501
|
Plastira I, Bernhart E, Goeritzer M, Reicher H, Kumble VB, Kogelnik N, Wintersperger A, Hammer A, Schlager S, Jandl K, Heinemann A, Kratky D, Malle E, Sattler W. 1-Oleyl-lysophosphatidic acid (LPA) promotes polarization of BV-2 and primary murine microglia towards an M1-like phenotype. J Neuroinflammation 2016; 13:205. [PMID: 27565558 PMCID: PMC5002165 DOI: 10.1186/s12974-016-0701-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/20/2016] [Indexed: 01/09/2023] Open
Abstract
Background Microglia, the immunocompetent cells of the CNS, rapidly respond to brain injury and disease by altering their morphology and phenotype to adopt an activated state. Microglia can exist broadly between two different states, namely the classical (M1) and the alternative (M2) phenotype. The first is characterized by the production of pro-inflammatory cytokines/chemokines and reactive oxygen and/or nitrogen species. In contrast, alternatively activated microglia are typified by an anti-inflammatory phenotype supporting wound healing and debris clearance. The objective of the present study was to determine the outcome of lysophosphatidic acid (LPA)-mediated signaling events on microglia polarization. Methods LPA receptor expression and cyto-/chemokine mRNA levels in BV-2 and primary murine microglia (PMM) were determined by qPCR. M1/M2 marker expression was analyzed by Western blotting, immunofluorescence microscopy, or flow cytometry. Cyto-/chemokine secretion was quantitated by ELISA. Results BV-2 cells express LPA receptor 2 (LPA2), 3, 5, and 6, whereas PMM express LPA1, 2, 4, 5, and 6. We show that LPA treatment of BV-2 and PMM leads to a shift towards a pro-inflammatory M1-like phenotype. LPA treatment increased CD40 and CD86 (M1 markers) and reduced CD206 (M2 marker) expression. LPA increased inducible nitric oxide synthase (iNOS) and COX-2 levels (both M1), while the M2 marker Arginase-1 was suppressed in BV-2 cells. Immunofluorescence studies (iNOS, COX-2, Arginase-1, and RELMα) extended these findings to PMM. Upregulation of M1 markers in BV-2 and PMM was accompanied by increased cyto-/chemokine transcription and secretion (IL-1β, TNFα, IL-6, CCL5, and CXCL2). The pharmacological LPA5 antagonist TCLPA5 blunted most of these pro-inflammatory responses. Conclusions LPA drives BV-2 and PMM towards a pro-inflammatory M1-like phenotype. Suppression by TCLPA5 indicates that the LPA/LPA5 signaling axis could represent a potential pharmacological target to interfere with microglia polarization in disease.
Collapse
Affiliation(s)
- Ioanna Plastira
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Eva Bernhart
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Madeleine Goeritzer
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Helga Reicher
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Vishwanath Bhat Kumble
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Nora Kogelnik
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Andrea Wintersperger
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Astrid Hammer
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Stefanie Schlager
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Katharina Jandl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21, 8010, Graz, Austria. .,BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
502
|
Jethwa SA, Leah EJ, Zhang Q, Bright NA, Oxley D, Bootman MD, Rudge SA, Wakelam MJO. Exosomes bind to autotaxin and act as a physiological delivery mechanism to stimulate LPA receptor signalling in cells. J Cell Sci 2016; 129:3948-3957. [PMID: 27557622 DOI: 10.1242/jcs.184424] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/14/2016] [Indexed: 12/23/2022] Open
Abstract
Autotaxin (ATX; also known as ENPP2), the lysophospholipase responsible for generating the lipid receptor agonist lysophosphatidic acid (LPA), is a secreted enzyme. Here we show that, once secreted, ATX can bind to the surface of cell-secreted exosomes. Exosome-bound ATX is catalytically active and carries generated LPA. Once bound to a cell, through specific integrin interactions, ATX releases the LPA to activate cell surface G-protein-coupled receptors of LPA; inhibition of signalling by the receptor antagonist Ki1642 suggests that these receptors are LPAR1 and LPAR3. The binding stimulates downstream signalling, including phosphorylation of AKT and mitogen-activated protein kinases, the release of intracellular stored Ca2+ and cell migration. We propose that exosomal binding of LPA-loaded ATX provides a means of efficiently delivering the lipid agonist to cell surface receptors to promote signalling. We further propose that this is a means by which ATX-LPA signalling operates physiologically.
Collapse
Affiliation(s)
- Susanna A Jethwa
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Emma J Leah
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Qifeng Zhang
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - David Oxley
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Martin D Bootman
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Simon A Rudge
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | |
Collapse
|
503
|
Fan Q, Cai Q, Xu Y. FOXM1 is a downstream target of LPA and YAP oncogenic signaling pathways in high grade serous ovarian cancer. Oncotarget 2016; 6:27688-99. [PMID: 26299613 PMCID: PMC4695018 DOI: 10.18632/oncotarget.4280] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/01/2015] [Indexed: 01/21/2023] Open
Abstract
Lysophosphatidic acid (LPA), a prototypical ligand for G protein coupled receptors, and Forkhead box protein M1 (FOXM1), a transcription factor that regulates expression of a wide array of genes involved in cancer initiation and progression, are two important oncogenic signaling molecules in human epithelial ovarian cancers (EOC). We conducted in vitro mechanistic studies using pharmacological inhibitors, genetic forms of the signaling molecules, and RNAi-mediated gene knock-down to uncover the molecular mechanisms of how these two molecules interact in EOC cells. Additionally, in vivo mouse studies were performed to confirm the functional involvement of FOXM1 in EOC tumor formation and progression. We show for the first time that LPA up-regulates expression of active FOXM1 splice variants in a time- and dose-dependent manner in the human EOC cell lines OVCA433, CAOV3, and OVCAR5. Gi-PI3K-AKT and G12/13-Rho-YAP signaling pathways were both involved in the LPA receptor (LPA1-3) mediated up-regulation of FOXM1 at the transcriptional level. In addition, down-regulation of FOXM1 in CAOV3 xenografts significantly reduced tumor and ascites formation, metastasis, and expression of FOXM1 target genes involved in cell proliferation, migration, or invasion. Collectively, our data link the oncolipid LPA, the oncogene YAP, and the central regulator of cell proliferation/mutagenesis FOXM1 in EOC cells. Moreover, these results provide further support for the importance of these pathways as potential therapeutic targets in EOC.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
504
|
Fleming JK, Glass TR, Lackie SJ, Wojciak JM. A novel approach for measuring sphingosine-1-phosphate and lysophosphatidic acid binding to carrier proteins using monoclonal antibodies and the Kinetic Exclusion Assay. J Lipid Res 2016; 57:1737-47. [PMID: 27444045 DOI: 10.1194/jlr.d068866] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Indexed: 01/01/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) are bioactive signaling lysophospholipids that activate specific G protein-coupled receptors on the cell surface triggering numerous biological events. In circulation, S1P and LPA associate with specific carrier proteins or chaperones; serum albumin binds both S1P and LPA while HDL shuttles S1P via interactions with apoM. We used a series of kinetic exclusion assays in which monoclonal anti-S1P and anti-LPA antibodies competed with carrier protein for the lysophospholipid to measure the equilibrium dissociation constants (Kd) for these carrier proteins binding S1P and the major LPA species. Fatty acid-free (FAF)-BSA binds these lysophospholipids with the following Kd values: LPA(16:0), 68 nM; LPA(18:1), 130 nM; LPA(18:2), 350 nM; LPA(20:4), 2.2 μM; and S1P, 41 μM. FAF human serum albumin binds each lysophospholipid with comparable affinities. By measuring the apoM concentration and expanding the model to include endogenous ligand, we were able to resolve the Kd values for S1P binding apoM in the context of human HDL and LDL particles (21 nM and 2.4 nM, respectively). The novel competitive assay and analysis described herein enables measurement of Kd values of completely unmodified lysophospholipids binding unmodified carrier proteins in solution, and thus provide insights into S1P and LPA storage in the circulation system and may be useful in understanding chaperone-dependent receptor activation and signaling.
Collapse
|
505
|
Chandrasekharan JA, Marginean A, Sharma-Walia N. An insight into the role of arachidonic acid derived lipid mediators in virus associated pathogenesis and malignancies. Prostaglandins Other Lipid Mediat 2016; 126:46-54. [PMID: 27450483 DOI: 10.1016/j.prostaglandins.2016.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/25/2016] [Accepted: 07/18/2016] [Indexed: 12/14/2022]
Abstract
Several studies shed light on the size and diversity of the lipidome, along with its role in physiological and pathological processes in human health. Besides that, lipids also function as important signaling mediators. This review focuses on discussing the role of arachidonic acid (AA) derived lipids as mediators in diseases with special emphasis on viral infections. Structurally, arachidonic acid derived lipids, also referred to as lipid mediators, can be classified into three specific classes: Class 1-eicosanoids derived from arachidonic acid metabolism; Class 2-lysophospholipids consisting of either a glycerol or a sphingosine backbone; Class 3-AA and ω-3 polyunsaturated fatty acid (PUFA) derivatives. Class 1 and 2 lipids are commonly referred to as pro-inflammatory molecules, which are found upregulated in diseases like cancer and viral infection. Class 3 lipids are anti-inflammatory molecules, which could be potentially used in treatment of diseases associated with inflammation. The function of each class has been elucidated as unique and contributory to an overall cellular homeostasis. Current work in this field is promising and will surely usher in a new era of lipid understanding and control not only at the molecular level, but also in terms of holistic patient care.
Collapse
Affiliation(s)
- Jayashree A Chandrasekharan
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alexandru Marginean
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
506
|
Sinderewicz E, Grycmacher K, Boruszewska D, Kowalczyk-Zięba I, Yamamoto Y, Yoshimoto Y, Woclawek-Potocka I. Lysophosphatidic Acid Synthesis and its Receptors' Expression in the Bovine Oviduct During the Oestrous Cycle. Reprod Domest Anim 2016; 51:541-9. [PMID: 27335048 DOI: 10.1111/rda.12717] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022]
Abstract
Lysophosphatidic acid (LPA) is a naturally occurring simple phospholipid which in the bovine reproductive system can be produced in the endometrium, corpus luteum, ovarian follicle and embryo. In this study, we examined the possibility that LPA receptors are expressed, and LPA synthesized, in the bovine oviduct. We found that the concentration of LPA was highest in infundibulum in the follicular phase of the oestrous cycle and was relatively high during the early-luteal phase in all examined parts of the oviduct. We also documented that LPA synthesis engages both available pathways for LPA production. The autotaxin (ATX) protein expression was significantly higher in the infundibulum compared to the isthmus during the follicular phase of the oestrous cycle. During the early-luteal phase of the oestrous cycle, ATX and phospholipase A2 (PLA2) protein expression was highest in ampulla, although the expression of LPARs was not as dynamic as LPA concentration in the oviduct tissue, and we presume that in the bovine oviduct, the most abundantly expressed receptor is LPAR2. In conclusion, our results indicate that the bovine oviduct is a site of LPA synthesis and a target for LPA action in the bovine reproductive tract. We documented that LPAR2 is the most abundantly expressed in the bovine oviduct. We hypothesize that in the bovine oviduct, LPA may be involved in the transport of gametes, fertilization and cellular signalling between the oviduct and cumulus-oocyte complex.
Collapse
Affiliation(s)
- E Sinderewicz
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - K Grycmacher
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - D Boruszewska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - I Kowalczyk-Zięba
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Y Yamamoto
- Department of Animal Science, Division of Agricultural and Life Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Y Yoshimoto
- Department of Animal Science, Division of Agricultural and Life Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - I Woclawek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
507
|
Abstract
Systemic sclerosis is a heterogeneous condition characterized by microvascular damage, dysregulation of the immune system, and progressive fibrosis affecting skin and internal organs. Currently, there are no approved disease-modifying therapies, and management mostly involves treatment of organ-specific complications. In recent years, major advances have greatly improved our understanding of the disease process, especially the molecular mechanisms by which fibrosis becomes self-sustaining. We discuss selected aspects of these mechanisms with a focus on those relevant to ongoing efforts to develop disease-modifying therapies. We also discuss advances in identification of patient subtypes, and selected examples of potential disease-modifying therapies in clinical development.
Collapse
|
508
|
Vázquez-Medina JP, Dodia C, Weng L, Mesaros C, Blair IA, Feinstein SI, Chatterjee S, Fisher AB. The phospholipase A2 activity of peroxiredoxin 6 modulates NADPH oxidase 2 activation via lysophosphatidic acid receptor signaling in the pulmonary endothelium and alveolar macrophages. FASEB J 2016; 30:2885-98. [PMID: 27178323 DOI: 10.1096/fj.201500146r] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/26/2016] [Indexed: 01/04/2023]
Abstract
Peroxiredoxin 6 (Prdx6) is essential for activation of NADPH oxidase type 2 (NOX2) in pulmonary microvascular endothelial cells (PMVECs), alveolar macrophages (AMs), and polymorphonuclear leukocytes. Angiotensin II and phorbol ester increased superoxide/H2O2 generation in PMVECs, AMs, and isolated lungs from wild-type (WT) mice, but had much less effect on cells or lungs from Prdx6-null or Prdx6-D140A-knock-in mice that lack the phospholipase A2 activity (PLA2) of Prdx6; addition of either lysophosphatidylcholine (LPC) or lysophosphatidic acid (LPA) to cells restored their oxidant generation. The generation of LPC by PMVECs required Prdx6-PLA2 We propose that Prdx6-PLA2 modulates NOX2 activation by generation of LPC that is converted to LPA by the lysophospholipase D activity of autotaxin (ATX/lysoPLD). Inhibition of lysoPLD with HA130 (cells,10 μM; lungs, 20 μM; IC50, 29 nM) decreased agonist-induced oxidant generation. LPA stimulates pathways regulated by small GTPases through binding to G-protein-coupled LPA receptors (LPARs). The LPAR blocker Ki16425 (cells, 10 μM; lungs, 25 μM; Ki, 0.34 μM) or cellular knockdown of LPAR type 1 decreased oxidant generation and blocked translocation of rac1 to plasma membrane. Thus, Prdx6-PLA2 modulates NOX2 activation through generation of LPC for conversion to LPA; binding of LPA to LPAR1 signals rac activation.-Vázquez-Medina, J. P., Dodia, C., Weng, L., Mesaros, C., Blair, I. A., Feinstein, S. I., Chatterjee, S., Fisher, A. B. The phospholipase A2 activity of peroxiredoxin 6 modulates NADPH oxidase 2 activation via lysophosphatidic acid receptor signaling in the pulmonary endothelium and alveolar macrophages.
Collapse
Affiliation(s)
- José Pablo Vázquez-Medina
- Institute for Environmental Medicine, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - Chandra Dodia
- Institute for Environmental Medicine, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - Liwei Weng
- Center for Cancer Pharmacology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA Center for Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clementina Mesaros
- Center for Cancer Pharmacology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA Center for Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian A Blair
- Center for Cancer Pharmacology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA Center for Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sheldon I Feinstein
- Institute for Environmental Medicine, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - Shampa Chatterjee
- Institute for Environmental Medicine, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - Aron B Fisher
- Institute for Environmental Medicine, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| |
Collapse
|
509
|
Steroid binding to Autotaxin links bile salts and lysophosphatidic acid signalling. Nat Commun 2016; 7:11248. [PMID: 27075612 PMCID: PMC4834639 DOI: 10.1038/ncomms11248] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 03/04/2016] [Indexed: 12/12/2022] Open
Abstract
Autotaxin (ATX) generates the lipid mediator lysophosphatidic acid (LPA). ATX-LPA signalling is involved in multiple biological and pathophysiological processes, including vasculogenesis, fibrosis, cholestatic pruritus and tumour progression. ATX has a tripartite active site, combining a hydrophilic groove, a hydrophobic lipid-binding pocket and a tunnel of unclear function. We present crystal structures of rat ATX bound to 7α-hydroxycholesterol and the bile salt tauroursodeoxycholate (TUDCA), showing how the tunnel selectively binds steroids. A structure of ATX simultaneously harbouring TUDCA in the tunnel and LPA in the pocket, together with kinetic analysis, reveals that bile salts act as partial non-competitive inhibitors of ATX, thereby attenuating LPA receptor activation. This unexpected interplay between ATX-LPA signalling and select steroids, notably natural bile salts, provides a molecular basis for the emerging association of ATX with disorders associated with increased circulating levels of bile salts. Furthermore, our findings suggest potential clinical implications in the use of steroid drugs. Autotaxin generates the bioactive lipid lysophosphatidic acid to regulate diverse biological processes. Here, the authors identify a role for bile salts as direct allosteric inhibitors of autotaxin activity, suggesting that steroids may function as regulators of lysophosphatidic acid signalling.
Collapse
|
510
|
Velasco M, O'Sullivan C, Sheridan GK. Lysophosphatidic acid receptors (LPARs): Potential targets for the treatment of neuropathic pain. Neuropharmacology 2016; 113:608-617. [PMID: 27059127 DOI: 10.1016/j.neuropharm.2016.04.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/15/2015] [Accepted: 04/04/2016] [Indexed: 01/08/2023]
Abstract
Neuropathic pain can arise from lesions to peripheral or central nerve fibres leading to spontaneous action potential generation and a lowering of the nociceptive threshold. Clinically, neuropathic pain can manifest in many chronic disease states such as cancer, diabetes or multiple sclerosis (MS). The bioactive lipid, lysophosphatidic acid (LPA), via activation of its receptors (LPARs), is thought to play a central role in both triggering and maintaining neuropathic pain. In particular, following an acute nerve injury, the excitatory neurotransmitters glutamate and substance P are released from primary afferent neurons leading to upregulated synthesis of lysophosphatidylcholine (LPC), the precursor for LPA production. LPC is converted to LPA by autotaxin (ATX), which can then activate macrophages/microglia and modulate neuronal functioning. A ubiquitous feature of animal models of neuropathic pain is demyelination of damaged nerves. It is thought that LPA contributes to demyelination through several different mechanisms. Firstly, high levels of LPA are produced following macrophage/microglial activation that triggers a self-sustaining feed-forward loop of de novo LPA synthesis. Secondly, macrophage/microglial activation contributes to inflammation-mediated demyelination of axons, thus initiating neuropathic pain. Therefore, targeting LPA production and/or the family of LPA-activated G protein-coupled receptors (GPCRs) may prove to be fruitful clinical approaches to treating demyelination and the accompanying neuropathic pain. This review discusses our current understanding of the role of LPA/LPAR signalling in the initiation of neuropathic pain and suggests potential targeted strategies for its treatment. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- María Velasco
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | | | - Graham K Sheridan
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK.
| |
Collapse
|
511
|
Chap H. Forty five years with membrane phospholipids, phospholipases and lipid mediators: A historical perspective. Biochimie 2016; 125:234-49. [PMID: 27059515 DOI: 10.1016/j.biochi.2016.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 01/02/2023]
Abstract
Phospholipases play a key role in the metabolism of phospholipids and in cell signaling. They are also a very useful tool to explore phospholipid structure and metabolism as well as membrane organization. They are at the center of this review, covering a period starting in 1971 and focused on a number of subjects in which my colleagues and I have been involved. Those include determination of phospholipid asymmetry in the blood platelet membrane, biosynthesis of lysophosphatidic acid, biochemistry of platelet-activating factor, first attempts to define the role of phosphoinositides in cell signaling, and identification of novel digestive (phospho)lipases such as pancreatic lipase-related protein 2 (PLRP2) or phospholipase B. Besides recalling some of our contributions to those various fields, this review makes an appraisal of the impressive and often unexpected evolution of those various aspects of membrane phospholipids and lipid mediators. It is also the occasion to propose some new working hypotheses.
Collapse
Affiliation(s)
- Hugues Chap
- Centre de Physiopathologie de Toulouse Purpan, Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France. hugues.chap.@univ-tlse3.fr
| |
Collapse
|
512
|
Castilla-Ortega E, Pavón FJ, Sánchez-Marín L, Estivill-Torrús G, Pedraza C, Blanco E, Suárez J, Santín L, Rodríguez de Fonseca F, Serrano A. Both genetic deletion and pharmacological blockade of lysophosphatidic acid LPA1 receptor results in increased alcohol consumption. Neuropharmacology 2016; 103:92-103. [DOI: 10.1016/j.neuropharm.2015.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/25/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022]
|
513
|
Jeong W, Seo H, Sung Y, Ka H, Song G, Kim J. Lysophosphatidic Acid (LPA) Receptor 3-Mediated LPA Signal Transduction Pathways: A Possible Relationship with Early Development of Peri-Implantation Porcine Conceptus. Biol Reprod 2016; 94:104. [PMID: 27030044 DOI: 10.1095/biolreprod.115.137174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/16/2016] [Indexed: 11/01/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a phospholipid with a variety of fatty acyl groups that mediates diverse biological effects on various types of cells through specific G protein-coupled receptors. LPA appears to play a significant role in many reproductive processes, including luteolysis, implantation, and placentation. Our previous study in pigs demonstrated that LPA and the LPA receptor system are present at the maternal-conceptus interface and that LPA increases uterine endometrial expression of prostaglandin-endoperoxide synthase 2 (PTGS2) through LPA receptor 3 (LPAR3). However, the role of LPA in conceptuses during early pregnancy has not been determined. Therefore, this study examined the effects of LPA in cell proliferation, migration, and activation of the intracellular signaling pathway in porcine conceptuses by using an established porcine trophectoderm (pTr) cell line isolated from Day 12 conceptuses. All examined LPA species with various fatty acid lengths increased proliferation and migration of pTr cells as the dosage increased. Immunoblot analyses found that LPA activated intracellular signaling molecules, extracellular signal-regulated kinase 1/2 (ERK1/2), ribosomal protein S6 kinase 90 kDa (P90RSK), ribosomal protein S6 (RPS6), and P38 in pTr cells. Furthermore, LPA increased expression of PTGS2 and urokinase-type plasminogen activator (PLAU), and the LPA-induced increases in PTGS2 and PLAU expression were inhibited by LPAR3 siRNA. Collectively, these results showed that LPA promotes proliferation, migration, and differentiation of pTr cells by activating the ERK1/2-P90RSK-RPS6 and P38 pathways, indicating that the LPA-LPAR3 system may be involved in the development of trophoblast during early pregnancy in pigs.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Heewon Seo
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Yujin Sung
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Hakhyun Ka
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jinyoung Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
514
|
Nishioka T, Arima N, Kano K, Hama K, Itai E, Yukiura H, Kise R, Inoue A, Kim SH, Solnica-Krezel L, Moolenaar WH, Chun J, Aoki J. ATX-LPA1 axis contributes to proliferation of chondrocytes by regulating fibronectin assembly leading to proper cartilage formation. Sci Rep 2016; 6:23433. [PMID: 27005960 PMCID: PMC4804234 DOI: 10.1038/srep23433] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/07/2016] [Indexed: 12/20/2022] Open
Abstract
The lipid mediator lysophosphatidic acid (LPA) signals via six distinct G protein-coupled receptors to mediate both unique and overlapping biological effects, including cell migration, proliferation and survival. LPA is produced extracellularly by autotaxin (ATX), a secreted lysophospholipase D, from lysophosphatidylcholine. ATX-LPA receptor signaling is essential for normal development and implicated in various (patho)physiological processes, but underlying mechanisms remain incompletely understood. Through gene targeting approaches in zebrafish and mice, we show here that loss of ATX-LPA1 signaling leads to disorganization of chondrocytes, causing severe defects in cartilage formation. Mechanistically, ATX-LPA1 signaling acts by promoting S-phase entry and cell proliferation of chondrocytes both in vitro and in vivo, at least in part through β1-integrin translocation leading to fibronectin assembly and further extracellular matrix deposition; this in turn promotes chondrocyte-matrix adhesion and cell proliferation. Thus, the ATX-LPA1 axis is a key regulator of cartilage formation.
Collapse
Affiliation(s)
- Tatsuji Nishioka
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Naoaki Arima
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Kotaro Hama
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Eriko Itai
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Hiroshi Yukiura
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan.,Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology (PRESTO), Kawaguchi City, Saitama 332-0012, Japan
| | - Seok-Hyung Kim
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wouter H Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA-92037, USA
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki-aza, Aoba-ku, Sendai, 980-8578, Japan.,Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-0004 Japan
| |
Collapse
|
515
|
Liu Y, An S, Ward R, Yang Y, Guo XX, Li W, Xu TR. G protein-coupled receptors as promising cancer targets. Cancer Lett 2016; 376:226-39. [PMID: 27000991 DOI: 10.1016/j.canlet.2016.03.031] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) regulate an array of fundamental biological processes, such as growth, metabolism and homeostasis. Specifically, GPCRs are involved in cancer initiation and progression. However, compared with the involvement of the epidermal growth factor receptor in cancer, that of GPCRs have been largely ignored. Recent findings have implicated many GPCRs in tumorigenesis, tumor progression, invasion and metastasis. Moreover, GPCRs contribute to the establishment and maintenance of a microenvironment which is permissive for tumor formation and growth, including effects upon surrounding blood vessels, signaling molecules and the extracellular matrix. Thus, GPCRs are considered to be among the most useful drug targets against many solid cancers. Development of selective ligands targeting GPCRs may provide novel and effective treatment strategies against cancer and some anticancer compounds are now in clinical trials. Here, we focus on tumor related GPCRs, such as G protein-coupled receptor 30, the lysophosphatidic acid receptor, angiotensin receptors 1 and 2, the sphingosine 1-phosphate receptors and gastrin releasing peptide receptor. We also summarize their tissue distributions, activation and roles in tumorigenesis and discuss the potential use of GPCR agonists and antagonists in cancer therapy.
Collapse
Affiliation(s)
- Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Richard Ward
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Wei Li
- Kidney Cancer Research, Diagnosis and Translational Technology Center of Yunnan Province, Department of Urology, The People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
516
|
Lysophosphatidic acid activates Arf6 to promote the mesenchymal malignancy of renal cancer. Nat Commun 2016; 7:10656. [PMID: 26854204 PMCID: PMC4748122 DOI: 10.1038/ncomms10656] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022] Open
Abstract
Acquisition of mesenchymal properties by cancer cells is critical for their malignant behaviour, but regulators of the mesenchymal molecular machinery and how it is activated remain elusive. Here we show that clear cell renal cell carcinomas (ccRCCs) frequently utilize the Arf6-based mesenchymal pathway to promote invasion and metastasis, similar to breast cancers. In breast cancer cells, ligand-activated receptor tyrosine kinases employ GEP100 to activate Arf6, which then recruits AMAP1; and AMAP1 then binds to the mesenchymal-specific protein EPB41L5, which promotes epithelial–mesenchymal transition and focal adhesion dynamics. In renal cancer cells, lysophosphatidic acid (LPA) activates Arf6 via its G-protein-coupled receptors, in which GTP-Gα12 binds to EFA6. The Arf6-based pathway may also contribute to drug resistance. Our results identify a specific mesenchymal molecular machinery of primary ccRCCs, which is triggered by a product of autotaxin and it is associated with poor outcome of patients. Acquisition of mesenchymal properties by cancer cells is a critical event for the development of malignancy. Here, the authors show that in renal cancer cells, lysosphosphatidic acid does not utilize the RhoA pathway but specifically activates the Arf6 mesenchymal pathway via its GPCRs and EFA6 to promote invasion, metastasis and drug resistance.
Collapse
|
517
|
Waite C, Mejia R, Ascoli M. Gq/11-Dependent Changes in the Murine Ovarian Transcriptome at the End of Gestation. Biol Reprod 2016; 94:62. [PMID: 26843449 PMCID: PMC4829089 DOI: 10.1095/biolreprod.115.136952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/01/2016] [Indexed: 11/26/2022] Open
Abstract
Parturition in rodents is highly dependent on the engagement of the luteal prostaglandin F2 alpha receptor, which, through activation of the Gq/11 family of G proteins, increases the expression of Akr1c18, leading to an increase in progesterone catabolism. To further understand the involvement of Gq/11 on luteolysis and parturition, we used microarray analysis to compare the ovarian transcriptome of mice with a granulosa/luteal cell-specific deletion of Galphaq/11 with their control littermates on Day 18 of pregnancy, when mice from both genotypes are pregnant, and on Day 22, when mice with a granulosa/luteal cell-specific deletion of Galphaq/11 are still pregnant but their control littermates are 1–2 days postpartum. Ovarian genes up-regulated at the end of gestation in a Galphaq/11 -dependent fashion include genes involved in focal adhesion and extracellular matrix interactions. Genes down-regulated at the end of gestation in a Galphaq/11-dependent manner include Serpina6 (which encodes corticosteroid-binding globulin); Enpp2 (which encodes autotaxin, the enzyme responsible for the synthesis of lysophosphatidic acid); genes involved in protein processing and export; reproductive genes, such as Lhcgr; the three genes needed to convert progesterone to estradiol (Cyp17a1, Hsd17b7, and Cyp19a1); and Inha. Activation of ovarian Gq/11 by engagement of the prostaglandin F2 alpha receptor on Day 18 of pregnancy recapitulated the regulation of many but not all of these genes. Thus, although the ovarian transcriptome at the end of gestation is highly dependent on the activation of Gq/11, not all of these changes are dependent on the actions of prostaglandin F2 alpha.
Collapse
Affiliation(s)
- Courtney Waite
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rachel Mejia
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Mario Ascoli
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
518
|
G-Protein-Coupled Lysophosphatidic Acid Receptors and Their Regulation of AKT Signaling. Int J Mol Sci 2016; 17:215. [PMID: 26861299 PMCID: PMC4783947 DOI: 10.3390/ijms17020215] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 12/13/2022] Open
Abstract
A hallmark of G-protein-coupled receptors (GPCRs) is their ability to recognize and respond to chemically diverse ligands. Lysophospholipids constitute a relatively recent addition to these ligands and carry out their biological functions by activating G-proteins coupled to a large family of cell-surface receptors. This review aims to highlight salient features of cell signaling by one class of these receptors, known as lysophosphatidic acid (LPA) receptors, in the context of phosphatidylinositol 3-kinase (PI3K)-AKT pathway activation. LPA moieties efficiently activate AKT phosphorylation and activation in a multitude of cell types. The interplay between LPA, its receptors, the associated Gαi/o subunits, PI3K and AKT contributes to the regulation of cell survival, migration, proliferation and confers chemotherapy-resistance in certain cancers. However, detailed information on the regulation of PI3K-AKT signals induced by LPA receptors is missing from the literature. Here, some urgent issues for investigation are highlighted.
Collapse
|
519
|
Ichiki T, Koga T, Okuno T, Saeki K, Yamamoto Y, Yamamoto H, Sakaguchi M, Yokomizo T. Modulation of leukotriene B
4
receptor 1 signaling by receptor for advanced glycation end products (RAGE). FASEB J 2016; 30:1811-22. [DOI: 10.1096/fj.201500117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/07/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Takako Ichiki
- Department of BiochemistryJuntendo University School of MedicineTokyoJapan
| | - Tomoaki Koga
- Department of BiochemistryJuntendo University School of MedicineTokyoJapan
| | - Toshiaki Okuno
- Department of BiochemistryJuntendo University School of MedicineTokyoJapan
| | - Kazuko Saeki
- Department of BiochemistryJuntendo University School of MedicineTokyoJapan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Masakiyo Sakaguchi
- Department of Cell BiologyOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Takehiko Yokomizo
- Department of BiochemistryJuntendo University School of MedicineTokyoJapan
| |
Collapse
|
520
|
Ackerman SJ, Park GY, Christman JW, Nyenhuis S, Berdyshev E, Natarajan V. Polyunsaturated lysophosphatidic acid as a potential asthma biomarker. Biomark Med 2016; 10:123-35. [PMID: 26808693 PMCID: PMC4881841 DOI: 10.2217/bmm.15.93] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022] Open
Abstract
Lysophosphatidic acid (LPA), a lipid mediator in biological fluids and tissues, is generated mainly by autotaxin that hydrolyzes lysophosphatidylcholine to LPA and choline. Total LPA levels are increased in bronchoalveolar lavage fluid from asthmatic lung, and are strongly induced following subsegmental bronchoprovocation with allergen in subjects with allergic asthma. Polyunsaturated molecular species of LPA (C22:5 and C22:6) are selectively synthesized in the airways of asthma subjects following allergen challenge and in mouse models of allergic airway inflammation, having been identified and quantified by LC/MS/MS lipidomics. This review discusses current knowledge of LPA production in asthmatic lung and the potential utility of polyunsaturated LPA molecular species as novel biomarkers in bronchoalveolar lavage fluid and exhaled breath condensate of asthma subjects.
Collapse
Affiliation(s)
- Steven J Ackerman
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Gye Young Park
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - John W Christman
- Department of Medicine, Ohio State University School of Medicine, Columbus, OH 43210, USA
| | - Sharmilee Nyenhuis
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Viswanathan Natarajan
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
521
|
Boruszewska D, Sinderewicz E, Kowalczyk-Zieba I, Grycmacher K, Woclawek-Potocka I. Studies on lysophosphatidic acid action during in vitro preimplantation embryo development. Domest Anim Endocrinol 2016; 54:15-29. [PMID: 26379100 DOI: 10.1016/j.domaniend.2015.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/18/2015] [Accepted: 08/18/2015] [Indexed: 11/24/2022]
Abstract
Assisted reproductive technologies, including in vitro embryo production (IVP), have been successfully used in animal reproduction to optimize breeding strategies for improved production and health in animal husbandry. Despite the progress in IVP techniques over the years, further improvements in in vitro embryo culture systems are required for the enhancement of oocyte and embryo developmental competence. One of the most important issues associated with IVP procedures is the optimization of the in vitro culture of oocytes and embryos. Studies in different species of animals and in humans have identified important roles for receptor-mediated lysophosphatidic acid (LPA) signaling in multiple aspects of human and animal reproductive tract function. The data on LPA signaling in the ovary and uterus suggest that LPA can directly contribute to embryo-maternal interactions via its influence on early embryo development beginning from the influence of the ovarian environment on the oocyte to the influence of the uterine environment on the preimplantation embryo. This review discusses the current status of LPA as a potential supplement in oocyte maturation, fertilization, and embryo culture media and current views on the potential involvement of the LPA signaling pathway in early embryo development.
Collapse
Affiliation(s)
- D Boruszewska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-748, Poland
| | - E Sinderewicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-748, Poland
| | - I Kowalczyk-Zieba
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-748, Poland
| | - K Grycmacher
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-748, Poland
| | - I Woclawek-Potocka
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-748, Poland.
| |
Collapse
|
522
|
Li YF, Li RS, Samuel SB, Cueto R, Li XY, Wang H, Yang XF. Lysophospholipids and their G protein-coupled receptors in atherosclerosis. Front Biosci (Landmark Ed) 2016; 21:70-88. [PMID: 26594106 DOI: 10.2741/4377] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lysophospholipids (LPLs) are bioactive lipid-derived signaling molecules generated by the enzymatic and chemical processes of regiospecific phospholipases on substrates such as membrane phospholipids (PLs) and sphingolipids (SLs). They play a major role as extracellular mediators by activating G-protein coupled receptors (GPCRs) and stimulating diverse cellular responses from their signaling pathways. LPLs are involved in various pathologies of the vasculature system including coronary heart disease and hypertension. Many studies suggest the importance of LPLs in their association with the development of atherosclerosis, a chronic and severe vascular disease. This paper focuses on the pathophysiological effects of different lysophospholipids on atherosclerosis, which may promote the pathogenesis of myocardial infarction and strokes. Their atherogenic biological activities take place in vascular endothelial cells, vascular smooth muscle cells, fibroblasts, monocytes and macrophages, dendritic cells, T-lymphocytes, platelets, etc.
Collapse
Affiliation(s)
- Ya-Feng Li
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA ; Department of Nephrology and Hemodialysis Center, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Rong-Shan Li
- Department of Nephrology and Hemodialysis Center, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Sonia B Samuel
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ramon Cueto
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xin-Yuan Li
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiao-Feng Yang
- Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
523
|
Carpéné C, Galitzky J, Sébastien Saulnier-Blache J. Short-term and rapid effects of lysophosphatidic acid on human adipose cell lipolytic and glucose uptake activities. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.2.222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
524
|
Howard P, Twycross R, Grove G, Charlesworth S, Mihalyo M, Wilcock A. Rifampin (INN Rifampicin). J Pain Symptom Manage 2015; 50:891-895. [PMID: 26432572 DOI: 10.1016/j.jpainsymman.2015.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 12/12/2022]
Abstract
Therapeutic Reviews aim to provide essential independent information for health professionals about drugs used in palliative and hospice care. Additional content is available on www.palliativedrugs.com. Country-specific books (Hospice and Palliative Care Formulary USA, and Palliative Care Formulary, British and Canadian editions) are also available and can be ordered from www.palliativedrugs.com. The series editors welcome feedback on the articles (hq@palliativedrugs.com).
Collapse
Affiliation(s)
- Paul Howard
- Earl Mountbatten Hospice, Isle of Wight, United Kingdom
| | | | - Graham Grove
- Earl Mountbatten Hospice, Isle of Wight, United Kingdom
| | | | - Mary Mihalyo
- Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
525
|
Stein AJ, Bain G, Prodanovich P, Santini AM, Darlington J, Stelzer NMP, Sidhu RS, Schaub J, Goulet L, Lonergan D, Calderon I, Evans JF, Hutchinson JH. Structural Basis for Inhibition of Human Autotaxin by Four Potent Compounds with Distinct Modes of Binding. Mol Pharmacol 2015; 88:982-92. [PMID: 26371182 DOI: 10.1124/mol.115.100404] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022] Open
Abstract
Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is a bioactive phospholipid that regulates diverse biological processes, including cell proliferation, migration, and survival/apoptosis, through the activation of a family of G protein-coupled receptors. The ATX-LPA pathway has been implicated in many pathologic conditions, including cancer, fibrosis, inflammation, cholestatic pruritus, and pain. Therefore, ATX inhibitors represent an attractive strategy for the development of therapeutics to treat a variety of diseases. Mouse and rat ATX have been crystallized previously with LPA or small-molecule inhibitors bound. Here, we present the crystal structures of human ATX in complex with four previously unpublished, structurally distinct ATX inhibitors. We demonstrate that the mechanism of inhibition of each compound reflects its unique interactions with human ATX. Our studies may provide a basis for the rational design of novel ATX inhibitors.
Collapse
Affiliation(s)
- Adam J Stein
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Gretchen Bain
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Pat Prodanovich
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Angelina M Santini
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Janice Darlington
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Nina M P Stelzer
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Ranjinder S Sidhu
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Jeffrey Schaub
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Lance Goulet
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Dave Lonergan
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Imelda Calderon
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Jilly F Evans
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - John H Hutchinson
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| |
Collapse
|
526
|
Reeves VL, Trybula JS, Wills RC, Goodpaster BH, Dubé JJ, Kienesberger PC, Kershaw EE. Serum Autotaxin/ENPP2 correlates with insulin resistance in older humans with obesity. Obesity (Silver Spring) 2015; 23:2371-6. [PMID: 26727116 PMCID: PMC4700540 DOI: 10.1002/oby.21232] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Autotaxin (ATX) is an adipocyte-derived lysophospholipase D that generates the lipid signaling molecule lysophosphatidic acid (LPA). The ATX/LPA pathway in adipose tissue has recently been implicated in obesity and insulin resistance in animal models, but the role of circulating ATX in humans remains unclear. The aim of the present study was to determine the relationship between serum ATX and insulin resistance. METHODS Older (60-75 years), nondiabetic human participants with overweight or obesity (BMI 25-37 kg m(-2) ) were characterized for metabolic phenotype including measures of energy, glucose, and lipid homeostasis. The relationship between serum ATX and metabolic parameters was then determined using correlative and predictive statistics. RESULTS Serum ATX was higher in females than in males. After controlling for sex, serum ATX correlated with multiple measures of adiposity and glucose homeostasis/insulin action. Serum ATX and BMI also independently predicted glucose infusion rate during a hyperinsulinemic euglycemic clamp and homeostatic model assessment of insulin resistance after controlling for sex and medication use. CONCLUSIONS Serum ATX correlates with and predicts measures of glucose homeostasis and insulin sensitivity in older humans, suggesting that it may be a potential pathogenic factor and/or diagnostic/therapeutic target for insulin resistance in this population.
Collapse
Affiliation(s)
- Valerie L. Reeves
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joy S. Trybula
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rachel C. Wills
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bret H. Goodpaster
- Diabetes and Obesity Research Center, Sanford Burnham Medical Research Institute, Orlando, FL 32827, USA
| | - John J. Dubé
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Petra C. Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie Medicine New Brunswick, Saint John, NB E2L4L5, Canada
| | - Erin E. Kershaw
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Corresponding author: Erin E. Kershaw, M.D., Division of Endocrinology, Department of Medicine, University of Pittsburgh, 200 Lothrop Street, BST E1140, Pittsburgh, PA 15261, USA, Telephone: 412-648-8454; Fax: 412-648-3290
| |
Collapse
|
527
|
Nan L, Wei J, Jacko AM, Culley MK, Zhao J, Natarajan V, Ma H, Zhao Y. Cross-talk between lysophosphatidic acid receptor 1 and tropomyosin receptor kinase A promotes lung epithelial cell migration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:229-35. [PMID: 26597701 DOI: 10.1016/j.bbamcr.2015.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/28/2015] [Accepted: 11/16/2015] [Indexed: 02/02/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid, which plays a crucial role in the regulation of cell proliferation, migration, and differentiation. LPA exerts its biological effects mainly through binding to cell-surface LPA receptors (LPA1-6), which belong to the G protein-coupled receptor (GPCR) family. Recent studies suggest that cross-talk between receptor tyrosine kinases (RTKs) and GPCRs modulates GPCRs-mediated signaling. Tropomyosin receptor kinase A (TrkA) is a RTK, which mediates nerve growth factor (NGF)-induced biological functions including cell migration in neuronal and non-neuronal cells. Here, we show LPA1 transactivation of TrkA in murine lung epithelial cells (MLE12). LPA induced tyrosine phosphorylation of TrkA in both time- and dose-dependent manners. Down-regulation of LPA1 by siRNA transfection attenuated LPA-induced phosphorylation of TrkA, suggesting a cross-talk between LPA1 and TrkA. To investigate the molecular regulation of the cross-talk, we focused on the interaction between LPA1 and TrkA. We found that LPA induced interaction between LPA1 and TrkA. The LPA1/TrkA complex was localized on the plasma membrane and in the cytoplasm. The C-terminus of LPA1 was identified as the binding site for TrkA. Inhibition of TrkA attenuated LPA-induced phosphorylation of TrkA and LPA1 internalization, as well as lung epithelial cell migration. These studies provide a molecular mechanism for the transactivation of TrkA by LPA, and suggest that the cross-talk between LPA1 and TrkA regulates LPA-induced receptor internalization and lung epithelial cell migration.
Collapse
Affiliation(s)
- Ling Nan
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China; Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jianxin Wei
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anastasia M Jacko
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Miranda K Culley
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jing Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States
| | - Haichun Ma
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China
| | - Yutong Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
528
|
The Role of Lysophosphatidic Acid on Airway Epithelial Cell Denudation in a Murine Heterotopic Tracheal Transplant Model. Transplant Direct 2015; 1:e35. [PMID: 27500235 PMCID: PMC4946481 DOI: 10.1097/txd.0000000000000542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/15/2015] [Indexed: 01/06/2023] Open
Abstract
Supplemental digital content is available in the text. Background Chronic rejection is the major leading cause of morbidity and mortality after lung transplantation. Obliterative bronchiolitis (OB), a fibroproliferative disorder of the small airways, is the main manifestation of chronic lung allograft rejection. However, there is currently no treatment for the disease. We hypothesized that lysophosphatidic acid (LPA) participates in the progression of OB. The aim of this study was to reveal the involvement of LPA on the lesion of OB. Methods Ki16198, an antagonist specifically for LPA1 and LPA3, was daily administered into the heterotopic tracheal transplant model mice at the day of transplantation. At days 10 and 28, the allografts were isolated and evaluated histologically. The messenger RNA levels of LPAR in microdissected mouse airway regions were assessed to reveal localization of lysophosphatidic acid receptors. The human airway epithelial cell was used to evaluate the mechanism of LPA-induced suppression of cell adhesion to the extracellular matrix (ECM). Results The administration of Ki16198 attenuated airway epithelial cell loss in the allograft at day 10. Messenger RNAs of LPA1 and LPA3 were detected in the airway epithelial cells of the mice. Lysophosphatidic acid inhibited the attachment of human airway epithelial cells to the ECM and induced cell detachment from the ECM, which was mediated by LPA1 and Rho-kinase pathway. However, Ki16198 did not prevent obliteration of allograft at day 28. Conclusions The LPA signaling is involved in the status of epithelial cells by distinct contribution in 2 different phases of the OB lesion. This finding suggests a role of LPA in the pathogenesis of OB.
Collapse
|
529
|
Phosphorylation and Internalization of Lysophosphatidic Acid Receptors LPA1, LPA2, and LPA3. PLoS One 2015; 10:e0140583. [PMID: 26473723 PMCID: PMC4608732 DOI: 10.1371/journal.pone.0140583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/27/2015] [Indexed: 12/31/2022] Open
Abstract
Results The lysophosphatidic acid receptors LPA1, LPA2, and LPA3 were individually expressed in C9 cells and their signaling and regulation were studied. Agonist-activation increases intracellular calcium concentration in a concentration-dependent fashion. Phorbol myristate acetate markedly inhibited LPA1- and LPA3-mediated effect, whereas that mediated by LPA2 was only partially diminished; the actions of the phorbol ester were inhibited by bisindolylmaleimide I and by overnight incubation with the protein kinase C activator, which leads to down regulation of this protein kinase. Homologous desensitization was also observed for the three LPA receptors studied, with that of LPA2 receptors being consistently of lesser magnitude; neither inhibition nor down-regulation of protein kinase C exerted any effect on homologous desensitization. Activation of LPA1–3 receptors induced ERK 1/2 phosphorylation; this effect was markedly attenuated by inhibition of epidermal growth factor receptor tyrosine kinase activity, suggesting growth factor receptor transactivation in this effect. Lysophosphatidic acid and phorbol myristate acetate were able to induce LPA1–3 phosphorylation, in time- and concentration-dependent fashions. It was also clearly observed that agonists and protein kinase C activation induced internalization of these receptors. Phosphorylation of the LPA2 subtype required larger concentrations of these agents and its internalization was less intense than that of the other subtypes. Conclusion Our data show that these three LPA receptors are phosphoproteins whose phosphorylation state is modulated by agonist-stimulation and protein kinase C-activation and that differences in regulation and cellular localization exist, among the subtypes.
Collapse
|
530
|
Si J, Su Y, Wang Y, Yan YL, Tang YL. Expressions of lysophosphatidic acid receptors in the development of human ovarian carcinoma. Int J Clin Exp Med 2015; 8:17880-17890. [PMID: 26770382 PMCID: PMC4694282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/05/2015] [Indexed: 06/05/2023]
Abstract
AIM To investigate the associations between the expressions of three lysophosphatidic acid (LPA) receptors (LPA1-3) and the development of ovarian carcinoma (OC). METHOD Ovarian tissue specimens, including normal ovarian epithelium tissues, benign ovarian tumor tissues and OC tissues were collected from patients who underwent surgical resections between March 2012 and December 2014. Immunohistochemical staining was used to detect LPA receptor expressions in ovarian tissues. Reverse transcription-polymerase chain reaction and Western blotting were used to detect mRNA and protein expression of LPA receptors, respectively. Association analysis between LPA receptors protein expression and clinical pathological characteristics was conducted. The value of LPA2 and LPA3 in discriminating OC was confirmed by receiver-operator characteristic (ROC) curves analysis. RESULTS The positive expression rates of LPA2 and LPA3 in OC group was obviously higher than normal control and benign groups. The LPA2 and LPA3 mRNA and protein levels in OC group were higher than in normal control and benign groups. LPA2 and LPA3 mRNA expression levels were positively correlated with LPA2 and LPA3 protein expression in OC group. ROC curve analysis revealed that LPA2 yield a specificity of 96.3% and a sensitivity of 97.9%, and LPA3 yield a specificity of 98.5% and a sensitivity of 97.9% for the detection of OC. CONCLUSION LPA2 and LPA3 were highly expressed in OC tissues, which may be involved in the development of OC. Further, LPA2 and LPA3 had higher sensitivity and specificity in distinguishing the OC from benign ovarian tumors, which could be potential diagnostic indictors in OC.
Collapse
Affiliation(s)
- Jinge Si
- Department of Gynecology and Obstetrics, Zhujiang Hospital of Southern Medical UniversityGuangzhou 510515, P. R. China
| | - Yuanyuan Su
- Department of Gynecology and Obstetrics, Zhongshan Affiliated Hospital of Sun Yat-Sen UniversityZhongshan 528403, P. R. China
| | - Yifeng Wang
- Department of Gynecology and Obstetrics, Zhujiang Hospital of Southern Medical UniversityGuangzhou 510515, P. R. China
| | - You-Liang Yan
- Department of Gynecology, Boai Hospital of ZhongshanZhongshan 528403, P. R. China
| | - Ya-Ling Tang
- Department of Gynecology, The First Affiliated Hospital of Xiamen UniversityXiamen 361003, P. R. China
| |
Collapse
|
531
|
Lee ST, Wiemels JL. Genome-wide CpG island methylation and intergenic demethylation propensities vary among different tumor sites. Nucleic Acids Res 2015; 44:1105-17. [PMID: 26464434 PMCID: PMC4756811 DOI: 10.1093/nar/gkv1038] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022] Open
Abstract
The epigenetic landscape of cancer includes both focal hypermethylation and broader hypomethylation in a genome-wide manner. By means of a comprehensive genomic analysis on 6637 tissues of 21 tumor types, we here show that the degrees of overall methylation in CpG island (CGI) and demethylation in intergenic regions, defined as ‘backbone’, largely vary among different tumors. Depending on tumor type, both CGI methylation and backbone demethylation are often associated with clinical, epidemiological and biological features such as age, sex, smoking history, anatomic location, histological type and grade, stage, molecular subtype and biological pathways. We found connections between CGI methylation and hypermutability, microsatellite instability, IDH1 mutation, 19p gain and polycomb features, and backbone demethylation with chromosomal instability, NSD1 and TP53 mutations, 5q and 19p loss and long repressive domains. These broad epigenetic patterns add a new dimension to our understanding of tumor biology and its clinical implications.
Collapse
Affiliation(s)
- Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, 120752, Republic of Korea
| | - Joseph L Wiemels
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, CA 94158, USA
| |
Collapse
|
532
|
Activation of Lysophosphatidic Acid Receptor Type 1 Contributes to Pathophysiology of Spinal Cord Injury. J Neurosci 2015; 35:10224-35. [PMID: 26180199 DOI: 10.1523/jneurosci.4703-14.2015] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Lysophosphatidic acid (LPA) is an extracellular lipid mediator involved in many physiological functions that signals through six known G-protein-coupled receptors (LPA1-LPA6). A wide range of LPA effects have been identified in the CNS, including neural progenitor cell physiology, astrocyte and microglia activation, neuronal cell death, axonal retraction, and development of neuropathic pain. However, little is known about the involvement of LPA in CNS pathologies. Herein, we demonstrate for the first time that LPA signaling via LPA1 contributes to secondary damage after spinal cord injury. LPA levels increase in the contused spinal cord parenchyma during the first 14 d. To model this potential contribution of LPA in the spinal cord, we injected LPA into the normal spinal cord, revealing that LPA induces microglia/macrophage activation and demyelination. Use of a selective LPA1 antagonist or mice lacking LPA1 linked receptor-mediated signaling to demyelination, which was in part mediated by microglia. Finally, we demonstrate that selective blockade of LPA1 after spinal cord injury results in reduced demyelination and improvement in locomotor recovery. Overall, these results support LPA-LPA1 signaling as a novel pathway that contributes to secondary damage after spinal cord contusion in mice and suggest that LPA1 antagonism might be useful for the treatment of acute spinal cord injury. SIGNIFICANCE STATEMENT This study reveals that LPA signaling via LPA receptor type 1 activation causes demyelination and functional deficits after spinal cord injury.
Collapse
|
533
|
Tveteraas IH, Aasrum M, Brusevold IJ, Ødegård J, Christoffersen T, Sandnes D. Lysophosphatidic acid induces both EGFR-dependent and EGFR-independent effects on DNA synthesis and migration in pancreatic and colorectal carcinoma cells. Tumour Biol 2015; 37:2519-26. [DOI: 10.1007/s13277-015-4010-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022] Open
|
534
|
No YR, Lee SJ, Kumar A, Yun CC. HIF1α-Induced by Lysophosphatidic Acid Is Stabilized via Interaction with MIF and CSN5. PLoS One 2015; 10:e0137513. [PMID: 26352431 PMCID: PMC4564097 DOI: 10.1371/journal.pone.0137513] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/18/2015] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that has broad effects on immune system and inflammatory response. A growing body of evidence implicates the role of MIF in tumor growth and metastasis. Lysophosphatidic acid (LPA), a bioactive lipid mediator, regulates colon cancer cell proliferation, invasion, and survival through LPA2 receptor. Loss of LPA2 results in decreased expression of MIF in a rodent model of colon cancer, but the mechanism of MIF regulation by LPA is yet to be determined. In this study, we show that LPA transcriptionally regulates MIF expression in colon cancer cells. MIF knockdown decreased LPA-mediated proliferation of HCT116 human adenocarcinoma cells without altering the basal proliferation rates. Conversely, extracellular recombinant MIF stimulated cell proliferation, suggesting that the effect of MIF may in part be mediated through activation of surface receptor. We have shown recently that LPA increases hypoxia-inducible factor 1α (HIF1α) expression. We found that MIF regulation by LPA was ablated by knockdown of HIF1α, indicating that MIF is a transcriptional target of HIF1α. Conversely, knockdown of MIF ablated an increase in HIF1α expression in LPA-treated cells, suggesting a reciprocal relationship between HIF1α and MIF. LPA stimulated co-immunoprecipitation of HIF1α and MIF, indicating that their association is necessary for stabilization of HIF1α. It has been shown previously that CSN9 signalosome subunit 5 (CSN5) interacts with HIF1α to stabilize HIF1α under aerobic conditions. We found that LPA did not alter expression of CSN5, but stimulated its interaction with HIF1α and MIF. Depletion of CSN5 mitigated the association between HIF1α and MIF, indicating that CSN5 acts as a physical link. We suggest that HIF1α, MIF, and CSN5 form a ternary complex whose formation is necessary to prevent degradation of HIF1α under aerobic conditions.
Collapse
Affiliation(s)
- Yi Ran No
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Sei-Jung Lee
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ajay Kumar
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America; Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
535
|
Benesch MGK, Tang X, Dewald J, Dong WF, Mackey JR, Hemmings DG, McMullen TPW, Brindley DN. Tumor-induced inflammation in mammary adipose tissue stimulates a vicious cycle of autotaxin expression and breast cancer progression. FASEB J 2015; 29:3990-4000. [DOI: 10.1096/fj.15-274480] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
|
536
|
Benesch MGK, Tang X, Venkatraman G, Bekele RT, Brindley DN. Recent advances in targeting the autotaxin-lysophosphatidate-lipid phosphate phosphatase axis in vivo. J Biomed Res 2015; 30:272-84. [PMID: 27533936 PMCID: PMC4946318 DOI: 10.7555/jbr.30.20150058] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022] Open
Abstract
Extracellular lysophosphatidate (LPA) is a potent bioactive lipid that signals through six G-protein-coupled receptors. This signaling is required for embryogenesis, tissue repair and remodeling processes. LPA is produced from circulating lysophosphatidylcholine by autotaxin (ATX), and is degraded outside cells by a family of three enzymes called the lipid phosphate phosphatases (LPPs). In many pathological conditions, particularly in cancers, LPA concentrations are increased due to high ATX expression and low LPP activity. In cancers, LPA signaling drives tumor growth, angiogenesis, metastasis, resistance to chemotherapy and decreased efficacy of radiotherapy. Hence, targeting the ATX-LPA-LPP axis is an attractive strategy for introducing novel adjuvant therapeutic options. In this review, we will summarize current progress in targeting the ATX-LPA-LPP axis with inhibitors of autotaxin activity, LPA receptor antagonists, LPA monoclonal antibodies, and increasing low LPP expression. Some of these agents are already in clinical trials and have applications beyond cancer, including chronic inflammatory diseases.
Collapse
Affiliation(s)
- Matthew G K Benesch
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Xiaoyun Tang
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Ganesh Venkatraman
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Raie T Bekele
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - David N Brindley
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada.
| |
Collapse
|
537
|
Song J, Guan M, Zhao Z, Zhang J. Type I Interferons Function as Autocrine and Paracrine Factors to Induce Autotaxin in Response to TLR Activation. PLoS One 2015; 10:e0136629. [PMID: 26313906 PMCID: PMC4552386 DOI: 10.1371/journal.pone.0136629] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 08/05/2015] [Indexed: 12/30/2022] Open
Abstract
Lysophosphatidic acid (LPA) is an important phospholipid mediator in inflammation and immunity. However, the mechanism of LPA regulation during inflammatory response is largely unknown. Autotaxin (ATX) is the key enzyme to produce extracellular LPA from lysophosphatidylcholine (LPC). In this study, we found that ATX was induced in monocytic THP-1 cells by TLR4 ligand lipopolysaccharide (LPS), TLR9 ligand CpG oligonucleotide, and TLR3 ligand poly(I:C), respectively. The ATX induction by TLR ligand was abolished by the neutralizing antibody against IFN-β or the knockdown of IFNAR1, indicating that type I IFN autocrine loop is responsible for the ATX induction upon TLR activation. Both IFN-β and IFN-α were able to induce ATX expression via the JAK-STAT and PI3K-AKT pathways but with different time-dependent manners. The ATX induction by IFN-β was dramatically enhanced by IFN-γ, which had no significant effect on ATX expression alone, suggesting a synergy effect between type I and type II IFNs in ATX induction. Extracellular LPA levels were significantly increased when THP-1 cells were treated with IFN-α/β or TLR ligands. In addition, the type I IFN-mediated ATX induction was identified in human monocyte-derived dendritic cells (moDCs) stimulated with LPS or poly(I:C), and IFN-α/β could induce ATX expression in human peripheral blood mononuclear cells (PBMCs) and monocytes isolated form blood samples. These results suggest that, in response to TLR activation, ATX is induced through a type I INF autocrine-paracrine loop to enhance LPA generation.
Collapse
Affiliation(s)
- Jianwen Song
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ming Guan
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
| | - Zhenwen Zhao
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
| | - Junjie Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
538
|
He P, Zhao L, Zhu L, Weinman EJ, De Giorgio R, Koval M, Srinivasan S, Yun CC. Restoration of Na+/H+ exchanger NHE3-containing macrocomplexes ameliorates diabetes-associated fluid loss. J Clin Invest 2015; 125:3519-31. [PMID: 26258413 DOI: 10.1172/jci79552] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 06/25/2015] [Indexed: 01/19/2023] Open
Abstract
Diarrhea is one of the troublesome complications of diabetes, and the underlying causes of this problem are complex. Here, we investigated whether altered electrolyte transport contributes to diabetic diarrhea. We found that the expression of Na+/H+ exchanger NHE3 and several scaffold proteins, including NHE3 regulatory factors (NHERFs), inositol trisphosphate (IP₃) receptor-binding protein released with IP₃ (IRBIT), and ezrin, was decreased in the intestinal brush border membrane (BBM) of mice with streptozotocin-induced diabetes. Treatment of diabetic mice with insulin restored intestinal NHE3 activity and fluid absorption. Molecular analysis revealed that NHE3, NHERF1, IRBIT, and ezrin form macrocomplexes, which are perturbed under diabetic conditions, and insulin administration reconstituted these macrocomplexes and restored NHE3 expression in the BBM. Silencing of NHERF1 or IRBIT prevented NHE3 trafficking to the BBM and insulin-dependent NHE3 activation. IRBIT facilitated the interaction of NHE3 with NHERF1 via protein kinase D2-dependent phosphorylation. Insulin stimulated ezrin phosphorylation, which enhanced the interaction of ezrin with NHERF1, IRBIT, and NHE3. Additionally, oral administration of lysophosphatidic acid (LPA) increased NHE3 activity and fluid absorption in diabetic mice via an insulin-independent pathway. Together, these findings indicate the importance of NHE3 in diabetic diarrhea and suggest LPA administration as a potential therapeutic strategy for management of diabetic diarrhea.
Collapse
|
539
|
LPA Promotes T Cell Recruitment through Synthesis of CXCL13. Mediators Inflamm 2015; 2015:248492. [PMID: 26339130 PMCID: PMC4539179 DOI: 10.1155/2015/248492] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 02/05/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid playing an important role in various inflammatory diseases by inducing expression and secretion of many inflammatory cytokines/chemokines. Here we report in a murine air pouch model of inflammation that LPA induced CXCL13 secretion in a time-dependent manner and with exacerbation of the response when LPA was administered after a pretreatment with TNF-α, a key inflammatory cytokine. LPA mediates recruitment of leukocytes, including that of CD3+ cells into unprimed and TNF-α-primed air pouches. CXCL13 neutralization using a blocking antibody injected into air pouches prior to administration of LPA into TNF-α-primed air pouches decreased CD3+ cell influx. Our data highlight that LPA-mediated CXCL13 secretion plays a role in T cell recruitment and participates in regulation of the inflammatory response.
Collapse
|
540
|
Mouratis MA, Magkrioti C, Oikonomou N, Katsifa A, Prestwich GD, Kaffe E, Aidinis V. Autotaxin and Endotoxin-Induced Acute Lung Injury. PLoS One 2015. [PMID: 26196781 PMCID: PMC4509763 DOI: 10.1371/journal.pone.0133619] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Acute Lung Injury (ALI) is a life-threatening, diffuse heterogeneous lung injury characterized by acute onset, pulmonary edema and respiratory failure. Lipopolysaccharide (LPS) is a common cause of both direct and indirect lung injury and when administered to a mouse induces a lung phenotype exhibiting some of the clinical characteristics of human ALI. Here, we report that LPS inhalation in mice results in increased bronchoalveolar lavage fluid (BALF) levels of Autotaxin (ATX, Enpp2), a lysophospholipase D largely responsible for the conversion of lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA) in biological fluids and chronically inflamed sites. In agreement, gradual increases were also detected in BALF LPA levels, following inflammation and pulmonary edema. However, genetic or pharmacologic targeting of ATX had minor effects in ALI severity, suggesting no major involvement of the ATX/LPA axis in acute inflammation. Moreover, systemic, chronic exposure to increased ATX/LPA levels was shown to predispose to and/or to promote acute inflammation and ALI unlike chronic inflammatory pathophysiological situations, further suggesting a differential involvement of the ATX/LPA axis in acute versus chronic pulmonary inflammation.
Collapse
Affiliation(s)
- Marios-Angelos Mouratis
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Christiana Magkrioti
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Nikos Oikonomou
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Aggeliki Katsifa
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Glenn D. Prestwich
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Eleanna Kaffe
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
- * E-mail:
| |
Collapse
|
541
|
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that is present in all tissues examined to date. LPA signals extracellularly via cognate G protein-coupled receptors to mediate cellular processes such as survival, proliferation, differentiation, migration, adhesion and morphology. These LPA-influenced processes impact many aspects of organismal development. In particular, LPA signalling has been shown to affect fertility and reproduction, formation of the nervous system, and development of the vasculature. Here and in the accompanying poster, we review the developmentally related features of LPA signalling.
Collapse
Affiliation(s)
- Xiaoyan Sheng
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yun C Yung
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Allison Chen
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
542
|
Xu K, Ma L, Li Y, Wang F, Zheng GY, Sun Z, Jiang F, Chen Y, Liu H, Dang A, Chen X, Chun J, Tian XL. Genetic and Functional Evidence Supports LPAR1 as a Susceptibility Gene for Hypertension. Hypertension 2015; 66:641-6. [PMID: 26123684 DOI: 10.1161/hypertensionaha.115.05515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/08/2015] [Indexed: 01/11/2023]
Abstract
Essential hypertension is a complex disease affected by genetic and environmental factors and serves as a major risk factor for cardiovascular diseases. Serum lysophosphatidic acid correlates with an elevated blood pressure in rats, and lysophosphatidic acid interacts with 6 subtypes of receptors. In this study, we assessed the genetic association of lysophosphatidic acid receptors with essential hypertension by genotyping 28 single-nucleotide polymorphisms from genes encoding for lysophosphatidic acid receptors, LPAR1, LPAR2, LPAR3, LPAR4, LPAR5, and LPAR6 and their flanking sequences, in 3 Han Chinese cohorts consisting of 2630 patients and 3171 controls in total. We identified a single-nucleotide polymorphism, rs531003 in the 3'-flanking genomic region of LPAR1, associated with hypertension (the Bonferroni corrected P=1.09×10(-5), odds ratio [95% confidence interval]=1.23 [1.13-1.33]). The risk allele C of rs531003 is associated with the increased expression of LPAR1 and the susceptibility of hypertension, particularly in those with a shortage of sleep (P=4.73×10(-5), odds ratio [95% confidence interval]=1.75 [1.34-2.28]). We further demonstrated that blood pressure elevation caused by sleep deprivation and phenylephrine-induced vasoconstriction was both diminished in LPAR1-deficient mice. Together, we show that LPAR1 is a novel susceptibility gene for human essential hypertension and that stress, such as shortage of sleep, increases the susceptibility of patients with risk allele to essential hypertension.
Collapse
Affiliation(s)
- Ke Xu
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Lu Ma
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Yang Li
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Fang Wang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Gu-Yan Zheng
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Zhijun Sun
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Feng Jiang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Yundai Chen
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Huirong Liu
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Aimin Dang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Xi Chen
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Jerold Chun
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Xiao-Li Tian
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.).
| |
Collapse
|
543
|
Igarashi H, Akahoshi N, Ohto-Nakanishi T, Yasuda D, Ishii S. The lysophosphatidic acid receptor LPA4 regulates hematopoiesis-supporting activity of bone marrow stromal cells. Sci Rep 2015; 5:11410. [PMID: 26090649 PMCID: PMC4473687 DOI: 10.1038/srep11410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/26/2015] [Indexed: 12/23/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a pleiotropic lipid mediator that acts through G protein-coupled receptors (LPA1-6). Although several biological roles of LPA4 are becoming apparent, its role in hematopoiesis has remained unknown. Here, we show a novel regulatory role for LPA4 in hematopoiesis. Lpar4 mRNA was predominantly expressed in mouse bone marrow (BM) PDGFRα+ stromal cells, known as the components of the hematopoietic stem/progenitor cell (HSPC) niche. Compared with wild-type mice, LPA4-deficient mice had reduced HSPC numbers in the BM and spleen and were hypersusceptible to myelosuppression, most likely due to impairments in HSPC recovery and stem cell factor production in the BM. Analysis of reciprocal BM chimeras (LPA4-deficient BM into wild-type recipients and vice versa) indicated that stromal cells likely account for these phenotypes. Consistently, LPA4-deficient BM stromal cells showed downregulated mRNA expression of stem cell factor and tenascin-c in vitro. Taken together, these results suggest a critical and novel role for the LPA/LPA4 axis in regulating BM stromal cells.
Collapse
Affiliation(s)
- Hidemitsu Igarashi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Noriyuki Akahoshi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takayo Ohto-Nakanishi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Daisuke Yasuda
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Satoshi Ishii
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
544
|
Wu C, Huang RT, Kuo CH, Kumar S, Kim CW, Lin YC, Chen YJ, Birukova A, Birukov KG, Dulin NO, Civelek M, Lusis AJ, Loyer X, Tedgui A, Dai G, Jo H, Fang Y. Mechanosensitive PPAP2B Regulates Endothelial Responses to Atherorelevant Hemodynamic Forces. Circ Res 2015; 117:e41-e53. [PMID: 26034042 DOI: 10.1161/circresaha.117.306457] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023]
Abstract
RATIONALE PhosPhatidic Acid Phosphatase type 2B (PPAP2B), an integral membrane protein known as lipid phosphate phosphatase (LPP3) that inactivates lysophosphatidic acid, was implicated in coronary artery disease (CAD) by genome-wide association studies. However, it is unclear whether genome-wide association studies-identified coronary artery disease genes, including PPAP2B, participate in mechanotransduction mechanisms by which vascular endothelia respond to local atherorelevant hemodynamics that contribute to the regional nature of atherosclerosis. OBJECTIVE To establish the critical role of PPAP2B in endothelial responses to hemodynamics. METHODS AND RESULTS Reduced PPAP2B was detected in vivo in mouse and swine aortic arch (AA) endothelia exposed to chronic disturbed flow, and in mouse carotid artery endothelia subjected to surgically induced acute disturbed flow. In humans, PPAP2B was reduced in the downstream part of carotid plaques where low shear stress prevails. In culture, reduced PPAP2B was measured in human aortic endothelial cells under atherosusceptible waveform mimicking flow in human carotid sinus. Flow-sensitive microRNA-92a and transcription factor KLF2 were identified as upstream inhibitor and activator of endothelial PPAP2B, respectively. PPAP2B suppression abrogated atheroprotection of unidirectional flow; inhibition of lysophosphatidic acid receptor 1 restored the flow-dependent, anti-inflammatory phenotype in PPAP2B-deficient cells. PPAP2B inhibition resulted in myosin light-chain phosphorylation and intercellular gaps, which were abolished by lysophosphatidic acid receptor 1/2 inhibition. Expression quantitative trait locus mapping demonstrated PPAP2B coronary artery disease risk allele is not linked to PPAP2B expression in various human tissues but significantly associated with reduced PPAP2B in human aortic endothelial cells. CONCLUSIONS Atherorelevant flows dynamically modulate endothelial PPAP2B expression through miR-92a and KLF2. Mechanosensitive PPAP2B plays a critical role in promoting anti-inflammatory phenotype and maintaining vascular integrity of endothelial monolayer under atheroprotective flow.
Collapse
Affiliation(s)
- Congqing Wu
- Department of Medicine, University of Chicago, Los Angeles
| | - Ru-Ting Huang
- Department of Medicine, University of Chicago, Los Angeles
| | | | - Sandeep Kumar
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Chan Woo Kim
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Yen-Chen Lin
- Department of Medicine, University of Chicago, Los Angeles
| | - Yen-Ju Chen
- Department of Medicine, University of Chicago, Los Angeles
| | - Anna Birukova
- Department of Medicine, University of Chicago, Los Angeles
| | | | | | - Mete Civelek
- Department of Medicine, University of California, Los Angeles
| | - Aldons J Lusis
- Department of Medicine, University of California, Los Angeles
| | - Xavier Loyer
- Paris-Cardiovascular Research Center, University Paris Descartes
| | - Alain Tedgui
- Paris-Cardiovascular Research Center, University Paris Descartes
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | - Hanjoong Jo
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Yun Fang
- Department of Medicine, University of Chicago, Los Angeles
| |
Collapse
|
545
|
Fukushima N, Ishii S, Tsujiuchi T, Kagawa N, Katoh K. Comparative analyses of lysophosphatidic acid receptor-mediated signaling. Cell Mol Life Sci 2015; 72:2377-94. [PMID: 25732591 PMCID: PMC11113652 DOI: 10.1007/s00018-015-1872-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/16/2015] [Accepted: 02/23/2015] [Indexed: 12/14/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid mediator that activates G protein-coupled LPA receptors to exert fundamental cellular functions. Six LPA receptor genes have been identified in vertebrates and are classified into two subfamilies, the endothelial differentiation genes (edg) and the non-edg family. Studies using genetically engineered mice, frogs, and zebrafish have demonstrated that LPA receptor-mediated signaling has biological, developmental, and pathophysiological functions. Computational analyses have also identified several amino acids (aa) critical for LPA recognition by human LPA receptors. This review focuses on the evolutionary aspects of LPA receptor-mediated signaling by comparing the aa sequences of vertebrate LPA receptors and LPA-producing enzymes; it also summarizes the LPA receptor-dependent effects commonly observed in mouse, frog, and fish.
Collapse
Affiliation(s)
- Nobuyuki Fukushima
- Division of Molecular Neurobiology, Department of Life Science, Kinki University, Higashiosaka, 577-8502 Japan
| | - Shoichi Ishii
- Division of Molecular Neurobiology, Department of Life Science, Kinki University, Higashiosaka, 577-8502 Japan
| | - Toshifumi Tsujiuchi
- Division of Cancer Biology and Bioinformatics, Department of Life Science, Kinki University, Higashiosaka, Japan
| | - Nao Kagawa
- Division of Animal Genetics, Department of Life Science, Kinki University, Higashiosaka, Japan
| | - Kazutaka Katoh
- Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871 Japan
- Computational Biology Research Center, The National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| |
Collapse
|
546
|
Khan M, Shunmugavel A, Dhammu TS, Matsuda F, Singh AK, Singh I. Oral administration of cytosolic PLA2 inhibitor arachidonyl trifluoromethyl ketone ameliorates cauda equina compression injury in rats. J Neuroinflammation 2015; 12:94. [PMID: 25971887 PMCID: PMC4436116 DOI: 10.1186/s12974-015-0311-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Phospholipase A2 (PLA2)-derived proinflammatory lipid mediators such as prostaglandin E2 (PGE2), leukotrienes B4 (LTB4), lysophosphatidylcholine (LPC), and free fatty acids (FFA) are implicated in spinal cord injury (SCI) pathologies. Reducing the levels of these injurious bioactive lipid mediators is reported to ameliorate SCI. However, the specific role of the group IVA isoform of PLA2 cytosolic PLA2 (cPLA2) in lumbar spinal canal stenosis (LSS) due to cauda equina compression (CEC) injury is not clear. In this study, we investigated the role of cPLA2 in a rat model of CEC using a non-toxic cPLA2-preferential inhibitor, arachidonyl trifluoromethyl ketone (ATK). METHODS LSS was induced in adult female rats by CEC procedure using silicone blocks within the epidural spaces of L4 to L6 vertebrae. cPLA2 inhibitor ATK (7.5 mg/kg) was administered by oral gavage at 2 h following the CEC. cPLA2-derived injurious lipid mediators and the expression/activity of cPLA2, 5-lipoxygenase (5-LOX), and cyclooxygenase-2 (COX-2) were assessed. ATK-treated (CEC + ATK) were compared with vehicle-treated (CEC + VEH) animals in terms of myelin levels, pain threshold, and motor function. RESULTS ATK treatment of CEC animals reduced the phosphorylation of cPLA2 (pcPLA2) determined by Western blot, improved locomotor function evaluated by rotarod task, and reduced pain threshold evaluated by mechanical hyperalgesia method. Levels of FFA and LPC, along with PGE2 and LTB4, were reduced in CEC + ATK compared with CEC + VEH group. However, ATK treatment reduced neither the activity/expression of 5-LOX nor the expression of COX-2 in CEC + VEH animals. Increased cPLA2 activity in the spinal cord from CEC + VEH animals correlated well with decreased spinal cord as well as cauda equina fiber myelin levels, which were restored after ATK treatment. CONCLUSION The data indicate that cPLA2 activity plays a significant role in tissue injury and pain after LSS. Reducing the levels of proinflammatory and tissue damaging eicosanoids and the deleterious lipid mediator LPC shows therapeutic potential. ATK inhibits cPLA2 activity, thereby decreasing the levels of injurious lipid mediators, reducing pain, improving functional deficits, and conferring protection against LSS injury. Thus, it shows potential for preclinical evaluation in LSS.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | | | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Fumiyo Matsuda
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA. .,School of Health Science, Kagoshima University, Kagoshima, Japan.
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA. .,Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
547
|
González de San Román E, Manuel I, Giralt MT, Chun J, Estivill-Torrús G, Rodríguez de Fonseca F, Santín LJ, Ferrer I, Rodríguez-Puertas R. Anatomical location of LPA1 activation and LPA phospholipid precursors in rodent and human brain. J Neurochem 2015; 134:471-85. [PMID: 25857358 DOI: 10.1111/jnc.13112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/02/2015] [Accepted: 03/31/2015] [Indexed: 12/29/2022]
Abstract
Lysophosphatidic acid (LPA) is a signaling molecule that binds to six known G protein-coupled receptors: LPA1 -LPA6 . LPA evokes several responses in the CNS, including cortical development and folding, growth of the axonal cone and its retraction process. Those cell processes involve survival, migration, adhesion proliferation, differentiation, and myelination. The anatomical localization of LPA1 is incompletely understood, particularly with regard to LPA binding. Therefore, we have used functional [(35) S]GTPγS autoradiography to verify the anatomical distribution of LPA1 binding sites in adult rodent and human brain. The greatest activity was observed in myelinated areas of the white matter such as corpus callosum, internal capsule and cerebellum. MaLPA1 -null mice (a variant of LPA1 -null) lack [(35) S]GTPγS basal binding in white matter areas, where the LPA1 receptor is expressed at high levels, suggesting a relevant role of the activity of this receptor in the most myelinated brain areas. In addition, phospholipid precursors of LPA were localized by MALDI-IMS in both rodent and human brain slices identifying numerous species of phosphatides and phosphatidylcholines. Both phosphatides and phosphatidylcholines species represent potential LPA precursors. The anatomical distribution of these precursors in rodent and human brain may indicate a metabolic relationship between LPA and LPA1 receptors. Lysophosphatidic acid (LPA) is a signaling molecule that binds to six known G protein-coupled receptors (GPCR), LPA1 to LPA6 . LPA evokes several responses in the central nervous system (CNS), including cortical development and folding, growth of the axonal cone and its retraction process. We used functional [(35) S]GTPγS autoradiography to verify the anatomical distribution of LPA1 -binding sites in adult rodent and human brain. The distribution of LPA1 receptors in rat, mouse and human brains show the highest activity in white matter myelinated areas. The basal and LPA-evoked activities are abolished in MaLPA1 -null mice. The phospholipid precursors of LPA are localized by MALDI-IMS. The anatomical distribution of LPA precursors in rodent and human brain suggests a relationship with functional LPA1 receptors.
Collapse
Affiliation(s)
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - María Teresa Giralt
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| | - Guillermo Estivill-Torrús
- UGC Intercentros de Neurociencias y UGC de Salud Mental, Instituto de Investigación Biomédica de Malaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Universidad de Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- UGC Intercentros de Neurociencias y UGC de Salud Mental, Instituto de Investigación Biomédica de Malaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Universidad de Málaga, Spain
| | - Luis Javier Santín
- Departmento de Psicobiología y Metodología de las Ciencias del Comportamiento. Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad of Málaga, Málaga, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, University Hospital Bellvitge, University of Barcelona, Ciberned, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| |
Collapse
|
548
|
Abstract
The brain is composed of many lipids with varied forms that serve not only as structural components but also as essential signaling molecules. Lysophosphatidic acid (LPA) is an important bioactive lipid species that is part of the lysophospholipid (LP) family. LPA is primarily derived from membrane phospholipids and signals through six cognate G protein-coupled receptors (GPCRs), LPA1-6. These receptors are expressed on most cell types within central and peripheral nervous tissues and have been functionally linked to many neural processes and pathways. This Review covers a current understanding of LPA signaling in the nervous system, with particular focus on the relevance of LPA to both physiological and diseased states.
Collapse
Affiliation(s)
- Yun C Yung
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole C Stoddard
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine, La Jolla, CA 92037, USA
| | - Hope Mirendil
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
549
|
Ishii S, Hirane M, Fukushima K, Tomimatsu A, Fukushima N, Tsujiuchi T. Diverse effects of LPA4, LPA5 and LPA6 on the activation of tumor progression in pancreatic cancer cells. Biochem Biophys Res Commun 2015; 461:59-64. [PMID: 25849892 DOI: 10.1016/j.bbrc.2015.03.169] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/29/2015] [Indexed: 01/05/2023]
Abstract
Lysophosphatidic acid (LPA) is an extracellular biological lipid which interacts with G protein-coupled LPA receptors (LPA1 to LPA6). LPA signaling via LPA receptors mediates several cellular responses. In the present study, to assess the roles of LPA4, LPA5 and LPA6 in cellular functions of pancreatic cancer cells, we generated LPA receptor knockdown cells from PANC-1 cells (PANC-sh4, PANC-sh5 and PANC-sh6 cells, respectively). In cell motility assay, PANC-sh4 and PANC-sh5 cells enhanced the cell motile activities, compared with control cells. In contrast, the cell motile activity of PANC-sh6 cells was suppressed. The invasive activities of PANC-sh4 and PANC-sh5 cells were markedly stimulated, while PANC-sh6 cells showed the low invasive activity. In colony assay, PANC-sh4 and PANC-sh5 cells formed the large sized colonies, but not PANC-sh6 cells. When endothelial cells were incubated with supernatants from PANC-sh4 and PANC-sh5 cells, the cell motility and tube formation of endothelial cells were significantly induced, but not PANC-sh6 cells. These results suggest that the diverse roles of LPA4, LPA5 and LPA6 are involved in the activation of tumor progression in pancreatic cancer cells.
Collapse
Affiliation(s)
- Shuhei Ishii
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Miku Hirane
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Kaori Fukushima
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Ayaka Tomimatsu
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Nobuyuki Fukushima
- Division of Molecular Neurobiology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Toshifumi Tsujiuchi
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kinki University, 3-4-1, Kowakae, Higashiosaka, Osaka 577-8502, Japan.
| |
Collapse
|
550
|
Barbayianni E, Kaffe E, Aidinis V, Kokotos G. Autotaxin, a secreted lysophospholipase D, as a promising therapeutic target in chronic inflammation and cancer. Prog Lipid Res 2015; 58:76-96. [DOI: 10.1016/j.plipres.2015.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/20/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
|