Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
J Pharmacol Sci ; 140(3): 242-247, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31421954

ABSTRACT

xCT, a well-known cystine transporter, is reported to be involved in the proliferation of various cells, such as cancer cells, immune cells, and fibroblasts. xCT inhibitor is expected to be a promising drug for cancer or immune diseases. However, there are little studies reporting that xCT inhibitors improve disease progression in vivo. To invent potent xCT inhibitors in vivo, we established a new in vivo model for assessing efficacy of xCT inhibition. dl-propargylglycine (PPG) was administered intraperitoneally to wild-type C57BL/6J mice. Concentration of cystathionine, another substrate of xCT, in the thymus and spleen was measured by LC-MS/MS. PPG increased cystathionine amounts in the thymus and spleen in a dose- and time-dependent manner. At 7 h after PPG administration, the efficacy of erastin, a representative xCT inhibitor, was clearly shown. We synthesized a new compound, Compound A, which had much higher inhibitory effect on xCT than erastin both in vitro and in vivo. We established a mouse model of PPG-induced cystathionine accumulation for assessing xCT inhibition in vivo. By using this model, we discovered that Compound A was approximately 15 times more effective in vivo than erastin.


Subject(s)
Alkynes/pharmacology , Amino Acid Transport System y+/antagonists & inhibitors , Glycine/analogs & derivatives , Animals , Cystathionine/metabolism , Female , Glycine/pharmacology , Mice , Mice, Inbred C57BL , Models, Animal , Piperazines/pharmacology , Spleen/drug effects , Spleen/metabolism , Tandem Mass Spectrometry/methods , Thymus Gland/drug effects , Thymus Gland/metabolism
2.
Sci Rep ; 9(1): 2662, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30804442

ABSTRACT

Lysophosphatidic acid (LPA) is a blood-derived bioactive lipid with numerous biological activities exerted mainly through six defined G protein-coupled receptors (LPA1-LPA6). LPA was first identified as a vasoactive compound because it induced transient hypertension when injected intravenously in rodents. Here, we examined the molecular mechanism underlying the LPA-induced hypertensive response. The LPA-induced hypertensive response was significantly attenuated by pretreatment with a Rho kinase inhibitor, which blocks Gα12/13 signaling. Consistent with this, the response was weakened in KO mice of LPA4, a Gα12/13-coupling LPA receptor. KO mice of another Gα12/13-coupling LPA receptor, LPA6, also showed an attenuated LPA-induced hypertensive response. However, LPA6 KO mice also displayed attenuated pressor responses to an adrenergic agent and abnormal blood vessel formation. Using several LPA analogs with varied affinity for each LPA receptor, we found a good correlation between the hypertensive and LPA4 agonistic activities. Incubated mouse plasma, which contained abundant LPA, also induced a hypertensive response. Interestingly the response was completely abolished when the plasma was incubated in the presence of an ATX inhibitor. Together, these results indicate that circulating LPA produced by ATX contributes to the elevation of blood pressure through multiple LPA receptors, mainly LPA4.


Subject(s)
Disease Susceptibility , Hypertension/etiology , Hypertension/metabolism , Lysophospholipids/adverse effects , Animals , Biomarkers , Disease Models, Animal , Fluorescent Antibody Technique , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Hypertension/physiopathology , Mice , Signal Transduction , rho-Associated Kinases/metabolism
3.
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
4.
J Cell Sci ; 128(21): 3871-7, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26345369

ABSTRACT

Lysophosphatidic acid (LPA) is emerging as an angiogenic factor, because knockdown of the enzyme that produces it (autotaxin, also known as ENPP2) and its receptors cause severe developmental vascular defects in both mice and fish. In addition, overexpression of autotaxin in mice causes similar vascular defects, indicating that the extracellular amount of LPA must be tightly regulated. Here, we focused on an LPA-degrading enzyme, lipid phosphate phosphatase 3 (LPP3, also known as PPAP2B), and showed that LPP3 was localized in specific cell-cell contact sites of endothelial cells and suppresses LPA signalling through the LPA6 receptor (also known as LPAR6). In HEK293 cells, overexpression of LPP3 dramatically suppressed activation of LPA6. In human umbilical vein endothelial cells (HUVECs), LPA induced actin stress fibre formation through LPA6, which was substantially upregulated by LPP3 knockdown. LPP3 was localized to cell-cell contact sites and was missing in non-contact sites to which LPA-induced actin stress fibre formation mediated by LPA6 was restricted. Interestingly, the expression of LPP3 in HUVECs was dramatically increased after forskolin treatment in a process involving Notch signalling. These results indicate that LPP3 regulates and localizes LPA signalling in endothelial cells, thereby stabilizing vessels through Notch signalling for proper vasculature.


Subject(s)
Lysophospholipids/metabolism , Phosphatidate Phosphatase/metabolism , Animals , Cell Line , Colforsin/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Phosphatidate Phosphatase/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
PLoS One ; 10(5): e0126734, 2015.
Article in English | MEDLINE | ID: mdl-25992708

ABSTRACT

Autotaxin (ATX) is a secretory protein, which converts lysophospholipids to lysophosphatidic acid (LPA), and is essential for embryonic vascular formation. ATX is abundantly detected in various biological fluids and its level is elevated in some pathophysiological conditions. However, the roles of elevated ATX levels remain to be elucidated. In this study, we generated conditional transgenic (Tg) mice overexpressing ATX and examined the effects of excess LPA signalling. We found that ATX overexpression in the embryonic period caused severe vascular defects and was lethal around E9.5. ATX was conditionally overexpressed in the neonatal period using the Cre/loxP system, which resulted in a marked increase in the plasma LPA level. This resulted in retinal vascular defects including abnormal vascular plexus and increased vascular regression. Our findings indicate that the ATX level must be carefully regulated to ensure coordinated vascular formation.


Subject(s)
Blood Vessels/abnormalities , Genes, Lethal , Phosphoric Diester Hydrolases/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoric Diester Hydrolases/genetics
6.
J Biochem ; 155(4): 235-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24451492

ABSTRACT

Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are second-generation lysophospholipid mediators that exert multiple biological functions through their own cognate receptors. They are both present in the blood stream, activate receptors with similar structures (endothelial differentiation gene receptors), have similar roles in the vasculature and are vasoactive. However, it is unclear whether these lysophospholipid mediators cross-talk downstream of each receptor. Here, we provide in vivo evidence that LPA signaling counteracted S1P signaling. When autotaxin (Atx), an LPA-producing enzyme, was overexpressed in zebrafish embryos by injecting atx mRNA, the embryos showed cardia bifida, a phenotype induced by down-regulation of S1P signaling. A similar cardiac phenotype was not induced when catalytically inactive Atx was introduced. The cardiac phenotype was synergistically enhanced when antisense morpholino oligonucleotides (MO) against S1P receptor (s1pr2/mil) or S1P transporter (spns2) was introduced together with atx mRNA. The Atx-induced cardia bifida was prominently suppressed when embryos were treated with an lpar1 receptor antagonist, Ki16425, or with MO against lpar1. These results provide the first in vivo evidence of cross-talk between LPA and S1P signaling.


Subject(s)
Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/enzymology , Heart Defects, Congenital/embryology , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Sphingosine/analogs & derivatives , Zebrafish/embryology , Animals , Down-Regulation/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , HEK293 Cells , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/pathology , Humans , Isoxazoles/pharmacology , Phenotype , Phosphoric Diester Hydrolases/genetics , Propionates/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects , Sphingosine/metabolism
7.
J Biol Chem ; 286(51): 43972-43983, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-21971049

ABSTRACT

Autotaxin (ATX) is a multifunctional ecto-type phosphodiesterase that converts lysophospholipids, such as lysophosphatidylcholine, to lysophosphatidic acid (LPA) by its lysophospholipase D activity. LPA is a lipid mediator with diverse biological functions, most of which are mediated by G protein-coupled receptors specific to LPA (LPA1-6). Recent studies on ATX knock-out mice revealed that ATX has an essential role in embryonic blood vessel formation. However, the underlying molecular mechanisms remain to be solved. A data base search revealed that ATX and LPA receptors are conserved in wide range of vertebrates from fishes to mammals. Here we analyzed zebrafish ATX (zATX) and LPA receptors both biochemically and functionally. zATX, like mammalian ATX, showed lysophospholipase D activity to produce LPA. In addition, all zebrafish LPA receptors except for LPA5a and LPA5b were found to respond to LPA. Knockdown of zATX in zebrafish embryos by injecting morpholino antisense oligonucleotides (MOs) specific to zATX caused abnormal blood vessel formation, which has not been observed in other morphant embryos or mutants with vascular defects reported previously. In ATX morphant embryos, the segmental arteries sprouted normally from the dorsal aorta but stalled in midcourse, resulting in aberrant vascular connection around the horizontal myoseptum. Similar vascular defects were not observed in embryos in which each single LPA receptor was attenuated by using MOs. Interestingly, similar vascular defects were observed when both LPA1 and LPA4 functions were attenuated by using MOs and/or a selective LPA receptor antagonist, Ki16425. These results demonstrate that the ATX-LPA-LPAR axis is a critical regulator of embryonic vascular development that is conserved in vertebrates.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation , Phosphoric Diester Hydrolases/chemistry , Receptors, Lysophosphatidic Acid/metabolism , Animals , HEK293 Cells , Humans , In Situ Hybridization , Lysophospholipids/metabolism , Mice , Microscopy, Fluorescence/methods , Neovascularization, Physiologic , Substrate Specificity , Time Factors , Zebrafish
8.
Biochimie ; 92(6): 698-706, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20417246

ABSTRACT

Lysophosphatidic acid (LPA) exhibits a wide variety of biological functions as a bio-active lysophospholipid through G-protein-coupled receptors specific to LPA. Currently at least six LPA receptors are identified, named LPA(1) to LPA(6), while the existence of other LPA receptors has been suggested. From studies on knockout mice and hereditary diseases of these LPA receptors, it is now clear that LPA is involved in various biological processes including brain development and embryo implantation, as well as patho-physiological conditions including neuropathic pain and pulmonary and renal fibrosis. Unlike sphingosine 1-phosphate, a structurally similar bio-active lysophospholipid to LPA and produced intracellularly, LPA is produced by multiple extracellular degradative routes. A plasma enzyme called autotaxin (ATX) is responsible for the most of LPA production in our bodies. ATX converts lysophospholipids such as lysophosphatidylcholine to LPA by its lysophospholipase D activity. Recent studies on ATX have revealed new aspects of LPA. In this review, we highlight recent advances in our understanding of LPA functions and several aspects of ATX, including its activity, expression, structure, biochemical properties, the mechanism by which it stimulates cell motility and its pahto-physiological function through LPA production.


Subject(s)
Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Animals , Cell Movement , Humans , Mice , Models, Biological , Phosphoric Diester Hydrolases/chemistry , Receptors, Lysophosphatidic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL