Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Cardiovasc Res ; 92(1): 149-58, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21632882

ABSTRACT

AIMS: We investigated the mechanisms of action of lysophosphatidic acid (LPA) to regulate vascular endothelial (VE)-cadherin dynamics and cell-cell contact. METHODS AND RESULTS: While a low concentration of LPA stimulated VE-cadherin internalization and subsequent cell-cell dissociation, a high concentration of LPA masked the disruptive actions on VE-cadherin and protected the barrier function in human vascular endothelial cells. Knockdown experiments of major LPA receptor subtypes, i.e. LPA(1) and p2y5 (also termed LPA(6)), with their specific small interfering RNAs, showed that LPA(1) and LPA(6) mediate the LPA-induced disruptive and protective actions on barrier integrity, respectively. LPA(6)-mediated tube formation, reflecting stabilization of barrier integrity, was confirmed by in vitro angiogenesis assay. The LPA(1)-mediated disruptive actions were inhibited by pertussis toxin, dominant-negative Rac1, and inhibitors for c-Jun NH(2)-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK), but not by dominant-negative RhoA. In contrast, the LPA(6)-mediated protective actions were associated with activation of Src and Rap1 and attenuated by abrogation of their activities. Further characterization showed that Rap1 is located downstream of Src and dependent on C3G, a Rap1 guanine nucleotide exchange factor. Finally, an LPA antagonist significantly inhibited lactic acid-induced limb lesions in vivo, which may be attributed to dysfunction of endothelial cells. CONCLUSION: LPA induced disruption and protection of VE-cadherin integrity through LPA(1)-G(i) protein-Rac1-JNK/p38MAPK and LPA(6)-G(12/13) protein-Src-C3G-Rap1 pathways, respectively.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTPase-Activating Proteins/physiology , Receptors, Lysophosphatidic Acid/physiology , Receptors, Purinergic P2/physiology , src-Family Kinases/physiology , Animals , Cells, Cultured , Humans , Lysophospholipids/pharmacology , MAP Kinase Kinase 4/physiology , Male , Protein Transport , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/physiology
2.
J Biol Chem ; 285(7): 4387-97, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20018878

ABSTRACT

The upstream signaling pathway leading to the activation of AMP-activated protein kinase (AMPK) by high density lipoprotein (HDL) and the role of AMPK in HDL-induced antiatherogenic actions were investigated. Experiments using genetic and pharmacological tools showed that HDL-induced activation of AMPK is dependent on both sphingosine 1-phosphate receptors and scavenger receptor class B type I through calcium/calmodulin-dependent protein kinase kinase and, for scavenger receptor class B type I system, additionally serine-threonine kinase LKB1 in human umbilical vein endothelial cells. HDL-induced activation of Akt and endothelial NO synthase, stimulation of migration, and inhibition of monocyte adhesion and adhesion molecule expression were dependent on AMPK activation. The inhibitory role of AMPK in the adhesion molecule expression and monocyte adhesion on endothelium of mouse aorta was confirmed in vivo and ex vivo. On the other hand, stimulation of ERK and proliferation were hardly affected by AMPK knockdown but completely inhibited by an N17Ras, whereas the dominant-negative Ras was ineffective for AMPK activation. In conclusion, dual HDL receptor systems differentially regulate AMPK activity through calcium/calmodulin-dependent protein kinase kinase and/or LKB1. Several HDL-induced antiatherogenic actions are regulated by AMPK, but proliferation-related actions are regulated by Ras rather than AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipoproteins, HDL/pharmacology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , Animals , Aorta/cytology , Aorta/metabolism , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line , Cell Proliferation , Endothelial Cells/cytology , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Humans , In Vitro Techniques , Male , Mice , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Umbilical Veins/cytology
3.
Carcinogenesis ; 30(3): 457-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129242

ABSTRACT

Malignant ascites from pancreatic cancer patients has been reported to stimulate migration of pancreatic cancer cells through lysophosphatidic acid (LPA) and LPA(1) receptors. Indeed, ascites- and LPA-induced migration was inhibited by Ki16425, an LPA(1) and LPA(3) antagonist, in Panc-1 cells. Unexpectedly, however, in the presence of Ki16425, ascites and LPA inhibited cell migration in response to epidermal growth factor (EGF). The inhibitory migratory response to ascites and LPA was also observed in the cells treated with pertussis toxin (PTX), a G(i) protein inhibitor, and attenuated by a small interfering RNA (siRNA) specific to the LPA(2) receptor. The inhibitory LPA action was reversed by the regulators of G-protein signaling domain of p115RhoGEF, dominant-negative RhoA or C3 toxin. Indeed, LPA activated RhoA, which was attenuated by the siRNA against the LPA(2) receptor. Moreover, LP-105, an LPA(2) agonist, also inhibited EGF-induced migration in the PTX-treated cells. A similar inhibitory migration response through LPA(2) receptors was also observed in YAPC-PD, BxPC-3, CFPAC-1 and PK-1 pancreatic cancer cell lines. LPA also inhibited the invasion of Panc-1 cells in the PTX-treated cells in the in vitro Matrigel invasion assay. We conclude that LPA(2) receptors are coupled to the G(12/13) protein/Rho-signaling pathway, leading to the inhibition of EGF-induced migration and invasion of pancreatic cancer cells.


Subject(s)
Ascites/metabolism , Cell Movement/drug effects , Lysophospholipids/pharmacology , Pancreatic Neoplasms/metabolism , Receptors, Lysophosphatidic Acid/physiology , Ascites/pathology , Cell Line, Tumor , Collagen , Drug Combinations , Epidermal Growth Factor/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Humans , Isoxazoles/pharmacology , Laminin , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Pertussis Toxin/pharmacology , Propionates/pharmacology , Proteoglycans , RNA, Small Interfering/genetics , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , rhoA GTP-Binding Protein/metabolism
4.
J Immunol ; 181(10): 7332-40, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981156

ABSTRACT

Changes in plasma lipoprotein profiles, especially low levels of high-density lipoprotein (HDL), are a common biomarker for several inflammatory and immune diseases, including atherosclerosis and rheumatoid arthritis. We examined the effect of simvastatin on HDL-induced anti-inflammatory actions. HDL and sphingosine 1-phosphate (S1P), a bioactive lipid component of the lipoprotein, inhibited TNF alpha-induced expression of VCAM-1, which was associated with NO synthase (NOS) activation, in human umbilical venous endothelial cells. The HDL- but not S1P-induced anti-inflammatory actions were enhanced by a prior treatment of the cells with simvastatin in a manner sensitive to mevalonic acid. Simvastatin stimulated the expression of scavenger receptor class B type I (SR-BI) and endothelial NOS. As for S1P receptors, however, the statin inhibited the expression of S1P(3) receptor mRNA but caused no detectable change in S1P(1) receptor expression. The reconstituted HDL, a stimulator of SR-BI, mimicked HDL actions in a simvastatin-sensitive manner. The HDL- and reconstituted HDL-induced actions were blocked by small interfering RNA specific to SR-BI regardless of simvastatin treatment. The statin-induced expression of SR-BI was attenuated by constitutively active RhoA and small interfering RNA specific to peroxisome proliferator-activated receptor-alpha. Administration of simvastatin in vivo stimulated endothelial SR-BI expression, which was accompanied by the inhibition of the ex vivo monocyte adhesion in aortas from TNF alpha-injected mice. In conclusion, simvastatin induces endothelial SR-BI expression through a RhoA- and peroxisome proliferator-activated receptor-alpha-dependent mechanism, thereby enhancing the HDL-induced activation of NOS and the inhibition of adhesion molecule expression.


Subject(s)
Endothelial Cells/metabolism , Hypolipidemic Agents/pharmacology , Inflammation/metabolism , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/biosynthesis , Simvastatin/pharmacology , Animals , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Immunoenzyme Techniques , Lysophospholipids/metabolism , Mice , Monocytes/drug effects , Monocytes/metabolism , Nitric Oxide Synthase/metabolism , PPAR alpha/drug effects , PPAR alpha/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Transfection , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/metabolism
5.
J Bone Miner Res ; 23(7): 1129-39, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18302504

ABSTRACT

Acidosis has been shown to induce depletion of bone calcium from the body. This calcium release process is thought to be partially cell mediated. In an organ culture of bone, acidic pH has been shown to induce cyclooxygenase-2 (COX-2) induction and prostaglandin E(2) (PGE(2)) production, resulting in stimulation of bone calcium release. However, the molecular mechanisms whereby osteoblasts sense acidic circumstances and thereby induce COX-2 induction and PGE(2) production remain unknown. In this study, we used a human osteoblastic cell line (NHOst) to characterize cellular activities, including inositol phosphate production, intracellular Ca(2+) concentration ([Ca(2+)](i)), PGE(2) production, and COX-2 mRNA and protein expression, in response to extracellular acidification. Small interfering RNA (siRNA) specific to the OGR1 receptor and specific inhibitors for intracellular signaling pathways were used to characterize acidification-induced cellular activities. We found that extracellular acidic pH induced a transient increase in [Ca(2+)](i) and inositol phosphate production in the cells. Acidification also induced COX-2 induction, resulting in PGE(2) production. These proton-induced actions were markedly inhibited by siRNA targeted for the OGR1 receptor and the inhibitors for G(q/11) protein, phospholipase C, and protein kinase C. We conclude that the OGR1/G(q/11)/phospholipase C/protein kinase C pathway regulates osteoblastic COX-2 induction and subsequent PGE(2) production in response to acidic circumstances.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Hydrogen-Ion Concentration , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/physiology , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Humans , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
6.
Am J Physiol Heart Circ Physiol ; 292(5): H2513-22, 2007 May.
Article in English | MEDLINE | ID: mdl-17237247

ABSTRACT

The migration of vascular smooth muscle cells (SMCs) is a hallmark of the pathogenesis of atherosclerosis and restenosis after angioplasty. Plasma low-density lipoprotein (LDL), but not high-density lipoprotein (HDL), induced the migration of human coronary artery SMCs (CASMCs). Among bioactive lipids postulated to be present in LDL, lysophosphatidic acid (LPA) appreciably mimicked the LDL action. In fact, the LDL-induced migration was markedly inhibited by pertussis toxin, an LPA receptor antagonist Ki-16425, and a small interfering RNA (siRNA) targeted for LPA(1) receptors. Moreover, LDL contains a higher amount of LPA than HDL does. HDL markedly inhibited LPA- and platelet-derived growth factor (PDGF)-induced migration, and sphingosine 1-phosphate (S1P), the content of which is about fourfold higher in HDL than in LDL, mimicked the HDL action. The inhibitory actions of HDL and S1P were suppressed by S1P(2) receptor-specific siRNA. On the other hand, the degradation of the LPA component of LDL by monoglyceride lipase or the antagonism of LPA receptors by Ki-16425 allowed LDL to inhibit the PDGF-induced migration. The inhibitory effect of LDL was again suppressed by S1P(2) receptor-specific siRNA. In conclusion, LPA/LPA(1) receptors and S1P/S1P(2) receptors mediate the stimulatory and inhibitory migration response to LDL and HDL, respectively. The balance of not only the content of LPA and S1P in lipoproteins but also the signaling activity between LPA(1) and S1P(2) receptors in the cells may be critical in determining whether the lipoprotein is a positive or negative regulator of CASMC migration.


Subject(s)
Coronary Vessels/physiology , Lipoproteins/metabolism , Lysophospholipids/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Adult , Cell Movement/physiology , Cells, Cultured , Humans , Male , Middle Aged
7.
J Biol Chem ; 281(49): 37457-67, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17046831

ABSTRACT

We characterized the molecular mechanisms by which high density lipoprotein (HDL) inhibits the expression of adhesion molecules, including vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, induced by sphingosine 1-phosphate (S1P) and tumor necrosis factor (TNF) alpha in endothelial cells. HDL inhibited S1P-induced nuclear factor kappaB activation and adhesion molecule expression in human umbilical vein endothelial cells. The inhibitory HDL actions were associated with nitric-oxide synthase (NOS) activation and were reversed by inhibitors for phosphatidylinositol 3-kinase and NOS. The HDL-induced inhibitory actions were also attenuated by the down-regulation of scavenger receptor class B type I (SR-BI) and its associated protein PDZK1. When TNFalpha was used as a stimulant, the HDL-induced NOS activation and the inhibitory action on adhesion molecule expression were, in part, attenuated by the down-regulation of the expression of S1P receptors, especially S1P(1), in addition to SR-BI. Reconstituted HDL composed mainly of apolipoprotein A-I and phosphatidylcholine mimicked the SR-BI-sensitive part of HDL-induced actions. Down-regulation of S1P(3) receptors severely suppressed the stimulatory actions of S1P. Although G(i/o) proteins may play roles in either stimulatory or inhibitory S1P actions, as judged from pertussis toxin sensitivity, the coupling of S1P(3) receptors to G(12/13) proteins may be critical to distinguish the stimulatory pathways from the inhibitory ones. In conclusion, even though S1P alone stimulates adhesion molecule expression, HDL overcomes S1P(3) receptor-mediated stimulatory actions through SR-BI/PDZK1-mediated signaling pathways involving phosphatidylinositol 3-kinase and NOS. In addition, the S1P component of HDL plays a role in the inhibition of TNFalpha-induced actions through S1P receptors, especially S1P(1).


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Receptors, Lysosphingolipid/metabolism , Scavenger Receptors, Class B/metabolism , Cell Adhesion Molecules/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Lipoproteins, HDL/pharmacology , Models, Biological , Nitric Oxide Synthase Type III/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
8.
Cell Signal ; 18(6): 841-50, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16111867

ABSTRACT

Sphingosine 1-phosphate (S1P) stimulates expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in human umbilical vein endothelial cells. S1P-induced actions were associated with nuclear factor kappa-B activation and inhibited by pertussis toxin as well as by antisense oligonucleotides specific to S1P receptors, especially, S1P(3). S1P also stimulated endothelial nitric oxide synthase (eNOS) and its activation was markedly inhibited by the antisense oligonucleotide for the S1P(1) receptor rather than that for the S1P(3) receptor. The dose-response curve of S1P to stimulate adhesion molecule expression was shifted to the left in the presence of the phosphatidylinositol 3-kinase inhibitor wortmannin and the NOS inhibitor Nomega-nitro-l-arginine methyl ester. NO donor S-nitroso-N-acetylpenicillamine inhibited S1P-induced adhesion molecule expression. Moreover, tumor necrosis factor-alpha-induced adhesion molecule expression was markedly inhibited by S1P in a manner sensitive to inhibitors for PI3-K and NOS. These results suggest that S1P receptors are coupled to both stimulatory and inhibitory pathways for adhesion molecule expression. The stimulatory pathway involves nuclear factor kappa-B and inhibitory one does phosphatidylinositol 3-kinase and NOS.


Subject(s)
Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Receptors, Lysosphingolipid/physiology , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Down-Regulation , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
9.
J Biol Chem ; 279(8): 6595-605, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14660630

ABSTRACT

Cytokines and growth factors in malignant ascites are thought to modulate a variety of cellular activities of cancer cells and normal host cells. The motility of cancer cells is an especially important activity for invasion and metastasis. Here, we examined the components in ascites, which are responsible for cell motility, from patients and cancer cell-injected mice. Ascites remarkably stimulated the migration of pancreatic cancer cells. This response was inhibited or abolished by pertussis toxin, monoglyceride lipase, an enzyme hydrolyzing lysophosphatidic acid (LPA), and Ki16425 and VPC12249, antagonists for LPA receptors (LPA1 and LPA3), but not by an LPA3-selective antagonist. These agents also inhibited the response to LPA but not to the epidermal growth factor. In malignant ascites, LPA is present at a high level, which can explain the migration activity, and the fractionation study of ascites by lipid extraction and subsequent thin-layer chromatography indicated LPA as an active component. A significant level of LPA1 receptor mRNA is expressed in pancreatic cancer cells with high migration activity to ascites but not in cells with low migration activity. Small interfering RNA against LPA1 receptors specifically inhibited the receptor mRNA expression and abolished the migration response to ascites. These results suggest that LPA is a critical component of ascites for the motility of pancreatic cancer cells and LPA1 receptors may mediate this activity. LPA receptor antagonists including Ki16425 are potential therapeutic drugs against the migration and invasion of cancer cells.


Subject(s)
Ascites/metabolism , Lysophospholipids/metabolism , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Adult , Animals , Blotting, Northern , Cell Adhesion , Cell Division , Cell Line, Tumor , Cell Movement , Chromatography, Thin Layer , Dose-Response Relationship, Drug , Epidermal Growth Factor/metabolism , Female , Humans , Isoxazoles/pharmacology , Lipids , Male , Mice , Mice, Inbred BALB C , Middle Aged , Monoacylglycerol Lipases/pharmacology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Pertussis Toxin/pharmacology , Propionates/pharmacology , RNA, Messenger/metabolism , Receptors, Lysophosphatidic Acid , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL