Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Trends Cancer ; 10(4): 283-285, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38494373

ABSTRACT

Secreted autotaxin (ATX) promotes tumor progression by producing the pleiotropic lipid mediator lysophosphatidic acid (LPA). In a recent Nature Cancer paper, Bhattacharyya et al. show that ATX/LPA signaling suppresses CCL11-driven infiltration of eosinophils into the pancreatic tumor microenvironment to facilitate tumor progression, thus revealing a new ATX-mediated immune escape mechanism and highlighting the antitumor potential of eosinophils.


Subject(s)
Neoplasms , Tumor Escape , Humans , Eosinophils , Tumor Microenvironment
2.
J Lipid Res ; 64(5): 100370, 2023 05.
Article in English | MEDLINE | ID: mdl-37059333

ABSTRACT

Conversion of lysophosphatidylcholine to lysophosphatidic acid (LPA) by autotaxin, a secreted phospholipase D, is a major pathway for producing LPA. We previously reported that feeding Ldlr-/- mice standard mouse chow supplemented with unsaturated LPA or lysophosphatidylcholine qualitatively mimicked the dyslipidemia and atherosclerosis induced by feeding a Western diet (WD). Here, we report that adding unsaturated LPA to standard mouse chow also increased the content of reactive oxygen species and oxidized phospholipids (OxPLs) in jejunum mucus. To determine the role of intestinal autotaxin, enterocyte-specific Ldlr-/-/Enpp2 KO (intestinal KO) mice were generated. In control mice, the WD increased enterocyte Enpp2 expression and raised autotaxin levels. Ex vivo, addition of OxPL to jejunum from Ldlr-/- mice on a chow diet induced expression of Enpp2. In control mice, the WD raised OxPL levels in jejunum mucus and decreased gene expression in enterocytes for a number of peptides and proteins that affect antimicrobial activity. On the WD, the control mice developed elevated levels of lipopolysaccharide in jejunum mucus and plasma, with increased dyslipidemia and increased atherosclerosis. All these changes were reduced in the intestinal KO mice. We conclude that the WD increases the formation of intestinal OxPL, which i) induce enterocyte Enpp2 and autotaxin resulting in higher enterocyte LPA levels; that ii) contribute to the formation of reactive oxygen species that help to maintain the high OxPL levels; iii) decrease intestinal antimicrobial activity; and iv) raise plasma lipopolysaccharide levels that promote systemic inflammation and enhance atherosclerosis.


Subject(s)
Anti-Infective Agents , Atherosclerosis , Dyslipidemias , Mice , Animals , Lysophosphatidylcholines , Enterocytes/metabolism , Lipopolysaccharides , Reactive Oxygen Species , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Diet, Western , Inflammation/genetics , Dyslipidemias/metabolism , Atherosclerosis/genetics
3.
Cell Chem Biol ; 30(1): 69-84.e14, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36640760

ABSTRACT

Autotaxin (ATX; ENPP2) produces the lipid mediator lysophosphatidic acid (LPA) that signals through disparate EDG (LPA1-3) and P2Y (LPA4-6) G protein-coupled receptors. ATX/LPA promotes several (patho)physiological processes, including in pulmonary fibrosis, thus serving as an attractive drug target. However, it remains unclear if clinical outcome depends on how different types of ATX inhibitors modulate the ATX/LPA signaling axis. Here, we show that the ATX "tunnel" is crucial for conferring key aspects of ATX/LPA signaling and dictates cellular responses independent of ATX catalytic activity, with a preference for activation of P2Y LPA receptors. The efficacy of the ATX/LPA signaling responses are abrogated more efficiently by tunnel-binding inhibitors, such as ziritaxestat (GLPG1690), compared with inhibitors that exclusively target the active site, as shown in primary lung fibroblasts and a murine model of radiation-induced pulmonary fibrosis. Our results uncover a receptor-selective signaling mechanism for ATX, implying clinical benefit for tunnel-targeting ATX inhibitors.


Subject(s)
Pulmonary Fibrosis , Mice , Animals , Pulmonary Fibrosis/drug therapy , Receptors, Lysophosphatidic Acid , Signal Transduction , Lysophospholipids/chemistry , Fibroblasts
4.
J Biol Chem ; 298(2): 101526, 2022 02.
Article in English | MEDLINE | ID: mdl-34958798

ABSTRACT

Ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP) family members (ENPP1-7) have been implicated in key biological and pathophysiological processes, including nucleotide and phospholipid signaling, bone mineralization, fibrotic diseases, and tumor-associated immune cell infiltration. ENPPs are single-pass transmembrane ecto-enzymes, with notable exceptions of ENPP2 (Autotaxin) and ENNP6, which are secreted and glycosylphosphatidylinositol (GPI)-anchored, respectively. ENNP1 and ENNP2 are the best characterized and functionally the most interesting members. Here, we review the structural features of ENPP1-7 to understand how they evolved to accommodate specific substrates and mediate different biological activities. ENPPs are defined by a conserved phosphodiesterase (PDE) domain. In ENPP1-3, the PDE domain is flanked by two N-terminal somatomedin B-like domains and a C-terminal inactive nuclease domain that confers structural stability, whereas ENPP4-7 only possess the PDE domain. Structural differences in the substrate-binding site endow each protein with unique characteristics. Thus, ENPP1, ENPP3, ENPP4, and ENPP5 hydrolyze nucleotides, whereas ENPP2, ENPP6, and ENNP7 evolved as phospholipases through adaptions in the catalytic domain. These adaptations explain the different biological and pathophysiological functions of individual members. Understanding the ENPP members as a whole advances our insights into common mechanisms, highlights their functional diversity, and helps to explore new biological roles.


Subject(s)
Phosphoric Diester Hydrolases , Pyrophosphatases , Catalytic Domain , Nucleotides/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Signal Transduction , Structure-Activity Relationship
5.
Cell Rep ; 37(7): 110013, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34788605

ABSTRACT

Autotaxin (ATX; ENPP2) produces lysophosphatidic acid (LPA) that regulates multiple biological functions via cognate G protein-coupled receptors LPAR1-6. ATX/LPA promotes tumor cell migration and metastasis via LPAR1 and T cell motility via LPAR2, yet its actions in the tumor immune microenvironment remain unclear. Here, we show that ATX secreted by melanoma cells is chemorepulsive for tumor-infiltrating lymphocytes (TILs) and circulating CD8+ T cells ex vivo, with ATX functioning as an LPA-producing chaperone. Mechanistically, T cell repulsion predominantly involves Gα12/13-coupled LPAR6. Upon anti-cancer vaccination of tumor-bearing mice, ATX does not affect the induction of systemic T cell responses but, importantly, suppresses tumor infiltration of cytotoxic CD8+ T cells and thereby impairs tumor regression. Moreover, single-cell data from melanoma tumors are consistent with intratumoral ATX acting as a T cell repellent. These findings highlight an unexpected role for the pro-metastatic ATX-LPAR axis in suppressing CD8+ T cell infiltration to impede anti-tumor immunity, suggesting new therapeutic opportunities.


Subject(s)
Lymphocytes, Tumor-Infiltrating/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemotaxis/physiology , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Neoplasms , Phosphoric Diester Hydrolases/physiology , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/physiology , Tumor Microenvironment
6.
J Cell Sci ; 133(3)2020 02 10.
Article in English | MEDLINE | ID: mdl-31932507

ABSTRACT

GDE2 (also known as GDPD5) is a multispanning membrane phosphodiesterase with phospholipase D-like activity that cleaves select glycosylphosphatidylinositol (GPI)-anchored proteins and thereby promotes neuronal differentiation both in vitro and in vivo GDE2 is a prognostic marker in neuroblastoma, while loss of GDE2 leads to progressive neurodegeneration in mice; however, its regulation remains unclear. Here, we report that, in immature neuronal cells, GDE2 undergoes constitutive endocytosis and travels back along both fast and slow recycling routes. GDE2 trafficking is directed by C-terminal tail sequences that determine the ability of GDE2 to cleave GPI-anchored glypican-6 (GPC6) and induce a neuronal differentiation program. Specifically, we define a GDE2 truncation mutant that shows aberrant recycling and is dysfunctional, whereas a consecutive deletion results in cell-surface retention and gain of GDE2 function, thus uncovering distinctive regulatory sequences. Moreover, we identify a C-terminal leucine residue in a unique motif that is essential for GDE2 internalization. These findings establish a mechanistic link between GDE2 neuronal function and sequence-dependent trafficking, a crucial process gone awry in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Neuroblastoma , Phospholipases , Animals , Cell Differentiation/genetics , Glycosylphosphatidylinositols/genetics , Mice , Phosphoric Diester Hydrolases/genetics
8.
FASEB J ; 33(3): 3623-3635, 2019 03.
Article in English | MEDLINE | ID: mdl-30481488

ABSTRACT

Autotaxin (ATX or ENPP2) is a secreted lysophospholipase D that produces lysophosphatidic acid (LPA), a pleiotropic lipid mediator acting on specific GPCRs. ATX and LPA have been implicated in key (patho)physiologic processes, including embryonic development, lymphocyte homing, inflammation, and cancer progression. Using LPA receptor knockout mice, we previously uncovered a role for LPA signaling in promoting colitis and colorectal cancer. Here, we examined the role of ATX in experimental colitis through inducible deletion of Enpp2 in adult mice. ATX expression was increased upon induction of colitis, whereas ATX deletion reduced the severity of inflammation in both acute and chronic colitis, accompanied by transient weight loss. ATX expression in lymphocytes was strongly reduced in Rag1-/- and µMT mice, suggesting B cells as a major ATX-producing source, which was validated by immunofluorescence and biochemical analyses. ATX secretion by B cells from control, but not Enpp2 knockout, mice led to ERK activation in colorectal cancer cells and promoted T cell migration. We conclude that ATX deletion suppresses experimental colitis and that B cells are a major source of ATX in the colon. Our study suggests that pharmacological inhibition of ATX could be a therapeutic strategy in colitis.-Lin, S., Haque, A., Raeman, R., Guo, L., He, P., Denning, T. L., El-Rayes, B., Moolenaar, W. H., Yun, C. C. Autotaxin determines colitis severity in mice and is secreted by B cells in the colon.


Subject(s)
B-Lymphocytes/metabolism , Colitis/metabolism , Colon/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , HCT116 Cells , Humans , Inflammation/metabolism , Lymphocytes/metabolism , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/physiology
9.
J Biol Chem ; 293(50): 19161-19176, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30381396

ABSTRACT

Chloride intracellular channel 4 (CLIC4) is a cytosolic protein implicated in diverse actin-based processes, including integrin trafficking, cell adhesion, and tubulogenesis. CLIC4 is rapidly recruited to the plasma membrane by RhoA-activating agonists and then partly colocalizes with ß1 integrins. Agonist-induced CLIC4 translocation depends on actin polymerization and requires conserved residues that make up a putative binding groove. However, the mechanism and significance of CLIC4 trafficking have been elusive. Here, we show that RhoA activation by either lysophosphatidic acid (LPA) or epidermal growth factor is necessary and sufficient for CLIC4 translocation to the plasma membrane and involves regulation by the RhoA effector mDia2, a driver of actin polymerization and filopodium formation. We found that CLIC4 binds the G-actin-binding protein profilin-1 via the same residues that are required for CLIC4 trafficking. Consistently, shRNA-induced profilin-1 silencing impaired agonist-induced CLIC4 trafficking and the formation of mDia2-dependent filopodia. Conversely, CLIC4 knockdown increased filopodium formation in an integrin-dependent manner, a phenotype rescued by wild-type CLIC4 but not by the trafficking-incompetent mutant CLIC4(C35A). Furthermore, CLIC4 accelerated LPA-induced filopodium retraction. We conclude that through profilin-1 binding, CLIC4 functions in a RhoA-mDia2-regulated signaling network to integrate cortical actin assembly and membrane protrusion. We propose that agonist-induced CLIC4 translocation provides a feedback mechanism that counteracts formin-driven filopodium formation.


Subject(s)
Carrier Proteins/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Profilins/metabolism , Pseudopodia/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Cell Membrane/metabolism , Chloride Channels/chemistry , Conserved Sequence , Crystallography, X-Ray , Enzyme Activation , Formins , HeLa Cells , Humans , Integrins/metabolism , Models, Molecular , Profilins/chemistry , Protein Binding , Protein Conformation , Protein Transport
10.
Int J Biochem Cell Biol ; 94: 71-78, 2018 01.
Article in English | MEDLINE | ID: mdl-29203233

ABSTRACT

Notch signaling plays an essential role in the proliferation, differentiation and cell fate determination of various tissues, including the developing pancreas. One regulator of the Notch pathway is GDE2 (or GDPD5), a transmembrane ecto-phosphodiesterase that cleaves GPI-anchored proteins at the plasma membrane, including a Notch ligand regulator. Here we report that Gdpd5-knockdown in zebrafish embryos leads to developmental defects, particularly, impaired motility and reduced pancreas differentiation, as shown by decreased expression of insulin and other pancreatic markers. Exogenous expression of human GDE2, but not catalytically dead GDE2, similarly leads to developmental defects. Human GDE2 restores insulin expression in Gdpd5a-depleted zebrafish embryos. Importantly, zebrafish Gdpd5 orthologues localize to the plasma membrane where they show catalytic activity against GPI-anchored GPC6. Thus, our data reveal functional conservation between zebrafish Gdpd5 and human GDE2, and suggest that strict regulation of GDE2 expression and catalytic activity is critical for correct embryonic patterning. In particular, our data uncover a role for GDE2 in regulating pancreas differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Organogenesis , Pancreas/metabolism , Phosphoric Diester Hydrolases/metabolism , Zebrafish Proteins/metabolism , Animals , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/diagnostic imaging , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Morpholinos/metabolism , Pancreas/diagnostic imaging , Pancreas/embryology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Phylogeny , Protein Domains , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
11.
Elife ; 62017 08 29.
Article in English | MEDLINE | ID: mdl-28849762

ABSTRACT

The urokinase receptor (uPAR) is a glycosylphosphatidylinositol (GPI)-anchored protein that promotes tissue remodeling, tumor cell adhesion, migration and invasion. uPAR mediates degradation of the extracellular matrix through protease recruitment and enhances cell adhesion, migration and signaling through vitronectin binding and interactions with integrins. Full-length uPAR is released from the cell surface, but the mechanism and significance of uPAR shedding remain obscure. Here we identify transmembrane glycerophosphodiesterase GDE3 as a GPI-specific phospholipase C that cleaves and releases uPAR with consequent loss of function, whereas its homologue GDE2 fails to attack uPAR. GDE3 overexpression depletes uPAR from distinct basolateral membrane domains in breast cancer cells, resulting in a less transformed phenotype, it slows tumor growth in a xenograft model and correlates with prolonged survival in patients. Our results establish GDE3 as a negative regulator of the uPAR signaling network and, furthermore, highlight GPI-anchor hydrolysis as a cell-intrinsic mechanism to alter cell behavior.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Phosphoric Diester Hydrolases/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Clustered Regularly Interspaced Short Palindromic Repeats , Female , Gene Knockout Techniques/methods , HEK293 Cells , Humans , Hydrolysis , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Phosphoric Diester Hydrolases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Tumor Burden , Vitronectin/genetics , Vitronectin/metabolism
13.
Oncotarget ; 8(4): 5672-5673, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28030823
15.
J Cell Sci ; 129(22): 4165-4174, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27852828

ABSTRACT

Cl- intracellular channels (CLICs) are a family of six evolutionary conserved cytosolic proteins that exist in both soluble and membrane-associated forms; however, their functions have long been elusive. Soluble CLICs adopt a glutathione S-transferase (GST)-fold, can induce ion currents in artificial membranes and show oxidoreductase activity in vitro, but there is no convincing evidence of CLICs having such activities in vivo. Recent studies have revealed a role for CLIC proteins in Rho-regulated cortical actin dynamics as well as vesicular trafficking and integrin recycling, the latter of which are under the control of Rab GTPases. In this Commentary, we discuss the emerging roles of CLIC proteins in these processes and the lessons learned from gene-targeting studies. We also highlight outstanding questions regarding the molecular function(s) of these important but still poorly understood proteins.


Subject(s)
Chloride Channels/metabolism , Intracellular Space/metabolism , Animals , Cell Membrane/metabolism , Endosomes/metabolism , Humans , Protein Transport
16.
Cancer Cell ; 30(4): 548-562, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27693046

ABSTRACT

Neuroblastoma is a pediatric embryonal malignancy characterized by impaired neuronal differentiation. A better understanding of neuroblastoma differentiation is essential for developing new therapeutic approaches. GDE2 (encoded by GDPD5) is a six-transmembrane-domain glycerophosphodiesterase that promotes embryonic neurogenesis. We find that high GDPD5 expression is strongly associated with favorable outcome in neuroblastoma. GDE2 induces differentiation of neuroblastoma cells, suppresses cell motility, and opposes RhoA-driven neurite retraction. GDE2 alters the Rac-RhoA activity balance and the expression of multiple differentiation-associated genes. Mechanistically, GDE2 acts by cleaving (in cis) and releasing glycosylphosphatidylinositol-anchored glypican-6, a putative co-receptor. A single point mutation in the ectodomain abolishes GDE2 function. Our results reveal GDE2 as a cell-autonomous inducer of neuroblastoma differentiation with prognostic significance and potential therapeutic value.


Subject(s)
Glypicans/metabolism , Neuroblastoma/enzymology , Neuroblastoma/pathology , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Differentiation/physiology , Chickens , Glycosylphosphatidylinositols/metabolism , HEK293 Cells , Humans , Prognosis
17.
Bioorg Med Chem Lett ; 26(22): 5403-5410, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27780639

ABSTRACT

The autotaxin-lysophosphatidic acid (ATX-LPA) axis has been implicated in several disease conditions including inflammation, fibrosis and cancer. This makes ATX an attractive drug target and its inhibition may lead to useful therapeutic agents. Through a high throughput screen (HTS) we identified a series of small molecule inhibitors of ATX which have subsequently been optimized for potency, selectivity and developability properties. This has delivered drug-like compounds such as 9v (CRT0273750) which modulate LPA levels in plasma and are suitable for in vivo studies. X-ray crystallography has revealed that these compounds have an unexpected binding mode in that they do not interact with the active site zinc ions but instead occupy the hydrophobic LPC pocket extending from the active site of ATX together with occupying the LPA 'exit' channel.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Lysophospholipase/antagonists & inhibitors , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/pharmacokinetics , Humans , Lysophospholipase/metabolism , Mice , Molecular Docking Simulation , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology
18.
Nat Commun ; 7: 11248, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27075612

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.


Subject(s)
Bile Acids and Salts/metabolism , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Steroids/metabolism , Animals , Bile Acids and Salts/chemistry , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/metabolism , Kinetics , Lysophospholipids/chemistry , Models, Molecular , Molecular Conformation , Molecular Structure , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Structure, Tertiary , Rats , Receptors, Lysophosphatidic Acid/metabolism , Steroids/chemistry , Taurochenodeoxycholic Acid/chemistry , Taurochenodeoxycholic Acid/metabolism
19.
Sci Rep ; 6: 23433, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27005960

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 ß1-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.


Subject(s)
Cartilage/metabolism , Chondrocytes/cytology , Fibronectins/metabolism , Osteochondrodysplasias/genetics , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cartilage/cytology , Cartilage/pathology , Cell Cycle , Cell Proliferation , Cells, Cultured , Chondrocytes/metabolism , Gene Targeting , Integrin beta1/metabolism , Lysophospholipids/metabolism , Mice , Osteochondrodysplasias/pathology , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
20.
J Biol Chem ; 291(9): 4323-33, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26740622

ABSTRACT

Invadosomes are actin-rich membrane protrusions that degrade the extracellular matrix to drive tumor cell invasion. Key players in invadosome formation are c-Src and Rho family GTPases. Invadosomes can reassemble into circular rosette-like superstructures, but the underlying signaling mechanisms remain obscure. Here we show that Src-induced invadosomes in human melanoma cells (A375M and MDA-MB-435) undergo rapid remodeling into dynamic extracellular matrix-degrading rosettes by distinct G protein-coupled receptor agonists, notably lysophosphatidic acid (LPA; acting through the LPA1 receptor) and endothelin. Agonist-induced rosette formation is blocked by pertussis toxin, dependent on PI3K activity and accompanied by localized production of phosphatidylinositol 3,4,5-trisphosphate, whereas MAPK and Ca(2+) signaling are dispensable. Using FRET-based biosensors, we show that LPA and endothelin transiently activate Cdc42 through Gi, concurrent with a biphasic decrease in Rac activity and differential effects on RhoA. Cdc42 activity is essential for rosette formation, whereas G12/13-mediated RhoA-ROCK signaling suppresses the remodeling process. Our results reveal a Gi-mediated Cdc42 signaling axis by which G protein-coupled receptors trigger invadosome remodeling, the degree of which is dictated by the Cdc42-RhoA activity balance.


Subject(s)
Endothelins/metabolism , Lysophospholipids/metabolism , Melanoma/metabolism , Podosomes/metabolism , Receptors, Lysophosphatidic Acid/agonists , cdc42 GTP-Binding Protein/agonists , rac1 GTP-Binding Protein/metabolism , Biomarkers/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fluorescence Resonance Energy Transfer , Humans , Hydrolysis , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Melanoma/enzymology , Melanoma/pathology , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Podosomes/enzymology , Podosomes/pathology , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time-Lapse Imaging , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/agonists , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL