Summary
Lysophosphatidic acid (LPA) is an endogenous bioactive lipid that is produced extracellularly and signals to cells via cognate LPA receptors, which are G-protein coupled receptors (GPCRs). Mature lymphocytes in mice and humans express three LPA receptors, LPA2, LPA5 and LPA6, and work from our group has determined that LPA5 signaling by T lymphocytes inhibits specific antigen-receptor signaling pathways that ultimately impair lymphocyte activation, proliferation and function. In this review, we discuss previous and ongoing work characterizing the ability of an LPA-LPA5 axis to serve as a peripheral immunological tolerance mechanism that restrains adaptive immunity but is subverted during settings of chronic inflammation. Specifically, LPA-LPA5 signaling is found to regulate effector cytotoxic CD8 T cells by (at least) two mechanisms: i) regulating the actin-microtubule cytoskeleton in a manner that impairs immunological synapse formation between an effector CD8 T cell and antigen-specific target cell, thus directly impairing cytotoxic activity, and ii) shifting T cell metabolism to depend on fatty-acid oxidation for mitochondrial respiration and reducing metabolic efficiency. The in vivo outcome of LPA5 inhibitory activity impairs CD8 T cell killing and tumor immunity in mouse models providing impetus to consider LPA5 antagonism for the treatment of malignancies and chronic infections.
Keywords: T cells, cytotoxic, lipid mediators, cytotoxicity, signal transduction cancer
Introduction
Adaptive immunity depends on the ability of naïve T and B lymphocytes to recognize foreign antigen by their antigen receptors that subsequently leads to the neutralization or elimination of the foreign antigen or infected, damaged or malignant cell. Critical to initiating this response are the signals transmitted by the T cell and B cell antigen receptors (TCR and BCR, respectively). These antigen receptor signaling events can be further positively or negatively regulated by cell intrinsic or extrinsic mechanisms, including the expression of cell-surface co-receptors (co-stimulatory or inhibitory) as well as paracrine signals from the microenvironment in which the cell activation occurs. Cancers and viruses that establish chronic infections often co-opt T cell inhibitory receptor signaling to evade T cell immunity by suppressing T cell functional activity. This review highlights work from our lab that has revealed the ability of the lysophosphatidic acid, or LPA, bioactive lysophospholipid to signal via the LPA5 G-protein-coupled receptor to antagonize T and B cell antigen receptor signaling, activation, proliferation and function while simultaneously regulating T cell metabolism. Through biochemical, molecular, cell biological, and metabolic analyses coupled with in vitro and in vivo models of lymphocyte function, this review summarizes our major findings on how lysophosphatidic acid, or LPA, regulates adaptive cytotoxic CD8 T cell immunity.
Lysophosphatidic acid (LPA) is a lysophospholipid which are a family of simple phospholipids comprised of a polar phosphate head group, glycerol backbone and a long-chain fatty acid that exists in distinct species based on carbon chain length and degree of saturation. Lysophospholipids are signaling lipids that bind surface and intracellular receptors to regulate a broad range of processes. LPA can be found both inside and outside cells where it is generated through different enzymatic pathways and subsequently signals in different manners.1–3 The ability of LPA to regulate lymphocyte activation, function, and adaptive immunity discussed in this review is restricted to extracellularly produced bioactive lipid (for references and review of LPA intracellular signaling see.2,4,5) Originally identified in mammalian and plant cells over 60 years ago, LPA was considered to be a metabolite involved in membrane phospholipid production.6 Approximately 20 years later a number of groups documented that LPA displayed bioactivity as an extracellular signaling molecule capable of acting on diverse cell types, including platelets and neutrophils. Examples of LPA bioactivity implicated LPA in inducing hypertension in rodents, promoting intracellular signaling, inducing morphological changes, promoting cell aggregation, and enhancing chemotaxis.7–13 Moolenaar and colleagues provided early and compelling evidence that LPA signaled in fibroblasts via a G-protein coupled receptor(s) (GPCRs) and by the mid-90s LPA was found to be the ligand for specific and distinct (orphan) G-protein coupled receptors (GPCRs) expressed by a variety of cell types.14,15 In the past 20 years considerable attention has been devoted towards investigating cell-type specific signaling including how LPA receptor (LPAR) signaling impacts the nervous, vascular and immune systems.3,16–20
Determining that LPA was a cognate GPCR ligand placed it in a class of other inflammatory lipids that signal through GPCRs including eicosanoids and sphingosine-1-phosphate (S1P).3,21 These signaling lipids regulate cell-type specific functions ranging from innate inflammation, hemostasis and angiogenesis and lymphocyte recirculation. Similar to LPA, S1P is also an endogenously-produced lysophospholipid and, as extracellular bioactive lipids, both lipids signal to cells via low nM affinity association with cognate GPCRs: six GPCRs for LPA, LPA1–6, and five GPCRs for S1P, S1P1–5.3,22 Initial findings with both LPA and S1P suggested that these lysophospholipids could induce cell migration, cell morphological alterations and cell proliferation in a variety of cell types.3 Notably, S1P signaling via S1P1 is required for the egress of immature T cells from the thymus23 and manipulation of S1P1 signaling is currently used in the clinic to treat multiple sclerosis.24 At present, a number of diverse activities have been attributed to LPA signaling in the nervous and vascular systems.3,17–19 Despite that all mature human and murine lymphocytes and other immune cell types express one or more of the six LPA receptors (LPA1–6)16,25–29, its role in innate and adaptive immunity has been less well studied relative to S1P.
LPA concentration in the blood of healthy individuals and wild type mice has been reported to range from high nM to low μM concentrations.30–35 This variability in the reporting of LPA levels is dependent not only on assay measurement (mass spectroscopy, ELISA, etc.) but also sample preparation. Accordingly, when samples collected for LPA measurement include active inhibition of both LPA production and degradation it appears the plasma from healthy individuals harbor on the order of 50nM LPA.3,36,37 However, systemic levels of extracellular LPA and autotaxin (ATX), the enzyme predominantly responsible for extracellularly produced LPA, are often found significantly elevated in human chronic inflammatory disorders such as chronic viral (HCV, HBV and EBV) infections38–41 autoimmune diseases42–44, obesity45–52 and a number of diverse cancers53–60. In these settings, serum LPA levels are considerably heightened (~2–10-fold) compared to healthy individuals and concentrations over 50μM have been reported in malignant effusions of ovarian cancer patients.61 It is further worth noting, however, that the membrane-associated phosphatidic-selective phospholipase A1a, PA-PLA1α, has also been shown to contribute to extracellular LPA production in certain microenvironments.3 How pathological systemic LPA levels impact diverse infections and disease is an area of active investigation.
Extracellular production of LPA as a bioactive GPCR signaling lipid
The vast majority of extracellular LPA is produced by ATX, a phospholipase D enzyme expressed and secreted by certain cell-types that hydrolyzes the abundantly available lysophosphatidylcholine (LPC) lipid to produce extracellular LPA.62,63 ATX, encoded by ENPP2 (Ectonucleotide Pyrophosphatase/ Phosphodiesterase 2), was initially isolated from conditioned media of the A2058 melanoma cell line and thought to be a motility factor that signaled in an autocrine manner via an unknown GPCR to promote directed (and random) melanoma migration.64 ATX was only later identified as a secreted phospholipase D whose enzymatic activity produces LPA through the hydrolysis of LPC. Since LPC is a highly abundant lipid, the rate-determining step of LPA synthesis is considered to be ATX catalysis. As a secreted enzyme, ATX has been shown to associate with surface integrins65,66 and has led to a current model in which ATX associates with integrins on a cell surface where it subsequently converts LPC to LPA that then signals to the ATX-expressing or nearby cells expressing LPARs (Figure 1). The association of LPA with integrins was initially thought to occur by an exposed RGD (arginine-glycine-aspartic acid) integrin ligand motif. However, structural analyses showed that ATX associates with integrins in an RGD-indpendent manner.66 Thus, the precise mechanism by which ATX associates with integrins requires further clarification. Regardless, physiological LPA signaling results from localized and directed autocrine/paracrine LPA production and subsequent GPCR-induced signaling65,67–69 allowing for local LPA concentrations to reach much higher concentrations than systemic levels. Extracellular LPA is quite labile with an in vivo half-life on the order of (3–5) minutes62,70 as a result of its rapid hydrolysis by lipid phosphate phosphohydrolase type 1 (LPP1) and LPP3 to monoacylglycerols (MAGs).70 These MAGs can be further catabolized into free fatty acids by MAG lipases. Indeed, the half-life of LPA increases 4 fold when LPA is intravenously introduced into LPP1-deficient mice and Lpp1–/– mice harbor elevated levels of LPA70. An ATX-deficiency resulting from homozygosity for null Enpp2 alleles (Enpp2−/−) results in embryonic lethality largely due to vascular and nervous system defects.63,71 However, heterozygous Enpp2+/− mice are viable and as adults are reported to harbor close to half the levels of systemic LPA as found in wild type mice.63,71
Figure 1.

Model of autocrine/paracrine LPA action. Autotaxin (ATX) is secreted by select cells and binds to integrins on the cell surface and hydrolyzes LPC to produce LPA. LPA then signals via nearby LPA G-protein-coupled receptors on the same or nearby cells.
While many diverse cell types express one or more of the six LPARs, the expression of the ATX phospholipase D responsible for LPA extracellular production is more restricted. Certain cell types such as fibroblastic reticular stromal cells72 and high endothelial venule cells associated with lymphoid organs constitutively express high levels of ATX where subsequent LPA production promotes chemokinetic activity and motility of T lymphocytes.65,67,73
Our interest in the ability of LPA to regulate adaptive immunity initiated with the appreciation that a structurally similar lysophospholipid, sphingosine-1-phosphate (S1P), was identified as a chemoattractant ligand for the S1P1 G-protein coupled receptor (GPCR) expressed by T and B lymphocytes. In particular, there are notable parallels in the developmental checkpoints that exist for both T and B lymphocytes in the thymus and bone marrow, respectively. Yet, an S1P1-deficiency selectively prevented immature T cells from exiting the thymus but not immature B cells from exiting the bone marrow.23 This apparent discrepancy led us to consider whether additional lipid GPCRs (e.g., LPARs) were expressed by immature B cells that might be alternatively used by immature B cells to promote marrow egress. While LPARs have not been demonstrated to contribute to B cell egress from the bone marrow, we determined that S1P3 was an additional S1P receptor contributing to immature B cell egress.74,75 There are five characterized S1P receptors, S1P1–5, and all are GPCRs that based on a shared homology belong to the Edg family of GPCRs; named initially as an induced endothelial differentiation gene. LPAR1–3 were the first LPARs to be identified and all three were shown to be members of the Edg family of GPCRs. By 2006 it was evident that LPA was a cognate ligand for three additional LPARs, LPA4, LPA5 and LPA6, that did not belong to the Edg family of receptors but nevertheless are cognate GPCRs for LPA. GPCRs signal via αβγ hetrotrimeric G-proteins and are typically characterized by the associated Gα subunit of which there are 4 families: Gαi, Gαs, Gαq and Gα12/13. The ability of individual LPARs to associate with a specific Gα family member but not others has been demonstrated using cell lines and overexpression approaches. However, whether these associations promote physiological in vivo LPAR signaling is less certain. In broad terms, the three Edg family members, LPA1–3, often associate with Gαi and Gαq heterotrimeric proteins and the non-Edg LPAR4–6 most often are found to associate with Gα12/13 heterotrimeric G-proteins.
The identification and molecular cloning of all six LPARs allowed us to survey the expression of the LPAR-encoding genes by quantitative real-time PCR and to determine that immature and mature B and T lymphocytes from both mice and humans expressed the same set of GPCRs specific for the LPA lysophospholipid, LPA2, LPA5 and LPA6.76–78 This review will highlight work from our group demonstrating the ability of LPA5 to antagonize antigen receptor signaling and function of T and B lymphocytes.
LPA suppresses lymphocyte antigen receptor-induced calcium stores release and signaling
S1P is a lysophospholipid chemoattractant that upon engaging the S1P1 GPCR signals directed cell migration via a Gαi associated heterotrimer signaling axis, similar to chemokine GPCRs. Despite similarities between S1P and LPA receptors, our initial experiments did not reveal any significant role for LPA as a chemoattractant for B or T lymphocytes ex vivo (although LPA has been shown to promote chemokinesis of T cells via LPA2 signaling65,67,73,79). Thus, we explored how LPA might influence other aspects of lymphocyte function. These studies soon revealed that when the antigen receptor on B and T cells were stimulated in the presence of pathological concentrations of LPA (1–20μM), antigen receptor-induced intracellular calcium mobilization was significantly attenuated indicating that LPA had the potential to suppress lymphocyte activation (Figure 2).76–78,80 As mentioned, a number of early studies had reported plasma LPA levels to be in the range of high nM to low μM concentrations.30–35 With the relatively recent understanding that LPA catabolism and production post-collection accounted for this variability in LPA concentrations37, it will be important to further determine if normal concentrations of 50–100nM LPA are able to impinge on antigen receptor signaling. In this regard, however, we note that in our studies (discussed below) we have found that an approximate 50% reduction of wild type LPA levels, as produced in Enpp2+/− mice, significantly improve in vivo T cell cytotoxicity suggesting that wild type LPA concentrations indeed normally influence adaptive immunity.
Figure 2.

LPA signals via LPA5 to impair IP3R activity and resulting in reduced intracellular calcium stores release and inhibited lymphocyte antigen receptor signaling. Top: The presence of LPA at the time of BCR- and TCR-induced signaling suppresses intracellular calcium stores release for follicular (FO) and marginal zone (MZ) B cells (left) and CD4 and CD8 T cells (right). Bottom: Schematic illustrating TCR- and BCR-proximal kinase and PLCg activity is intact in the presence of LPA5 signaling while IP3R activity is depressed resulting in reduced antigen receptor-induced cytosolic calcium levels.
Antigen receptor signaling by T and B lymphocytes initiates with tyrosine phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) within the cytoplasmic domains of CD3 and CD79, respectively. T and B cell-specific tyrosine kinases phosphorylate ITAMs that recruit Zap70 and Syk kinases, respectively, that in turn are also activated by tyrosine phosphorylation. Activated Zap70/Syk subsequently promote canonical signaling cascades that lead to the tyrosine phosphorylation and activation of PLCγ1 in T cells and PLCγ2 in B cells by Tec-family kinases.81 Activated PLCγ hydrolyzes phosphatidylinositol 4,5 bisphosphate producing diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). IP3 subsequently engages IP3R calcium release channels expressed on the surface of the end reticulum (ER) to release intracellular stores of calcium into the cytosol (Figure 2). Calcium levels within the ER are monitored by the ER resident proteins STIM1 and STIM2, which upon depletion of ER calcium stores, oligomerize and stimulate the ORAI calcium channel within the plasma membrane to facilitate extracellular calcium entry.82–84 Resulting overall elevated levels of cytosolic calcium are required for a number of important lymphocyte biological processes such as cytokine secretion and cytolytic granule exocytosis but also the activation of transcription factors (e.g, NFAT) that initiate maturational transcriptional programs. Our finding that LPA impaired intracellular calcium mobilization initiated by antigen receptor signaling indicated that this lysophospholipid may contribute to the regulation of these processes.
Antigen receptor-proximal kinase signaling is not perturbed by LPA
To determine more precisely where LPA, signaling via an LPAR (LPA2, LPA5 and/or LPA6), suppresses antigen receptor-induced calcium intracellular stores release, primary mouse splenic B cells and human T cells isolated from healthy peripheral blood were stimulated via the antigen receptor in the presence and absence of LPA and evaluated by western blot analyses for activation of lymphocyte-specific kinases and PLCγ. The results of these experiments demonstrated that in the presence of LPA, LPAR signaling did not perturb TCR or BCR receptor-proximal signaling events. For mouse B cells stimulated with anti-IgM F(ab’)2, we found equivalent amounts of pSyk, pBtk and pPLCγ2 in the presence and absence of 20μM LPA76; for human T cells we found that stimulation with anti-CD3 and anti-CD28 in the presence and absence of LPA led to equivalent increases in total tyrosine phosphorylation of cellular proteins over 5 minutes and similar increases in pCD3ζ and pPLCγ180. Thus, LPA treatment of antigen receptor-stimulated mouse and human lymphocytes did not alter antigen receptor-proximal kinase signaling events, including the activation of PLCγ.
LPAR signaling impairs IP3R activity and inhibits intracellular calcium stores release in lymphocytes
PLCγ activity produces IP3 which engages the IP3R calcium channel subsequently promoting the release of ER calcium stores into the cytosol. Cytosolic calcium release is rapidly followed by extracellular calcium influx via ORAI channels with resulting cytosolic calcium levels reaching low micromolar concentrations critical for efficient antigen receptor signaling. PLCγ activation was found to be intact in the presence of LPA, as indicated by tyrosine phosphorylation, suggesting that LPAR signaling was inhibiting elevation in cytosolic calcium either by impeding IP3R-mediated calcium release from intracellular stores and/or the ORAI-mediated extracellular calcium entry through the plasma membrane. To evaluate these two possibilities, we measured calcium mobilization using flow cytometry after antigen receptor stimulation in the absence and presence of LPA with the A20 B cell line and primary mouse splenic T cells, with both cell types displaying similar results. Specifically, when lymphocytes were stimulated via the antigen receptor in the absence or presence of 1–20μM LPA, and ethylene glycol-bis(2-aminoethylether)-N,N,N′,N′-tetraacetic acid (EGTA) was present in the medium to prevent extracellular calcium influx, we observed a dose-dependent inhibition of calcium release from intracellular stores which strongly suggested that LPAR signaling was impeding IP3R activity.76,80 Because we were concerned that incomplete calcium ER stores release may not fully activate ORAI activity at the plasma membrane, we directly evaluated whether LPAR signaling impaired ORAI activity. To accomplish this, we used thapsigargin, a plant toxin that specifically inhibits the sarcoplasmic-ER calcium ATPase (SERCA), which is an ER calcium pump whose function is to constitutively replenish ER calcium stores with cytosolic calcium.85 Lymphocytes treated with thapsigargin fully release calcium into the cytosol from ER stores (albeit with slower kinetics than antigen-receptor stimulation) and promote STIM1/2 to fully activate the ORAI channel. Once calcium stores release is complete, additional calcium given in the medium overcomes EGTA chelation and permits extracellular calcium entry via ORAI channels. Neither the A20 B cell line nor primary splenic T cells demonstrate any impairment of the ORAI channel when LPA is included in these analyses, suggesting that LPAR signaling impaired intracellular calcium mobilization via selectively inhibiting IP3R activity.76,80
The results from two additional experiments further confirmed IP3R activity induced by antigen receptor engagement is inhibited by LPAR signaling. In the first, A20 B cells were loaded with a membrane-permeable caged-IP3 that was released upon exposure to UV light. When, A20 B cells were loaded with caged-IP3 and either treated or not with 5μM LPA immediately prior to UV excitation, LPA treated cells displayed reduced calcium release from intracellular stores, demonstrating that LPA inhibition of calcium mobilization occurs downstream of IP3 generation.76 Further, with primary splenic T cells, adenophostin A, an IP3R-specific agonist more potent than IP386, was able to fully rescue the IP3R-mediated calcium release from intracellular stores that was otherwise significantly reduced in the presence of LPA.80 Considered in aggregate, these data provide strong evidence that the presence of LPA impairs antigen receptor-induced IP3R activity leading to reduced intracellular calcium mobilization and subsequent signaling in lymphocytes. Cell biological and molecular studies described below address how LPAR signaling impacts cytotoxic CD8 T cell immune synapse formation and demonstrate that as a consequence of LPAR-mediated cytoskeletal reorganization, IP3R localization during synapse formation is perturbed possibly accounting for LPA-induced suppression of IP3R activity.
Importantly, we recognize that LPAR signaling in different cell types can mobilize calcium from internal stores including in immune cells such as NK cells, eosinophils, dendritic cells and the Jurkat leukemia cell line87–96 but is not observed with T or B lymphocytes,76,77 likely reflecting differences in expression of PLC isoforms and Gα family heterotrimeric G-proteins between cell types. In particular, lymphocytes express relatively high levels of PLCγ isoforms (PLCγ1 by T cells and PLCγ2 by B cells) whereas PLCβ isoforms are more broadly expressed and display reduced levels in T and B lymphocytes relative to PLCγ.97,98
TCR-mediated stimulation of ERK activity is inhibited by LPA
TCR signaling activates PLCγ1 which hydrolyzes membrane-bound PIP2 to produce IP3 and DAG resulting in a bifurcation of the signaling pathway. As discussed above, IP3 production leads to elevated cytosolic calcium levels necessary to promote transcriptional activity and maturational programs. DAG production as a product of TCR signaling promotes the activation of the mitogen-activated protein kinase (MAPK) pathway involving Ras-Erk pathway via inducing PKCθ and RasGRP.99–102 Thus, it was of interest to understand if LPA also regulated TCR signaling downstream of DAG production since both calcium mobilization and Erk activity are required for important lymphocyte-specific processes such as cytotoxicity103–105 and cytokine secretion106–111. Accordingly, we also evaluated Erk activity as measured by levels of pERK (Erk1/2; Thr202/Tyr204) after stimulation of the TCR with and without LPA present. Both primary mouse and human T cells isolated from peripheral blood stimulated with anti-CD3 in the presence of 10μM LPA revealed a diminution in the activation of Erk that, for mouse T cells, was shown to depend on LPA5 expression78 as described below. Thus, as further demonstrated in the next section, LPA5 signaling negatively impacts acute TCR-induced signaling pathways that include the calcium mobilization and ERK activation that bifurcate after PLCγ1 hydrolysis of PIP2.
An LPA5-Gα13-Arhgef1 signaling axis inhibits antigen receptor mediated signaling
To determine the LPAR(s) responsible for the observed LPA-induced suppression of antigen receptor signaling, short hairpin RNAs (shRNAs) specific for Lpar genes were initially used and expressed in mature primary B cells to demonstrate that LPA-mediated suppression of BCR signaling was dependent on LPA5 as this inhibition was lost when Lpar5 expression was silenced by RNA interference76. The conclusion that an LPA-LPA5 axis suppresses antigen receptor signaling of intracellular calcium was further genetically confirmed in mouse T lymphocytes using splenic T cells individually deficient for LPA2, LPA5 and LPA6. That is, LPA-mediated antagonism of TCR-induced calcium mobilization was maintained in Lpar2−/− and Lpar6−/− T cells but lost in Lpar5−/− T cells.76–78 Importantly, human T cell TCR-mediated calcium mobilization was also inhibited by LPA and this inhibition was relieved with an LPA5-specific antagonist indicating that LPA impairment of human TCR-induced calcium mobilization is also dependent on LPA5 signaling.78
LPA5 receptor-proximal signaling events leading to antigen receptor suppression was characterized using both biochemical analyses of the A20 mature B cell line and genetic in vivo analyses of mature B cells from mouse mutants.76 Specifically, retroviral-mediated expression of shRNAs specific for the Gα proteins that associate with LPA5, Gαq and Gα12/133,17, in A20 B cells revealed that Gα12/13, but not Gαq, expression was required for LPAR-mediated suppression of BCR-induced calcium mobilization.76 Furthermore, Arhgef1, a hematopoietically-restricted protein able to regulate GPCR signaling and activate the RhoA GTPase, was also further determined to be required for LPA5 suppression as evidenced by the inability of LPA to suppress BCR signaling in Arhgef1−/− B cells.76 In particular, Arhgef1 harbors a regulator of G protein signaling (RGS) domain previously shown to associate with (active) GTP-bound Gα12 and Gα13 G-proteins and accelerate their GTPase activity thereby aiding in return to the inactive GDP-bound state and terminating GPCR signaling.112 In addition, Arhgef1 is also a guanine nucleotide exchange factor (GEF) and association with Gα13 stimulates its RhoA GEF activity.113 As a consequence of LPA5-Gα12/13 mediated signaling, Arhgef1 is expected to attenuate LPA5 signaling and subsequently activate RhoA; a well-characterized and critical regulator of the actin cytoskeleton. Indeed, LPA treatment alone of primary human T cells leads to activation of RhoA.80
LPA suppresses T lymphocyte activation, proliferation and effector functions
TCR-mediated calcium mobilization and ERK activation are important for cytokine production by T cells109,114,115 and cytotoxic activity by CD8 T cell effectors.103–105 The ability of LPA to suppress both TCR-induced ERK activity and calcium mobilization suggested that LPA suppression of CD8 T cell TCR signaling would be observed in in vitro and in vivo assays of lymphocyte activation, proliferation and function. The ability of LPA to suppress TCR-mediated cell activation and function was characterized using primary mouse T cells stimulated by antigen presenting cells (APCs) presenting peptide in the context of major histocompatibility complex (MHC). To accomplish this, we relied on OT-I TCR transgenic T cells with specificity for the chicken ovalbumin (Ova)-derived SIINFEKL peptide presented by MHC class I H-2b-expressing APCs.116 Furthermore, LPA is a relatively labile lipid that is quickly degraded in vitro and physiologically.62,70 Accordingly, many of these in vitro and in vivo experiments exploited a metabolically stable LPA mimic, octodecenyl thiophosphate (OTP)117–119, developed and provided by a long-standing collaborator in our LPA studies, Gabor Tigyi, PhD at the University of Tennessee Health Sciences Center.
Antigen-specific TCR-mediated induction of activation marker expression and cell proliferation are suppressed by LPA in vitro and in vivo
Initial in vitro experiments evaluated whether LPA, or the metabolically-stable LPA analog, OTP, suppressed the ability of SIINFEKL-presenting splenocytes to stimulate naïve OT-I CD8 T cells.77 Experimental readouts in these diverse experiments were TCR-induced changes in the surface expression of the CD25, CD69, CD62L and CD44 activation markers in addition to proliferation.77,78 Consistent with the ability of LPA5 to inhibit TCR-induced calcium mobilization and Erk activation, in each case we found the presence of LPA/OTP significantly diminished expression of the CD25, CD69 and CD44 activation markers, impaired down-regulation of CD62L and potently suppressed proliferation by in vitro TCR-stimulated naïve CD8 T cells. Importantly, OTP did not perturb in vitro T cell viability over the range of concentrations used in these studies, suggesting that OTP in vivo treatment did not impact T cell survival.77,78
To promote in vivo LPAR signaling by OT-I CD8 T cells during antigen-specific TCR stimulation, we delivered OTP (or a vehicle control) subcutaneously every 8 hours for 3 days (OTP has an approximate 5.5 hour in vivo half-life; G. Tigyi, personal communication). OTP treatment was initiated one day after the intravenous adoptive transfer of naïve CFSE-labeled OT-I CD8 T cells and one hour before the subcutaneous transfer of SIINFEKL-pulsed bone marrow-derived dendritic cells (BMDCs).77 As expected, in the absence of experimentally-induced LPA signaling by OTP, three days after peptide-pulsed BMDC transfer, OT-I CD8 T cells in the draining lymph node were CD25+, had extensively diluted CFSE and accumulated to considerable numbers. Strikingly, OTP treatment during this period suppressed CD25 (mean fluorescent intensity) expression, and CFSE dilution by OT-I CD8 T cells and their cumulative numbers were 10-fold reduced compared to vehicle treated mice. Further, compared to wild type, virtually all LPA5-deficient OT-I CD8 T cells stimulated in vivo by peptide-pulsed BMDCs had divided and accumulated to six-fold higher numbers in the draining lymph node three days after stimulation.77 These findings provide abundant demonstration that LPA5 signaling inhibits CD8 T cell antigen-specific TCR signaling, activation and proliferation in vitro and in vivo.
LPA suppresses in vitro and in vivo CD8 T cell cytotoxic function
LPA5 in vivo signaling suppresses antigen-specific T cell activation leading to a reduced accumulation of antigen-specific T cells in the draining lymph node77 and we were interested to understand if LPA signaling via LPA5 further altered effector CD8 T cell function. Initial assessment of effector CD8 T cell function relied on intracellular expression of IFNγ and TNFα after in vitro antigen-specific stimulation and did not reveal any substantial expression differences between Lpar5−/− and wild type T cells.77 A similar cytokine analysis of in vivo tumor-specific CD8 T cells harvested from a subcutaneously implanted melanoma tumor revealed comparable expression levels between Lpar5−/− and wild type T cells.77 Thus, early findings did not suggest that LPA5 signaling regulated effector CD8 T cell function as measured by induced intracellular cytokine expression, however, as described in a later section, LPA is found to impair cytokine secretion as a result of LPA-driven modulation of the cytoskeleton. Further, when effector CD8 T cell cytotoxic activity was directly tested, LPAR signaling again suppressed T cell killing activity in vitro and, in vivo, this suppression was shown to be LPA5-dependent.120
The ability of LPA5 to regulate cytotoxic killing by CD8 T cells was first investigated with in vitro killing assays relying on quantitative real time imaging (Incucyte Live-Cell analysis) of effector CD8 T cells actively killing antigen-specific B16 melanoma cells over 24 hours.78 Specifically, primary naïve OT-I CD8 T cells were stimulated in vitro with SIINFEKL-pulsed APCs to generate Ova-specific cytotoxic effector CD8 T cells that were assayed for killing activity towards B16 melanoma cells either stably expressing chicken Ova (B16.cOVA) or pulsed with SIINFEKL peptide.78 Here, in a dose-dependent manner, OTP inhibited both cytotoxic OT-I CD8 T cell Ova-specific killing and peripheral blood-derived human CD8 T cell allogeneic killing of a breast cancer cell line.78
The ability of LPAR signaling to impair CD8 cytotoxic T cell target cell killing in vitro is supported by in vivo experiments and further provided strong evidence that LPA5 inhibitory signaling is driven by endogenous systemic LPA levels78,120. This was quantitatively demonstrated using an in vivo cytotoxic killing assay121 which measured the ability of Ova-specific CD8 T cells, generated in vivo after Ova immunization, to eliminate adoptively-transferred (antigen-specific) SIINFEKL peptide-presenting APCs. In one approach, CD8 T cell in vivo killing was measured in Enpp2+/− mice, which harbor 50% LPA systemic levels compared to wild type.63,71 Here, wild type and Enpp2+/− mice were immunized with Ova and found to elicit comparable frequencies of SIINFEKL-tetramer-positive Ova-specific CD8 T cells 4 days later.78 SIINFEKL-pulsed and non-pulsed fluorescently-distinguishable target cells were co-transferred into immunized mice and specific killing measured (compared to killing of irrelevant (HSV1) peptide-pulsed and non-pulsed target cell transfers) one day later as measured as loss of the antigen-specific target cell population. These experiments revealed that endogenously-generated Ova-specific CD8 T cells in Enpp2+/− mice, harboring reduced LPA systemic levels, but (presumably) the same wild type T cell repertoire, killed twice the frequency of target cells compared with Ova-specific CD8 T cells from wild type mice.78 This indicates that reducing the systemic levels of LPA by half results in a two-fold increase in vivo antigen-specific killing. In additional approaches, wild type and Lpar5−/− mice were immunized with Ova (again generating comparable frequencies of Ova-tetramer-specific CD8 T cells four days post-immunization) and SIINFEKL-pulsed and irrelevant peptide-pulsed antigen presenting cells transferred to immunized mice. Here, Ova-specific Lpar5−/− CD8 T cell in vivo killing activity was significantly better than wild type.120 The final iteration of this approach involved the adoptive transfer of either wild type or Lpar5−/− OT-I CD8 T cells into a wild type host immediately prior to Ova immunization. Five days later peptide-pulsed and non-pulsed target cells were transferred and two hours later cytotoxic Lpar5−/− OT-I CD8 T cells were found to display significantly better target cell killing compared to wild type OT-I CD8 T cells. 120 In aggregate these in vitro and in vivo studies not only show that systemic LPA levels limit antigen-specific CD8 T cell killing via LPA5 but also provide strong evidence that elevated systemic LPA levels, as occurs with cancers and chronic viral infections, would robustly impair adaptive cellular immunity.
As discussed above, the extracellular LPA lysophospholipid signals to T cells via LPA5 to inhibit CD8 T cell TCR signaling, activation, proliferation and cytotoxic activity, at least in part, by impairing TCR-induced IP3R activity.76,80 Mature T (and B) lymphocytes in both humans and mice express LPA2, LPA5, and LPA6, but LPAR expression is not restricted to lymphocytes or immune cells as LPARs are widely expressed by diverse cell types.122,123 This raises the question of why does LPAR signaling regulate CD8 T cell TCR signaling and cytotoxic activity and how is this accomplished? Currently, we propose systemic LPA signaling via LPA5 expressed by lymphocytes acts as a form of peripheral tolerance that contributes to the suppression of basal and potentially energetically-wasteful polyreactive or weak self-reactive T and B cell antigen receptor signaling. In the following sections, we discuss in greater detail how specifically LPA serves as a tolerizing mechanism in CD8 T cells by impairing cytoskeletal reorganization and bioenergetics, which are both required for antigen-specific killing.
LPA obstructs TCR signaling and CD8 T cell effector cell killing ability by subverting the actin and microtubule cytoskeleton
An LPA5-Gα13-Arhgef1 signaling axis impedes IP3R activity and Arhgef1, as a RhoGEF, would be expected to promote RhoA activation, possibly to regulate the actin cytoskeleton. Thus, an immunofluorescence microscopy study was undertaken to understand how LPAR signaling altered the cytoskeleton during the formation of an immunological synapse (IS) between a cytotoxic CD8 T cell and antigen-specific target cell. Here, attention was focused on the formation and function of the IS that not only defines the location of TCR recognition of target cells but also where cytolytic granules (and cytokines) are secreted to kill a target cell.80 The ability of CD8 T cells to kill an infected or transformed cell requires that killing activity be very carefully and specifically controlled to be directed precisely towards the target cell and avoiding collateral damage of other nearby host cells.124,125 Further, naïve CD8 T cells do not exert cytotoxic activity on initial specific antigen encounter but instead become activated, proliferate and develop into effector cytotoxic CD8 T cells expressing perforin and granzyme B stored within cytolytic granules. Thus, IS formation and function were assessed in an antigen-specific manner using primary effector CD8 T cells:target cells. This study revealed that the presence of LPA, at the time antigen-specific CD8 T cell:target cell conjugates were established, significantly perturbed IS formation in several important features that rely on the actin and microtubule cytoskeleton (Figure 3).80
Figure 3.

LPA alters the regulation of the cytoskeleton during CD8 T cell:target cell IS formation in manner feasibly accounting for the resulting impairment in TCR signaling and killing activity. Top: Immunofluorescence microscopy of primary cytotoxic CD8 T cells (T; cell on right) shortly after immune synapse formation with antigen-specific target B cell (TC: cell on left). IS were established in the presence of vehicle or with wild type cells (left 3 images) or in the presence of LPA (OTP) or with mDia1−/− T cells (right 3 images). Staining: DAPI (blue) F-actin (green), perforin (red) and IP3R1, mDia1, IFNγ (yellow in top 4 rows). Bottom left: Schematic of an IS between an antigen-specific CD8 T cell:target cell showing IP3R, RhoA mDia1, polymerized actin and stable microtubules positioned at the IS where cytotoxic granules and cytokines are transported to be secreted. Bottom right: Presence of LPA results in inefficient IP3R localization to the IS and altered localization of RhoA and F-actin and perturbed microtubule detyrosination that impairs TCR signaling (see text), perforin and cytokine targeted release.80
LPA and TCR induction of the RhoA GTPase and actin polymerization in CD8 T cells
The rapid reorganization of both the actin and microtubule cytoskeleton in effector CD8 T cells upon target cell contact has long been appreciated126–132 and is of particular importance during physiological CD8 T cell TCR signaling and killing initiated by immune synapse formation with an antigen-presenting target cell.125,133–135 TCR signaling initiated via synapse formation with an APC promotes actin and microtubule polymerization and reorientation of the centrosome, or microtubule-organizing center (MTOC),134–136 which is necessary for the transport of cytolytic granules and cytokines to the IS for secretion.136,137 The RhoA GTPase is considered to play a major role in regulating the actin cytoskeleton, but RhoA also contributes to regulating microtubule dynamics by stabilizing microtubules through its effector, Diaphanous-related formin-1 (mDia1).138–143 It is thus not surprising, but also not well appreciated, that TCR signaling induced by anti-CD3 stimulation of purified single T cells in suspension promotes RhoA activation80,144 as well as actin polymerization, which has been more broadly characterized.125,144–147 Furthermore, both RhoA activity and polymerized actin are not only required for antigen-specific APC-induced TCR signaling but also for a number of important T cell biological processes that include integrin adhesion, immune synapse formation and target cell killing.135,145–150
On IS formation between a T cell and target cell, filamentous (F-) actin quickly accumulates within the T cell along the juxtaposed plasma membranes and is clearly observed by immunofluorescence microscopy.80,134 TCR signaling also induces RhoA activation and both total and active (GTP-bound) RhoA are not only found at the antigen-specific IS but also at the uropod of the polarized cytotoxic T cell.80 In agreement with the ability of T cell-expressed LPARs (LPA2, LPA5 and LPA6) to signal via a Gα13-RhoA axis151, LPA alone activated RhoA and the presence of LPA during the formation of T cell:target cell conjugates led to significant changes in the cellular locations of both RhoA and polymerized actin.80 Although cytotoxic T cell:target cell conjugates established in the presence of LPA revealed a similar localization of RhoA and F-actin along the IS, when LPA was present, F-actin further localized along the cell body accumulating at the uropod whereas RhoA (total and active) expression was precluded from uropod positioning in cell conjugates and was restricted to the IS.80 Thus, concurrent LPAR signaling at the time of conjugate formation perturbed the localization of both RhoA and polymerized actin in cytotoxic CD8 T cells.
Again, consistent with an LPA5-Gα13-Arhgef1 axis, LPA treatment of human primary T cells alone in the absence of TCR signaling induces significant levels of (active) GTP-RhoA that are considerably elevated compared with those induced by TCR stimulation alone.80 Intriguingly, simultaneous TCR and LPAR signaling by naïve T cells results in active RhoA levels that are slightly but significantly elevated relative to TCR signaling alone but not near the levels induced by LPAR signaling alone. This suggests that when both TCR and LPAR signaling occurs simultaneously, the cellular levels of RhoA required by TCR signaling supersede those needed for LPAR signaling. Regardless, TCR signaling promotes both RhoA activity and actin polymerization whereas LPAR signaling induces RhoA activation but fails to induce rapid actin polymerization in T cells.80 This suggests that early induction of RhoA activity in T cells by LPAR signaling is not devoted to actin polymerization.
LPA regulates the microtubule cytoskeleton and impairs mDia1 and IP3R positioning to the IS
In T cells, microtubules have been shown to be acetylated and detyrosinated, post-translational modifications associated with microtubule stabilization to facilitate the transport of the TCR, ZAP70, and other cargo to the immunological synapse.152–154 In other cell types LPA has been shown to activate RhoA and its effector mDia, a formin which not only promotes actin polymerization but also regulates microtubule stability through detyrosination.138,142,155–157 mDia is expressed by T cells and, in its absence, T cells display impaired chemotaxis and trafficking to secondary lymphoid organs in addition to reduced TCR-induced actin polymerization, cell polarity and proliferation.158,159 When we evaluated microtubule acetylation and detyrosination in T cell conjugates established in the presence or absence of LPA, we found LPA did not appear to influence microtubule acetylation but did impair microtubule detyrosination. Furthermore, in wild type CD8 T cell:target cell conjugates, mDia1 is found in the central region of the IS. However, in the presence of LPA, mDia1 was not centrally located but was instead enriched at the periphery of the IS.80 Together these observations reveal that LPAR signaling at the time of effector CD8 T cell and target cell immune synapse formation leads to altered localization of RhoA, mDia1, actin polymerization and microtubule detyrosination.80 Thus, LPAR signaling alters the regulation of both the actin and microtubule cytoskeleton leading to impaired TCR signaling and immune synapse formation.
As discussed, LPA signaling through LPA5 suppresses TCR signaling by impairing IP3R activity.76,80 IP3R1 has previously been shown to co-localize with the TCR in anti-CD3 stimulated Jurkat T cells where the Fyn tyrosine kinase phosphorylates IP3R1 (Tyr 353), thereby enhancing its activity and increasing sensitivity to IP3.160,161 Thus, it was not surprising to find that the IP3R1 calcium channel also rapidly and exclusively positioned at the immune synapse in antigen-specific wild type CD8 T cell:target cell conjugates.80 The positioning of the IP3R close to the IS at the site of TCR-peptide/MHC interaction would be advantageous for several reasons. Not only would this allow the IP3R calcium channel to provide intracellular calcium in proximity to the TCR signalosome and effectors that are dependent on calcium for activity, but this positioning would also be important for maximizing IP3R activation by Fyn and placing the IP3R in close proximity to where PLCγ1 is generating IP3. Notably, however, when LPA was present during conjugate formation, IP3R1 location to the IS was considerably diminished and instead IP3R1 was now enriched at the periphery of the IS and at the distal uropod end of the cell. The positioning of IP3R1 at the synapse is dependent on TCR-induced mDia1 activity as IP3R1 was also aberrantly positioned in mDia1−/− OT-I CD8 T cell:target cell conjugates, but not in Fmnl1-deficient OT-I CD8 T cells that lack a different T cell formin.80 Indeed, LPAR signaling during IS formation led to altered positioning not only of IP3R1 but also mDia and IP3R1 positioning was similar whether conjugates were established in the presence of LPA or with T cells lacking mDia1. Finally, positioning of the IP3R to the IS in wild type T cell:target cell conjugates was dependent on both actin and microtubule polymerization as pharmacological inhibitors of polymerization impaired the position of the IP3R to the IS.80 These findings show that LPAR signaling during T cell target cell killing subverts mDia1 localization and activity needed for IP3R positioning at the IS.
Considered together, these data suggest that full IP3R activity depends on its location requiring the calcium channel to be in close proximity of the IS at the plasma membrane where TCR-associated Fyn phosphorylates and fully activates IP3R1 and where PLCγ1 is generating IP3 via hydrolysis of membrane PIP2. LPA5 regulation of the cytoskeleton prevents efficient IP3R positioning to the IS where its ligand is being produced and, likely as a result of inefficient IP3R activation and IP3 diffusion, leads to inefficient signaling by those IP3Rs not at the synapse. Whether LPA5-induced signaling effectors further impair IP3R activity by a more direct mechanism cannot be ruled out. However, LPA5 inhibition of calcium stores release is overcome with a (potent) IP3R agonist that has a higher affinity for IP3R1 than IP3. This argues that LPA-driven impairment of IP3R activity is likely not regulated by post-translational or protein-association inhibitory mechanisms but is compatible with LPA-mediated cytoskeletal changes that physically constrain IP3R from being fully activated and with full access to lP3.
LPA-mediate reorganization of the cytoskeleton impedes cytolytic granule and cytokine secretion at the IS
A critical functional consequence of LPA-mediated subversion of the actin and microtubule cytoskeleton during target cell killing is that perforin-containing cytolytic granules are not delivered efficiently to the IS78 (Figure 3) and ultimately impair both in vitro and in vivo CD8 T cells effector cytotoxicity.78,120 Furthermore, IL-2 and IFNγ are directionally secreted in CD4 T cells137 and in cytotoxic CD8 T cell:target cell conjugates where both IL-2 and IFNγ are transported to the IS for secretion80. In contrast, TNFα is secreted in a multi-dimensional direction in both CD4 and CD8 T cells80,137. Importantly, LPAR signaling by effector CD8 T cells does not impact the expression of either IL-2, IFNγ or TNFα as determined by intracellular cytokine staining.77,80 However, LPAR signaling impedes the directional secretion of IL-2 and IFNγ, but not the secretion of TNFα.80
LPA regulation of ciliogenesis and CD8 T cell immune synapse formation
It is worth noting that immune synapse formation by cytotoxic CD8 T cells has been recognized to have possible evolutionary origins in primary cilia formation.162–165 Primary (non-motile) cilia are microtubule-based structures that emanate from the plasma membrane and serve as sensory receptors or ‘antennae’ for most vertebrate cells, but evidently are not formed in T cells.166 Similar to cilia formation where the centrosome, or MTOC, docks close to the plasma membrane and directs microtubule assembly of the cilium, on establishing an IS the MTOC of the T cell also polarizes close to the plasma membrane at the immune synapse where it directs microtubule assembly for the release of cytolytic granules and secretory vesicle cargo, such as cytokines, at the cell-cell interface.166 Moreover, the intraflaggelar transport protein, IFT20, characterized in contributing to cilia assembly also positions to the IS in cytotoxic T cell conjugates.163 Of interest, as sensory receptors, a number different receptors are located on the primary cilium including a number of GPCRs167 that respond and integrate information from diverse stimuli such as chemical, mechanical, light, temperature and osmolality to regulate tissue homeostasis. Defects in primary ciliogenesis lead to multisystemic genetic disorders, known as ciliopathies. Recently, variants in CCDC28B (Coiled-coil domain-containing protein 28B), a gene encoding a cilia-associated protein, was found in a subset of common-variable immune deficiency patients whose T cells were impaired in IS formation.168 Further, an LPA-LPA1 axis has been reported by several groups to promote disassembly of cilium in cultured human retinal pigment epithelial cells or human primary astrocytes thereby promoting cilia disassembly.169–171 Thus, LPAR regulation of IS formation has evolutionary origins in LPA regulation of the primary cilium.
LPA5 expression and signaling by CD8 T cells impedes tumor immunity
Diverse cancer cells often aberrantly turn on expression of ENPP2172 and systemic levels of LPA are often elevated with certain cancers.173 Indeed, a large body of literature has provided evidence that LPA is able to promote cell proliferation and facilitate tumor growth via different mechanisms that largely depend on signaling by LPA1–3 Edg family members.174,175 The notion that ATX expression and LPA1–3 signaling promote tumorigenesis is strongly supported by a mouse model of enforced transgenic expression of either ATX or LPA1, LPA2, or LPA3 in mammary breast epithelium.174 Transgenic lines for all three transgenes frequently developed late-onset, estrogen receptor (ER)-positive, invasive, and metastatic mammary cancer.174 Thus, abundant data supports the notion that an ATX-LPA axis promotes tumorigenesis that, in turn, can increase systemic levels of LPA. In our studies we have observed that physiological levels of LPA (i.e., not intentionally manipulated) signaling via LPA5 interferes with both cellular and humoral antigen-specific immunity.76,78,120 As example, T cell immunization of Enpp2+/− mouse mutants, which express approximately half the levels of systemic LPA as wild type mice63,71, leads to a two-fold increase of in vivo antigen-specific target cell killing78, and Ova-specific Lpar5−/− CD8 T cells, when directly compared to Ova-specific wild type CD8 T cells, kill adoptively-transferred Ova peptide presenting target cells at higher and significant frequencies.120 Further, immunization of Lpar5−/− and wild type mice with a model antigen results in heighted antigen-specific antibody responses by LPA5-deficient B cells.76 With this in mind, we addressed whether LPA5-mediated in vivo signaling tempers tumor-specific CD8 T cell anti-tumor immunity using mouse models of tumor growth and metastasis.
For these tumor-specific in vivo experiments, OT-I TCR transgenic T cells and B16 melanoma cells stably-expressing chicken ovalbumin (B16.cOva) were employed. B16.cOva melanoma cells express and process ovalbumin followed by the presentation of SIINFEKL with MHC class I that is recognized by adoptively transferred mature ‘tumor-specific’ wild type and Lpar5−/− OT-I CD8 T cells. B16.cOva cells were implanted subcutaneously in the flank of mouse cohorts and allowed to grow for 5 days and measured daily. At day five, when tumors were palpable, either C57BL/6 or Lpar5−/− OT-I CD8 T cells were adoptive-transferred intravenously and mice monitored for an additional 8 days77 and again one week later78 (day 20 of tumor implantation). Beginning five days after implant and at the time of adoptive T cell transfer, B16.cOva tumor size increased at a nearly linear rate over the next eight days in wild type hosts and this growth rate was not changed with the transfer of wild type OT-I CD8 T cells reflecting the inability of a C57BL/6 wild type host to control this aggressive syngeneic melanoma cell line.176 Transfer of Lpar5−/− OT-I CD8 T cells 5 days after tumor implant, in contrast, significantly reduced tumor growth at six and eight days post T cell transfer and B16.cOva tumors were significantly reduced in mass while harboring higher numbers of OT-I CD8 T cells compared to wild type T cell transfers.77
In another approach, B16.cOva melanoma cells were intravenously injected into C57BL/6 wild type hosts followed immediately with the adoptive transfer of either wild type or Lpar5−/− OT-I CD8 T cells. Twenty days after tumor cells were injected and T cells transferred, mice that received LPA5-deficient tumor-specific T cells harbored significantly fewer B16.cOva-derived tumors in the lungs that were not only significantly smaller in size but also contained considerably more LPA5-deficient OT-I CD8 T cells relative to than tumors transferred with wild type CD8 T cells (Figure 4).120 Impressively, Lpar5−/− cytotoxic CD8 T cells also expressed reduced levels of the Tim-3 and Lag-3 inhibitory receptors and the Tox transcription factor120, which taken together with the observed differences in tumor burden and improved tumor immunity, strongly suggest that Lpar5−/− tumor-specific CD8 T cells were less dysfunctional.
Figure 4.

Lpar5−/− tumor-specific CD8 T cells provide better control of B16 lung tumors than wild type CD8 T cells. A) Representative hematoxylin & eosin (H&E) histology images of day 20 B16.cOVA tumors after i.v. transfer together with either wild type (left) or Lpar5−/− (right) OT-I CD8 T cells. Scale bars=100 μm. Right: Quantification of B16.cOva tumor size after transfer of B6 (filled bar) or Lpar5−/− (open bar) OT-I CD8 T cells. B) Number of B6 (filled bar) or Lpar5−/− (open bar) OT-I CD8 T cells found in lung tumors. Student’s t test *p < 0.05 and **p < 0.005.120
These proof-of-principle studies, while contrived, provide strong in vivo evidence that LPA5 inhibitory signaling by bona fide cytotoxic CD8 T cells, and in response to endogenously produced LPA, impairs tumor immunity. Further, ATX is often aberrantly expressed by diverse cancers to promote cell proliferation and tumorigenesis and is often accompanied by heightened systemic LPA levels. Thus, this suggests that the ATX-LPA axis serves to promote tumorigenesis (via induced ATX and LPA1–3 expression and resulting LPA autocrine activity) while simultaneously this tumor-derived LPA exerts paracrine activity suppresses cytotoxic CD8 T cell tumor immunity (Figure 5).
Figure 5.

Schematic of induction of ENPP2 expression and subsequent ATX production by a transformed cell leading to local LPA production. LPA then signals via LPA1–3 expressed by malignant cells to promote autocrine cell proliferation and tumorigenesis (left) or LPA5 expressed by tumor-specific CD8 T cells to suppress tumor immunity.
LPA5 signaling by effector CD8 T cells modifies cellular metabolic energy source and efficiency
This review has highlighted in vitro and in vivo studies documenting CD8 T cell TCR signaling and killing activity are dysregulated if LPA simultaneously engages LPA5 during T cell recognition of tumor-specific antigen. However, as a bioactive lysophospholipid, LPA levels are not only increased with obesity but LPA has also been reported to alter mitochondrial metabolism in skeletal muscle and induce de novo lipid synthesis in ovarian cancer cells.45,46,50,177 Thus, it was hypothesized that LPA may also signal to T cells to regulate metabolic fitness and, consequently, functional activity. Consistent with this notion, treatment of primary cytotoxic OT-I CD8 T cells with a physiological concentration (1μM) of LPA for up to four hours significantly modulated T cell metabolism.120 This is illustrated, in part, by the evaluation of cytotoxic CD8 T cells treated in vitro with LPA followed by the measurement of oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using the Seahorse Cell Mito Stress test to assess mitochondrial function. These results revealed that LPA treatment of OT-I primary CD8 T cells led to significant increases in both basal T cell respiration and maximal respiratory capacity in effector CD8 T cells and, importantly, the ability to achieve maximal respiratory capacity was dependent on LPA5 (Figure 6 and ref. 119). Intriguingly, while promoting an increase in respiratory capacity of effector CD8 T cells, LPA treatment also promoted an increase in extracellular acidification of the media indicating that glycolysis was ongoing and generating and releasing lactate from the cell further suggesting an alternate non-glycolytic endogenous energy source was being used to increase mitochondrial metabolism (Figure 6). One likely non-glycolytic energy source are fatty-acid lipids, which are often stored in lipid droplets, a cell reservoir of neutral lipids produced by most cells and whose presence can reflect metabolic changes and access to nutrients.178 Whether LPA-mediated regulation of T cell metabolism also changed the presence of lipid droplets was tested with BODIPY staining of neutral lipids and revealed a significant loss of lipid droplet content after 30 min – four hours of LPA treatment.120 Moreover, preventing fatty-acid transport into the T cell mitochondria with etomoxir (1μM), a pharmacological inhibitor of carnitine palmitoyltransferase-1 (CPT-1), also led to an immediate reduction in maximal respiratory capacity.120 These findings thus show that despite ongoing aerobic glycolysis, LPA signaling via LPA5 shifts CD8 T cell metabolism to consume fatty-acids for mitochondrial respiration,120 and consistent with a previous finding showing that PD-1 promotes fatty-acid oxidation of endogenous lipids.179 Moreover, as a ubiquitous bioactive lipid signaling via GPCR cognate receptors, LPA participates in regulating the cytoskeleton and metabolism of CD8 T cells in a manner that also would be predicted to negatively impact T cell adaptive immunity.180
Figure 6.

LPA promotes an increase in basal T cell respiration, maximal respiratory capacity, proton leak and transiently elevates ATP production. Naïve (A) and effector (B) OT-I CD8 T cells were cultured in vitro with media (red) or 1 μM LPA for 30 minutes (green), 2 hrs (blue) or 4 hrs (teal) and oxygen consumption rate (OCR; left graphs) and extracellular acidification rate (ECAR; right graphs) measured. Assay was performed with injections of oligomycin (oligo), (4-(trifluoromethoxy) phenyl) carbonohydrazonoyl dicyanide (FCCP), antimycin A (ant), and rotenone (rot) at 18-minute intervals in media supplemented with 25 mM glucose. Data are n=6 technical replicates.120
LPA, tumor immunity and cancer
There is considerable interest in the ATX-LPA axis from the cancer field where different cancers are often found to aberrantly express ENPP2 thus producing extracellular LPA for autocrine LPAR signaling by tumor cells to promote cell proliferation and facilitate tumorigenesis. Evaluation of all curated non-redundant studies in cBioPortal181,182 (a cancer database harboring multidimensional genomics data sets from individuals with different cancers) reveals that individuals with cancers in which ENPP2 has been amplified have significantly worse progression-free survival and also when compared to patients with MYC amplification (Figure 7)120, a gene amplification known to be associated with poor outcomes.183 Notably, plasma LPA levels (16:0 species) are significantly higher in melanoma patients that do not respond to checkpoint blockade immunotherapy compared to melanoma patients that positively respond to checkpoint blockade therapy (Figure 7).120 Within this data, there was a single outlier in the non-responder group that did not respond to upfront immunotherapy treatment. However, when followed-up it was determined this patient was later treated with a bispecific anti-PD/ICOS antibody and is currently disease free.120 Thus, plasma LPA levels may not only serve as a prognostic indicator for therapy treatment, but antagonism of an ATX-LPA-LPAR axis may be an attractive therapeutic intervention for certain cancers.
Figure 7.

Lysophosphatidic acid as a potential prognostic marker in melanoma. A) Data analysis from The Cancer Genome Atlas (TCGA) on progression-free survival. Data was from pan-cancer data from all solid tumors in cBioPortal from the complete curated non-redundant studies (as of June 18, 2021). Cohorts were stratified based on genomic status of amplification of ENNP2, MYC, or wild type for both genes. B) Relative abundance of LPA 16:0 in stage IV melanoma responder patients (blue; complete and partial response) and non-responder patients (red; stable disease and progressive disease) measured both pre- and post-therapy treatment. Unpaired Student’s t-test where p<0.05.120
Our studies document the ability of LPA5 signaling to suppress acute in vivo TCR signaling by naïve CD4 and CD8 T cells and in response to physiological endogenous LPA levels. Based on this, in vivo LPA5 antagonism would be expected to lower the threshold for activation and proliferation of naïve CD8 T cells responding to tumor antigen resulting in a higher number of responding tumor-specific T cells, as shown in mouse tumor models and with LPA5-deficient CD8 T cells.77,120 This is also consistent with the ability of a high affinity antigen to elicit an increased number of antigen-specific CD8 T cells when compared to a lower affinity antigen.184 Further, unmutated tumor antigens are self-antigens and any endogenous naïve CD8 T cell capable of recognizing tumor self-antigen will almost certainly express low-affinity TCRs185 as high-affinity CD8 T cells are eliminated during central tolerance induction in the thymus. If, as we hypothesize, LPA5-mediated suppression of TCR and BCR signaling has evolved as a mechanism to restrain activation of lymphocytes with self- and poly-reactive antigen receptors, then LPA5 antagonism might prove further useful as a cancer therapeutic in promoting anti-tumor responses from self (tumor) antigen-specific CD8 T cells. Of course, this would need to be carefully considered as autoimmune T cell responses may also be facilitated. Along these lines, it should nevertheless be noted that for studies presented here, the OT-I TCR affinity for the SIINFEKL peptide (KD ~ 6μM) is more reflective of TCR affinities that are usually higher for pathogen-derived peptides compared to the weaker affinities normally demonstrated for bona fide tumor antigens.185
LPA, through its regulation of the actin and microtubule skeleton also impedes TCR signaling at the immunological synapse as cytotoxic effector CD8 T cells are actively engaged in killing antigen-specific tumor cells or virally infected cells. LPA5 antagonism could thus feasibly not only promote TCR signaling by naïve T cells on initial encounter with tumor antigen but also enhance tumor killing by weak-affinity cytotoxic effector CD8 T cells. Notably, current approved checkpoint blockade immunotherapy in the clinic is focused on promoting killing activity from tumor-specific effector T cells as most of the targeted inhibitory receptors (PD-1186,187, CTLA-4188,189, Tim3190,191, Lag3192,193 and TIGIT194,195) are not expressed by naïve T cells but instead their expression is induced only after initial T cell recognition of tumor antigen.
Finally, the LPA1 Edg family member GPCR has been shown to promote the disassembly of primary cilium in cultured human retinal epithelial cells and primary astrocytes.169–171 Primary cilium are found in non-cycling quiescent cells as cilia assembly has been long-recognized to be incompatible with cell proliferation,196 and primary cilia are often not expressed by cancer cells.197 Indeed, it has been suggested that newly transformed cells may need to disassemble primary cilia in order to proliferate, thus, LPA antagonism might additionally prevent cilia disassembly and proliferation by a number of different cancer cells.173
Summary and concluding remarks
The studies described in this review demonstrate that active LPA5 signaling during CD8 T cell antigen-specific recognition of a target cell competes with TCR signaling for reorganization of the actin and microtubule cytoskeleton and thus restrains the recruitment of critical effector molecules to the immunological synapse. As a result, TCR-induced intracellular calcium stores release and transport of cytolytic granules and cytokines to the IS are impaired. Further, LPA induced LPA5 signaling also regulates T cell metabolism by promoting fatty-acid oxidation for mitochondrial respiration and in a manner independent from LPA5 regulation of TCR signaling. Thus, LPA signaling, at least in part via LPA5, regulates both the cytoskeleton and metabolism of effector CD8 T cells in a manner that ultimately impinges on CD8 T cell antigen-receptor signaling and killing activity.
Our findings suggest that LPA, under normal physiological conditions, acts as an inhibitory signal to restrain T cell activation thus contributing to peripheral immunological tolerance. Tumor- and chronic infection-induced increases in systemic LPA levels would be expected to raise the TCR signaling threshold to limit CD8 T cell adaptive immunity by not only inhibiting the activation of naïve T cells to tumor or viral antigens but also by directly impeding killing activity. LPA also promotes T cell chemokinesis and movement into, and within, the lymph node 72,73,79 that likely contributes to the ability of T cells to scan APCs for possible antigen-specific interactions. Accordingly, the ability of a T cell to be fully activated is dependent on the levels of LPA present in the environment as TCR and LPAR signaling compete for the same signaling molecules to achieve their specific end.
Most LPA cognate GPCRs are able to associate with Gα12/13 and thus regulate RhoA GTPase activity in reorganizing the actin and microtubule cytoskeleton. Accordingly, a major function of systemic LPA signaling via LPA receptors may lie in cell type-specific regulation of the cytoskeleton necessary not only for cell shape but also cell division, polarity, migration and secretion. In particular, LPA receptors that associate with and signal via Gα12/13 heterotrimeric G-proteins have also been shown to engage the Hippo pathway198, an evolutionary conserved signaling pathway that responds to the cell microenvironment and regulates cell proliferation and tissue homeostasis, including organ size.199 Thus, the ability of LPA to also further participate in controlling cellular metabolism is not surprising and further suggests that LPA-LPAR cell type specific signaling may not only reorganize the cytoskeleton in a variety of diverse cells but also may regulate metabolism in response to the environment.
The therapeutic interference of the ATX-LPA axis has attracted industry attention and currently there are 14 phase I and II clinical trials evaluating ATX and LPA1 inhibitors for the treatment of systemic sclerosis, rheumatoid arthritis, pulmonary fibrosis, chronic liver disease, ovarian cancer and metastatic pancreatic cancer. Our findings would suggest ATX inhibition, which quickly reduces systemic LPA levels, would also promote tumor-specific CD8 T cell immunity. The studies presented here would also suggest LPA5 antagonism may be a useful therapeutic approach for promoting T cell immunity to certain cancers and would require the identification of effective small molecule inhibitors for this receptor. Indeed, LPA5 antagonists have been evaluated for alleviating microglia inflammatory cascades.200 Notably, LPARs are all GPCRs, which currently comprise over one-third of all small molecule drugs approved for use by the US Food and Drug Administration201,202 and drugs targeting both S1P1 and S1P5 GPCRs are in current use in the clinic for the treatment of multiple sclerosis.24 It may also be useful to investigate the potential for LPA5 agonism to suppress autoreactive lymphocyte responses in autoimmune diseases.43
Acknowledgements
We thank Moriah Castleman for comments on the manuscript, past Torres lab members, especially Jiancheng Hu (National Cancer Centre Singapore), Shannon Oda (Seattle Children’s Research Institute/UW Pediatrics) and Divij Mathew (University of Pennsylvania) for their contributions. This work was supported by NIH grants R01AI143261, R21AA02923, R01AI136534 (RMT) and R01AI152535, R21AI156232 (RP), and T32AI007405 (MD) and a Hertz Graduate Fellowship (JAT).
Footnotes
Conflict of Interest Statement
The authors declare no competing interests.
References
- 1.Aoki J, Inoue A, Okudaira S. Two pathways for lysophosphatidic acid production. Biochim Biophys Acta. 2008;1781(9):513–518. [DOI] [PubMed] [Google Scholar]
- 2.Xu Y Targeting Lysophosphatidic Acid in Cancer: The Issues in Moving from Bench to Bedside. Cancers (Basel). 2019;11(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kano K, Aoki J, Hla T. Lysophospholipid Mediators in Health and Disease. Annu Rev Pathol. 2022;17:459–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.McIntyre TM, Pontsler AV, Silva AR, et al. Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARgamma agonist. Proc Natl Acad Sci U S A. 2003;100(1):131–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang C, Baker DL, Yasuda S, et al. Lysophosphatidic acid induces neointima formation through PPARgamma activation. J Exp Med. 2004;199(6):763–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lipton JH, McMurray WC. Mitochondrial biogenesis in cultured mammalian cells. III. Synthesis of mitochondrial phospholipids by subcellular fractions isolated from normal and chloramphenicol-treated BHK-21 cells. Biochim Biophys Acta. 1977;486(2):228–242. [DOI] [PubMed] [Google Scholar]
- 7.Tokumura A, Fukuzawa K, Tsukatani H. Effects of synthetic and natural lysophosphatidic acids on the arterial blood pressure of different animal species. Lipids. 1978;13(8):572–574. [DOI] [PubMed] [Google Scholar]
- 8.Gerrard JM, Kindom SE, Peterson DA, Peller J, Krantz KE, White JG. Lysophosphatidic acids. Influence on platelet aggregation and intracellular calcium flux. Am J Pathol. 1979;96(2):423–438. [PMC free article] [PubMed] [Google Scholar]
- 9.Gerrard JM, Clawson CC, White JG. Lysophosphatidic acids: III. Enhancement of neutrophil chemotaxis. Am J Pathol. 1980;100(3):609–618. [PMC free article] [PubMed] [Google Scholar]
- 10.Tokumura A, Fukuzawa K, Yamada S, Tsukatani H. Stimulatory effect of lysophosphatidic acids on uterine smooth muscles of non-pregant rats. Arch Int Pharmacodyn Ther. 1980;245(1):74–83. [PubMed] [Google Scholar]
- 11.Lapetina EG, Billah MM, Cuatrecasas P. Lysophosphatidic acid potentiates the thrombin-induced production of arachidonate metabolites in platelets. J Biol Chem. 1981;256(23):11984–11987. [PubMed] [Google Scholar]
- 12.Watson SP, McConnell RT, Lapetina EG. Decanoyl lysophosphatidic acid induces platelet aggregation through an extracellular action. Evidence against a second messenger role for lysophosphatidic acid. Biochem J. 1985;232(1):61–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Moolenaar WH, van der Bend RL, van Corven EJ, Jalink K, Eichholtz T, van Blitterswijk WJ. Lysophosphatidic acid: a novel phospholipid with hormone- and growth factor-like activities. Cold Spring Harb Symp Quant Biol. 1992;57:163–167. [DOI] [PubMed] [Google Scholar]
- 14.Hecht JH, Weiner JA, Post SR, Chun J. Ventricular zone gene-1 (vzg-1) encodes a lysophosphatidic acid receptor expressed in neurogenic regions of the developing cerebral cortex. J Cell Biol. 1996;135(4):1071–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hla T, Lee MJ, Ancellin N, Paik JH, Kluk MJ. Lysophospholipids--receptor revelations. Science. 2001;294(5548):1875–1878. [DOI] [PubMed] [Google Scholar]
- 16.Georas SN. Lysophosphatidic acid and autotaxin: emerging roles in innate and adaptive immunity. Immunol Res. 2009;45(2–3):229–238. [DOI] [PubMed] [Google Scholar]
- 17.Sheng X, Yung YC, Chen A, Chun J. Lysophosphatidic acid signalling in development. Development. 2015;142(8):1390–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yung YC, Stoddard NC, Mirendil H, Chun J. Lysophosphatidic Acid signaling in the nervous system. Neuron. 2015;85(4):669–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Smyth SS, Kraemer M, Yang L, Van Hoose P, Morris AJ. Roles for lysophosphatidic acid signaling in vascular development and disease. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865(8):158734. [DOI] [PubMed] [Google Scholar]
- 20.Lee SC, Dacheux MA, Norman DD, et al. Regulation of Tumor Immunity by Lysophosphatidic Acid. Cancers (Basel). 2020;12(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tang X, Hou Y, Schwartz TW, Haeggstrom JZ. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids. Biochem Pharmacol. 2022;204:115208. [DOI] [PubMed] [Google Scholar]
- 22.Kihara Y, Maceyka M, Spiegel S, Chun J. Lysophospholipid receptor nomenclature review: IUPHAR Review 8. Br J Pharmacol. 2014;171(15):3575–3594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427(6972):355–360. [DOI] [PubMed] [Google Scholar]
- 24.Burg N, Salmon JE, Hla T. Sphingosine 1-phosphate receptor-targeted therapeutics in rheumatic diseases. Nat Rev Rheumatol. 2022;18(6):335–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Goetzl EJ, Kong Y, Voice JK. Cutting edge: differential constitutive expression of functional receptors for lysophosphatidic acid by human blood lymphocytes. J Immunol. 2000;164(10):4996–4999. [DOI] [PubMed] [Google Scholar]
- 26.Goetzl EJ, Rosen H. Regulation of immunity by lysosphingolipids and their G protein-coupled receptors. J Clin Invest. 2004;114(11):1531–1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wang L, Knudsen E, Jin Y, Gessani S, Maghazachi AA. Lysophospholipids and chemokines activate distinct signal transduction pathways in T helper 1 and T helper 2 cells. Cell Signal. 2004;16(9):991–1000. [DOI] [PubMed] [Google Scholar]
- 28.Blaho VA, Hla T. Regulation of mammalian physiology, development, and disease by the sphingosine 1-phosphate and lysophosphatidic acid receptors. Chem Rev. 2011;111(10):6299–6320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Choi JW, Herr DR, Noguchi K, et al. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol. 2010;50:157–186. [DOI] [PubMed] [Google Scholar]
- 30.Eichholtz T, Jalink K, Fahrenfort I, Moolenaar WH. The bioactive phospholipid lysophosphatidic acid is released from activated platelets. Biochem J. 1993;291 ( Pt 3)(Pt 3):677–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen YL, Xu Y. Determination of lysophosphatidic acids by capillary electrophoresis with indirect ultraviolet detection. J Chromatogr B Biomed Sci Appl. 2001;753(2):355–363. [DOI] [PubMed] [Google Scholar]
- 32.Baker DL, Morrison P, Miller B, et al. Plasma lysophosphatidic acid concentration and ovarian cancer. JAMA. 2002;287(23):3081–3082. [DOI] [PubMed] [Google Scholar]
- 33.Kishimoto T, Matsuoka T, Imamura S, Mizuno K. A novel colorimetric assay for the determination of lysophosphatidic acid in plasma using an enzymatic cycling method. Clin Chim Acta. 2003;333(1):59–67. [DOI] [PubMed] [Google Scholar]
- 34.Murph M, Tanaka T, Pang J, et al. Liquid chromatography mass spectrometry for quantifying plasma lysophospholipids: potential biomarkers for cancer diagnosis. Methods Enzymol. 2007;433:1–25. [DOI] [PubMed] [Google Scholar]
- 35.Hosogaya S, Yatomi Y, Nakamura K, et al. Measurement of plasma lysophosphatidic acid concentration in healthy subjects: strong correlation with lysophospholipase D activity. Ann Clin Biochem. 2008;45(Pt 4):364–368. [DOI] [PubMed] [Google Scholar]
- 36.Nakamura K, Kishimoto T, Ohkawa R, et al. Suppression of lysophosphatidic acid and lysophosphatidylcholine formation in the plasma in vitro: proposal of a plasma sample preparation method for laboratory testing of these lipids. Anal Biochem. 2007;367(1):20–27. [DOI] [PubMed] [Google Scholar]
- 37.Kano K, Matsumoto H, Kono N, Kurano M, Yatomi Y, Aoki J. Suppressing postcollection lysophosphatidic acid metabolism improves the precision of plasma LPA quantification. J Lipid Res. 2021;62:100029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Baumforth KR, Flavell JR, Reynolds GM, et al. Induction of autotaxin by the Epstein-Barr virus promotes the growth and survival of Hodgkin lymphoma cells. Blood. 2005;106(6):2138–2146. [DOI] [PubMed] [Google Scholar]
- 39.Watanabe N, Ikeda H, Nakamura K, et al. Both plasma lysophosphatidic acid and serum autotaxin levels are increased in chronic hepatitis C. J Clin Gastroenterol. 2007;41(6):616–623. [DOI] [PubMed] [Google Scholar]
- 40.Joshita S, Ichikawa Y, Umemura T, et al. Serum autotaxin is a useful liver fibrosis marker in patients with chronic hepatitis B virus infection. Hepatol Res. 2018;48(4):275–285. [DOI] [PubMed] [Google Scholar]
- 41.Kostadinova L, Shive CL, Anthony DD. Elevated Autotaxin and LPA Levels During Chronic Viral Hepatitis and Hepatocellular Carcinoma Associate with Systemic Immune Activation. Cancers (Basel). 2019;11(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nikitopoulou I, Oikonomou N, Karouzakis E, et al. Autotaxin expression from synovial fibroblasts is essential for the pathogenesis of modeled arthritis. J Exp Med. 2012;209(5):925–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schmitz K, Brunkhorst R, de Bruin N, et al. Dysregulation of lysophosphatidic acids in multiple sclerosis and autoimmune encephalomyelitis. Acta Neuropathol Commun. 2017;5(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nojiri T, Kurano M, Araki O, et al. Serum autotaxin levels are associated with Graves’ disease. Endocr J. 2019;66(5):409–422. [DOI] [PubMed] [Google Scholar]
- 45.Ferry G, Tellier E, Try A, et al. Autotaxin is released from adipocytes, catalyzes lysophosphatidic acid synthesis, and activates preadipocyte proliferation. Up-regulated expression with adipocyte differentiation and obesity. J Biol Chem. 2003;278(20):18162–18169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kulkarni P, Getzenberg RH. High-fat diet, obesity and prostate disease: the ATX-LPA axis? Nat Clin Pract Urol. 2009;6(3):128–131. [DOI] [PubMed] [Google Scholar]
- 47.Federico L, Ren H, Mueller PA, et al. Autotaxin and its product lysophosphatidic acid suppress brown adipose differentiation and promote diet-induced obesity in mice. Mol Endocrinol. 2012;26(5):786–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Reeves VL, Trybula JS, Wills RC, et al. Serum Autotaxin/ENPP2 correlates with insulin resistance in older humans with obesity. Obesity (Silver Spring). 2015;23(12):2371–2376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Orosa B, Garcia S, Conde C. The autotaxin-lysophosphatidic acid pathway in pathogenesis of rheumatoid arthritis. Eur J Pharmacol. 2015;765:228–233. [DOI] [PubMed] [Google Scholar]
- 50.D’Souza K, Nzirorera C, Cowie AM, et al. Autotaxin-LPA signaling contributes to obesity-induced insulin resistance in muscle and impairs mitochondrial metabolism. J Lipid Res. 2018;59(10):1805–1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.D’Souza K, Paramel GV, Kienesberger PC. Lysophosphatidic Acid Signaling in Obesity and Insulin Resistance. Nutrients. 2018;10(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Brandon JA, Kraemer M, Vandra J, et al. Adipose-derived autotaxin regulates inflammation and steatosis associated with diet-induced obesity. PLoS One. 2019;14(2):e0208099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sasagawa T, Okita M, Murakami J, Kato T, Watanabe A. Abnormal serum lysophospholipids in multiple myeloma patients. Lipids. 1999;34(1):17–21. [DOI] [PubMed] [Google Scholar]
- 54.Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer. 2003;3(8):582–591. [DOI] [PubMed] [Google Scholar]
- 55.Chun J, Rosen H. Lysophospholipid receptors as potential drug targets in tissue transplantation and autoimmune diseases. Curr Pharm Des. 2006;12(2):161–171. [DOI] [PubMed] [Google Scholar]
- 56.Cooper AB, Wu J, Lu D, Maluccio MA. Is autotaxin (ENPP2) the link between hepatitis C and hepatocellular cancer? J Gastrointest Surg. 2007;11(12):1628–1634; discussion 1634–1625. [DOI] [PubMed] [Google Scholar]
- 57.Masuda A, Nakamura K, Izutsu K, et al. Serum autotaxin measurement in haematological malignancies: a promising marker for follicular lymphoma. Br J Haematol. 2008;143(1):60–70. [DOI] [PubMed] [Google Scholar]
- 58.Braddock DT. Autotaxin and lipid signaling pathways as anticancer targets. Curr Opin Investig Drugs. 2010;11(6):629–637. [PubMed] [Google Scholar]
- 59.Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br J Cancer. 2010;102(6):941–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gotoh M, Fujiwara Y, Yue J, et al. Controlling cancer through the autotaxin-lysophosphatidic acid receptor axis. Biochem Soc Trans. 2012;40(1):31–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xiao YJ, Schwartz B, Washington M, et al. Electrospray ionization mass spectrometry analysis of lysophospholipids in human ascitic fluids: comparison of the lysophospholipid contents in malignant vs nonmalignant ascitic fluids. Anal Biochem. 2001;290(2):302–313. [DOI] [PubMed] [Google Scholar]
- 62.Albers HM, Dong A, van Meeteren LA, et al. Boronic acid-based inhibitor of autotaxin reveals rapid turnover of LPA in the circulation. Proc Natl Acad Sci U S A. 2010;107(16):7257–7262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pamuklar Z, Federico L, Liu S, et al. Autotaxin/lysopholipase D and lysophosphatidic acid regulate murine hemostasis and thrombosis. J Biol Chem. 2009;284(11):7385–7394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Stracke ML, Krutzsch HC, Unsworth EJ, et al. Identification, purification, and partial sequence analysis of autotaxin, a novel motility-stimulating protein. J Biol Chem. 1992;267(4):2524–2529. [PubMed] [Google Scholar]
- 65.Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. Autotaxin, an ectoenzyme that produces lysophosphatidic acid, promotes the entry of lymphocytes into secondary lymphoid organs. Nat Immunol. 2008;9(4):415–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hausmann J, Kamtekar S, Christodoulou E, et al. Structural basis of substrate discrimination and integrin binding by autotaxin. Nat Struct Mol Biol. 2011;18(2):198–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Nakasaki T, Tanaka T, Okudaira S, et al. Involvement of the lysophosphatidic acid-generating enzyme autotaxin in lymphocyte-endothelial cell interactions. Am J Pathol. 2008;173(5):1566–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Moolenaar WH, Perrakis A. Insights into autotaxin: how to produce and present a lipid mediator. Nat Rev Mol Cell Biol. 2011;12(10):674–679. [DOI] [PubMed] [Google Scholar]
- 69.Tabchy A, Tigyi G, Mills GB. Location, location, location: a crystal-clear view of autotaxin saturating LPA receptors. Nat Struct Mol Biol. 2011;18(2):117–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Tomsig JL, Snyder AH, Berdyshev EV, et al. Lipid phosphate phosphohydrolase type 1 (LPP1) degrades extracellular lysophosphatidic acid in vivo. Biochem J. 2009;419(3):611–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.van Meeteren LA, Ruurs P, Stortelers C, et al. Autotaxin, a secreted lysophospholipase D, is essential for blood vessel formation during development. Mol Cell Biol. 2006;26(13):5015–5022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Takeda A, Kobayashi D, Aoi K, et al. Fibroblastic reticular cell-derived lysophosphatidic acid regulates confined intranodal T-cell motility. Elife. 2016;5:e10561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhang Y, Chen YC, Krummel MF, Rosen SD. Autotaxin through lysophosphatidic acid stimulates polarization, motility, and transendothelial migration of naive T cells. J Immunol. 2012;189(8):3914–3924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Donovan EE, Pelanda R, Torres RM. S1P3 confers differential S1P-induced migration by autoreactive and non-autoreactive immature B cells and is required for normal B-cell development. Eur J Immunol. 2010;40(3):688–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Pereira JP, Xu Y, Cyster JG. A role for S1P and S1P1 in immature-B cell egress from mouse bone marrow. PLoS One. 2010;5(2):e9277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hu J, Oda SK, Shotts K, et al. Lysophosphatidic acid receptor 5 inhibits B cell antigen receptor signaling and antibody response. J Immunol. 2014;193(1):85–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Oda SK, Strauch P, Fujiwara Y, et al. Lysophosphatidic acid inhibits CD8 T cell activation and control of tumor progression. Cancer Immunol Res. 2013;1(4):245–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Mathew D, Kremer KN, Strauch P, Tigyi G, Pelanda R, Torres RM. LPA(5) Is an Inhibitory Receptor That Suppresses CD8 T-Cell Cytotoxic Function via Disruption of Early TCR Signaling. Front Immunol. 2019;10:1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Knowlden SA, Capece T, Popovic M, et al. Regulation of T cell motility in vitro and in vivo by LPA and LPA2. PLoS One. 2014;9(7):e101655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kremer KN, Buser A, Thumkeo D, et al. LPA suppresses T cell function by altering the cytoskeleton and disrupting immune synapse formation. Proc Natl Acad Sci U S A. 2022;119(15):e2118816119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Berg LJ, Finkelstein LD, Lucas JA, Schwartzberg PL. Tec family kinases in T lymphocyte development and function. Annu Rev Immunol. 2005;23:549–600. [DOI] [PubMed] [Google Scholar]
- 82.Cahalan MD. STIMulating store-operated Ca(2+) entry. Nat Cell Biol. 2009;11(6):669–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hogan PG, Lewis RS, Rao A. Molecular basis of calcium signaling in lymphocytes: STIM and ORAI. Annu Rev Immunol. 2010;28:491–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hogan PG, Rao A. Store-operated calcium entry: Mechanisms and modulation. Biochem Biophys Res Commun. 2015;460(1):40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Trebak M, Putney JW Jr. ORAI Calcium Channels. Physiology (Bethesda). 2017;32(4):332–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mak DO, McBride S, Foskett JK. ATP-dependent adenophostin activation of inositol 1,4,5-trisphosphate receptor channel gating: kinetic implications for the durations of calcium puffs in cells. J Gen Physiol. 2001;117(4):299–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.An S, Bleu T, Zheng Y, Goetzl EJ. Recombinant human G protein-coupled lysophosphatidic acid receptors mediate intracellular calcium mobilization. Mol Pharmacol. 1998;54(5):881–888. [DOI] [PubMed] [Google Scholar]
- 88.Young KW, Bootman MD, Channing DR, et al. Lysophosphatidic acid-induced Ca2+ mobilization requires intracellular sphingosine 1-phosphate production. Potential involvement of endogenous EDG-4 receptors. J Biol Chem. 2000;275(49):38532–38539. [DOI] [PubMed] [Google Scholar]
- 89.Panther E, Idzko M, Corinti S, et al. The influence of lysophosphatidic acid on the functions of human dendritic cells. J Immunol. 2002;169(8):4129–4135. [DOI] [PubMed] [Google Scholar]
- 90.Hasegawa Y, Erickson JR, Goddard GJ, et al. Identification of a phosphothionate analogue of lysophosphatidic acid (LPA) as a selective agonist of the LPA3 receptor. J Biol Chem. 2003;278(14):11962–11969. [DOI] [PubMed] [Google Scholar]
- 91.Jin Y, Knudsen E, Wang L, Maghazachi AA. Lysophosphatidic acid induces human natural killer cell chemotaxis and intracellular calcium mobilization. Eur J Immunol. 2003;33(8):2083–2089. [DOI] [PubMed] [Google Scholar]
- 92.Idzko M, Laut M, Panther E, et al. Lysophosphatidic acid induces chemotaxis, oxygen radical production, CD11b up-regulation, Ca2+ mobilization, and actin reorganization in human eosinophils via pertussis toxin-sensitive G proteins. J Immunol. 2004;172(7):4480–4485. [DOI] [PubMed] [Google Scholar]
- 93.Lee CW, Rivera R, Dubin AE, Chun J. LPA(4)/GPR23 is a lysophosphatidic acid (LPA) receptor utilizing G(s)-, G(q)/G(i)-mediated calcium signaling and G(12/13)-mediated Rho activation. J Biol Chem. 2007;282(7):4310–4317. [DOI] [PubMed] [Google Scholar]
- 94.Lichte K, Rossi R, Danneberg K, et al. Lysophospholipid receptor-mediated calcium signaling in human keratinocytes. J Invest Dermatol. 2008;128(6):1487–1498. [DOI] [PubMed] [Google Scholar]
- 95.Choi JW, Lim S, Oh YS, et al. Subtype-specific role of phospholipase C-beta in bradykinin and LPA signaling through differential binding of different PDZ scaffold proteins. Cell Signal. 2010;22(7):1153–1161. [DOI] [PubMed] [Google Scholar]
- 96.Jans R, Mottram L, Johnson DL, et al. Lysophosphatidic acid promotes cell migration through STIM1- and Orai1-mediated Ca2+(i) mobilization and NFAT2 activation. J Invest Dermatol. 2013;133(3):793–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Miscia S, Di Baldassarre A, Cataldi A, et al. Immunocytochemical localization of phospholipase C isozymes in cord blood and adult T-lymphocytes. J Histochem Cytochem. 1999;47(7):929–936. [DOI] [PubMed] [Google Scholar]
- 98.Heng TS, Painter MW, Immunological Genome Project C. The Immunological Genome Project: networks of gene expression in immune cells. Nat Immunol. 2008;9(10):1091–1094. [DOI] [PubMed] [Google Scholar]
- 99.Kortum RL, Rouquette-Jazdanian AK, Samelson LE. Ras and extracellular signal-regulated kinase signaling in thymocytes and T cells. Trends Immunol. 2013;34(6):259–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Roose JP, Mollenauer M, Gupta VA, Stone J, Weiss A. A diacylglycerol-protein kinase C-RasGRP1 pathway directs Ras activation upon antigen receptor stimulation of T cells. Mol Cell Biol. 2005;25(11):4426–4441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dower NA, Stang SL, Bottorff DA, et al. RasGRP is essential for mouse thymocyte differentiation and TCR signaling. Nat Immunol. 2000;1(4):317–321. [DOI] [PubMed] [Google Scholar]
- 102.Izquierdo M, Leevers SJ, Marshall CJ, Cantrell D. p21ras couples the T cell antigen receptor to extracellular signal-regulated kinase 2 in T lymphocytes. J Exp Med. 1993;178(4):1199–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lyubchenko TA, Wurth GA, Zweifach A. Role of calcium influx in cytotoxic T lymphocyte lytic granule exocytosis during target cell killing. Immunity. 2001;15(5):847–859. [DOI] [PubMed] [Google Scholar]
- 104.Fierro AF, Wurth GA, Zweifach A. Cross-talk with Ca(2+) influx does not underlie the role of extracellular signal-regulated kinases in cytotoxic T lymphocyte lytic granule exocytosis. J Biol Chem. 2004;279(24):25646–25652. [DOI] [PubMed] [Google Scholar]
- 105.Pores-Fernando AT, Zweifach A. Calcium influx and signaling in cytotoxic T-lymphocyte lytic granule exocytosis. Immunol Rev. 2009;231(1):160–173. [DOI] [PubMed] [Google Scholar]
- 106.Feske S, Wulff H, Skolnik EY. Ion channels in innate and adaptive immunity. Annu Rev Immunol. 2015;33:291–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Oh-Hora M, Yamashita M, Hogan PG, et al. Dual functions for the endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat Immunol. 2008;9(4):432–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Koike T, Yamagishi H, Hatanaka Y, et al. A novel ERK-dependent signaling process that regulates interleukin-2 expression in a late phase of T cell activation. J Biol Chem. 2003;278(18):15685–15692. [DOI] [PubMed] [Google Scholar]
- 109.Egerton M, Fitzpatrick DR, Kelso A. Activation of the extracellular signal-regulated kinase pathway is differentially required for TCR-stimulated production of six cytokines in primary T lymphocytes. Int Immunol. 1998;10(2):223–229. [DOI] [PubMed] [Google Scholar]
- 110.Fitzpatrick DR, Kelso A. Independent regulation of cytokine genes in T cells: the paradox in the paradigm. Transplantation. 1998;65(1):1–5. [DOI] [PubMed] [Google Scholar]
- 111.Egerton M, Fitzpatrick DR, Catling AD, Kelso A. Differential activation of T cell cytokine production by the extracellular signal-regulated kinase (ERK) signaling pathway. Eur J Immunol. 1996;26(10):2279–2285. [DOI] [PubMed] [Google Scholar]
- 112.Kozasa T, Jiang X, Hart MJ, et al. p115 RhoGEF, a GTPase activating protein for Galpha12 and Galpha13. Science. 1998;280(5372):2109–2111. [DOI] [PubMed] [Google Scholar]
- 113.Hart MJ, Jiang X, Kozasa T, et al. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Galpha13. Science. 1998;280(5372):2112–2114. [DOI] [PubMed] [Google Scholar]
- 114.Jorritsma PJ, Brogdon JL, Bottomly K. Role of TCR-induced extracellular signal-regulated kinase activation in the regulation of early IL-4 expression in naive CD4+ T cells. J Immunol. 2003;170(5):2427–2434. [DOI] [PubMed] [Google Scholar]
- 115.Whitehurst CE, Geppert TD. MEK1 and the extracellular signal-regulated kinases are required for the stimulation of IL-2 gene transcription in T cells. J Immunol. 1996;156(3):1020–1029. [PubMed] [Google Scholar]
- 116.Hogquist KA, Jameson SC, Heath WR, Howard JL, Bevan MJ, Carbone FR. T cell receptor antagonist peptides induce positive selection. Cell. 1994;76(1):17–27. [DOI] [PubMed] [Google Scholar]
- 117.Deng W, Shuyu E, Tsukahara R, et al. The lysophosphatidic acid type 2 receptor is required for protection against radiation-induced intestinal injury. Gastroenterology. 2007;132(5):1834–1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kiss GN, Fells JI, Gupte R, et al. Virtual screening for LPA2-specific agonists identifies a nonlipid compound with antiapoptotic actions. Mol Pharmacol. 2012;82(6):1162–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kosanam H, Ma F, He H, et al. Development of an LC-MS/MS assay to determine plasma pharmacokinetics of the radioprotectant octadecenyl thiophosphate (OTP) in monkeys. J Chromatogr B Analyt Technol Biomed Life Sci. 2010;878(26):2379–2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Turner J, Fredrickson M, D’Antonio M, et al. Lysophosphatidic acid modulates CD8 T cell immunosurveillance, metabolism, and anti-tumor immunity. 2023, Nat Comm, In press, 10.21203/rs.3.rs-2058360/v1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Barber DL, Wherry EJ, Ahmed R. Cutting edge: rapid in vivo killing by memory CD8 T cells. J Immunol. 2003;171(1):27–31. [DOI] [PubMed] [Google Scholar]
- 122.Choi JW, Herr DR, Noguchi K, et al. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol. 2010;50:157–186. [DOI] [PubMed] [Google Scholar]
- 123.Ishii S, Noguchi K, Yanagida K. Non-Edg family lysophosphatidic acid (LPA) receptors. Prostaglandins Other Lipid Mediat. 2009;89(3–4):57–65. [DOI] [PubMed] [Google Scholar]
- 124.Cassioli C, Baldari CT. The Expanding Arsenal of Cytotoxic T Cells. Front Immunol. 2022;13:883010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ritter AT, Angus KL, Griffiths GM. The role of the cytoskeleton at the immunological synapse. Immunol Rev. 2013;256(1):107–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Geiger B, Rosen D, Berke G. Spatial relationships of microtubule-organizing centers and the contact area of cytotoxic T lymphocytes and target cells. J Cell Biol. 1982;95(1):137–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ryser JE, Rungger-Brandle E, Chaponnier C, Gabbiani G, Vassalli P. The area of attachment of cytotoxic T lymphocytes to their target cells shows high motility and polarization of actin, but not myosin. J Immunol. 1982;128(3):1159–1162. [PubMed] [Google Scholar]
- 128.Kupfer A, Dennert G. Reorientation of the microtubule-organizing center and the Golgi apparatus in cloned cytotoxic lymphocytes triggered by binding to lysable target cells. J Immunol. 1984;133(5):2762–2766. [PubMed] [Google Scholar]
- 129.Pardi R, Inverardi L, Rugarli C, Bender JR. Antigen-receptor complex stimulation triggers protein kinase C-dependent CD11a/CD18-cytoskeleton association in T lymphocytes. J Cell Biol. 1992;116(5):1211–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.DeBell KE, Conti A, Alava MA, Hoffman T, Bonvini E. Microfilament assembly modulates phospholipase C-mediated signal transduction by the TCR/CD3 in murine T helper lymphocytes. J Immunol. 1992;149(7):2271–2280. [PubMed] [Google Scholar]
- 131.Valitutti S, Dessing M, Aktories K, Gallati H, Lanzavecchia A. Sustained signaling leading to T cell activation results from prolonged T cell receptor occupancy. Role of T cell actin cytoskeleton. J Exp Med. 1995;181(2):577–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kuhn JR, Poenie M. Dynamic polarization of the microtubule cytoskeleton during CTL-mediated killing. Immunity. 2002;16(1):111–121. [DOI] [PubMed] [Google Scholar]
- 133.Vicente-Manzanares M, Sanchez-Madrid F. Role of the cytoskeleton during leukocyte responses. Nat Rev Immunol. 2004;4(2):110–122. [DOI] [PubMed] [Google Scholar]
- 134.Ritter AT, Asano Y, Stinchcombe JC, et al. Actin depletion initiates events leading to granule secretion at the immunological synapse. Immunity. 2015;42(5):864–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Hammer JA, Wang JC, Saeed M, Pedrosa AT. Origin, Organization, Dynamics, and Function of Actin and Actomyosin Networks at the T Cell Immunological Synapse. Annu Rev Immunol. 2019;37:201–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. Centrosome polarization delivers secretory granules to the immunological synapse. Nature. 2006;443(7110):462–465. [DOI] [PubMed] [Google Scholar]
- 137.Huse M, Lillemeier BF, Kuhns MS, Chen DS, Davis MM. T cells use two directionally distinct pathways for cytokine secretion. Nat Immunol. 2006;7(3):247–255. [DOI] [PubMed] [Google Scholar]
- 138.Palazzo AF, Cook TA, Alberts AS, Gundersen GG. mDia mediates Rho-regulated formation and orientation of stable microtubules. Nat Cell Biol. 2001;3(8):723–729. [DOI] [PubMed] [Google Scholar]
- 139.Wen Y, Eng CH, Schmoranzer J, et al. EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration. Nat Cell Biol. 2004;6(9):820–830. [DOI] [PubMed] [Google Scholar]
- 140.Gundersen GG, Wen Y, Eng CH, et al. Regulation of microtubules by Rho GTPases in migrating cells. Novartis Found Symp. 2005;269:106–116; discussion 116–126, 223–130. [PubMed] [Google Scholar]
- 141.Eng CH, Huckaba TM, Gundersen GG. The formin mDia regulates GSK3beta through novel PKCs to promote microtubule stabilization but not MTOC reorientation in migrating fibroblasts. Mol Biol Cell. 2006;17(12):5004–5016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yamana N, Arakawa Y, Nishino T, et al. The Rho-mDia1 pathway regulates cell polarity and focal adhesion turnover in migrating cells through mobilizing Apc and c-Src. Mol Cell Biol. 2006;26(18):6844–6858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.DeWard AD, Alberts AS. Microtubule stabilization: formins assert their independence. Curr Biol. 2008;18(14):R605–608. [DOI] [PubMed] [Google Scholar]
- 144.Gomez TS, Billadeau DD. T cell activation and the cytoskeleton: you can’t have one without the other. Adv Immunol. 2008;97:1–64. [DOI] [PubMed] [Google Scholar]
- 145.Beemiller P, Krummel MF. Mediation of T-cell activation by actin meshworks. Cold Spring Harb Perspect Biol. 2010;2(9):a002444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kumari S, Curado S, Mayya V, Dustin ML. T cell antigen receptor activation and actin cytoskeleton remodeling. Biochim Biophys Acta. 2014;1838(2):546–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Comrie WA, Burkhardt JK. Action and Traction: Cytoskeletal Control of Receptor Triggering at the Immunological Synapse. Front Immunol. 2016;7:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Angkachatchai V, Finkel TH. ADP-ribosylation of rho by C3 ribosyltransferase inhibits IL-2 production and sustained calcium influx in activated T cells. J Immunol. 1999;163(7):3819–3825. [PubMed] [Google Scholar]
- 149.Lang P, Guizani L, Vitte-Mony I, et al. ADP-ribosylation of the ras-related, GTP-binding protein RhoA inhibits lymphocyte-mediated cytotoxicity. J Biol Chem. 1992;267(17):11677–11680. [PubMed] [Google Scholar]
- 150.Joseph N, Reicher B, Barda-Saad M. The calcium feedback loop and T cell activation: how cytoskeleton networks control intracellular calcium flux. Biochim Biophys Acta. 2014;1838(2):557–568. [DOI] [PubMed] [Google Scholar]
- 151.Xiang SY, Dusaban SS, Brown JH. Lysophospholipid receptor activation of RhoA and lipid signaling pathways. Biochim Biophys Acta. 2013;1831(1):213–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Andres-Delgado L, Anton OM, Alonso MA. Centrosome polarization in T cells: a task for formins. Front Immunol. 2013;4:191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hashimoto-Tane A, Yokosuka T, Sakata-Sogawa K, et al. Dynein-driven transport of T cell receptor microclusters regulates immune synapse formation and T cell activation. Immunity. 2011;34(6):919–931. [DOI] [PubMed] [Google Scholar]
- 154.Serrador JM, Cabrero JR, Sancho D, Mittelbrunn M, Urzainqui A, Sanchez-Madrid F. HDAC6 deacetylase activity links the tubulin cytoskeleton with immune synapse organization. Immunity. 2004;20(4):417–428. [DOI] [PubMed] [Google Scholar]
- 155.Nagasaki T, Gundersen GG. Depletion of lysophosphatidic acid triggers a loss of oriented detyrosinated microtubules in motile fibroblasts. J Cell Sci. 1996;109 ( Pt 10):2461–2469. [DOI] [PubMed] [Google Scholar]
- 156.Cook TA, Nagasaki T, Gundersen GG. Rho guanosine triphosphatase mediates the selective stabilization of microtubules induced by lysophosphatidic acid. J Cell Biol. 1998;141(1):175–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Bartolini F, Gundersen GG. Formins and microtubules. Biochim Biophys Acta. 2010;1803(2):164–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Eisenmann KM, West RA, Hildebrand D, et al. T cell responses in mammalian diaphanous-related formin mDia1 knock-out mice. J Biol Chem. 2007;282(34):25152–25158. [DOI] [PubMed] [Google Scholar]
- 159.Sakata D, Taniguchi H, Yasuda S, et al. Impaired T lymphocyte trafficking in mice deficient in an actin-nucleating protein, mDia1. J Exp Med. 2007;204(9):2031–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.deSouza N, Cui J, Dura M, McDonald TV, Marks AR. A function for tyrosine phosphorylation of type 1 inositol 1,4,5-trisphosphate receptor in lymphocyte activation. J Cell Biol. 2007;179(5):923–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Liang J, Lyu J, Zhao M, et al. Tespa1 regulates T cell receptor-induced calcium signals by recruiting inositol 1,4,5-trisphosphate receptors. Nat Commun. 2017;8:15732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Stinchcombe JC, Griffiths GM. Secretory mechanisms in cell-mediated cytotoxicity. Annu Rev Cell Dev Biol. 2007;23:495–517. [DOI] [PubMed] [Google Scholar]
- 163.Finetti F, Paccani SR, Riparbelli MG, et al. Intraflagellar transport is required for polarized recycling of the TCR/CD3 complex to the immune synapse. Nat Cell Biol. 2009;11(11):1332–1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.de la Roche M, Asano Y, Griffiths GM. Origins of the cytolytic synapse. Nat Rev Immunol. 2016;16(7):421–432. [DOI] [PubMed] [Google Scholar]
- 165.Cassioli C, Baldari CT. A Ciliary View of the Immunological Synapse. Cells. 2019;8(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Douanne T, Stinchcombe JC, Griffiths GM. Teasing out function from morphology: Similarities between primary cilia and immune synapses. J Cell Biol. 2021;220(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Hilgendorf KI, Johnson CT, Jackson PK. The primary cilium as a cellular receiver: organizing ciliary GPCR signaling. Curr Opin Cell Biol. 2016;39:84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Capitani N, Onnis A, Finetti F, et al. A CVID-associated variant in the ciliogenesis protein CCDC28B disrupts immune synapse assembly. Cell Death Differ. 2022;29(1):65–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Loskutov YV, Griffin CL, Marinak KM, et al. LPA signaling is regulated through the primary cilium: a novel target in glioblastoma. Oncogene. 2018;37(11):1457–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Walia V, Cuenca A, Vetter M, et al. Akt Regulates a Rab11-Effector Switch Required for Ciliogenesis. Dev Cell. 2019;50(2):229–246 e227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Hu HB, Song ZQ, Song GP, et al. LPA signaling acts as a cell-extrinsic mechanism to initiate cilia disassembly and promote neurogenesis. Nat Commun. 2021;12(1):662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Willier S, Butt E, Grunewald TG. Lysophosphatidic acid (LPA) signalling in cell migration and cancer invasion: a focussed review and analysis of LPA receptor gene expression on the basis of more than 1700 cancer microarrays. Biol Cell. 2013;105(8):317–333. [DOI] [PubMed] [Google Scholar]
- 173.Balijepalli P, Sitton CC, Meier KE. Lysophosphatidic Acid Signaling in Cancer Cells: What Makes LPA So Special? Cells. 2021;10(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Liu S, Umezu-Goto M, Murph M, et al. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell. 2009;15(6):539–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br J Cancer. 2010;102(6):941–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Fidler IJ. Biological behavior of malignant melanoma cells correlated to their survival in vivo. Cancer Res. 1975;35(1):218–224. [PubMed] [Google Scholar]
- 177.Mukherjee A, Wu J, Barbour S, Fang X. Lysophosphatidic acid activates lipogenic pathways and de novo lipid synthesis in ovarian cancer cells. J Biol Chem. 2012;287(30):24990–25000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Olzmann JA, Carvalho P. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol. 2019;20(3):137–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Patsoukis N, Bardhan K, Chatterjee P, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015;6:6692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Reina-Campos M, Scharping NE, Goldrath AW. CD8(+) T cell metabolism in infection and cancer. Nat Rev Immunol. 2021;21(11):718–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Kalkat M, De Melo J, Hickman KA, et al. MYC Deregulation in Primary Human Cancers. Genes (Basel). 2017;8(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Zehn D, Lee SY, Bevan MJ. Complete but curtailed T-cell response to very low-affinity antigen. Nature. 2009;458(7235):211–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Aleksic M, Liddy N, Molloy PE, et al. Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur J Immunol. 2012;42(12):3174–3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8(5):765–772. [DOI] [PubMed] [Google Scholar]
- 187.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–3895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Brunet JF, Denizot F, Luciani MF, et al. A new member of the immunoglobulin superfamily--CTLA-4. Nature. 1987;328(6127):267–270. [DOI] [PubMed] [Google Scholar]
- 189.Freeman GJ, Lombard DB, Gimmi CD, et al. CTLA-4 and CD28 mRNA are coexpressed in most T cells after activation. Expression of CTLA-4 and CD28 mRNA does not correlate with the pattern of lymphokine production. J Immunol. 1992;149(12):3795–3801. [PubMed] [Google Scholar]
- 190.Monney L, Sabatos CA, Gaglia JL, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415(6871):536–541. [DOI] [PubMed] [Google Scholar]
- 191.Hastings WD, Anderson DE, Kassam N, et al. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur J Immunol. 2009;39(9):2492–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Baixeras E, Roman-Roman S, Jitsukawa S, et al. Cloning and expression of a lymphocyte activation gene (LAG-1). Mol Immunol. 1990;27(11):1091–1102. [DOI] [PubMed] [Google Scholar]
- 193.Triebel F, Jitsukawa S, Baixeras E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393–1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Boles KS, Vermi W, Facchetti F, et al. A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC. Eur J Immunol. 2009;39(3):695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Levin SD, Taft DW, Brandt CS, et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur J Immunol. 2011;41(4):902–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Kasahara K, Inagaki M. Primary ciliary signaling: links with the cell cycle. Trends Cell Biol. 2021;31(12):954–964. [DOI] [PubMed] [Google Scholar]
- 197.Plotnikova OV, Golemis EA, Pugacheva EN. Cell cycle-dependent ciliogenesis and cancer. Cancer Res. 2008;68(7):2058–2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Yu FX, Zhao B, Panupinthu N, et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150(4):780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Yu FX, Zhao B, Guan KL. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell. 2015;163(4):811–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Plastira I, Joshi L, Bernhart E, et al. Small-Molecule Lysophosphatidic Acid Receptor 5 (LPAR5) Antagonists: Versatile Pharmacological Tools to Regulate Inflammatory Signaling in BV-2 Microglia Cells. Front Cell Neurosci. 2019;13:531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Congreve M, de Graaf C, Swain NA, Tate CG. Impact of GPCR Structures on Drug Discovery. Cell. 2020;181(1):81–91. [DOI] [PubMed] [Google Scholar]
- 202.Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov. 2017;16(12):829–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
