Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biol. Res ; 50: 1, 2017. tab, graf
Article in English | LILACS | ID: biblio-838973

ABSTRACT

BACKGROUND: Pathologic vascular smooth muscle cell (VSMC) proliferation and migration after vascular injury promotes the development of occlusive vascular disease. Therefore, an effective chemical agent to suppress aberrant proliferation and migration of VSMCs can be a potential therapeutic modality for occlusive vascular disease such as atherosclerosis and restenosis. To find an anti-proliferative chemical agent for VSMCs, we screened an in-house small molecule library, and the selected small molecule was further validated for its anti-proliferative effect on VSMCs using multiple approaches, such as cell proliferation assays, wound healing assays, transwell migration assays, and ex vivo aortic ring assay. RESULTS: Among 43 initially screened small molecule inhibitors of kinases that have no known anti-proliferative effect on VSMCs, a spleen tyrosine kinase (Syk) inhibitor (BAY61-3606) showed significant anti-proliferative effect on VSMCs. Further experiments indicated that BAY61 attenuated the VSMC proliferation in both concentration- and time-dependent manner, and it also significantly suppressed the migration of VSMCs as assessed by both wound healing assays and transwell assays. Additionally, BAY61 suppressed the sprouting of VSMCs from endothelium-removed aortic rings. CONCLUSION: The present study identified a Syk kinase inhibitor as a potent VSMC proliferation and migration inhibitor and warrants further studies to elucidate its underlying molecular mechanisms, such as its primary target, and to validate its in vivo efficacy as a therapeutic agent for restenosis and atherosclerosis.


Subject(s)
Animals , Rats , Pyrimidines/pharmacology , Cell Movement/drug effects , Niacinamide/analogs & derivatives , Myocytes, Smooth Muscle/drug effects , Cell Proliferation/drug effects , Syk Kinase/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Aorta, Thoracic/drug effects , Time Factors , Wound Healing/drug effects , Cells, Cultured , Blotting, Western , Reproducibility of Results , Rats, Sprague-Dawley , Niacinamide/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Cell Migration Assays , Muscle, Smooth, Vascular/cytology
2.
Biol. Res ; 50: 23, 2017. graf
Article in English | LILACS | ID: biblio-950874

ABSTRACT

BACKGROUND: Airway remodeling is a key feature of asthma, characterized by increased proliferation of airway smooth muscle cells (ASMCs). S100A8 is a calcium-binding protein with a potential to regulate cell proliferation. Here, the effect of exogenous S100A8 protein on the proliferation of ASMCs induced by platelet-derived growth factor (PDGF) and the underlying molecular mechanism was investigated. METHODS: Rat ASMCs were cultured with or without a neutralizing antibody to the receptor for advanced glycation end-products (RAGE), a potential receptor for S100A8 protein. Purified recombinant rat S100A8 protein was then added into the cultured cells, and the proliferation of ASMCs induced by PDGF was detected by colorimetric-based WST-8 assay and ampedance-based xCELLigence proliferation assay. The expression levels of RAGE in ASMCs were analyzed using western blotting assay. RESULTS: Results showed that exogenous S100A8 inhibited the PDGF-induced proliferation of rat ASMCs in a dose- dependent manner with the maximal effect at 1 µg/ml in vitro. Furthermore, when ASMCs was pre-treated with anti-RAGE neutralizing antibody, the inhibitory effect of S100A8 on PDGF-induced proliferation was significantly suppressed. In addition, neither the treatment with S100A8 or PDGF alone nor the pre-treatment with rS100A8 followed by PDGF stimulation affected the expression levels of RAGE. CONCLUSIONS: Our study demonstrated that S100A8 inhibits PDGF-induced ASMCs proliferation in a manner dependent on membrane receptor RAGE.


Subject(s)
Animals , Rats , Platelet-Derived Growth Factor/agonists , Myocytes, Smooth Muscle/drug effects , Calgranulin A/administration & dosage , Cell Proliferation/drug effects , Receptor for Advanced Glycation End Products/drug effects , Cells, Cultured
3.
Indian J Biochem Biophys ; 2013 Oct; 50(5): 419-427
Article in English | IMSEAR | ID: sea-150251

ABSTRACT

There is growing evidence that ouabain, a cardiotonic steroid may promote growth of cardiac and vascular myocytes, indicating its novel role in cell growth and proliferation, without appreciable inhibition of the sodium pump. The mechanism(s) by which low dose of ouabain produces pulmonary artery smooth muscle cell proliferation, a prerequisite for right ventricular hypertrophy, is currently unknown. Here, we analyzed the effects of low dose of ouabain (10 nM) on increase in [Ca2+]i, m-calpain and protein kinase C (PKC) activities on pulmonary artery smooth muscle cell proliferation and determined their sequential involvement in this scenario. We treated bovine pulmonary artery smooth muscle cells with a low dose of ouabain (10 nM) and determined [Ca2+]i in the cells by fluorometric assay using fura2-AM, m-calpain activity by fluorometric assay using SLLVY-AMC as the substrate, PKC activity using an assay kit and assay of Na+/K+ATPase activity spectrophotometrically. We purified m-calpain and PKCα by standard chromatographic procedure by HPLC and then studied cleavage of the purified PKCα by m-calpain using Western immunoblot method. Subsequently, we performed cell proliferation assay utilizing the redox dye resazunin. We used selective inhibitors of [Ca2+]i (BAPTA-AM), m-calpain (MDL28170), PKCα (Go6976) and determined their involvement in ouabain (10 nM)-mediated smooth muscle cell proliferation. Our results suggested that treatment of bovine pulmonary artery smooth muscle cells with a low dose of ouabain (10 nM) increased [Ca2+]i and subsequently stimulated m-calpain activity and proteolytically activated PKCα in caveolae (signaling microdomain also known as signalosomes) of the cells. Upon activation, PKCα increased the smooth muscle cell proliferation via Go/G1 to S/G2-M phase transition. Thus, [Ca2+]i-mCalpain-PKCα signaling axis plays a crucial role during low dose of ouabain-mediated pulmonary artery smooth muscle cell proliferation.


Subject(s)
Amino Acid Sequence , Animals , Calpain/metabolism , Cattle , Caveolae/drug effects , Caveolae/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Ouabain/pharmacology , Protein Kinase C-alpha/chemistry , Protein Kinase C-alpha/metabolism , Proteolysis/drug effects , Pulmonary Artery/cytology , Sodium-Potassium-Exchanging ATPase/metabolism
4.
Experimental & Molecular Medicine ; : e65-2013.
Article in English | WPRIM | ID: wpr-152459

ABSTRACT

Vascular smooth muscle cells (VSMCs) undergo phenotypic changes in response to vascular injury such as angioplasty. Protein kinase G (PKG) has an important role in the process of VSMC phenotype switching. In this study, we examined whether rosiglitazone, a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, could modulate VSMC phenotype through the PKG pathway to reduce neointimal hyperplasia after angioplasty. In vitro experiments showed that rosiglitazone inhibited the phenotype change of VSMCs from a contractile to a synthetic form. The platelet-derived growth factor (PDGF)-induced reduction of PKG level was reversed by rosiglitazone treatment, resulting in increased PKG activity. This increased activity of PKG resulted in phosphorylation of vasodilator-stimulated phosphoprotein at serine 239, leading to inhibited proliferation of VSMCs. Interestingly, rosiglitazone did not change the level of nitric oxide (NO) or cyclic guanosine monophosphate (cGMP), which are upstream of PKG, suggesting that rosiglitazone influences PKG itself. Chromatin immunoprecipitation assays for the PKG promoter showed that the activation of PKG by rosiglitazone was mediated by the increased binding of Sp1 on the promoter region of PKG. In vivo experiments showed that rosiglitazone significantly inhibited neointimal formation after balloon injury. Immunohistochemistry staining for calponin and thrombospondin showed that this effect of rosiglitazone was mediated by modulating VSMC phenotype. Our findings demonstrate that rosiglitazone is a potent modulator of VSMC phenotype, which is regulated by PKG. This activation of PKG by rosiglitazone results in reduced neointimal hyperplasia after angioplasty. These results provide important mechanistic insight into the cardiovascular-protective effect of PPARgamma.


Subject(s)
Animals , Rats , Aorta/injuries , Calcium-Binding Proteins/genetics , Cell Proliferation , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Hyperplasia/metabolism , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , PPAR gamma/agonists , Promoter Regions, Genetic , Rats, Sprague-Dawley , Sp1 Transcription Factor/metabolism , Thiazolidinediones/pharmacology , Thrombospondins/genetics , Tunica Intima/metabolism , Vascular System Injuries/metabolism
5.
Experimental & Molecular Medicine ; : e67-2013.
Article in English | WPRIM | ID: wpr-83998

ABSTRACT

Serotonin (5-hydroxytryptamine (5-HT)) is a neurotransmitter that regulates a variety of functions in the nervous, gastrointestinal and cardiovascular systems. Despite such importance, 5-HT signaling pathways are not entirely clear. We demonstrated previously that 4-aminopyridine (4-AP)-sensitive voltage-gated K+ (Kv) channels determine the resting membrane potential of arterial smooth muscle cells and that the Kv channels are inhibited by 5-HT, which depolarizes the membranes. Therefore, we hypothesized that 5-HT contracts arteries by inhibiting Kv channels. Here we studied 5-HT signaling and the detailed role of Kv currents in rat mesenteric arteries using patch-clamp and isometric tension measurements. Our data showed that inhibiting 4-AP-sensitive Kv channels contracted arterial rings, whereas inhibiting Ca2+-activated K+, inward rectifier K+ and ATP-sensitive K+ channels had little effect on arterial contraction, indicating a central role of Kv channels in the regulation of resting arterial tone. 5-HT-induced arterial contraction decreased significantly in the presence of high KCl or the voltage-gated Ca2+ channel (VGCC) inhibitor nifedipine, indicating that membrane depolarization and the consequent activation of VGCCs mediate the 5-HT-induced vasoconstriction. The effects of 5-HT on Kv currents and arterial contraction were markedly prevented by the 5-HT2A receptor antagonists ketanserin and spiperone. Consistently, alpha-methyl 5-HT, a 5-HT2 receptor agonist, mimicked the 5-HT action on Kv channels. Pretreatment with a Src tyrosine kinase inhibitor, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine, prevented both the 5-HT-mediated vasoconstriction and Kv current inhibition. Our data suggest that 4-AP-sensitive Kv channels are the primary regulator of the resting tone in rat mesenteric arteries. 5-HT constricts the arteries by inhibiting Kv channels via the 5-HT2A receptor and Src tyrosine kinase pathway.


Subject(s)
Animals , Male , Rats , 4-Aminopyridine/pharmacology , Action Potentials , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cells, Cultured , Ketanserin/pharmacology , Mesenteric Arteries/drug effects , Muscle Contraction , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Nifedipine/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Spiperone/pharmacology , Vasoconstriction , src-Family Kinases/antagonists & inhibitors
6.
Experimental & Molecular Medicine ; : 21-29, 2010.
Article in English | WPRIM | ID: wpr-104282

ABSTRACT

Angiotensin II is a major effector molecule in the development of cardiovascular disease. In vascular smooth muscle cells (VSMCs), angiotensin II promotes cellular proliferation and extracellular matrix accumulation through the upregulation of plasminogen activator inhibitor-1 (PAI-1) expression. Previously, we demonstrated that small heterodimer partner (SHP) represses PAI-1 expression in the liver through the inhibition of TGF-beta signaling pathways. Here, we investigated whether SHP inhibited angiotensin II-stimulated PAI-1 expression in VSMCs. Adenovirus-mediated overexpression of SHP (Ad-SHP) in VSMCs inhibited angiotensin II- and TGF-beta-stimulated PAI-1 expression. Ad-SHP also inhibited angiotensin II-, TGF-beta- and Smad3-stimulated PAI-1 promoter activity, and angiotensin II-stimulated AP-1 activity. The level of PAI-1 expression was significantly higher in VSMCs of SHP-/- mice than wild type mice. Moreover, loss of SHP increased PAI-1 mRNA expression after angiotensin II treatment. These results suggest that SHP inhibits PAI-1 expression in VSMCs through the suppression of TGF-beta/Smad3 and AP-1 activity. Thus, agents that target the induction of SHP expression in VSMCs might help prevent the development and progression of atherosclerosis.


Subject(s)
Animals , Humans , Mice , Rats , Adenoviridae/genetics , Angiotensin II/pharmacology , Blotting, Northern , Cells, Cultured , Electrophoretic Mobility Shift Assay , Genetic Vectors/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Transforming Growth Factor beta/pharmacology
7.
Experimental & Molecular Medicine ; : 757-764, 2009.
Article in English | WPRIM | ID: wpr-71507

ABSTRACT

Monocyte chemoattractant protein-1 (MCP1) plays a key role in monocyte/macrophage infiltration to the sub-endothelial space of the blood vessel wall, which is a critical initial step in atherosclerosis. In this study, we examined the intracellular signaling pathway of IL-1beta-induced MCP1 expression using various chemical inhibitors. The pretreatment of a phosphatidylcholine (PC)-specific PLC (PC-PLC) inhibitor (D609), PKC inhibitors, or an NF-kappaB inhibitor completely suppressed the IL-1beta-induced MCP1 expression through blocking NF-kappaB translocation to the nucleus. Pretreatment with inhibitors of tyrosine kinase or PLD partially suppressed MCP1 expression and failed to block nuclear NF-kappaB translocation. These results suggest that IL-1beta induces MCP1 expression through activation of NF-kappaB via the PC-PLC/PKC signaling pathway.


Subject(s)
Humans , Active Transport, Cell Nucleus/drug effects , Aorta/pathology , Atherosclerosis/immunology , Bridged-Ring Compounds/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Chemokine CCL2/biosynthesis , Estrenes/pharmacology , Genistein/pharmacology , Interleukin-1beta/metabolism , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Phospholipases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrrolidinones/pharmacology , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Thiones/pharmacology
8.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 27-32, 2008.
Article in English | WPRIM | ID: wpr-634576

ABSTRACT

In order to investigate the effects of puerarin on pulmonary vascular remodeling and protein kinase C-alpha (PKC-alpha) in chronic exposure smoke rats, 54 male Wistar rats were randomly divided into 7 groups: control group (C group), smoke exposure groups (S(4w) group, S(8w) group), puerarin groups (P(4w) group, P(8w) group), propylene glycol control groups (PC(4w) group, PC(8w) group). Rats were exposed to cigarette smoke or air for 4 to 8 weeks. Rats in puerarin groups also received puerarin. To evaluate vascular remodeling, alpha-smooth muscle actin (alpha-SM-actin) staining was used to count the percentage of completely muscularised vessels to intraacinar pulmonary arteries (CMA/IAPA) which was determined by morphometric analysis of histological sections. Pulmonary artery smooth muscle cell (PASMC) apoptosis was detected by in situ end labeling technique (TUNEL), and proliferation by proliferating cell nuclear antigen (PCNA) staining. Reverse transcription-polymerase chain reaction (RT-PCR), immunofluorescence staining and Western blot analysis were done to detect the PKC-alpha mRNA and protein expression in pulmonary arteries. The results showed that in cigarette smoke-exposed rats the percentage of CMA/IAPA and alpha-SM-actin expression were increased greatly, PASMC apoptosis was increased and proliferation was markedly increased; Apoptosis indices (AI) and proliferation indices (PI) were higher than in C group; AI and PI were correlated with vascular remodeling indices; The expression of PKC-alpha mRNA and protein in pulmonary arteries was significantly higher than in C group. In rats treated with puerarin, the percentage of CMA/IAPA and cell proliferation was reduced, whereas PASMC apoptosis was increased; The expression levels of PKC-alpha mRNA and protein were lower than in smoke exposure rats. There was no difference among all these data between S groups and PC groups. These findings suggested that cigarette smoke-induced pulmonary vascular remodeling was most likely an effect of the imbalance of PASMC proliferation and apoptosis. Puerarin appears to be able to reduce cell proliferation and vascular remodeling possibly through PKC signaling transduction pathway.


Subject(s)
Apoptosis , Cell Proliferation , Endothelium, Vascular/drug effects , Isoflavones/pharmacology , Lung/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Protein Kinase C-alpha/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Rats, Wistar , Smoking , Tobacco Smoke Pollution , Vasodilator Agents/pharmacology
9.
Yonsei Medical Journal ; : 230-236, 2008.
Article in English | WPRIM | ID: wpr-187376

ABSTRACT

PURPOSE: This study was designed to investigate the change of peroxisome proliferator-activated receptor gamma (PPARgamma) after the infection of the human coronary artery smooth muscle cells (HCSMCs) with Chlamydia pneumoniae (C. pneumoniae) and the effect of PPARgamma agonist on the expression of PPARgamma of C. pneumoniae-infected HCSMCs. MATERIALS AND METHODS: To determine the effect of PPARgamma agonist on the proliferation of C. pneumoniae-infected HCSMCs, rosiglitazone at various concentrations was applied 1 hour before inoculation of HCSMCs. RESULTS: The expression of PPARgamma mRNA in HCSMCs increased from 3 hours after C. pneumoniae infection and reached that of noninfected HCSMCs at 24 hours (p < 0.05). The expression of PPARgamma protein in HCSMCs also increased from 3 hours after C. pneumoniae and persisted until 24 hours as compared with that of noninfected HCSMCs (p < 0.05). The pretreatment of HCSMCs with rosiglitazone followed by the infection with C. pneumoniae augmented the expression of PPARgamma mRNA and protein (p < 0.05) and decreased cell proliferation. CONCLUSION: Our results showed that the expression of PPARgamma increases in response to C. pneumoniae infection and rosiglitazone further augmented the expression of PPARgamma. It is suggested that rosiglitazone could ameliorate the chronic inflammation in the vessel wall induced by C. pneumoniae by augmenting PPARgamma expression.


Subject(s)
Humans , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Chlamydophila pneumoniae/growth & development , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , PPAR gamma/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thiazolidinediones/pharmacology
10.
Yonsei Medical Journal ; : 592-600, 2008.
Article in English | WPRIM | ID: wpr-167115

ABSTRACT

PURPOSE: Thiazolidinediones (TZDs) are known to inhibit the proliferation of vascular smooth muscle cell (VSMC) by increasing the activity of p27(Kip1) and retinoblastoma protein (RB). However, the upstream signaling mechanisms associated with this pathway have not been elucidated. The Akt-mTOR-P70S6 kinase pathway is the central regulator of cell growth and proliferation, and increases cell proliferation by inhibiting the activities of p27(Kip1) and retinoblastoma protein (RB). Therefore, we hypothesized in this study that rosiglitazone inhibits VSMC proliferation through the inhibition of the Akt-TOR-P70S6K signaling pathway. MATERIALS and METHODS: Rat aortic smooth muscle cells (RAoSMCs) were treated with 10microM of rosiglitazone 24 hours before the addition of insulin as a mitogenic stimulus. Western blot analysis was performed to determine the inhibitory effect of rosiglitazone treatment on the Akt-mTOR-P70S6K signaling pathway. Carotid balloon injury was also performed in Otsuka Long-Evans Tokushima Fatty (OLETF) diabetic rats that were pretreated with 3 mg/kg of rosiglitazone. RESULTS: Western blot analysis demonstrated significant inhibition of activation of p-Akt, p-m-TOR, and p-p70S6K in cells treated with rosiglitazone. The inhibition of the activation of the p-mTOR-p-p70S6K pathway seemed to be mediated by both the upstream PI3K pathway and MEK-ERK complex. CONCLUSION: The inhibitory effect of rosiglitazone on RAoSMC proliferation in vitro and in vivo is mediated by the inhibition of the Akt-mTOR-P70S6K pathway.


Subject(s)
Animals , Male , Rats , Cell Proliferation/drug effects , Cells, Cultured , Cytoprotection/drug effects , Enzyme Activation/drug effects , Insulin/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Thiazolidinediones/pharmacology
11.
Experimental & Molecular Medicine ; : 267-277, 2007.
Article in English | WPRIM | ID: wpr-201428

ABSTRACT

In vascular smooth muscle cells (VSMCs), induction of the heme oxygenase-1 (HO-1) confers vascular protection against cellular proliferation mainly via its up-regulation of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) that is involved in negative regulation of cellular proliferation. In the present study, we investigated whether the phytochemical curcumin and its metabolite tetrahydrocurcumin could induce HO-1 expression and growth inhibition in rat VSMCs and, if so, whether their antiproliferative effect could be mediated via HO-1 expression. At non-toxic concentrations, curcumin possessing two Michael-reaction acceptors induced HO-1 expression by activating antioxidant response element (ARE) through translocation of the nuclear transcription factor E2-related factor-2 (Nrf2) into the nucleus and also inhibited VSMC growth triggered by 5% FBS in a dose-dependent manner. In contrast, tetrahydrocurcumin lacking Michael-reaction acceptor showed no effect on HO-1 expression, ARE activation and VSMC growth inhibition. The antiproliferative effect of curcumin in VSMCs was accompanied by the increased expression of p21(WAF1/CIP1). Inhibition of VSMC growth and expression of p21(WAF1/CIP1) by curcumin were partially, but not completely, abolished when the cells were co- incubated with the HO inhibitor tin protoporphyrin. In human aortic smooth muscle cells (HASMCs), curcumin also inhibited growth triggered by TNF-alpha and increased p21(WAF1/CIP1) expression via HO-1-dependent manner. Our findings suggest that curcumin has an ability to induce HO-1 expression, presumably through Nrf2-dependent ARE activation, in rat VSMCs and HASMCs, and provide evidence that the antiproliferative effect of curcumin is considerably linked to its ability to induce HO-1 expression.


Subject(s)
Animals , Humans , Rats , Active Transport, Cell Nucleus , Aorta/cytology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Curcumin/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1/biosynthesis , Metalloporphyrins/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/metabolism , Protoporphyrins/pharmacology , Regulatory Sequences, Nucleic Acid , Response Elements , Tumor Necrosis Factor-alpha/pharmacology
12.
Braz. j. med. biol. res ; 39(6): 739-748, June 2006. graf
Article in English | LILACS | ID: lil-428284

ABSTRACT

Store-operated Ca2+ entry plays an important role in Ca2+ homeostasis in cells but the mechanisms of control of these channels are not completely understood. We describe an investigation of the role of the CD38-cyclic-ADP-ribose (cADPR)-ryanodine-channel (RyR) signaling pathway in store-operated Ca2+ entry in human smooth muscle. We observed that human myometrial cells have a functional store-operated Ca2+ entry mechanism. Furthermore, we observed the presence of transient receptor potential 1, 3, 4, 5, and 6 ion channels in human myometrial cells. Store-operated Ca2+ transient was inhibited by at least 50-70 percent by several inhibitors of the RyR, including ryanodine (10 µM), dantrolene (10 µM), and ruthenium red (10 µM). Furthermore, the cell permeable inhibitor of the cADPR-system, 8-Br-cADPR (100 µM), is a potent inhibitor of the store-operated entry, decreasing the store operated entry by 80 percent. Pre-incubation of cells with 100 µM cADPR and the hydrolysis-resistant cADPR analog 3-deaza-cADPR (50 µM), but not with ADP-ribose (ADPR) leads to a 1.6-fold increase in the store-operated Ca2+ transient. In addition, we observed that nicotinamide (1-10 mM), an inhibitor of cADPR synthesis, also leads to inhibition of the store-operated Ca2+ transient by 50-80 percent. Finally, we observed that the transient receptor potential channels, RyR, and CD38 can be co-immunoprecipitated, indicating that they interact in vivo. Our observations clearly implicate the CD38-cADPR-ryanodine signaling pathway in the regulation of store-operated Ca2+ entry in human smooth muscle cells.


Subject(s)
Female , Humans , Calcium/metabolism , Cyclic ADP-Ribose/metabolism , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , Blotting, Western , Calcium/pharmacology , Cyclic ADP-Ribose/antagonists & inhibitors , Cyclic ADP-Ribose/pharmacology , Immunohistochemistry , Immunoprecipitation , Myocytes, Smooth Muscle/drug effects , Myometrium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction , Transient Receptor Potential Channels/metabolism
13.
Yakhteh Medical Journal. 2006; 8 (1): 1-6
in Persian | IMEMR | ID: emr-81573

ABSTRACT

The aim of this study is to investigate the effect of one nitric oxide inhibitor on the thickness and number of circular smooth muscle cells of pylor in rat embryo. To determine the influence of nitric oxide reduction on the muscular layer of pylor in rat embryo, in the present study, pregnant Sprague-Dawley rats received the NO synthase inhibitor N-nitro-L-arginine methyl ester [L-NAME]. A dose of 80mg/kg of L-NAME solution in saline was injected to the rats intraperitoneally [IP] during the middle week and last week of the pregnancy period per day. The embryos were removed on the expected day of delivery. Their stomach and duodenum were dissected, fixed by Bouin solution and tissue processing was carried out. By using a rotary microtome 5 micro serial cross sections were obtained and stained with Trichrom-Mason and Pop-Nicola. Then sections were evaluated for thickness and number of circular smooth muscle cells under a light microscope using a scaled lens and a checkered lens eye-piece. Statistical analysis [One-Way ANOVA- Duncan]of light microscopic findings indicated that 80mg/kg of L-NAME in the last week of pregnancy [the first trial group] results in pyloric hypertrophy and hyperplasia. On the basis of these results we believe that reductions of nitric oxide production in the third trimester of pregnancy could be one of the reasons of pyloric stenosis in infants


Subject(s)
Animals, Laboratory , Myocytes, Smooth Muscle/drug effects , Muscle, Smooth , Pylorus/drug effects , Embryonic Structures/drug effects , Rats, Sprague-Dawley , Nitric Oxide , Nitric Oxide Synthase
14.
Experimental & Molecular Medicine ; : 153-161, 2006.
Article in English | WPRIM | ID: wpr-15696

ABSTRACT

Adhesion and migration of vascular smooth muscle cells (VSMCs) play an important role in the pathogenesis of atherosclerosis. These processes involve the interaction of VSMCs with extracellular matrix proteins. Here, we investigated integrin isoforms and signaling pathways mediating the adhesion and migration of VSMCs on betaig-h3, a transforming growth factor (TGF)-beta-inducible extracellular matrix protein that is elevated in atherosclerotic plaques. Adhesion assays showed that the alphavbeta5 integrin is a functional receptor for the adhesion of aortic VSMCs to betaig-h3. An YH18 motif containing amino acids between 563 and 580 of betaig-h3 was an essential motif for the adhesion and growth of VSMCs. Interaction between the YH18 motif and the alphavbeta5 integrin was responsible for the migration of VSMCs on betaig-h3. Inhibitors of phosphatidylinositide 3-kinase, extracellular signal-regulated kinase (ERK), and Src kinase reduced the adhesion and migration of VSMCs on betaig-h3. betaig-h3 triggered phosphorylation and activation of AKT, ERK, focal adhesion kinase, and paxillin mediating the adhesion and migration of VSMCs. Taken together, these results suggest that betaig-h3 and alphavbeta5 integrin play a role in the adhesion and migration of VSMCs during the pathogenesis of atherosclerosis.


Subject(s)
Humans , Animals , src-Family Kinases/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Signal Transduction/physiology , Receptors, Vitronectin/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Paxillin/metabolism , Myocytes, Smooth Muscle/drug effects , Muscle, Smooth, Vascular/cytology , Morpholines/pharmacology , Molecular Sequence Data , Integrins/genetics , Flavonoids/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Enzyme Inhibitors/pharmacology , Chromones/pharmacology , Cells, Cultured , Cell Movement/physiology , Cell Adhesion/physiology , Amino Acid Sequence , Amino Acid Motifs/genetics , Phosphatidylinositol 3-Kinase/antagonists & inhibitors
15.
Asian Pac J Allergy Immunol ; 2005 Jun-Sep; 23(2-3): 107-13
Article in English | IMSEAR | ID: sea-36897

ABSTRACT

Vascular replacement in vital organs is sometimes necessary for human life for example because of atherosclerosis. Blood vessel tissue engineering is applied for autologous transplantations to avoid graft rejections. Stem cells are used for blood vessel tissue engineering because they are the origin of smooth muscle cells, endothelial cells and fibroblasts. This paper shows that bone marrow stromal cells (BMSCs) can be induced to differentiate into the early stage of smooth muscle cells by using 0.01 microM retinoic acid. The differentiation of BMSCs to smooth muscle cells was detected by the expression of smooth muscle alpha actin (SM alpha-actin), the earliest smooth muscle cell marker. The SM alpha-actin marker expression was demonstrated using indirect immunofluorescence technique and Western blot analysis. The induction of BMSC to form early stages of smooth muscle cells in this study is appropriate for blood vessel tissue engineering because the early stage smooth muscle cells may be stimulated to develop vascular walls with endothelial cells using a co-culture system.


Subject(s)
Actins/drug effects , Blotting, Western , Bone Marrow Cells/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Humans , Keratolytic Agents/administration & dosage , Myocytes, Smooth Muscle/drug effects , Stromal Cells/drug effects , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Tretinoin/administration & dosage
16.
The Korean Journal of Gastroenterology ; : 388-395, 2005.
Article in Korean | WPRIM | ID: wpr-165587

ABSTRACT

BACKGROUND/AIMS: Tamoxifen is a widely used anticancer drug for breast cancer with frequent gastrointestinal side effects. Changes in gastrointestinal motility is associated with altered activities of membrane ion channels. Ion channels have important role in regulating membrane potential and cell excitability. This study was performed to investigate the effects of tamoxifen on the membrane ionic currents in colonic smooth muscle cells. METHODS: Murine colonic smooth muscle cells were isolated from the proximal colon using collagenase, and the membrane currents were recorded using a whole-cell patch clamp technique. RESULTS: Two types of voltage-dependent K+ currents were recorded (A-type and delayed rectifier K+ currents). Tamoxifen inhibited both types of voltage-dependent K+ currents in a dose-dependent manner. However, tamoxifen did not change the half-inactivation potential and the recovery time of voltage-dependent K+ currents. Chelerythrine, a protein kinase C inhibitor or phorbol 12, 13-dibutyrate, a protein kinase C activator did not affect the voltage-dependent K+ currents. Guanosine 5'-O-(2-thio-diphosphate) did not affect the tamoxifen-induced inhibition of voltage-dependent K+ currents. Tamoxifen inhibited voltage-dependent Ca2+ currents completely in whole-test ranges. CONCLUSIONS: These results suggest that tamoxifen can alter various membrane ionic currents in smooth muscle cells and cause some adverse effects on the gastrointestinal motility.


Subject(s)
Animals , Mice , Antineoplastic Agents, Hormonal/pharmacology , Calcium Channels/drug effects , Colon/drug effects , English Abstract , In Vitro Techniques , Membrane Potentials , Myocytes, Smooth Muscle/drug effects , Potassium Channels/drug effects , Tamoxifen/pharmacology
17.
Biocell ; 28(1): 21-30, Apr. 2004.
Article in English | LILACS | ID: lil-384231

ABSTRACT

The flutamide antiandrogenic effects on the Guinea pig male prostate morphology in puberal, post-puberal and adult ages were evaluated in the present study. Daily-treated group animals received flutamide subcutaneous injection at a dose of 10 mg/Kg body weight for 10 days. The control group animals received a pharmacological vehicle under the same conditions. The lateral prostate was removed, fixed and processed for light and transmission electron microscopy. The results revealed an increase of the acinus diameter in the treated puberal animals and straitness in the stromal compartment around the acini. The epithelial cells exhibited cubic phenotype. In the post-puberal and adult animals, a decrease of the acinus diameter was observed, as well as an increase of the smooth muscle layer and presence of the folds at epithelium. The ultrastructural evaluation of the secretory cells in the treated group demonstrated endomembrane enlargement, mainly in the rough endoplasmic reticulum and Golgi apparatus. In addition, a decrease of the microvilli and alterations in the distribution patterns and density of the stromal fibrillar components were observed. In conclusion, the flutamide treatment exerts tissue effects on the lateral prostate, promoting stroma/epithelium alterations.


Subject(s)
Androgen Antagonists/pharmacology , Epithelial Cells/drug effects , Flutamide/pharmacology , Prostate/drug effects , Age Factors , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Epithelial Cells/ultrastructure , Stromal Cells/drug effects , Stromal Cells/ultrastructure , Guinea Pigs , Microscopy, Electron , Microvilli/drug effects , Microvilli/ultrastructure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/ultrastructure , Prostate/ultrastructure , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Sexual Maturation , Cell Size/drug effects , Cell Size/physiology
18.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 356-8, 368, 2003.
Article in English | WPRIM | ID: wpr-640963

ABSTRACT

We investigated the expression of heme oxygenase-1 (HO-1) gene and production of endogenous carbon monoxide (CO) in the rat lung tissue at different time points of chronic hypoxic pulmonary hypertension and the effect of hemin on the expression of HO-1 gene and pulmonary hypertension. A rat model of hypoxic pulmonary hypertension was recreated by exposure to intermittent normobaric hypoxic environment (10% O2). Reverse transcriptase polymerase chain reaction (RT-PCR) was performed to determine the level of HO-1 mRNA in the rat lung tissue and double wave length spectrophotometry was used to evaluate the quantity of COHb in arterial blood. Cardiac catheterization was employed to measure the right ventricular systolic pressure (RVSP) and HE staining was performed in dissected lung tissue to observe the pathological changes of the intra-acinar pulmonary arteries (IAPA). It was found that (1) There was a low level of HO-1 mRNA in normal rat lung tissue, but the level of HO-1 mRNA increased by 2-4 times in the lung tissue of hypoxic rats (P<0.01). The quantity of COHb was 2-3 times those of control group (P<0.01 or P<0.05). These were accompanied by the increased of RVSP and the thickened IAPA; (2) Hemin could keep the HO-1 mRNA and COHb in the hypoxic rat lung tissue at a high level, and partially suppressed the increase of rat RVSP, thereby ameliorating the pathological changes of IAPA. In conclusion, the upregulation of the expression of HO-1 gene and production of CO in the rat lung of hypoxic pulmonary hypertension plays a role of inhibition in the development of hypoxic pulmonary hypertension. Hemin has a therapeutic effect on hypoxic pulmonary hypertension.


Subject(s)
Hypoxia/complications , Carbon Monoxide/metabolism , Carbon Monoxide/physiology , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Lung/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL