Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2021 May 26;246(16):1802–1809. doi: 10.1177/15353702211019283

Revisiting the role of lysophosphatidic acid in stem cell biology

Gábor Tigyi 1, Kuan-Hung Lin 1, Il Ho Jang 2,3, Sue Chin Lee 1,
PMCID: PMC8381701  PMID: 34038224

Abstract

Stem cells possess unique biological characteristics such as the ability to self-renew and to undergo multilineage differentiation into specialized cells. Whereas embryonic stem cells (ESC) can differentiate into all cell types of the body, somatic stem cells (SSC) are a population of stem cells located in distinct niches throughout the body that differentiate into the specific cell types of the tissue in which they reside in. SSC function mainly to restore cells as part of normal tissue homeostasis or to replenish cells that are damaged due to injury. Cancer stem-like cells (CSC) are said to be analogous to SSC in this manner where tumor growth and progression as well as metastasis are fueled by a small population of CSC that reside within the corresponding tumor. Moreover, emerging evidence indicates that CSC are inherently resistant to chemo- and radiotherapy that are often the cause of cancer relapse. Hence, major research efforts have been directed at identifying CSC populations in different cancer types and understanding their biology. Many factors are thought to regulate and maintain cell stemness, including bioactive lysophospholipids such as lysophosphatidic acid (LPA). In this review, we discuss some of the newly discovered functions of LPA not only in the regulation of CSC but also normal SSC, the similarities in these regulatory functions, and how these discoveries can pave way to the development of novel therapies in cancer and regenerative medicine.

Keywords: Cancer stem cells, therapy resistance, chemoresistance, metastasis, lysophosphatidic acid, autotaxin, mesenchymal stem cells, somatic stem cells, intestinal stem cells, stem cell therapy

Impact statement

The evolving concepts of SSC provide insights into the highly dynamic nature of stem cell plasticity, a phenomenon that is often observed in CSC as well. LPA has been documented to play a prominent role in regulating stem cell biology in multiple levels; ESC, SSC and CSC. In particular, several recent studies have shown that targeting the LPA signaling axis could have new therapeutic applications in the area of dentistry as well as gastrointestinal and neurodegenerative diseases. Despite this, pharmacological agents targeting the autotaxin (ATX)-LPA receptor (LPAR) signaling axis for use in regenerative medicine and cancer has not been explored fully.

Introduction

Lysophosphatidic acid (LPA) is a bioactive lipid mediator that regulates many cellular functions such as cell proliferation, survival, differentiation, migration and invasion. There are several pathways that lead to the generation of LPA: (1) hydrolysis of lysophosphatidylcholine (LPC) by autotaxin (ATX), which produces the bulk of circulating LPA in biological fluids, and (2) actions by the enzymes glycerol-3-phosphate acyltransferase, phospholipase A, and acylglycerol kinase, which are responsible for the intracellular production of LPA.1,2 LPA exerts many of its biological functions via six extracellular G protein-coupled receptors (termed LPAR1-6), and the intracellular nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ). 2 The physiological levels of LPA in the plasma ranges from 0.1 to 1 µM. In serum, concentrations above 10 µM have been reported as well.3,4 Dysregulation of LPA signaling leads to many pathological disorders ranging from neuroinflammatory and cardiovascular diseases to fibrosis, bone disorders, obesity, and cancer. 5 In many instances, LPA levels are found to be elevated in these pathological conditions.69

Regulation of embryonic stem cells by LPA

Early efforts in generating ATX knockout mice revealed the importance of LPA signaling in embryonic development. In particular, deletion of ATX (encoded by the gene Enpp2) in mice led to death at embryonic days E9.5 to E10.5 as a result of severe abnormalities in the vascular and neural systems.10,11 Paradoxically, overexpression of ATX at the embryonic stage can also result in severe vascular defects that leads to embryonic death, suggesting that the expression of ATX and hence LPA signaling, must be tightly regulated during embryonic development to ensure proper vascular development. 12 Consistent with these findings, mice lacking lipid phosphatase 3 (LPP3), the enzyme that catalyzes LPA degradation, also suffer the same fate of vascular abnormality and embryonic lethality. 13 Based on the critical role of LPA signaling in embryonic development, it is not surprising to find that mouse embryonic stem cells (ESC) express almost all LPA receptors—LPAR1-3 14 and 5, 15 whereas human ESC and induced pluripotent stem cells (iPSC) express LPAR1-6.1621 LPA via activation of LPAR initiate a variety of downstream signaling pathways such as phospholipase C (PLC), extracellular signal regulated kinase (ERK), c-jun N-terminal kinase (JNK), p38, and signal transducer and activator of transcription 3 (STAT3) to regulate ESC proliferation, survival, self-renewal, and pluripotency.14,22,23 More recently, LPA has been shown to modulate the Hippo signaling pathway by activating downstream transcription cofactors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) to induce naïve pluripotency in human ESC and iPSC.24,25 Both YAP and TAZ are gaining recognition as essential drivers of stemness not only in ESC, but also in somatic stem cells (SSC) and cancer stem-like cells (CSC). 26 As the role of LPA on the regulation of ESC has been covered extensively in Lidgerwood et al., 27 we focus herein on other aspects of LPA regulation—stemness of SSC and CSC.

Regulation of somatic stem cells by LPA

Our understanding of stem cell biology is constantly evolving, especially in the case of SSC, also known as adult stem cells. Traditionally, our view of SSC biology is based solely on the hematopoietic stem cell (HSC) system, which suggests that stem cell population is rare, largely quiescent and long-lived in nature. HSC undergo asymmetric division giving rise to one daughter stem cell and a progenitor stem cell. Dividing progenitor stem cells then proceed through a unidirectional and well-structured hierarchical differentiation pathway which concludes once terminally differentiated cell types of the blood are formed. 28 However, the discovery of SSC in organs such as the intestinal tract, 29 esophagus 30 and testis 31 have unveiled mechanisms that differ from the HSC system. For example, the pool of SSC in certain niches can be abundant, short-lived and rapidly proliferating. In the following section, we discuss how LPA signaling regulates different pools of SSC in various tissues and highlight its therapeutic applicability in these areas.

Hematopoietic stem cells

HSC pass through a series of proliferation and differentiation processes to give rise to all cellular components of blood. It is highly regulated by numerous factors present in hematopoietic organs such as the bone marrow and spleen. Interestingly, high concentrations of LPA, ATX, and lipid phosphate phosphatases have been identified in the microvessels of human bone marrow, where they are known to promote early stages of myeloid differentiation. Moreover, gene profiling results showed that hematopoietic progenitors express LPAR1-6 at various levels in a manner that is dependent on the stage of differentiation. 32 In particular, LPA-LPAR4 axis has been shown to indirectly regulate early stages of HSC differentiation by affecting stromal cell activity in the bone marrow. 33 With regard to HSC mobility, LPA has been reported to increase HSC proliferation and rate of migration across the stromal cell layer. 34 So far, many evidence suggested that LPA regulates HSC commitment toward myeloid lineage.27,35 For example, LPAR1 has been reported to promote myeloid differentiation in CD34+ HSC, 32 leukemia cells, 36 and erythroid-megakaryocytic progenitors. 37 In both human and mouse, activation of the LPA-PPARγ signaling axis promoted the differentiation of monocyte into macrophage. 38 Furthermore, LPAR2 and LPAR3 are differentially expressed at various stages of hematopoiesis and play key roles in deciding the fate of the myeloid-erythroid-megakaryocytic lineage.39,40 Finally, studies using LPAR agonist in a murine anemia model demonstrated the potential therapeutic utility of targeting the LPA-LPAR signaling axis for the treatment of blood disease.39,41 These findings clearly highlight the critical roles of the LPA-LPAR signaling axis during early and late stages of hematopoiesis through precise regulation of HSC niches.

Neural stem cells

Neural stem cells (NSC) are present not only in the embryonic stage but also in adult brain and function in the renewal of neurons throughout life. Recent advances in imaging mass spectrometry showed variations in lipid composition in the human subventricular zone of the brain where NSC and neural stem progenitor cells (NSPC) reside and participate in the regulation of adult neurogenesis. 42 For instance, LPA is a potent neuromodulator that regulates either positively or negatively various aspects of neurogenesis ranging from NSPC proliferation,4345 migration 46 to differentiation.4547 The reported contradictory effects that LPA has on NSPC could be attributed to differences in species, cell-origins, and/or concentrations of LPA used that could impact the overall fate of NSPC. NSPC derived from human PSC are found to express LPAR1-5,45,47 whereas LPAR1-4 are differentially expressed in murine NSPC in a manner that is dependent on the developmental stage.46,48,49 LPA has been reported to induced dysregulation of LPA-LPAR signaling not only led to neurodevelopmental defects such as fetal hydrocephalus 50 but can also affect adult hippocampal neurogenesis, a process that plays a critical role in establishing and maintaining memories. This is particularly important in the context of addiction in which the generation of new hippocampal neurons from NSPC could help reverse the long-term cognitive defects induced by cocaine. Interestingly, a recent study demonstrated that repeated intracerebroventricular infusion of LPA improved contextual memory in cocaine-treated wild type mice, but not in LPAR1 knockout mice. 51 These findings highlight the critical role that LPA-LPAR1 signaling axis play in adult hippocampal neurogenesis and hippocampal-dependent memory functions and could potentially serve as a novel therapy to treat cognitive defects associated with cocaine addiction.

Mesenchymal stem cells

Mesenchymal stem cells (MSC) are present in the bone marrow, adipose tissue, lungs, teeth, Wharton’s jelly, and umbilical cord blood. Undeniably, these multipotent stem cells have gained clinical interest for use in regenerative medicine due to their ability to differentiate into multilineage cell types (e.g. chrondocytes, osteoblasts, adipocytes, and myocytes). Moreover, in the bone marrow, MSC secrete soluble factors that supports HSC maintenance and differentiation into mature blood cells. 52 Human MSC derived from various sources (bone marrow, adipose, and blood cord) express LPAR1-6 receptors at varying degrees,5355 whereas bone marrow-derived murine MSC express LPAR1, 4, and 6.33,56 LPAR display differential, sometimes opposing functions in MSC biology. For instance, in skeletal bone activation of LPAR1 has been shown to promote MSC differentiation into osteoblasts, whereas LPAR4 was found to be inhibitory. 57 Likewise, differentiation of MSC into myofibroblasts in the lungs was also mediated via LPAR1. 58 In terms of migration, LPA via activation of LPAR1 induced the recruitment of bone marrow-derived MSC into the synovial fluid of patients with rheumatoid arthritis 59 and promoted the migration of lung-resident MSC to the site of inflammation in response to lung injury. 60 In addition, LPA protected MSC against apoptosis induced by serum deprivation, hypoxia, and lipopolysaccharide by activating prosurvival pathways such as ERK 1/2 and AKT.55,56 Taken together, these findings highlight a profound role of LPA in the proliferation, survival, migration, and differentiation of MSC.

Periodontal ligament stem cells

Periodontal ligament stem cells (PDLSC) are multipotent SSC that reside in the perivascular space of the periodontium. Similar to MSC, PDLSC have the ability to differentiate into multilineage specialized cells ranging from periodontal ligament, alveolar bone and cementum to adipocytes, neurons, and blood vessels.61,62 Because of the multipotent nature of PDLSC and the ease of obtaining these cells (less invasive dental procedure compared to isolation of MSC from bone marrow), they are considered to be an excellent source of MSC for use in a wide range of regenerative therapy not limited to dental applications. However, one major drawback of PDLSC is the low yield of stem cells, requiring substantial in vitro expansion that may result in the loss of stem cell properties. 63 In this regard, LPA has recently been shown to promote the proliferation PDLSC in culture, suggesting that LPA could potentially be used as a mitogenic growth factor in the expansion of PDLSC. 64 In fact, LPA is present in normal human saliva and its levels can increase up to 10-fold in patients with moderate to severe periodontitis. 65 LPA appears to modulate periodontal inflammation and wound healing by regulating the expression of pro- and anti-inflammatory genes in gingival fibroblasts. 66 Using Porphyromonas gingivalis-derived lipopolysaccharide to mimic periodontitis in vitro, Kim et al., 64 demonstrated that blocking LPAR1 with the LPAR1 antagonist AM095 decreased the expression of proinflammatory cytokines and promoted the survival and osteogenic differentiation of PDLSC, suggesting that therapeutic targeting of LPAR1 could be explored for the treatment of periodontitis. An intriguing observation from this study is that treatment of PDLSC with the LPAR2 antagonist AMGEN35 significantly reduced the viability of PDLSC in culture, 67 which indicate that the proliferative effects of LPA in PDLSC reported earlier in Kim et al. 64 could potentially be mediated via LPAR2. Other studies have shown that LPA promoted the adhesion and migration of dental pulp cells to the site of injury, thus facilitating dental pulp repair. This process appears to be mediated via the Rho/Rho-associated kinase pathway. 68 LPA has also been reported to protect dental pulp cells from ischemia-induced apoptosis; clearly establishing a regulatory role of LPA in oral tissues. 69

Intestinal stem cells

The intestinal epithelium is known to continuously renew itself every four to five days via a specific pool of SSC located at the base of the crypt that express Leu-rich repeat containing G protein-coupled receptor 5 (Lgr5). Unlike the quiescent nature of HSC, Lgr5+ intestinal stem cells (ISC) are highly proliferative and, functioning like a conveyor belt, differentiate into epithelial cells as they move towards the tip of the villi where they undergo apoptosis and shed off into the lumen. 29 Lgr5+ ISC are known to be highly sensitive to radiation and are rapidly depleted following exposure to ionizing radiation. 70 We have recently showed that LPA via activation of LPAR2 protects Lgr5+ ISC from radiation-induced apoptosis, allowing for the survival and subsequent expansion of enteroids. 71 Previously, we reported that radiation or chemotherapy increase the expression of both ATX, the lysophospholipase that generates LPA, and LPAR2, 72 which could reflect a feed forward protective mechanism initiated by cells in an attempt to survive genotoxic insults. We found this mechanism to also exist in Lgr5+ ISC. Moreover, treatment with a nonlipid agonist of the LPAR2, termed Radioprotectin-1 alone led to an increase in LPAR2 expression as well. 71 In fact, Radioprotectin-1 when given at 24 h after exposure to sublethal dose of ionizing radiation decreased mortality in mice by 51% compared to vehicle treated mice. 71 The unique anti-apoptotic actions of LPAR2 is attributed to its C-terminal region, which contains LIM- and PDZ-binding domains that interacts with several key players such as Siva-1, TRIP6 and NHERF2. These interactions enhance prosurvival signaling pathways including ERK1/2 and AKT, and contribute to the arrest of apoptotic progression following genotoxic injury. 73 Moreover, activation of LPAR2 promotes DNA damage repair by accelerating the resolution of DNA double strand breaks.72,74 Thus, targeting LPAR2 could potentially be a novel strategy to mitigate the negative effects of radiation by promoting DNA damage repair and survival of the gastrointestinal epithelium. The differential roles of LPA-LPAR signaling axis in SSC biology are summarized in Figure 1.

Figure 1.

Figure 1.

Regulation of SSC pools by the ATX-LPA-LPAR signaling axis. The ATX-LPA-LPAR signaling pathway regulates the proliferation, migration, differentiation, and prosurvival of various SSC pools within the body. ATX: autotaxin; LPC: lysophosphatidylcholine; LPA: lysophosphatidic acid; HSC: hematopoietic stem cell; MSC: mesenchymal stem cells; NSC: neural stem cells; ISC: intestinal stem cells; PDLSC: periodontal ligament stem cells. (A color version of this figure is available in the online journal.)

Potential roles of LPA on cancer stem-like cells

Only a handful of studies reported a role of LPA in the regulation of CSC. Seo et al. demonstrated that the ATX-LPA-LPAR1 signaling axis is involved in the maintenance of ovarian CSC stemness. Specifically, treatment of ovarian CSC with LPA augmented CSC characteristics such as inducing the expression of stemness-related genes (e.g. OCT4, SOX2, and ALDH1), enhancing sphere-forming abilities, promoting resistance to chemotherapeutics, and increasing the tumor-initiating potential in mice. All these observations were abrogated when LPAR1 was silenced either via pharmacological or genetic interference. 75 In another study, Fan et al., demonstrated that activation of the nuclear receptor PPARγ by LPA led to the increase in the expression of ZIP4, an oncogene responsible for the maintenance of stemness in ovarian CSC. 76 In therapy-resistant breast CSC, ATX was found to be the second most upregulated gene, while lipid phosphate phosphatase 2 (LPP2) the enzyme that degrades LPA, is the most downregulated gene when compared to chemosensitive cancer cells, suggesting that the levels of LPA may be critical in promoting therapeutic resistance. 77 In fact, we have recently showed that radiation or chemotherapeutics increase the expression of ATX and LPAR2 in murine breast CSC; similar to what we have seen in the Lgr5+ ISC. Moreover, treatment of murine breast CSC with either ATX inhibitor or LPAR2 antagonist reduced the number of CSC spheres formed compared to vehicle treated control. 78 Collectively, these studies points to a key regulatory role of the ATX-LPA signaling axis in the self-renewal, therapeutic resistance, and maintenance of CSC.

CSC are also key to the seeding of metastasis. In this context, we showed that downregulation of ATX by shRNA reduces the number of B16-F10 melanoma metastasis to the lungs. 79 Seeding of pulmonary melanoma metastasis by CSC is also affected by LPAR expressed in the lung stroma. We found that the number of B16-F10 cells detected 24 h after intravenous inoculation is significantly reduced in LPAR1 and LPAR5 knockout mice. 79 Inhibition of stromal ATX using pharmacological blockade of this enzyme reduced the number of B16-F10 lung metastases in mice.79,80 Taken together, the ATX-LPAR axis appears to play a profound role in the metastatic process by affecting the tumor cell—microenvironment interaction.

The plasticity nature of SSC and CSC

A pool of intestinal stem cells known as the +4 stem cells (designated based on their location within the crypt) constitute a reserve pool of stem cells that are largely quiescent and give rise to the highly proliferative Lgr5+ ISC periodically or after injury-induced cell loss. However, recent studies uncovered that these reserve +4 stem cells are, in fact, precursor cells that are committed to terminally differentiate into secretory cells of the Paneth and enteroendocrine lineage. Yet, sudden loss of Lgr5+ ISC following intestinal injury can cause +4 precursor cells to reacquire a multipotent Lgr5+ stem cell phenotype and repopulate the Lgr5+ ISC pool.81,82 Such plasticity goes against the unidirectional hierarchical differentiation system seen in HSC. In fact, similar observations were reported in other organs such as the lungs where differentiated airway epithelial cells can reacquire a multipotent stem cell phenotype following the ablation of basal stem cell pool. 83

This phenomenon appears to play out in cancer as well. For example, de Sousa e Melo et al. have elegantly demonstrated that CSC hierarchies may be much more plastic and dynamic than previously appreciated. By crossing a mouse that models the human colon cancer with one that expresses diphtheria toxin receptor fused to GFP under the endogenous regulation of Lgr5, the authors were able to selectively track and ablate Lgr5+ CSC upon treatment with diphtheria toxin. Intriguingly, selective ablation of Lgr5+ CSC resulted only in the restriction of primary tumor growth and not complete tumor regression. A surprising observation was that tumor growth was maintained by Lgr5-cells, working continuously to repopulate the loss of Lgr5+ CSC pool. This led to rapid regrowth of tumors in mice upon withdrawal of diphtheria toxin treatment. 84 Similarly, Shimokawa et al. demonstrated that following depletion of Lgr5+ CSC pool, differentiated colorectal cancer cells (i.e. non-CSC pool) can revert to a CSC phenotype upon residing in the emptied niche previously occupied by Lgr5+ CSC. 85

In a separate study, Gunjal et al. sorted cells from the human ovarian A2780 cancer cell line into four distinct groups based on the surface expression of stem cell markers; (1) CD24CD44, (2) CD24+CD44, (3) CD24CD44+, and (4) CD24+CD44+. Single cell representing each phenotype were then cultured under limiting dilution conditions. Surprisingly, all single cell gave rise to clones that expressed surface markers comparable to that of the parental cell line. More importantly, all four phenotypes could be detected in the expanded clones arising from group 1 (i.e. single cell that lack both CD24CD44 stem cell surface markers at the time of isolation). 86 Taken together, these studies lead to the proposition of a secondary CSC concept known as the stochastic model in which stemness within a tumor is not hardwired but rather fluctuates in response to cell expansion, competition for space, injury or in response to cues from local niches. In no way does the stochastic model of CSC meant to disregard the existence of “true” CSC or cells with intrinsic stem-like traits, but rather highlight the dynamic nature of stem cell plasticity that could emerge in a context dependent manner influenced not only by intrinsic properties but also by extrinsic factors in the tumor microenvironment (TME)—much like in the regeneration of the intestinal epithelium following injury. In this context, the ATX-LPA-LPAR signaling axis has been shown to regulate not only the functions of CSC and cancer cells, but also various stromal cells within the TME such as fibroblasts, adipocytes, and immune cells (Figure 2).78,87 Dysregulation of the ATX-LPA-LPAR signaling pathway in each component of the TME spurs tumor progression, metastasis, therapy resistance and immune evasion. For example, breast cancer cells have been reported to reprogram adjacent adipocytes to increase ATX expression and further promote cancer progression. 88 In ovarian cancer, tumor cell-derived LPA can induce aerobic glycolysis in normal fibroblasts, a metabolic event which mediates the priming from normal fibroblasts to activated cancer-associated fibroblasts. 89 Furthermore, the ATX-LPA-LPAR5 signaling axis has been demonstrated to play a prominent role in mediating cancer immune evasion by inhibiting the cytotoxic effector function of CD8 T cells as discussed extensively in Lee et al. 87 Thus, a comprehensive understanding on the role of the ATX-LPA-LPAR signaling axis in regulating the TME, stem cell niche, and CSC; how one component influences and shapes the plasticity of another and vice versa, is of paramount importance.

Figure 2.

Figure 2.

Regulation of CSC, non-CSC and different stromal cells within the TME by the ATX-LPA-LPAR signaling axis. The ATX-LPA-LPAR signaling pathway is commonly upregulated in cancer cells, CSC, and in response to chemo- and radiotherapy. This signaling pathway is also dysregulated in adipocytes, CAF, TAM, and T cells, which functions to further drive malignancy in cancer. CAF: cancer-associated fibroblasts; NK: natural killer cells; TAM: tumor-associated macrophages; ECM: extracellular matrix; ATX: autotaxin; LPC: lysophosphatidylcholine; LPA: lysophosphatidic acid. (A color version of this figure is available in the online journal.)

Conclusions

There remains much to learn about the biological nature of stem cells, particularly in the context of stem cell plasticity where loss of SSC in a niche can result in the rapid replacement by differentiated daughter cells that reacquire stem-like traits. In a way, cancer can be viewed as defective or misappropriated tissue regeneration. The sobering observation that stemness is not hardwired but highly plastic further complicates the identification and eradication of CSC. Despite significant progress being made in the molecular analysis of markers that could help identify distinct CSC populations, the more pressing and important questions on the biology underlying CSC and the contributions of non-CSC and the TME to the growth and progression of cancer remain to be addressed.

Footnotes

Authors’ contributions: GT, KHL, IHJ, and SCL wrote the manuscript.

Authors’ note: All colored figures were created with BioRender.com.

DECLARATION OF CONFLICTING INTERESTS: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

FUNDING: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the William and Ella Owens Medical Research Foundation (SCL), the University of Tennessee CORNET grant (SCL), the National Cancer Institute grant CA092160 (GT), the National Institute of Allergy and Infectious Diseases grant AI107331 and 1U19AI150574 (GT), the Harriet Van Vleet endowment (GT), the Memphis Institute of Regenerative Medicine (MIRM) Institute Project 4 pilot grant (GT), and MOST-109–2917-I-564–029 (KHL).

References

  • 1.Aoki J. Two pathways for lysophosphatidic acid production. Biochimica et Biophysica Acta (BBA)—Mol Cell Biol Lipids 2008; 1781:513–8 [DOI] [PubMed] [Google Scholar]
  • 2.Geraldo LHM, Spohr T, Amaral RFD, Fonseca ACCD, Garcia C, Mendes FA, Freitas C, dosSantos MF, Lima FRS. Role of lysophosphatidic acid and its receptors in health and disease: novel therapeutic strategies. Sig Transduct Target Ther 2021; 6. doi: 10.1038/s41392-020-00367-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.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; 277:48737–44 [DOI] [PubMed] [Google Scholar]
  • 4.Michalczyk A, Budkowska M, Dołȩgowska B, Chlubek D, Safranow K. Lysophosphatidic acid plasma concentrations in healthy subjects: circadian rhythm and associations with demographic, anthropometric and biochemical parameters. Lipids Health Dis 2017; 16. doi: 10.1186/s12944-017-0536-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res 2014; 55:1192–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Arun P, Rossetti F, DeMar JC, Wang Y, Batuure AB, Wilder DM, Gist ID, Morris AJ, Sabbadini RA, Long JB. Antibodies against lysophosphatidic acid protect against Blast-Induced ocular injuries. Front Neurol 2020; 11. doi: 10.3389/fneur.2020.611816 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schober A, Siess W. Lysophosphatidic acid in atherosclerotic diseases. Br J Pharmacol 2012; 167:465–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Xu Y. Lysophosphatidic acid as a potential biomarker for ovarian and other gynecologic cancers. JAMA 1998; 280:719–23 [DOI] [PubMed] [Google Scholar]
  • 9.Kaffe M, A. Deregulated lysophosphatidic acid metabolism and signaling in liver cancer. Cancers 2019; 11. doi: 10.3390/cancers11111626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradère JP, Pettit TR, Wakelam MJO, Saulnier-Blache JSb Mummery CL, Moolenaar WH, Jonkers J. Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Mol Cell Biol 2006; 26:5015–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Moolenaar WH, Houben AJS, Lee S-J, van Meeteren LA. Autotaxin in embryonic development. Biochimica et Biophysica Acta (BBA)—Mol Cell Biol Lipids 2013; 1831:13–9 [DOI] [PubMed] [Google Scholar]
  • 12.Yue J, Yukiura H, Kano K, Kise R, Inoue A, Aoki J. Autotaxin overexpression causes embryonic lethality and vascular defects. PLoS One 2015; 10. doi: 10.1371/journal.pone.0126734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Escalante-Alcalde D, Hernandez L, Le Stunff H, Maeda R, Lee H-S, Jr, Gang C, Sciorra VA, Daar I, Spiegel S, Morris AJ, Stewart CL. The lipid phosphatase LPP3 regulates extra-embryonic vasculogenesis and axis patterning. Development 2003; 130:4623–37 [DOI] [PubMed] [Google Scholar]
  • 14.Todorova MG, Fuentes E, Soria B, Nadal A, Quesada I. Lysophosphatidic acid induces Ca2+ mobilization and c-Myc expression in mouse embryonic stem cells via the phospholipase C pathway. Cell Signall 2009; 21:523–8 [DOI] [PubMed] [Google Scholar]
  • 15.Lee C-W, Rivera R, Gardell S, Dubin AE, Chun J. GPR92 as a new G12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. J Biol Chem 2006; 281:23589–97 [DOI] [PubMed] [Google Scholar]
  • 16.Pitson SM, Eacute, Bay A. Regulation of stem cell pluripotency and neural differentiation by lysophospholipids. Neurosignals 2009; 17:242–54 [DOI] [PubMed] [Google Scholar]
  • 17.Ermakov A, Pells S, Freile P, Ganeva VV, Wildenhain J, Bradley M, Pawson A, Millar R, De Sousa PA. A role for intracellular calcium downstream of G-protein signaling in undifferentiated human embryonic stem cell culture. Stem Cell Res 2012; 9:171–84 [DOI] [PubMed] [Google Scholar]
  • 18.Assou S, Le Carrour T, Tondeur S, Ström S, Gabelle A, Marty S, Nadal L, Pantesco V, Réme T, Hugnot J-P, Gasca S, Hovatta O, Hamamah S, Klein B, De Vos J. A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells 2007; 25:961–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pébay A, Wong RCB, Pitson SM, Wolvetang EJ, Peh GSL, Filipczyk A, Koh KLL, Tellis I, Nguyen LTV, Pera MF. Essential roles of sphingosine-1-phosphate and platelet-derived growth factor in the maintenance of human embryonic stem cells. Stem Cells 2005; 23:1541–8 [DOI] [PubMed] [Google Scholar]
  • 20.Kleger A, Liebau S, Lin Q, von Wichert G, Seufferlein T. The impact of bioactive lipids on cardiovascular development. Stem Cells Int 2011; 2011:1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lidgerwood GE, Morris AJ, Conquest A, Daniszewski M, Rooney LA, Lim SY, Hernández D, Liang HH, Allen P, Connell PP, Guymer RH, Hewitt AW, Pébay A. Role of lysophosphatidic acid in the retinal pigment epithelium and photoreceptors. Biochimica et Biophysica Acta (BBA)—Mol Cell Biol Lipids 2018; 1863:750–61 [DOI] [PubMed] [Google Scholar]
  • 22.Schuck S, Soloaga A, Schratt G, Arthur JSC, Nordheim A. The kinase MSK1 is required for induction of c-fos by lysophosphatidic acid in mouse embryonic stem cells. BMC Mol Biol 2003; 4. doi: 10.1186/1471-2199-4-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kime C, Sakaki-Yumoto M, Goodrich L, Hayashi Y, Sami S, Derynck R, Asahi M, Panning B, Yamanaka S, Tomoda K. Autotaxin-mediated lipid signaling intersects with LIF and BMP signaling to promote the naive pluripotency transcription factor program. Proc Natl Acad Sci 2016; 113:12478–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.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. Yap induces human naive pluripotency. Cell Rep 2016; 14:2301–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hsiao C, Lampe M, Nillasithanukroh S, Han W, Lian X, Palecek SP. Human pluripotent stem cell culture density modulates Yap signaling. Biotechnol J 2016; 11:662–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mo JS, Park HW, Guan KL. The hippo signaling pathway in stem cell biology and cancer. EMBO Rep 2014; 15:642–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lidgerwood GE, Pitson SM, Bonder C, Pébay A. Roles of lysophosphatidic acid and sphingosine-1-phosphate in stem cell biology. Progr Lipid Res 2018; 72:42–54 [DOI] [PubMed] [Google Scholar]
  • 28.Clevers H. What is an adult stem cell? Science 2015; 350:1319–20 [DOI] [PubMed] [Google Scholar]
  • 29.Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell 2013; 154:274–84 [DOI] [PubMed] [Google Scholar]
  • 30.Doupe DP, Alcolea MP, Roshan A, Zhang G, Klein AM, Simons BD, Jones PH. A single progenitor population switches behavior to maintain and repair esophageal epithelium. Science 2012; 337:1091–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Klein AM, Nakagawa T, Ichikawa R, Yoshida S, Simons BD. Mouse germ line stem cells undergo rapid and stochastic turnover. Cell Stem Cell 2010; 7:214–24 [DOI] [PubMed] [Google Scholar]
  • 32.Gibson SB, Evseenko D, Latour B, Richardson W, Corselli M, Sahaghian A, Cardinal S, Zhu Y, Chan R, Dunn B, Crooks GM. Lysophosphatidic acid mediates myeloid differentiation within the human bone marrow microenvironment. PLoS One 2013; 8. doi: 10.1371/journal.pone.0063718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Igarashi H, Akahoshi N, Ohto-Nakanishi T, Yasuda D, Ishii S. The lysophosphatidic acid receptor LPA4 regulates hematopoiesis-supporting activity of bone marrow stromal cells. Sci Rep 2015; 5. doi: 10.1038/srep11410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yanai N, Matsui N, Furusawa T, Okubo T, Obinata M. Sphingosine-1-phosphate and lysophosphatidic acid trigger invasion of primitive hematopoietic cells into stromal cell layers. Blood 2000; 96:139–44 [PubMed] [Google Scholar]
  • 35.Lin K-H, Chiang J-C, Ho Y-H, Yao C-L, Lee H. Lysophosphatidic acid and hematopoiesis: from microenvironmental effects to intracellular signaling. Int J Mol Sci 2020; 21. doi: 10.3390/ijms21062015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Okabe-Kado J, Hagiwara-Watanabe Y, Niitsu N, Kasukabe T, Kaneko Y. NM23 downregulation and lysophosphatidic acid receptor EDG2/lpa1 upregulation during myeloid differentiation of human leukemia cells. Leukem Res 2018; 66:39–48 [DOI] [PubMed] [Google Scholar]
  • 37.Chiang C-L, Chen S-SA, Lee SJ, Tsao K-C, Chu P-L, Wen C-H, Hwang S-M, Yao C-L, Lee H. Lysophosphatidic acid induces erythropoiesis through activating lysophosphatidic acid receptor 3. Stem Cells 2011; 29:1763–73 [DOI] [PubMed] [Google Scholar]
  • 38.Ray R, Rai V. Lysophosphatidic acid converts monocytes into macrophages in both mice and humans. Blood 2017; 129:1177–83 [DOI] [PubMed] [Google Scholar]
  • 39.Chiang J-C, Chen W-M, Lin K-H, Hsia K, Ho Y-H, Lin Y-C, Shen T-L, Lu J-H, Chen S-K, Yao C-L, Chen BPC, Lee H. Lysophosphatidic acid receptors 2 and 3 regulate erythropoiesis at different hematopoietic stages. Biochimica et Biophysica Acta (BBA)—Mol Cell Biol Lipids 2021; 1866. doi: 10.1016/j.bbalip.2020.158818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lin K-H, Chiang J-C, Chen W-M, Ho Y-H, Yao C-L, Lee H. Transcriptional regulation of lysophosphatidic acid receptors 2 and 3 regulates myeloid commitment of hematopoietic stem cells. Am J Physiol Cell Physiol 2021; 320. doi: 10.1152/ajpcell.00506.2020 [DOI] [PubMed] [Google Scholar]
  • 41.Lin K-H, Ho Y-H, Chiang J-C, Li M-W, Lin S-H, Chen W-M, Chiang C-L, Lin Y-N, Yang Y-J, Chen C-N, Lu J, Huang C-J, Tigyi G, Yao C-L, Lee H. Pharmacological activation of lysophosphatidic acid receptors regulates erythropoiesis. Sci Rep 2016; 6. doi: 10.1038/srep27050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hunter M, Demarais NJ, Faull RLM, Grey AC, Curtis MA. Layer-specific lipid signatures in the human subventricular zone demonstrated by imaging mass spectrometry. Sci Rep 2018; 8. doi: 10.1038/s41598-018-20793-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hurst JH, Mumaw J, Machacek DW, Sturkie C, Stice SL, Callihan P, Hooks SB. Human neural progenitors express functional lysophospholipid receptors that regulate cell growth and morphology. BMC Neurosci 2008; 9. doi: 10.1186/1471-2202-9-118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Matas-Rico E, García-Diaz B, Llebrez-Zayas P, López-Barroso D, Santín L, Pedraza C, Smith-Fernández A, Fernández-Llebrez P, Tellez T, Redondo M, Chun J, De Fonseca FR, Estivill-Torrús G. Deletion of lysophosphatidic acid receptor LPA1 reduces neurogenesis in the mouse dentate gyrus. Mol Cell Neurosci 2008; 39:342–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Frisca F, Crombie DE, Dottori M, Goldshmit Y, Pébay A. Rho/ROCK pathway is essential to the expansion, differentiation, and morphological rearrangements of human neural stem/progenitor cells induced by lysophosphatidic acid. J Lipid Res 2013; 54:1192–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kimura A, Ohmori T, Ohkawa R, Madoiwa S, Mimuro J, Murakami T, Kobayashi E, Hoshino Y, Yatomi Y, Sakata Y. Essential roles of sphingosine 1-phosphate/S1P1Receptor axis in the migration of neural stem cells toward a site of spinal cord injury. Stem Cells 2007; 25:115–24 [DOI] [PubMed] [Google Scholar]
  • 47.Dottori M, Leung J, Turnley AM, Pébay A. Lysophosphatidic acid inhibits neuronal differentiation of neural stem/progenitor cells derived from human embryonic stem cells. Stem Cells 2008; 26:1146–54 [DOI] [PubMed] [Google Scholar]
  • 48.Jin Rhee H, Nam J-S, Sun Y, Kim M-J, Choi H-K, Han D-H, Kim N-H, Huh S-O. Lysophosphatidic acid stimulates cAMP accumulation and cAMP response element-binding protein phosphorylation in immortalized hippocampal progenitor cells. NeuroReport 2006; 17:523–6 [DOI] [PubMed] [Google Scholar]
  • 49.Walker TL, Overall RW, Vogler S, Sykes AM, Ruhwald S, Lasse D, Ichwan M, Fabel K, Kempermann G. Lysophosphatidic acid receptor is a functional marker of adult hippocampal precursor cells. Stem Cell Rep 2016; 6:552–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yung YC, Mutoh T, Lin ME, Noguchi K, Rivera RR, Choi JW, Kingsbury MA, Chun J. Lysophosphatidic acid signaling may initiate fetal hydrocephalus. Sci Translat Med 2011; 3:99ra87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ladrón de Guevara‐Miranda D, Moreno‐Fernández RD, Gil‐Rodríguez S, Rosell‐Valle C, Estivill‐Torrús G, Serrano A, Pavón FJ, Rodríguez de Fonseca F, Santín LJ, Castilla‐Ortega E. Lysophosphatidic acid‐induced increase in adult hippocampal neurogenesis facilitates the forgetting of cocaine‐contextual memory. Addic Biol 2018; 24:458–70 [DOI] [PubMed] [Google Scholar]
  • 52.Williams AR, Hare JM, Dimmeler S, Losordo D. Mesenchymal stem cells. Circulat Res 2011; 109:923–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu D, Wang Y, Ye Y, Yin G, Chen L. Distinct molecular basis for endothelial differentiation: Gene expression profiles of human mesenchymal stem cells versus umbilical vein endothelial cells. Cell Immunol 2014; 289:7–14 [DOI] [PubMed] [Google Scholar]
  • 54.Almeida-Porada G, Binder BYK, Sondergaard CS, Nolta JA, Leach JK. Lysophosphatidic acid enhances stromal Cell-Directed angiogenesis. PLoS One 2013; 8. doi: 10.1371/journal.pone.0082134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li N, Yan Y-L, Fu S, Li R-J, Zhao P-F, Xu X-Y, Yang J-P, Damirin A. Lysophosphatidic acid enhances human umbilical cord mesenchymal stem cell viability without differentiation via LPA receptor mediating manner. Apoptosis 2017; 22:1296–309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Chen J, Baydoun AR, Xu R, Deng L, Liu X, Zhu W, Shi L, Cong X, Hu S, Chen X. Lysophosphatidic acid protects mesenchymal stem cells against hypoxia and serum Deprivation-Induced apoptosis. Stem Cells 2008; 26:135–45 [DOI] [PubMed] [Google Scholar]
  • 57.Liu Y-B, Kharode Y, Bodine PVN, Yaworsky PJ, Robinson JA, Billiard J. LPA induces osteoblast differentiation through interplay of two receptors: LPA1 and LPA4. J Cell Biochem 2010; 109:794–800. doi: 10.1016/j.bbalip.2020.158818 [DOI] [PubMed] [Google Scholar]
  • 58.Tang N, Zhao Y, Feng R, Liu Y, Wang S, Wei W, Ding Q, An M, Wen J, Li L. Lysophosphatidic acid accelerates lung fibrosis by inducing differentiation of mesenchymal stem cells into myofibroblasts. J Cell Mol Med 2013; 18:156–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Song HY, Lee MJ, Kim MY, Kim KH, Lee IH, Shin SH, Lee JS, Kim JH. Lysophosphatidic acid mediates migration of human mesenchymal stem cells stimulated by synovial fluid of patients with rheumatoid arthritis. Biochimica et Biophysica Acta (BBA)—Mol Cell Biol Lipids 2010; 1801:23–30 [DOI] [PubMed] [Google Scholar]
  • 60.Badri L, Lama VN. Lysophosphatidic acid induces migration of human lung-resident mesenchymal stem cells through the β-catenin pathway. Stem Cells 2012; 30:2010–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Seo B-M, Miura M, Gronthos S, Mark Bartold P, Batouli S, Brahim J, Young M, Gehron Robey P, Wang CY, Shi S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004; 364:149–55 [DOI] [PubMed] [Google Scholar]
  • 62.Zhu W, Liang M. Periodontal ligament stem cells: current status, concerns, and future prospects. Stem Cells Int 2015; 2015:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Iwata T, Yamato M, Zhang Z, Mukobata S, Washio K, Ando T, Feijen J, Okano T, Ishikawa I. Validation of human periodontal ligament-derived cells as a reliable source for cytotherapeutic use. J Clin Periodontol 2010; 37:1088–99 [DOI] [PubMed] [Google Scholar]
  • 64.Kim BC, Song JI, So KH, Hyun SH. Effects of lysophosphatidic acid on human periodontal ligament stem cells from teeth extracted from dental patients. J Biomed Res 2019; 33:122–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bathena SP, Huang J, Nunn ME, Miyamoto T, Parrish LC, Lang MS, McVaney TP, Toews ML, Cerutis DR, Alnouti Y. Quantitative determination of lysophosphatidic acids (LPAs) in human saliva and gingival crevicular fluid (GCF) by LC–MS/MS. J Pharmaceut Biomed Anal 2011; 56:402–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cerutis DR, Weston MD, Alnouti Y, Bathena SP, Nunn ME, Ogunleye AO, McVaney TP, Headen KV, Miyamoto T. A major human oral lysophosphatidic acid species, LPA 18:1, regulates novel genes in human gingival fibroblasts. J Periodontol 2015; 86:713–25 [DOI] [PubMed] [Google Scholar]
  • 67.Kim DH, Seo EJ, Tigyi GJ, Lee BJ, Jang IH. The role of lysophosphatidic acid receptor 1 in inflammatory response induced by lipopolysaccharide from Porphyromonas gingivalis in human periodontal ligament stem cells. Intern J Oral Biol 2020; 45:42–50 [Google Scholar]
  • 68.Cheng R, Cheng L, Shao M-y, Yang H, Wang F-M, Hu T, Zhou X-D. Roles of lysophosphatidic acid and the rho-associated kinase pathway in the migration of dental pulp cells. Exp Cell Res 2010; 316:1019–27 [DOI] [PubMed] [Google Scholar]
  • 69.Pan H, Cheng L, Yang H, Zou W, Cheng R, Hu T. Lysophosphatidic acid rescues human dental pulp cells from ischemia-induced apoptosis. J Endodont 2014; 40:217–22 [DOI] [PubMed] [Google Scholar]
  • 70.Andersson-Rolf A, Zilbauer M, Koo B-K, Clevers H. Stem cells in repair of gastrointestinal epithelia. Physiology 2017; 32:278–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kuo B, Szabó E, Lee SC, Balogh A, Norman D, Inoue A, Ono Y, Aoki J, Tigyi G. The LPA2 receptor agonist radioprotectin-1 spares Lgr5-positive intestinal stem cells from radiation injury in murine enteroids. Cell Signall 2018; 51:23–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Balogh A, Shimizu Y, Lee SC, Norman DD, Gangwar R, Bavaria M, Moon C, Shukla P, Rao R, Ray R, Naren AP, Banerje S, Miller DD, Balazs L, Pelus L, Tigyi G. The autotaxin–LPA 2 GPCR axis is modulated by γ-irradiation and facilitates DNA damage repair. Cell Signall 2015; 27:1751–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.ES, Lai Y-J, Tsukahara R, Chen C-S, Fujiwara Y, Yue J, Yu J-H, Guo H, Kihara A, Tigyi G, Lin F-T. Lysophosphatidic acid 2 receptor-mediated supramolecular complex formation regulates its antiapoptotic effect. J Biol Chem 2009; 284:14558–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Tigyi GJ, Johnson LR, Lee SC, Norman DD, Szabo E, Balogh A, Thompson K, Boler A, McCool WS. Lysophosphatidic acid type 2 receptor agonists in targeted drug development offer broad therapeutic potential. J Lipid Res 2019; 60:464–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Seo EJ, Kwon YW, Jang IH, Kim DK, Lee SI, Choi EJ, Kim K-H, Suh D-S, Lee JH, Choi KU, Lee JW, Mok HJ, Kim KP, Matsumoto H, Aoki J, Kim JH. Autotaxin regulates maintenance of ovarian cancer stem cells through lysophosphatidic acid-mediated autocrine mechanism. Stem Cells 2016; 34:551–64 [DOI] [PubMed] [Google Scholar]
  • 76.Fan Q, Cai Q, Li P, Wang W, Wang J, Gerry E, Wang T-L, Shih I-M, Nephew KP, Xu Y. The novel ZIP4 regulation and its role in ovarian cancer. Oncotarget 2017; 8:90090–107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, Lander ES. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 2009; 138:645–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tigyi GJ, Yue J, Norman DD, Szabo E, Balogh A, Balazs L, Zhao G, Lee SC. Regulation of tumor cell—microenvironment interaction by the autotaxin-lysophosphatidic acid receptor axis. Adv Biol Regulat 2019; 71:183–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee SC, Fujiwara Y, Liu J, Yue J, Shimizu Y, Norman DD, Wang Y, Tsukahara R, Szabo E, Patil R, Banerjee S, Miller DD, Balazs L, Ghosh MC, Waters CM, Oravecz T, Tigyi GJ. Autotaxin and LPA1 and LPA5 receptors exert disparate functions in tumor cells versus the host tissue microenvironment in melanoma invasion and metastasis. Mol Cancer Res 2015; 13:174–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.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; 40:31–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Buczacki SJA, Zecchini HI, Nicholson AM, Russell R, Vermeulen L, Kemp R, Winton DJ. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 2013; 495:65–69 [DOI] [PubMed] [Google Scholar]
  • 82.van Es JH, Sato T, van de Wetering M, Lyubimova A, Yee Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J, Martens ACM, Barker N, van Oudenaarden A, Clevers H. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell Biol 2012; 14:1099–104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tata PR, Mou H, Pardo-Saganta A, Zhao R, Prabhu M, Law BM, Vinarsky V, Cho JL, Breton S, Sahay A, Medoff BD, Rajagopal J. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 2013; 503:218–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.de Sousa e Melo F, Kurtova AV, Harnoss JM, Kljavin N, Hoeck JD, Hung J, Anderson JE, Storm EE, Modrusan Z, Koeppen H, Dijkgraaf GJP, Piskol R, de Sauvage FJ. A distinct role for Lgr5+ stem cells in primary and metastatic colon cancer. Nature 2017; 543:676–80 [DOI] [PubMed] [Google Scholar]
  • 85.Shimokawa M, Ohta Y, Nishikori S, Matano M, Takano A, Fujii M, Date S, Sugimoto S, Kanai T, Sato T. Visualization and targeting of LGR5+ human colon cancer stem cells. Nature 2017; 545:187–92 [DOI] [PubMed] [Google Scholar]
  • 86.Gunjal P, Pedziwiatr D, Ismail AA, Kakar SS, Ratajczak MZ. An emerging question about putative cancer stem cells in established cell lines—are they true stem cells or a fluctuating cell phenotype? J Cancer Stem Cell Res 2015; 3. doi: 10.14343/jcscr.2015.3e1004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lee SC, Dacheux MA, Norman DD, Balázs L, Torres RM, Augelli-Szafran CE, Tigyi GJ. Regulation of tumor immunity by lysophosphatidic acid. Cancers 2020; 12. doi: 10.3390/cancers12051202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Brindley DN, Tang X, Meng G, Benesch MGK. Role of adipose tissue-derived autotaxin, lysophosphatidate signaling, and inflammation in the progression and treatment of breast cancer. Int J Mol Sci 2020; 21. doi: 10.3390/ijms21165938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Radhakrishnan R, Ha JH, Jayaraman M, Liu J, Moxley KM, Isidoro C, Sood AK, Song YS, Dhanasekaran DN. Ovarian cancer cell-derived lysophosphatidic acid induces glycolytic shift and cancer-associated fibroblast-phenotype in normal and peritumoral fibroblasts. Cancer Lett 2019; 442:464–74 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES