor metastasis in vivo. Cancer Res. 66:7983–7990. DOI: 10.1158/0008-5472.CAN-05-4381. PMID: 16912173.
crossref
93. Yu S, Murph MM, Lu Y, Liu S, Hall HS, Liu J, Stephens C, Fang X, Mills GB. 2008; Lysophosphatidic acid receptors determine tumorigenicity and aggressiveness of ovarian cancer cells. J Natl Cancer Inst. 100:1630–1642. DOI: 10.1093/jnci/djn378. PMID: 19001604. PMCID: PMC2720766.
crossref
94. van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradère 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. DOI: 10.1128/MCB.02419-05. PMID: 16782887. PMCID: PMC1489177.
crossref
95. Stracke ML, Krutzsch HC, Unsworth EJ, Arestad A, Cioce V, Schiffmann E, Liotta LA. 1992; Identification, purification, and partial sequence analysis of autotaxin, a novel motility-stimulating protein. J Biol Chem. 267:2524–2529. PMID: 1733949.
crossref
96. Lee H, Goetzl EJ, An S. 2000; Lysophosphatidic acid and sphingosine 1-phosphate stimulate endothelial cell wound healing. Am J Physiol Cell Physiol. 278:C612–C618. DOI: 10.1152/ajpcell.2000.278.3.C612. PMID: 10712250.
97. Gustin C, Van Steenbrugge M, Raes M. 2008; LPA modulates monocyte migration directly and via LPA-stimulated endothelial cells. Am J Physiol Cell Physiol. 295:C905–C914. DOI: 10.1152/ajpcell.00544.2007. PMID: 18632732.
crossref
98. Kim J, Keys JR, Eckhart AD. 2006; Vascular smooth muscle migration and proliferation in response to lysophosphatidic acid (LPA) is mediated by LPA receptors coupling to Gq. Cell Signal. 18:1695–1701. DOI: 10.1016/j.cellsig.2006.01.009. PMID: 16504475.
crossref
99. 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. DOI: 10.1161/CIRCRESAHA.108.180778. PMID: 18703779. PMCID: PMC2637300.
crossref
100. Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, Tigyi G, Siess W. 2003; Subtype-selective antagonists of lysophosphatidic acid receptors inhibit platelet activation triggered by the lipid core of atherosclerotic plaques. Circulation. 108:741–747. DOI: 10.1161/01.CIR.0000083715.37658.C4. PMID: 12885756.
crossref
101. 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. DOI: 10.1074/jbc.M505790200. PMID: 16278219.
crossref
102. Wu WT, Chen CN, Lin CI, Chen JH, Lee H. 2005; Lysophospholipids enhance matrix metalloproteinase-2 expression in human endothelial cells. Endocrinology. 146:3387–3400. DOI: 10.1210/en.2004-1654. PMID: 15878967.
crossref
103. Braet F, Wisse E. 2002; Structural and functional aspects of liver sinusoidal endothelial cell fenestrae: a review. Comp Hepatol. 1:1. DOI: 10.1186/1476-5926-1-1. PMID: 12437787. PMCID: PMC131011.
104. DeLeve LD. 2013; Liver sinusoidal endothelial cells and liver regeneration. J Clin Invest. 123:1861–1866. DOI: 10.1172/JCI66025. PMID: 23635783. PMCID: PMC3635729.
crossref
105. Chiu JJ, Chien S. 2011; Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 91:327–387. DOI: 10.1152/physrev.00047.2009. PMID: 21248169. PMCID: PMC3844671.
crossref
106. Robinson MW, Harmon C, O'Farrelly C. 2016; Liver immunology and its role in inflammation and homeostasis. Cell Mol Immunol. 13:267–276. DOI: 10.1038/cmi.2016.3. PMID: 27063467. PMCID: PMC4856809.
crossref
107. Chou CH, Lai SL, Ho CM, Lin WH, Chen CN, Lee PH, Peng FC, Kuo SH, Wu SY, Lai HS. 2015; Lysophosphatidic acid alters the expression profiles of angiogenic factors, cytokines, and chemokines in mouse liver sinusoidal endothelial cells. PLoS One. 10:e0122060. DOI: 10.1371/journal.pone.0122060. PMID: 25822713. PMCID: PMC4379007.
crossref
108. Schulze C, Smales C, Rubin LL, Staddon JM. 1997; Lysophosphatidic acid increases tight junction permeability in cultured brain endothelial cells. J Neurochem. 68:991–1000. DOI: 10.1046/j.1471-4159.1997.68030991.x. PMID: 9048744.
crossref
109. Zheng Y, Kong Y, Goetzl EJ. 2001; Lysophosphatidic acid receptor-selective effects on Jurkat T cell migration through a Matrigel model basement membrane. J Immunol. 166:2317–2322. DOI: 10.4049/jimmunol.166.4.2317. PMID: 11160288.
crossref
110. Stam JC, Michiels F, van der Kammen RA, Moolenaar WH, Collard JG. 1998; Invasion of T-lymphoma cells: cooperation between Rho family GTPases and lysophospholipid receptor signaling. EMBO J. 17:4066–4074. DOI: 10.1093/emboj/17.14.4066. PMID: 9670021. PMCID: PMC1170739.
crossref
111. Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. 2008; Autotaxin, an ectoenzyme that produces lysophosphatidic acid, promotes the entry of lymphocytes into secondary lymphoid organs. Nat Immunol. 9:415–423. DOI: 10.1038/ni1573. PMID: 18327261. PMCID: PMC2783613.
crossref
112. Jin Y, Knudsen E, Wang L, Maghazachi AA. 2003; Lysophosphatidic acid induces human natural killer cell chemotaxis and intracellular calcium mobilization. Eur J Immunol. 33:2083–2089. DOI: 10.1002/eji.200323711. PMID: 12884281.
crossref
113. Bagga S, Price KS, Lin DA, Friend DS, Austen KF, Boyce JA. 2004; Lysophosphatidic acid accelerates the development of human mast cells. Blood. 104:4080–4087. DOI: 10.1182/blood-2004-03-1166. PMID: 15319282.
crossref
114. Lundequist A, Boyce JA. 2011; LPA5 is abundantly expressed by human mast cells and important for lysophosphatidic acid induced MIP-1β release. PLoS One. 6:e18192. DOI: 10.1371/journal.pone.0018192. PMID: 21464938. PMCID: PMC3065470.
crossref
115. Hashimoto T, Yamashita M, Ohata H, Momose K. 2003; 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. 91:8–14. DOI: 10.1254/jphs.91.8. PMID: 12686725.
crossref
116. Rahaman M, Costello RW, Belmonte KE, Gendy SS, Walsh MT. 2006; Neutrophil sphingosine 1-phosphate and lysophosphatidic acid receptors in pneumonia. Am J Respir Cell Mol Biol. 34:233–241. DOI: 10.1165/rcmb.2005-0126OC. PMID: 16224106.
crossref
117. Koh JS, Lieberthal W, Heydrick S, Levine JS. 1998; Lysophosphatidic acid is a major serum noncytokine survival factor for murine macrophages which acts via the phosphatidylinositol 3-kinase signaling pathway. J Clin Invest. 102:716–727. DOI: 10.1172/JCI1002. PMID: 9710440. PMCID: PMC508934.
crossref
118. Rosskopf D, Daelman W, Busch S, Schurks M, Hartung K, Kribben A, Michel MC, Siffert W. 1998; Growth factor-like action of lysophosphatidic acid on human B lymphoblasts. Am J Physiol. 274:C1573–C1582. DOI: 10.1152/ajpcell.1998.274.6.C1573. PMID: 9611122.
crossref
119. Knowlden S, Georas SN. 2014; The autotaxin-LPA axis emerges as a novel regulator of lymphocyte homing and inflammation. J Immunol. 192:851–857. DOI: 10.4049/jimmunol.1302831. PMID: 24443508. PMCID: PMC3905607.
crossref
120. Gardell SE, Dubin AE, Chun J. 2006; Emerging medicinal roles for lysophospholipid signaling. Trends Mol Med. 12:65–75. DOI: 10.1016/j.molmed.2005.12.001. PMID: 16406843.
crossref
121. Ye X, Ishii I, Kingsbury MA, Chun J. 2002; Lysophosphatidic acid as a novel cell survival/apoptotic factor. Biochim Biophys Acta. 1585:108–113. DOI: 10.1016/S1388-1981(02)00330-X. PMID: 12531543.
crossref

Fig. 1
LPA signaling pathways. LPA activates G-protein-coupled receptors and initiates various downstream signaling cascades. LPA influences subsequent cellular processes such as proliferation, survival, apoptosis, morphological change, and migration, as well as brain organization within the nervous system.
IJSC-13-182-f1.tif
Fig. 2
LPA in the nervous system. (A) In neural progenitor cells, LPA promotes the survival of neural progenitor cells, as well as an increase in terminal mitosis. (B) In neurons, LPA changes to cell morphology and promotes growth cone collapse. (C) In oligodendrocytes, LPA promotes maturation and myelination. (D) In astrocytes, LPA activates the proliferation of astrocytes.
IJSC-13-182-f2.tif
TOOLS
Similar articles