Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Heart Vessels ; 30(3): 406-9, 2015 May.
Article in English | MEDLINE | ID: mdl-24477746

ABSTRACT

A 78-year-old man with unstable angina showed 90% stenosis in the proximal left anterior descending artery. Pre-procedural intravascular ultrasound revealed ruptured plaque and attenuated plaque in the lesion. Under these conditions, two overlapping sirolimus-eluting stent (SES) implantation in this lesion resulted in slow flow which was recovered by intracoronary nitrates, nicorandil, and nitroprusside without further complications. When the patient showed up again 5 years later with recurrence of angina pectoris, angiography revealed a hazy ulcerated in-stent restenosis (ISR) at the site of the SES. Pre-procedural optical coherence tomography (OCT) imaging revealed multiple intimal ruptures, cavity formation behind the stent struts, a thin-cap fibroatheroma containing neointima surrounded by signal-poor, lipid-rich area in the proximal SES, suggesting the progression of neoatherosclerosis within SES. Importantly, there occurred slow flow again after balloon angioplasty for this lesion. We would suggest careful OCT examination is warranted to confirm development of neoatherosclerosis within the stent, and distal protection device should be considered to prevent slow flow phenomenon even in a patient with very late ISR.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Cardiovascular Agents/administration & dosage , Coronary Restenosis/therapy , Coronary Stenosis/therapy , Drug-Eluting Stents , No-Reflow Phenomenon/etiology , Sirolimus/administration & dosage , Aged , Angioplasty, Balloon, Coronary/adverse effects , Coronary Angiography , Coronary Circulation , Coronary Restenosis/diagnosis , Coronary Restenosis/etiology , Coronary Restenosis/physiopathology , Coronary Stenosis/diagnosis , Coronary Stenosis/physiopathology , Denture, Partial, Temporary , Humans , Male , No-Reflow Phenomenon/diagnosis , No-Reflow Phenomenon/physiopathology , Recurrence , Retreatment , Time Factors , Tomography, Optical Coherence , Treatment Outcome , Ultrasonography, Interventional
3.
Int J Cardiol ; 166(2): 448-52, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-22112682

ABSTRACT

BACKGROUND: At the intervention for cardiovascular disease (CVD), albuminuria is a new pivotal target. Calcium channel blocker (CCB) is one of the most expected agents. Currently CCBs have been classified by delivery system, half-life and channel types. We tested anti-albuminuric effect among 4 types of CCBs. METHODS: Subjects were 50 hypertensives (SBP/DBP 164.7±17.1/92.3±12.2mmHg, s-Cr 0.81±0.37mg/dl, urinary albumin excretion (UAE) 69.4 (33.5-142.6) mg/gCr). Four CCBs were administered in a crossover setting: nifedipine CR, a long biological half-life L type by controlled release; cilnidipine, an N/L type; efonidipine, a T/L type; and amlodipine, a long biological half-life L type. RESULTS: Comparable BP reductions were obtained. UAE at endpoints ware as follows (mg/gCr, *P<0.01): nifedipine CR 30.8 (17.3-81.1),* cilnidipine 33.9 (18.0-67.7),* efonidipine 51.0 (21.2-129.8), amlodipine 40.6 (18.7-94.7). By all agents, significant augmentations were observed in PRA, angiotensin I and angiotensin II (AngII). AngII at cilnidipine was significantly lower than that at amlodipine. PAC at cilnidipine and efonidipine was significantly lower than that at amlodipine. Nifedipine CR significantly reduced ANP concentration. CONCLUSIONS: It is revealed that only nifedipine CR and cilnidipine could reduce albuminuria statistically. Thus, it is suggested that the 2 CCBs might be favorable for organ protection in hypertensives.


Subject(s)
Albuminuria/drug therapy , Calcium Channel Blockers/therapeutic use , Cardiovascular Diseases/drug therapy , Hypertension/drug therapy , Aged , Aged, 80 and over , Albuminuria/diagnosis , Albuminuria/urine , Biomarkers/urine , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/urine , Cross-Over Studies , Dihydropyridines/therapeutic use , Female , Humans , Hypertension/diagnosis , Hypertension/urine , Male , Middle Aged , Nifedipine/therapeutic use
4.
J Steroid Biochem Mol Biol ; 129(3-5): 145-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22212769

ABSTRACT

Angiogenesis plays a pivotal role in cardiovascular diseases such as ischemic heart disease, limb ischemia and heart failure, and has recently been shown to mediate various biological activities related to the pathogenesis of these diseases. In the present study, we evaluated the role of aldosterone in angiogenesis. Tube formation assay on Matrigel using human umbilical vein endothelial cells (HUVEC) revealed that aldosterone inhibited endothelial morphogenesis in a manner sensitive to eplerenone, a selective mineralocorticoid receptor antagonist. The anti-angiogenic effect of aldosterone was further confirmed by an in vivo angiogenesis assay using a Matrigel plug model in mice. Reverse transcription-mediated polymerase chain reaction and immunoblotting demonstrated that aldosterone downregulated the expression levels of vascular endothelial growth factor receptor-2 (VEGFR-2) and peroxisome proliferators-activated receptor gamma (PPAR gamma). VEGFR-2 expression was found to be enhanced in response to PPAR gamma activation by troglitazone, and attenuated by GW9662, a specific antagonist of PPAR gamma. In the tube formation assay, endothelial morphogenesis was stimulated by troglitazone, and inhibited by GW9662, indicating that PPAR gamma activation mediates positive regulation of angiogenesis through enhancement of VEGFR-2 expression. These data suggest that aldosterone inhibits angiogenesis through VEGFR-2 downregulation, subsequent to, at least in part, attenuation of PPAR gamma expression. The present findings provide a new insight into the possible therapeutic application of mineralocorticoid receptor blockade to various cardiovascular diseases.


Subject(s)
Aldosterone/pharmacology , Angiogenesis Inhibitors/pharmacology , Down-Regulation/drug effects , Neovascularization, Physiologic/drug effects , PPAR gamma/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Collagen/pharmacology , Drug Combinations , Endothelium/growth & development , Eplerenone , Human Umbilical Vein Endothelial Cells , Humans , Laminin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/pharmacology , Morphogenesis/drug effects , PPAR gamma/metabolism , Proteoglycans/pharmacology , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
J Clin Invest ; 120(11): 3979-95, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20978351

ABSTRACT

Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid that has pleiotropic effects in a variety of cell types including ECs, SMCs, and macrophages, all of which are central to the development of atherosclerosis. It may therefore exert stimulatory and inhibitory effects on atherosclerosis. Here, we investigated the role of the S1P receptor S1PR2 in atherosclerosis by analyzing S1pr2-/- mice with an Apoe-/- background. S1PR2 was expressed in macrophages, ECs, and SMCs in atherosclerotic aortas. In S1pr2-/-Apoe-/- mice fed a high-cholesterol diet for 4 months, the area of the atherosclerotic plaque was markedly decreased, with reduced macrophage density, increased SMC density, increased eNOS phosphorylation, and downregulation of proinflammatory cytokines compared with S1pr2+/+Apoe-/- mice. Bone marrow chimera experiments indicated a major role for macrophage S1PR2 in atherogenesis. S1pr2-/-Apoe-/- macrophages showed diminished Rho/Rho kinase/NF-κB (ROCK/NF-κB) activity. Consequently, they also displayed reduced cytokine expression, reduced oxidized LDL uptake, and stimulated cholesterol efflux associated with decreased scavenger receptor expression and increased cholesterol efflux transporter expression. S1pr2-/-Apoe-/- ECs also showed reduced ROCK and NF-κB activities, with decreased MCP-1 expression and elevated eNOS phosphorylation. Pharmacologic S1PR2 blockade in S1pr2+/+Apoe-/- mice diminished the atherosclerotic plaque area in aortas and modified LDL accumulation in macrophages. We conclude therefore that S1PR2 plays a critical role in atherogenesis and may serve as a novel therapeutic target for atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/immunology , Atherosclerosis/pathology , Macrophages/immunology , Receptors, Lysosphingolipid/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Becaplermin , Cells, Cultured , Female , Lysophospholipids/metabolism , Macrophages/cytology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , NF-kappa B/genetics , NF-kappa B/metabolism , Plaque, Atherosclerotic/pathology , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
6.
J Hypertens ; 28(10): 2156-60, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20625317

ABSTRACT

OBJECTIVE: Calcium channel blocker (CCB) is one of the most useful antihypertensive agents. However, the activation of the renin-angiotensin system (RAS) is an unfavorable characteristic. N-type calcium channel is thought to be involved in catecholamine's release. Accordingly, N/L-type CCB has a probability of less activation of the RAS. We substantiated the hypothesis that N/L-type CCB, cilnidipine, leads to less activation of the RAS compared with conventional L-type CCB, amlodipine. DESIGN: Randomized, cross-over study. SETTING: Outpatient study. PARTICIPANTS: Participants were 110 hypertensive patients [male/female 46/64, age 66.3 ± 10.8 years, systolic blood pressure (SBP)/diastolic blood pressure (DBP) 161.8 ± 16.9/92.9 ± 12.4 mmHg, s-Cr 0.77 ± 0.32 mg/dl, plasma renin activity (PRA) 0.65 ± 0.63 ng/ml per h, angiotensin I (AngI) 70.5 ± 77.3 pg/ml, angiotensin II (AngII) 5.2 ± 3.9 pg/ml, plasma aldosterone concentration (PAC) 76.3 ± 35.9 pg/ml, urinary albumin excretion (UAE) 108.1 ± 284.2 mg/gCr]. Amlodipine besilate or cilnidipine was administered for 12 weeks in a cross-over manner as a monotherapy with an intention-to-treat fashion by titrating doses. Final doses of amlodipine besilate and cilnidipine were 6.6 ± 2.7 and 13.7 ± 5.1 mg/day, respectively. MAIN OUTCOME MEASURES: Changes in blood pressure, PRA, AngI, AngII, PAC, UAE of baseline and each end of amlodipine besilate and cilnidipine administration. RESULTS: Results were as follows (amlodipine vs. cilnidipine): SBP/DBP (mmHg): 135.2 ± 11.7/79.8 ± 9.6 vs. 136.7 ± 13.2/79.5 ± 10.9, P = 0.22/0.74; PRA (ng/ml per h): 1.16 ± 1.03 vs. 0.95 ± 0.78, P < 0.01; AngI (pg/ml): 155.0 ± 306.4 vs. 101.8 ± 92.0, P < 0.05; AngII (pg/ml): 12.0 ± 12.3 vs. 7.1 ± 4.5, P < 0.001; PAC (pg/ml): 81.6 ± 37.9 vs. 74.3 ± 36.2, P < 0.05; UAE (mg/gCr): 145.4 ± 424.5 vs. 58.8 ± 125.1, P < 0.05. Thus, in spite of the comparable blood pressure reductions, each level of components of the RAS at cilnidipine administration was significantly lower than those at amlodipine. Apart from this, UAE at cilnidipine administration was also significantly lower than that at amlodipine. CONCLUSION: It is suggested that cilnidipine leads to less activation of the RAS compared with amlodipine for the first time in human clinical patients and therefore cilnidipine might be expected to be superior in organ protection in addition to the antialbuminuric effect.


Subject(s)
Amlodipine/pharmacology , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Hypertension/physiopathology , Renin-Angiotensin System/drug effects , Aged , Albuminuria/physiopathology , Aldosterone/blood , Amlodipine/therapeutic use , Angiotensin I/blood , Angiotensin II/blood , Blood Pressure/drug effects , Blood Pressure/physiology , Calcium Channel Blockers/therapeutic use , Cross-Over Studies , Dihydropyridines/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Hypertension/blood , Hypertension/drug therapy , Male , Middle Aged , Renin/blood , Renin-Angiotensin System/physiology
7.
Diabetes Care ; 32(8): 1485-90, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19509012

ABSTRACT

OBJECTIVE Recent studies have proven the favorable effects of angiotensin receptor blockers (ARBs) on cardiovascular and renal disorders. However, determinants of the response to ARBs remain unclear. We substantiated the hypothesis that genetic variants of the renin-angiotensin system (RAS) have significant impacts on the response to ARBs. RESEARCH DESIGN AND METHODS Subjects comprised 231 consecutively enrolled hypertensive individuals including 45 type 2 diabetic subjects. Five genetic variants of the RAS, i.e., renin (REN) C-5312T, ACE insertion/deletion, angiotensinogen M235T, angiotensin II type 1 receptor A1166C, and angiotensin II type 2 receptor C3123A were assayed by PCR and restriction fragment-length polymorphism. A dose of 40-160 mg/day of valsartan was administered for 3 months as a monotherapy. RESULTS Changes in diastolic blood pressure significantly differed between genotypes of REN C-5312T: 10.7-mmHg reduction (from 95.9 +/- 12.9 to 85.2 +/- 11.4) in CC versus 7.0-mmHg reduction (from 94.7 +/- 14.0 to 87.7 +/- 12.6) in CT/TT (P = 0.02 for interactive effects of valsartan and genotype). Responder rates also differed between the genotypes: 72.8% in CC versus 58.0% in CT/TT (P = 0.03). Univariate analysis indicated a significant association of response to valsartan with blood pressure, diabetes, plasma aldosterone concentration, and CC homozygotes of REN C-5312T. Finally, multiple logistic regression analysis revealed that systolic blood pressure, CC homozygotes of REN C-5312T, and diabetes were independent predictors for responders with odds ratios (95% CI) of 2.49 (1.41-4.42), 2.03 (1.10-3.74), and 0.48 (0.24-0.96), respectively. CONCLUSIONS This study provides strong support that a genetic variant of REN C-5312T and diabetes contribute to the effects of ARBs and are independent predictors for responder. Thus, in treatment of hypertension with ARBs, a new possibility for personalized medicine has been shown.


Subject(s)
Angiotensin Receptor Antagonists , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Genetic Variation , Hypertension/drug therapy , Receptor, Angiotensin, Type 2/genetics , Renin-Angiotensin System/genetics , Blood Pressure/drug effects , Body Mass Index , Creatinine/blood , DNA Primers , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/physiopathology , Female , Gene Deletion , Glomerular Filtration Rate , Glycated Hemoglobin/metabolism , Humans , Hypertension/genetics , Male , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Receptor, Angiotensin, Type 1/genetics , Renin/genetics
8.
Biochem Biophys Res Commun ; 346(1): 293-300, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16756949

ABSTRACT

We speculated that the sphingosine-1-phosphate (S1P) receptor S1P(2), which uniquely inhibits cell migration, might mediate inhibitory effects on endothelial cell migration and angiogenesis, different from S1P(1) and S1P(3). Mouse vascular endothelial cells, which endogenously express S1P(2) and S1P(3), but not S1P(1), responded to S1P and epidermal growth factor (EGF) with stimulation of Rac, migration, and the formation of tube-like structures on the Matrigel. The S1P(3)-antagonist VPC-23019 abolished S1P-induced, G(i)-dependent Rac stimulation, cell migration, and tube formation, whereas the S1P(2)-antagonist JTE-013 enhanced these S1P-induced responses, suggesting that S1P(2) exerts inhibitory effects on endothelial Rac, migration, and angiogenesis. S1P(2) overexpression markedly augmented S1P-induced, G(i)-independent inhibition of EGF-induced migration and tube formation. Finally, the blockade of S1P(2) by JTE-013 potentiated S1P-induced stimulation of angiogenesis in vivo in the Matrigel implant assay. These observations indicate that in contrast to S1P(1) and S1P(3), S1P(2) negatively regulates endothelial morphogenesis and angiogenesis most likely through down-regulating Rac.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/physiology , Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Receptors, Lysosphingolipid/physiology , Animals , Cell Migration Inhibition , Endothelial Cells/drug effects , Endothelium/growth & development , Mice , Stimulation, Chemical , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/physiology
9.
Biochem J ; 394(Pt 3): 581-92, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16336212

ABSTRACT

We demonstrated previously that membrane depolarization and excitatory receptor agonists such as noradrenaline induce Ca2+-dependent Rho activation in VSM (vascular smooth muscle), resulting in MP (myosin phosphatase) inhibition through the mechanisms involving Rho kinase-mediated phosphorylation of its regulatory subunit MYPT1. In the present study, we show in de-endothelialized VSM strips that the PI3K (phosphoinositide 3-kinase) inhibitors LY294002 and wortmannin inhibited KCl membrane depolarization- and noradrenaline-induced Rho activation and MYPT1 phosphorylation, with concomitant inhibition of MLC (20-kDa myosin light chain) phosphorylation and contraction. LY294002 also augmented de-phosphorylation of MLC and resultantly relaxation in KCl-contracted VSM, whereas LY294002 was much less effective or ineffective under the conditions in which MP was inhibited by either a phosphatase inhibitor or a phorbol ester in Rho-independent manners. VSM express at least four PI3K isoforms, including the class I enzymes p110alpha and p110beta and the class II enzymes PI3K-C2alpha and -C2beta. The dose-response relationships of PI3K-inhibitor-induced inhibition of Rho, MLC phosphorylation and contraction were similar to that of PI3K-C2alpha inhibition, but not to that of the class I PI3K inhibition. Moreover, KCl and noradrenaline induced stimulation of PI3K-C2alpha in a Ca2+-dependent manner, but not of p110alpha or p110beta. Down-regulation of PI3K-C2alpha expression by siRNA (small interfering RNA) inhibited contraction and phosphorylation of MYPT1 and MLC in VSM cells. Finally, intravenous wortmannin infusion induced sustained hypotension in rats, with inhibition of PI3K-C2alpha activity, GTP-loading of Rho and MYPT1 phosphorylation in the artery. These results indicate the novel role of PI3K-C2alpha in Ca2+-dependent Rho-mediated negative control of MP and thus VSM contraction.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Phosphatase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Aorta/metabolism , Calcium/metabolism , Class II Phosphatidylinositol 3-Kinases , Down-Regulation , Gene Expression Regulation, Enzymologic , Male , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , RNA Interference , RNA, Small Interfering , Rabbits , Rats , Rats, Wistar
10.
Arthritis Res Ther ; 7(6): R1158-73, 2005.
Article in English | MEDLINE | ID: mdl-16277668

ABSTRACT

ADAMs (a disintegrin and metalloproteinases) comprise a new gene family of metalloproteinases, and may play roles in cell-cell interaction, cell migration, signal transduction, shedding of membrane-anchored proteins and degradation of extracellular matrix. We screened the mRNA expression of 10 different ADAMs with a putative metalloproteinase motif in synovial tissues from patients with rheumatoid arthritis (RA) or osteoarthritis (OA). Reverse transcription PCR and real-time quantitative PCR analyses indicated that among the ADAMs, ADAM15 mRNA was more frequently expressed in the RA samples and its expression level was significantly 3.8-fold higher in RA than in OA (p < 0.01). In situ hybridization, immunohistochemistry and immunoblotting demonstrated that ADAM15 is expressed in active and precursor forms in the synovial lining cells, endothelial cells of blood vessels and macrophage-like cells in the sublining layer of RA synovium. There was a direct correlation between ADAM15 mRNA expression levels and vascular density in the synovial tissues (r = 0.907, p < 0.001; n = 20). ADAM15 was constitutively expressed in RA synovial fibroblasts and human umbilical vein endothelial cells (HUVECs), and the expression level was increased in HUVECs by treatment with vascular endothelial growth factor (VEGF)165. On the other hand, ADAM15 expression in RA synovial fibroblasts was enhanced with VEGF165 only if vascular endothelial growth factor receptor (VEGFR)-2 expression was induced by treatment with tumor necrosis factor-alpha, and the expression was blocked with SU1498, a specific inhibitor of VEGFR-2. These data demonstrate that ADAM15 is overexpressed in RA synovium and its expression is up-regulated by the action of VEGF165 through VEGFR-2, and suggest the possibility that ADAM15 is involved in angiogenesis in RA synovium.


Subject(s)
ADAM Proteins/metabolism , Arthritis, Rheumatoid/enzymology , Membrane Proteins/metabolism , Neovascularization, Pathologic/enzymology , Synovial Membrane/enzymology , Vascular Endothelial Growth Factor A/metabolism , ADAM Proteins/genetics , Aged , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cytokines/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/drug effects , Humans , In Situ Hybridization , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/genetics , Middle Aged , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/blood supply , Synovial Membrane/pathology , Up-Regulation , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Lab Invest ; 85(12): 1489-506, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16200075

ABSTRACT

Matrix metalloproteinase-7 (MMP-7) (also known as matrilysin-1) is secreted as a proenzyme (proMMP-7) and plays a key role in the degradation of various extracellular matrix (ECM) and non-ECM molecules after activation. To identify the binding proteins related to proMMP-7 activation, a human lung cDNA library was screened by yeast two-hybrid system using proMMP-7 as bait. We identified a candidate molecule CD151, which is a member of the transmembrane 4 superfamily. Complex formation of proMMP-7 with CD151 was demonstrated by immunoprecipitation of the molecules from CaR-1 cells, a human rectal carcinoma cell line, expressing both proMMP-7 and CD151, and CD151 stable transfectants incubated with proMMP-7. Yeast two-hybrid assays using deletion mutants of proMMP-7 and CD151 suggested an interaction between the propeptide of proMMP-7 and the COOH-terminal extracellular loop of CD151. The binding activity of (125)I-labeled proMMP-7 to CD151 on the cell membranes was shown with CD151 stable transfectants. Laser-scanning confocal microscopy demonstrated that proMMP-7 colocalizes with CD151 on the cell membranes of CD151 stable transfectants and CaR-1 cells. In situ zymography using crosslinked carboxymethylated transferrin, a substrate of MMP-7, demonstrated proteinase activity on and around CD151 stable transfectants and CaR-1 cells, while the activity was abolished by their treatment with MMP inhibitors, anti-MMP-7 antibody or anti-CD151 antibody. In human lung adenocarcinoma tissues, colocalization of MMP-7 and CD151 was demonstrated on the carcinoma cells. Metalloproteinase activity was present in these tissues and could be inhibited by antibodies to MMP-7 or CD151. These data demonstrate for the first time that proMMP-7 is captured and activated on the cell membranes through interaction with CD151, and suggest the possibility that similar to the MT1-MMP/MMP-2 system, MMP-7 is involved in the pericellular activation mechanism mediated by CD151, a crucial step in proteolysis on the cell membranes under various pathophysiological conditions including cancer invasion and metastasis.


Subject(s)
Adenocarcinoma/enzymology , Antigens, CD/metabolism , Cell Membrane/enzymology , Lung Neoplasms/enzymology , Matrix Metalloproteinase 7/metabolism , Rectal Neoplasms/enzymology , Adenocarcinoma/pathology , Antibodies, Blocking/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Cell Line, Tumor , Cell Membrane/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/immunology , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Tetraspanin 24 , Transfection , Two-Hybrid System Techniques
12.
Biochem J ; 374(Pt 3): 715-22, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12803545

ABSTRACT

We have recently reported that S1P (sphingosine-1-phosphate) differentially regulates cellular Rac activity and cell migration in either a positive or a negative direction via distinct G-protein-coupled receptor subtypes, i.e. S1P1/Edg1 (endothelial differentiation gene) and S1P2/Edg5 respectively, when each of the S1P receptor subtypes is expressed in CHO (Chinese-hamster ovary) cells. In B16F10 mouse melanoma cells, in which S1P2, but not the other S1P receptor subtypes, is endogenously expressed, S1P inhibited cell migration with concomitant inhibition of Rac and stimulation of RhoA in dose-dependent manners. Overexpression of S1P2 in the melanoma cells resulted in potentiation of S1P inhibition of both Rac and cell migration. In contrast, overexpression of S1P1 led to stimulation of cell migration, particularly at the lower S1P concentrations. Treatment of B16F10 cells with S1P inhibited lung metastasis 3 weeks after injection into mouse tail veins. Intriguingly, overexpression of S1P2 greatly potentiated the inhibition of metastasis by S1P, whereas that of S1P1 resulted in aggravation of metastasis. Suppression of cellular Rac activity by adenovirus-transduced expression of N17Rac, but not N19RhoA, strongly inhibited cell migration in vitro and lung metastasis in vivo. These results provide the first evidence that G-protein-coupled receptors could participate in the regulation of metastasis, in which ligand-dependent, subtype-specific regulation of the cellular Rac activity is probably critically involved as a mechanism.


Subject(s)
Cell Migration Inhibition , Down-Regulation , Lysophospholipids , Melanoma, Experimental/secondary , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/physiology , Up-Regulation , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/biosynthesis , Animals , CHO Cells , Calcium Signaling/genetics , Cricetinae , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Receptors, Lysophospholipid , Tumor Cells, Cultured , Up-Regulation/genetics , rac GTP-Binding Proteins/physiology
13.
Am J Pathol ; 162(1): 171-81, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12507900

ABSTRACT

To assess the possible involvement of vascular endothelial growth factor (VEGF) in the pathology of osteoarthritic (OA) cartilage, we examined the expression of VEGF isoforms and their receptors in the articular cartilage, and the effects of VEGF on the production of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in OA chondrocytes. Reverse transcriptase-polymerase chain reaction analyses demonstrated that mRNAs for three VEGF isoforms (VEGF(121), VEGF(165), and VEGF(189)) are detectable in all of the OA and normal (NOR) cartilage samples. However, the mRNA expression of their receptors (VEGFR-1 = Flt-1, VEGFR-2 = KDR and neuropilin-1) was recognized only in the OA samples. The protein expression of VEGFR-1 and VEGFR-2 in OA chondrocytes was also demonstrated by immunohistochemistry of the OA cartilage tissue and cultured OA chondrocytes. In situ hybridization and immunohistochemistry indicated that VEGF is expressed in the chondrocytes in the superficial and transitional zones of OA cartilage. A linear correlation was obtained between VEGF immunoreactivity and Mankin scores in the cartilage (r = 0.906, P < 0.001). The production levels of VEGF determined by enzyme-linked immunosorbent assay were significantly 3.3-fold higher in OA than in NOR samples (P < 0.001). Among MMP-1, -2, -3, -7, -8, -9, and -13, TIMP-1 and -2 measured by their sandwich enzyme immunoassay systems, the production of MMP-1 and MMP-3 but not TIMP-1 or TIMP-2 was significantly enhanced by the treatment of cultured OA chondrocytes with VEGF (P < 0.05), whereas no such effect was obtained with cultured NOR chondrocytes. These results demonstrate that VEGF and its receptors are expressed in OA cartilage, and suggest the possibility that VEGF is implicated for the destruction of OA articular cartilage through the increased production of MMPs.


Subject(s)
Cartilage, Articular/metabolism , Endothelial Growth Factors/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Lymphokines/biosynthesis , Osteoarthritis/metabolism , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Aged , Cartilage, Articular/pathology , Cell Division/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Culture Media, Conditioned/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Humans , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/genetics , Lymphokines/pharmacology , Matrix Metalloproteinases/metabolism , Osteoarthritis/pathology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , Receptors, Vascular Endothelial Growth Factor/genetics , Reference Values , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Biol Chem ; 277(39): 36288-95, 2002 Sep 27.
Article in English | MEDLINE | ID: mdl-12114504

ABSTRACT

Vascular endothelial growth factor (VEGF), a potent angiogenic mitogen, plays a crucial role in angiogenesis under various pathophysiological conditions. We have recently demonstrated that VEGF(165), one of the VEGF isoforms, binds connective tissue growth factor (CTGF) and that its angiogenic activity is inhibited in the VEGF(165).CTGF complex form (Inoki, I., Shiomi, T., Hashimoto, G., Enomoto, H., Nakamura, H., Makino, K., Ikeda, E., Takata, S., Kobayashi, K. and Okada, Y. (2002) FASEB J. 16, 219-221). In the present study, we further examined the susceptibility of the VEGF(165).CTGF complex to matrix metalloproteinases (MMP-1, -2, -3, -7, -9, and -13), ADAMTS4 (aggrecanase-1), and serine proteinases, and evaluated the recovery of the angiogenic activity of VEGF(165) after the treatment. Among the MMPs, MMP-1, -3, -7, and -13 processed CTGF of the complex into the major NH(2)- and COOH-terminal fragments, whereas VEGF(165) was completely resistant to the MMPs. On the other hand, elastase and plasmin cleaved both CTGF and VEGF(165) of the complex, but they were completely resistant to ADAMTS4. By digestion of the immobilized VEGF(165).CTGF complex with MMP-3 or MMP-7, both NH(2)- and COOH-terminal fragments of CTGF were dissociated and released from the complex into the liquid phase. The in vitro angiogenic activity of VEGF(165) blocked in the VEGF(165).CTGF complex was reactivated to original levels after CTGF digestion of the complex with MMP-1, -3, and -13. Recovery of angiogenic activity was further confirmed by in vivo angiogenesis assay using a Matrigel injection model in mice. These results demonstrate for the first time that CTGF is a substrate of MMPs and that the angiogenic activity of VEGF(165) suppressed by the complex formation with CTGF is recovered through the selective degradation of CTGF by MMPs. MMPs may play a novel role through CTGF degradation in VEGF-induced angiogenesis during embryonic development, tissue maintenance, and/or pathological processes of various diseases.


Subject(s)
Endothelial Growth Factors/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Aorta/cytology , Binding Sites , Cattle , Collagenases/metabolism , Connective Tissue Growth Factor , DNA, Complementary/metabolism , Endothelium, Vascular/cytology , Fibrinolysin/metabolism , Humans , Immunoblotting , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 7/metabolism , Pancreatic Elastase/metabolism , Protein Binding , Recombinant Proteins/metabolism , Serine Endopeptidases/metabolism , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
FASEB J ; 16(2): 219-21, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11744618

ABSTRACT

Vascular endothelial growth factor (VEGF) is a strong angiogenic mitogen and plays important roles in angiogenesis under various pathophysiological conditions. The in vivo angiogenic activity of secreted VEGF may be regulated by extracellular inhibitors, because it is also produced in avascular tissues such as the cartilage. To seek the binding inhibitors against VEGF, we screened the chondrocyte cDNA library by a yeast two-hybrid system by using VEGF165 as bait and identified connective tissue growth factor (CTGF) as a candidate. The complex formation of VEGF165 with CTGF was first established by immunoprecipitation from the cells overexpressing both binding partners. A competitive affinity-binding assay also demonstrated that CTGF binds specifically to VEGF165 with two classes of binding sites (Kd = 26 +/- 11 nM and 125 +/- 38 nM). Binding assay using deletion mutants of CTGF indicated that the thrombospondin type-1 repeat (TSP-1) domain of CTGF binds to the exon 7-coded region of VEGF165 and that the COOH-terminal domain preserves the affinity to both VEGF165 and VEGF121. The interaction of VEGF165 with CTGF inhibited the binding of VEGF165 to the endothelial cells and the immobilized KDR/IgG Fc; that is, a recombinant protein for VEGF165 receptor. By in vitro tube formation assay of endothelial cells, full-length CTGF and the deletion mutant possessing the TSP-1 domain inhibited VEGF165-induced angiogenesis significantly in the complex form. This antiangiogenic activity of CTGF was demonstrated further by in vivo angiogenesis assay by using Matrigel injection model in mice. These data demonstrate for the first time that VEGF165 binds to CTGF through a protein-to-protein interaction and suggest that the angiogenic activity of VEGF165 is regulated negatively by CTGF in the extracellular environment.


Subject(s)
Endothelial Growth Factors/metabolism , Growth Substances/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Lymphokines/metabolism , Neovascularization, Physiologic/physiology , Animals , Blood Vessels/chemistry , Blood Vessels/drug effects , Blood Vessels/growth & development , Cell Line , Connective Tissue Growth Factor , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Growth Substances/genetics , Growth Substances/pharmacology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Injections, Subcutaneous , Lymphokines/genetics , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Skin/blood supply , Skin/chemistry , Skin/drug effects , Two-Hybrid System Techniques , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , von Willebrand Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...