Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Circ Res ; 125(2): 152-166, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31096851

ABSTRACT

RATIONALE: Neointimal hyperplasia is characterized by excessive accumulation of vascular smooth muscle cells (SMCs) leading to occlusive disorders, such as atherosclerosis and stenosis. Blood vessel injury increases growth factor secretion and matrix synthesis, which promotes SMC proliferation and neointimal hyperplasia via FAK (focal adhesion kinase). OBJECTIVE: To understand the mechanism of FAK action in SMC proliferation and neointimal hyperplasia. METHODS AND RESULTS: Using combined pharmacological FAK catalytic inhibition (VS-4718) and SMC-specific FAK kinase-dead (Myh11-Cre-ERT2) mouse models, we report that FAK regulates SMC proliferation and neointimal hyperplasia in part by governing GATA4- (GATA-binding protein 4) cyclin D1 signaling. Inhibition of FAK catalytic activity facilitates FAK nuclear localization, which is required for proteasome-mediated GATA4 degradation in the cytoplasm. Chromatin immunoprecipitation identified GATA4 binding to the mouse cyclin D1 promoter, and loss of GATA4-mediated cyclin D1 transcription diminished SMC proliferation. Stimulation with platelet-derived growth factor or serum activated FAK and redistributed FAK from the nucleus to cytoplasm, leading to concomitant increase in GATA4 protein and cyclin D1 expression. In a femoral artery wire injury model, increased neointimal hyperplasia was observed in parallel with elevated FAK activity, GATA4 and cyclin D1 expression following injury in control mice, but not in VS-4718-treated and SMC-specific FAK kinase-dead mice. Finally, lentiviral shGATA4 knockdown in the wire injury significantly reduced cyclin D1 expression, SMC proliferation, and neointimal hyperplasia compared with control mice. CONCLUSIONS: Nuclear enrichment of FAK by inhibition of FAK catalytic activity during vessel injury blocks SMC proliferation and neointimal hyperplasia through regulation of GATA4-mediated cyclin D1 transcription.


Subject(s)
Cell Proliferation , Cyclin D1/metabolism , Focal Adhesion Kinase 1/metabolism , GATA4 Transcription Factor/metabolism , Myocytes, Smooth Muscle/metabolism , Tunica Intima/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cyclin D1/genetics , Focal Adhesion Kinase 1/antagonists & inhibitors , Hyperplasia/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/physiology , Tunica Intima/pathology
2.
Am J Physiol Heart Circ Physiol ; 311(4): H904-H912, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27521420

ABSTRACT

Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-ß-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.


Subject(s)
Aorta/metabolism , Contractile Proteins/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/drug effects , Blotting, Western , Cattle , Chromatography, Liquid , Contractile Proteins/drug effects , Contractile Proteins/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , Immunoprecipitation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/drug effects , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Rats , Serum Response Factor/drug effects , Serum Response Factor/genetics , Serum Response Factor/metabolism , Tandem Mass Spectrometry , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Am J Physiol Cell Physiol ; 307(7): C648-56, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25099734

ABSTRACT

The low-voltage-activated T-type Ca(2+) channels play an important role in mediating the cellular responses to altered oxygen tension. Among three T-type channel isoforms, α1G, α1H, and α1I, only α1H was found to be upregulated under hypoxia. However, mechanisms underlying such hypoxia-dependent isoform-specific gene regulation remain incompletely understood. We, therefore, studied the hypoxia-dependent transcriptional regulation of α1G and α1H gene promoters with the aim to identify the functional hypoxia-response elements (HREs). In rat pulmonary artery smooth muscle cells (PASMCs) and pheochromocytoma (PC12) cells after hypoxia (3% O2) exposure, we observed a prominent increase in α1H mRNA at 12 h along with a significant rise in α1H-mediated T-type current at 24 and 48 h. We then cloned two promoter fragments from the 5'-flanking regions of rat α1G and α1H gene, 2,000 and 3,076 bp, respectively, and inserted these fragments into a luciferase reporter vector. Transient transfection of PASMCs and PC12 cells with these recombinant constructs and subsequent luciferase assay revealed a significant increase in luciferase activity from the reporter containing the α1H, but not α1G, promoter fragment under hypoxia. Using serial deletion and point mutation analysis strategies, we identified a functional HRE at site -1,173cacgc-1,169 within the α1H promoter region. Furthermore, an electrophoretic mobility shift assay using this site as a DNA probe demonstrated an increased binding activity to nuclear protein extracts from the cells after hypoxia exposure. Taken together, these findings indicate that hypoxia-induced α1H upregulation involves binding of hypoxia-inducible factor to an HRE within the α1H promoter region.


Subject(s)
Calcium Channels, T-Type/genetics , Transcription, Genetic , Animals , Binding Sites , Calcium Channels, T-Type/metabolism , Cell Hypoxia , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , PC12 Cells , Pulmonary Artery/metabolism , Pulmonary Veins/metabolism , RNA, Messenger/metabolism , Rats , Response Elements , Time Factors , Transfection , Up-Regulation
4.
Biochim Biophys Acta ; 1823(10): 1666-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22766303

ABSTRACT

The transcriptional activator ß-catenin is a key mediator of the canonical Wnt signaling pathway. ß-catenin itself does not bind DNA but functions via interaction with T-cell factor (TCF)/lymphoid-enhancing factor (LEF) transcription factors. Thus, in the case of active Wnt signaling, ß-catenin, in cooperation with TCF/LEF proteins family, activates the expression of a wide variety of genes. To date, the list of established ß-catenin interacting targets is far from complete. In this study, we aimed to establish the interaction between ß-catenin and transcription factors that might affect TCF activity. We took advantage of EMSA, using TCF as a probe, to screen oligonucleotides known to bind specific transcription factors that might dislodge or antagonize ß-catenin/TCF binding. We found that Sox9 and KLF4 antagonize ß-catenin/TCF binding in HEK293, A549, SW480, and T47D cells. This inhibition of TCF binding was concentration-dependent and correlated to the in vitro TCF-luciferase functional assays. Overexpression of Sox9 and KLF4 transcription factors in cancer cells shows a concentration-dependent reduction of TCF-luciferase as well as the TCF-binding activities. In addition, we demonstrated that both Sox9 and KLF4 interact with ß-catenin in an immunoprecipitation assay and reduce its binding to TCF4. Together, these results demonstrate that Sox9 and KLF4 transcription factors antagonize ß-catenin/TCF in cancer cells.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Kruppel-Like Transcription Factors/metabolism , SOX9 Transcription Factor/metabolism , Transcription Factors/metabolism , beta Catenin/antagonists & inhibitors , Binding, Competitive/drug effects , Cell Line, Tumor , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Luciferases/metabolism , Oligonucleotides/pharmacology , Protein Binding/drug effects , Transcription Factor 4 , beta Catenin/metabolism
5.
Am J Physiol Regul Integr Comp Physiol ; 304(9): R734-43, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23447135

ABSTRACT

Chronic hypoxia attenuates soluble guanylate cyclase-induced vasorelaxation in serotonin (5-HT)-contracted ovine carotid arteries. Because protein kinase G (PKG) mediates many effects of soluble guanylate cyclase activation through phosphorylation of multiple kinase targets in vascular smooth muscle, we tested the hypothesis that chronic hypoxia reduces the ability of PKG to phosphorylate its target proteins, which attenuates the ability of PKG to induce vasorelaxation. We also tested the hypothesis that hypoxia attenuates PKG expression and/or activity. Arteries from normoxic and chronically hypoxic (altitude of 3,820 m for 110 days) fetal and adult sheep were denuded of endothelium and equilibrated with 95% O2-5% CO2 in the presence of nitro-l-arginine methyl ester (l-NAME) and N(G)-nitro-l-arginine (l-NNA) to inhibit residual endothelial nitric oxide synthase. Concentration-response relations for 5-HT were determined in the presence of prazosin to minimize activation of α-adrenergic receptors. The PKG activator 8-(p-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate (8-pCTP-cGMP) reduced agonist binding affinity of the 5-HT receptor in a concentration-dependent manner that was attenuated by hypoxia. Expression and activity of PKG-I was not significantly affected by chronic hypoxia in either fetal or adult arteries, although PKG-I abundance was greater in fetal arteries. Pretreatment with the large conductance calcium-sensitive potassium channel (BK) inhibitor iberiotoxin attenuated the vasorelaxation induced by 8-pCPT-cGMP in normoxic but not chronically hypoxic arteries. These results support the hypothesis that hypoxia attenuates the vasorelaxant effects of PKG through suppression of the ability of PKG to activate large conductance calcium-sensitive potassium channels in arterial smooth muscle. The results also reveal that this hypoxic effect is greater in fetal than adult arteries and that chronic maternal hypoxia can profoundly affect fetal vascular function.


Subject(s)
Carotid Arteries/drug effects , Cyclic GMP-Dependent Protein Kinases/physiology , Hypoxia/physiopathology , Muscle, Smooth, Vascular/drug effects , Serotonin/physiology , Aging/physiology , Animals , Blotting, Western , Chronic Disease , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dose-Response Relationship, Drug , Endpoint Determination , Female , Fetus/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Phosphorylation , Pregnancy , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/physiology , Serotonin/pharmacology , Serotonin Receptor Agonists/metabolism , Serotonin Receptor Agonists/pharmacology , Sheep , Thionucleotides/pharmacology
6.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L64-74, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22582112

ABSTRACT

Persistent hypoxic pulmonary vasoconstriction (HPV) plays a significant role in the pathogenesis of pulmonary hypertension, which is an emerging clinical problem around the world. We recently showed that hypoxia-induced activation of glucose-6-phosphate dehydrogenase (Glc-6-PD) in pulmonary artery smooth muscle links metabolic changes within smooth muscle cells to HPV and that inhibition of Glc-6PD reduces acute HPV. Here, we demonstrate that exposing pulmonary arterial rings to hypoxia (20-30 Torr) for 12 h in vitro significantly (P < 0.05) reduces (by 30-50%) SM22α and smooth muscle myosin heavy chain expression and evokes HPV. Glc-6-PD activity was also elevated in hypoxic pulmonary arteries. Inhibition of Glc-6-PD activity prevented the hypoxia-induced reduction in SM22α expression and inhibited HPV by 80-90% (P < 0.05). Furthermore, Glc-6-PD and protein kinase G (PKG) formed a complex in pulmonary artery, and Glc-6-PD inhibition increased PKG-mediated phosphorylation of VASP (p-VASP). In turn, increasing PKG activity upregulated SM22α expression and attenuated HPV evoked by Glc-6-PD inhibition. Increasing passive tension (from 0.8 to 3.0 g) in hypoxic arteries for 12 h reduced Glc-6-PD, increased p-VASP and SM22α levels, and inhibited HPV. The present findings indicate that increases in Glc-6-PD activity influence PKG activity and smooth muscle cell phenotype proteins, all of which affect pulmonary artery contractility and remodeling.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/physiology , Animals , Biomarkers/metabolism , Cattle , Cell Hypoxia/physiology , Down-Regulation , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Phosphorylation/genetics , Pulmonary Artery/metabolism , Vasoconstriction/genetics , Vasoconstriction/physiology
7.
Mol Cell Biochem ; 368(1-2): 27-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22618526

ABSTRACT

Degradation and resynthesis of the extracellular matrix (ECM) are essential during tissue remodeling. Expansion of the vascular intima in atherosclerosis and restenosis following injury is dependent upon smooth muscle cell (SMC) proliferation and migration. The migration of SMC from media to intima critically depends on degradation of ECM protein by matrix metalloproteinases (MMPs). MMP inhibitors and eNOS gene transfer have been shown to inhibit SMC migration in vitro and neointima formation in vivo. Nitric oxide (NO) and cyclic-GMP have been implicated in the inhibition of VSMC migration. But, there are few studies addressing the role of NO signaling pathways on the expression of MMPs. Here we reported the involvement of cyclic-GMP-dependent protein kinase (PKG) (an important mediator of NO and cGMP signaling pathway in VSMC) on MMP-2 expression in rat aortic SMC. The goal of the present study was to gain insight into the possible involvement of PKG on MMP-2 in rat aortic SMC. MMP-2 protein and mRNA level and activity were downregulated in PKG-expressing cells as compared to PKG-deficient cells. In addition, the secretion of tissue inhibitor of metalloproteinase-2 (TIMP-2) was increased in PKG-expressing cells as compared to PKG-deficient cells. PKG-specific membrane permeable peptide inhibitor (DT-2) reverses the process. Interestingly, little or no changes of MMP-9 were observed throughout the study. Taken together our data suggest the possible role of PKG in the suppression of MMP-2.


Subject(s)
Aorta/enzymology , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Matrix Metalloproteinase 2/biosynthesis , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Aorta/cytology , Cell Movement/physiology , Cell Proliferation , Cyclic GMP/metabolism , Male , Matrix Metalloproteinase 9/biosynthesis , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/blood , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-2/biosynthesis
8.
Am J Physiol Cell Physiol ; 298(5): C1188-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20164378

ABSTRACT

Diabetes is a major predictor of in-stent restenosis, which is associated with fibroproliferative remodeling of the vascular wall due to increased transforming growth factor-beta (TGF-beta) action. It is well established that thrombospondin1 (TSP1) is a major regulator of TGF-beta activation in renal and cardiac complications of diabetes. However, the role of the TSP1-TGF-beta pathway in macrovascular diabetic complications, including restenosis, has not been addressed. In mesangial cells, high glucose concentrations depress protein kinase G (PKG) activity, but not PKG-I protein, thereby downregulating transcriptional repression of TSP1. Previously, we showed that high glucose downregulates PKG-I protein expression by vascular smooth muscle cells (VSMCs) through altered NADPH oxidase signaling. In the present study, we investigated whether high glucose regulation of PKG protein and activity in VSMCs similarly regulates TSP1 expression and downstream TGF-beta activity. These studies showed that high glucose stimulates both TSP1 expression and TGF-beta bioactivity in primary murine aortic smooth muscle cells (VSMCs). TSP1 is responsible for the increased TGF-beta bioactivity under high glucose conditions, because treatment with anti-TSP1 antibody, small interfering RNA-TSP1, or an inhibitory peptide blocked glucose-mediated increases in TGF-beta activity and extracellular matrix protein (fibronectin) expression. Overexpression of constitutively active PKG, but not the PKG-I protein, inhibited glucose-induced TSP1 expression and TGF-beta bioactivity, suggesting that PKG protein expression is insufficient to regulate TSP1 expression. Together, these data establish that glucose-mediated downregulation of PKG levels stimulates TSP1 expression and enhances TGF-beta activity and matrix protein expression, which can contribute to vascular remodeling in diabetes.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Glucose/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/genetics , Diabetes Mellitus , Down-Regulation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Enzymologic , Mice , Mice, Inbred C57BL , NADPH Oxidases , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/genetics , Up-Regulation
9.
Am J Physiol Heart Circ Physiol ; 299(5): H1660-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20802137

ABSTRACT

Although the regulation of smooth muscle cell (SMC) gene expression by cGMP-dependent protein kinase (PKG) is now recognized, the mechanisms underlying these effects are not fully understood. In this study, we report that PKG-I stimulates myocardin/serum response factor (SRF)-dependent gene expression in vascular SMCs. The expression of PKG in PKG-deficient cells enhanced myocardin-induced SM22 promoter activity in a concentration-dependent fashion. However, neither SRF nor myocardin expression was affected. To investigate alternative mechanisms, we examined whether PKG affects the phosphorylation of E26-like protein-1 (Elk-1), a SRF/myocardin transcription antagonist. The activation of PKG caused an increase in a higher molecular mass form of phospho-Elk-1 that was determined to be small ubiquitin-related modifier (sumo)ylated Elk-1. PKG increased Elk-1 sumoylation twofold compared with the PKG-deficient cells, and Elk-1 sumoylation was reduced using dominant-negative sumo-conjugating enzyme, DN-Ubc9, confirming PKG-dependent sumoylation of phospho-Elk-1 in vascular SMCs. In addition, PKG stimulated Elk-1 sumoylation in COS-7 cells overexpressing Elk-1, sumo-1, and PKG-I. The increased expression of PKG in vascular SMCs inhibited Elk-1 binding to SMC-specific promoters, SM22 and smooth muscle myosin heavy chain, as measured by EMSA and chromatin immunoprecipitation assay, and PKG suppressed the Elk-1 inhibition of SM22 reporter gene expression. Taken together, these data suggest that PKG-I decreases Elk-1 activity by sumo modification of Elk-1, thereby increasing myocardin-SRF activity on SMC-specific gene expression.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/pharmacology , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Expression Regulation/physiology , Microfilament Proteins/metabolism , Models, Animal , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Trans-Activators/metabolism
10.
J Appl Physiol (1985) ; 107(1): 192-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19407253

ABSTRACT

A broad variety of evidence obtained largely in pulmonary vasculature suggests that chronic hypoxia modulates vasoreactivity to nitric oxide (NO). The present study explores the general hypothesis that chronic hypoxia also modulates cerebrovascular reactivity to NO, and does so by modulating the activity of soluble guanylate cyclase (sGC), the primary target for NO in vascular smooth muscle. Pregnant and nonpregnant ewes were maintained at either sea level or at 3,820 m for the final 110 days of gestation, at which time middle cerebral arteries from term fetal lambs and nonpregnant adults were harvested. In both fetal and adult arteries, NO-induced vasodilatation was attenuated by chronic hypoxia and completely inhibited by 10 microM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), a selective inhibitor of sGC. sGC abundance (in ng sGC/mg protein) measured via Western immunoblots was approximately 10-fold greater in fetal (17.6 +/- 1.6) than adult (1.7 +/- 0.3) arteries but was not affected by chronic hypoxia. The specific activity of sGC (in pmol cGMP.microg sGC(-1).min(-1)) was similar in fetal (255 +/- 64) and adult (280 +/- 75) arteries and was inhibited by chronic hypoxia in both fetal (120 +/- 10) and adult (132 +/- 26) arteries. Rates of cGMP degradation (in pmol cGMP.mg protein(-1).min(-1)) were similar in fetal (159 +/- 59) and adult (134 +/- 36) arteries but were not significantly depressed by chronic hypoxia in either fetal (115 +/- 25) or adult (108 +/- 25) arteries. The cGMP analog 8-(p-chlorophenylthio)-cGMP was a more potent vasorelaxant in fetal (pD(2) = 4.7 +/- 0.1) than adult (pD(2) = 4.3 +/- 0.1) arteries, but its ability to promote vasodilatation was not affected by chronic hypoxia in either age group. Together, these results reveal that hypoxic inhibition of NO-induced vasodilatation is attributable largely to attenuation of the specific activity of sGC and does not involve significant changes in sGC abundance, cGMP-phosphodiesterase activity, or the vasorelaxant activity of protein kinase G.


Subject(s)
Cerebral Arteries/enzymology , Fetus/blood supply , Guanylate Cyclase/metabolism , Hypoxia/enzymology , Receptors, Cytoplasmic and Nuclear/metabolism , Sheep/physiology , Altitude Sickness/complications , Altitude Sickness/enzymology , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/physiology , Chronic Disease , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endothelium-Dependent Relaxing Factors/pharmacology , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , Hypoxia/etiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Nitric Oxide/pharmacology , Oxadiazoles/pharmacology , Pregnancy , Quinoxalines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Vasodilation/drug effects , Vasodilation/physiology
11.
Circ Res ; 97(4): 329-36, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-16037573

ABSTRACT

Ischemic and pharmacological preconditioning can be triggered by an intracellular signaling pathway in which Gi-coupled surface receptors activate a cascade including phosphatidylinositol 3-kinase, endothelial nitric oxide synthase, guanylyl cyclase, and protein kinase G (PKG). Activated PKG opens mitochondrial KATP channels (mitoKATP) which increase production of reactive oxygen species. Steps between PKG and mitoKATP opening are unknown. We describe effects of adding purified PKG and cGMP on K+ transport in isolated mitochondria. Light scattering and respiration measurements indicate PKG induces opening of mitoKATP similar to KATP channel openers like diazoxide and cromakalim in heart, liver, and brain mitochondria. This effect was blocked by mitoKATP inhibitors 5-hydroxydecanoate, tetraphenylphosphonium, and glibenclamide, PKG-selective inhibitor KT5823, and protein kinase C (PKC) inhibitors chelerythrine, Ro318220, and PKC-epsilon peptide antagonist epsilonV(1-2). MitoKATP are opened by the PKC activator 12-phorbol 13-myristate acetate. We conclude PKG is the terminal cytosolic component of the trigger pathway; it transmits the cardioprotective signal from cytosol to inner mitochondrial membrane by a pathway that includes PKC-epsilon.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Cytosol/metabolism , Ischemic Preconditioning, Myocardial , Mitochondria, Heart/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/pharmacology , Animals , Brain/metabolism , Cyclic GMP/physiology , Male , Mitochondria, Liver/metabolism , Oxygen Consumption , Potassium Channels/physiology , Protein Kinase C/physiology , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology , Tetraethylammonium Compounds/pharmacology
12.
Front Biosci ; 11: 356-67, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16146737

ABSTRACT

This basic science review examines the role of cGMP and cGMP-dependent protein kinase (PKG) in the regulation of vascular smooth muscle cell (VSMC) phenotype. The first such studies suggested a role for nitric oxide (NO) and atrial natriuretic peptides (ANP), and the downstream second messenger cGMP, in the inhibition of VSMC proliferation. Subsequently, many laboratories confirmed the anti-proliferative effects of the cGMP pathway in cultured cells and the anti-atherosclerotic effects of the pathway in in vivo animal models. Other studies suggested that the cGMP target, PKG, mediated the anti-proliferative effects of cGMP although other laboratories have not consistently observed these effects. On the other hand, PKG mediates cGMP-dependent increases in smooth muscle-specific gene expression, and in vivo studies suggest that PKG expression itself reduces vascular lesions. The mechanisms by which PKG regulates gene expression are addressed, but it still unknown how the cGMP-PKG pathway is involved in smooth muscle-specific gene expression and phenotype.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation , Myocytes, Smooth Muscle/metabolism , Actins/chemistry , Animals , Aorta/metabolism , Atrial Natriuretic Factor/chemistry , Blotting, Western , Calcium-Binding Proteins/chemistry , Calmodulin-Binding Proteins/chemistry , Cell Proliferation , Collagen/chemistry , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/cytology , Humans , Integrins/metabolism , Microfilament Proteins/chemistry , Models, Biological , Muscle, Smooth/metabolism , Muscle, Smooth, Vascular/cytology , Myosins/metabolism , Nitric Oxide/chemistry , Phenotype , Plasminogen/chemistry , Protein Conformation , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction , Calponins
13.
Diabetes ; 52(8): 2144-50, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12882934

ABSTRACT

Hyperglycemia is a crucial factor in the development of diabetic nephropathy. We previously showed that high glucose upregulates thrombospondin 1 (TSP1)-dependent transforming growth factor (TGF)-beta activation by altering cGMP-dependent protein kinase (PKG) activity as a result of decreased nitric oxide signaling. In the present study, we showed that high glucose concentrations significantly reduced endogenous PKG activity. To further examine the mechanisms by which PKG regulates TSP1 expression and TSP1-dependent TGF-beta activation, we generated stably transfected rat mesangial cells (RMCs) with inducible expression tetracycline-induced gene expression of the catalytic domain of PKG. After tetracycline induction, the catalytic domain of PKG is expressed as a cGMP-independent active kinase. Expression of the catalytic domain prevented high glucose-mediated increases in transcription of the TSP1 gene with no alteration in TSP1 mRNA stability. Glucose stimulation of TSP1 protein expression and TGF-beta bioactivity were also downregulated. TGF-beta-dependent fibronectin and type IV collagen expression under high glucose conditions were significantly reduced upon catalytic domain expression in transfected RMCs. These results show that constitutively active PKG inhibits the fibrogenic potential of high glucose through repression of TSP1-dependent TGF-beta bioactivity, suggesting that gene transfer of the catalytic domain of PKG might provide a new strategy for treatment of diabetic renal fibrosis.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Hyperglycemia/metabolism , Thrombospondin 1/genetics , Transforming Growth Factor beta/metabolism , Animals , Anti-Bacterial Agents , Catalytic Domain/genetics , Cell Line , Collagen/metabolism , Diabetic Nephropathies/metabolism , Enzyme Activation/drug effects , Extracellular Matrix/enzymology , Fibronectins/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glomerular Mesangium/cytology , Glucose/pharmacology , Humans , Rats , Tetracycline , Transfection
14.
Tissue Eng ; 10(1-2): 189-99, 2004.
Article in English | MEDLINE | ID: mdl-15009945

ABSTRACT

Rat smooth muscle cells (SMCs) stably transfected with the gene for the phenotype regulating protein cyclic guanosine monophosphate-dependent protein kinase (PKG) were used as a cell source in the preparation of three-dimensional (3D) collagen type I vascular constructs. PKG-transfected cells expressed severalfold higher levels of the contractile protein smooth muscle alpha-actin (SMA), relative to untransfected SMCs, both in monolayer culture and in 3D gels. The proliferation rate of PKG-transfected cells was lower than that of untransfected cells in both culture geometries. Three-dimensional collagen constructs made with PKG-transfected cells compacted to a similar degree as those made with untransfected cells, and this compaction could be augmented by biochemical stimulation with platelet-derived growth factor BB (PDGF) or transforming growth factor beta(1) (TGF). Application of cyclic mechanical strain to tubular collagen gels seeded with PKG-transfected cells resulted in a higher degree of gel compaction and circumferential matrix alignment, relative to statically grown controls, but cell proliferation and SMA expression were not affected. These results show that genetic modification of SMCs can be used as a tool to control cell function in vascular tissue engineering, and that the function of such cells can be further modulated by application of biochemical and mechanical stimulation.


Subject(s)
Aorta/physiology , Myocytes, Smooth Muscle/physiology , Tissue Engineering , Transfection , Animals , Biocompatible Materials , Collagen , Phenotype , Rats , Time Factors
16.
Cardiovasc Res ; 97(2): 200-7, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23139241

ABSTRACT

The ability of the endothelium to produce nitric oxide, which induces generation of cyclic guanosine monophosphate (cGMP) that activates cGMP-dependent protein kinase (PKG-I), in vascular smooth muscle cells (VSMCs), is essential for the maintenance of vascular homeostasis. Yet, disturbance of this nitric oxide/cGMP/PKG-I pathway has been shown to play an important role in many cardiovascular diseases. In the last two decades, in vitro and in vivo models of vascular injury have shown that PKG-I is suppressed following nitric oxide, cGMP, cytokine, and growth factor stimulation. The molecular basis for these changes in PKG-I expression is still poorly understood, and they are likely to be mediated by a number of processes, including changes in gene transcription, mRNA stability, protein synthesis, or protein degradation. Emerging studies have begun to define mechanisms responsible for changes in PKG-I expression and have identified cis- and trans-acting regulatory elements, with a plausible role being attributed to post-translational control of PKG-I protein levels. This review will focus mainly on recent advances in understanding of the regulation of PKG-I expression in VSMCs, with an emphasis on the physiological and pathological significance of PKG-I down-regulation in VSMCs in certain circumstances.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/genetics , Gene Expression Regulation, Enzymologic , Transcription, Genetic , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Cyclic GMP-Dependent Protein Kinase Type I/physiology , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology
17.
J Biochem ; 149(4): 433-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21292805

ABSTRACT

The type-I cGMP-dependent protein kinase (PKG-I) expression regulation is not yet completely understood. In this study, we examined the role of 3'-untranslated region (3'UTR)-PKG-I messenger RNA (mRNA) in the control of PKG-I expression in vascular smooth muscle cells (VSMCs). Using a 3'-rapid amplification of cDNA ends (RACE) for the amplification of complementary DNA (cDNA) ends, we generated and cloned a 1.2-kb-3'UTR mRNA PKG-I in pGL3 control vector downstream of the luciferase reporter gene. Serial deletions and functional studies revealed that among the deleted constructs, only the 1.2-kb-3'UTR PKG-I mRNA possesses the highest activity in transfected VSMC. Kinetic luciferase assays in the presence of actinomycin D showed that this construct stabilizes luciferase activity compared to the control vector. Sequence analysis of 3'UTR-PKG-I mRNA revealed the existence of four AU-rich regions (AU1 through AU4) in addition to a potential poly(A) site. Different riboprobes were generated either by 5'-end-labelling of designed ribonucleotides, containing individual AU-rich regions or by in vitro transcription assay using cloned 1.2-kb cDNA as a template. RNA-electrophoretic mobility shift assay (EMSA) and ultra-violet cross-linking (UV-CL) assays showed that AU1, AU3, AU4 and 1.2-kb probes were able to retard cytosolic and nuclear proteins. Taken together, these data suggest that PKG-I expression is subjected to post-transcriptional regulation in VSMC through the 3'UTR of its mRNA.


Subject(s)
3' Untranslated Regions/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Muscle, Smooth, Vascular/enzymology , RNA, Messenger/genetics , Animals , Cattle , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism
18.
Cell Signal ; 21(6): 859-66, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19168131

ABSTRACT

Type I cGMP-dependent protein kinase (PKG-I) mediates nitric oxide (NO) and hormone dependent smooth muscle relaxation and stimulates smooth muscle cell-specific gene expression. Expression of PKG-I in cultured smooth muscle cells depends on culture conditions and is inhibited by inflammatory cytokines such as interleukin-I and tumor necrosis factor-alpha, which are known to stimulate Type II NO synthase (iNOS) expression. We report here that the suppression of PKG-I protein levels in smooth muscle cells is triggered by the ubiquitin/26S proteasome pathway. Incubation of vascular smooth muscle cells with phosphodiesterase-resistant cyclic GMP analogs (e.g., 8-bromo-cGMP) decreases PKG-I protein level in a time- and concentration-dependent manner. To study this process, we tested the effects of 8-Br-cGMP on PKG-I protein level in Cos7 cells, which do not express endogenous type I PKG mRNA. 8-Br-cGMP induced the ubiquitination and down-regulation of PKG-Ialpha, but not PKG-Ibeta. Treatment of cells with the 26S proteasome inhibitor, MG-132, increased ubiquitination of PKG. Blocking PKG-I catalytic activity using the cell-permeant specific PKG-I inhibitor, DT-2, inhibited cGMP-induced PKG-I ubiquitination and down-regulation, suggesting that PKG catalytic activity and autophosphorylation were required for suppression of PKG-I level. Mutation of the known autophosphorylation sites of PKG-Ialpha to alanine uncovered a specific role for autophosphorylation of serine-64 in cGMP-dependent ubiquitination and suppression of PKG-I level. The results suggest that chronic elevation of cGMP, as seen in inflammatory conditions, triggers ubiquitination and degradation of PKG-Ialpha in smooth muscle.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/pharmacology , Ubiquitination/drug effects , Animals , COS Cells , Cattle , Chlorocebus aethiops , Cyclic GMP/analogs & derivatives , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Down-Regulation/drug effects , Humans , Isoenzymes/metabolism , Mice , Models, Biological , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Nitric Oxide/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Rats , Rats, Sprague-Dawley , Transfection
19.
J Biol Chem ; 280(18): 18425-33, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15741164

ABSTRACT

Cyclic GMP-dependent protein kinase I plays a pivotal role in regulating smooth muscle cell relaxation, growth, and differentiation. Expression of the enzyme varies greatly in smooth muscle and in other tissues and cell types, yet little is known regarding the mechanisms regulating cGMP-dependent protein kinase gene expression. The present work was undertaken to characterize the mechanisms controlling kinase gene expression in vascular smooth muscle cells. A 2-kb human cGMP-dependent protein kinase I 5'-noncoding promoter sequence was characterized by serial deletion, and functional studies demonstrated that a 591-bp 5'-promoter construct possessed the highest activity compared with all other constructs generated from the larger promoter. Analysis of the sequence between -472 and -591 bp from the transcriptional start site revealed the existence of two E-like boxes known to bind upstream stimulatory factors. Electrophoretic mobility shift assays and functional studies using luciferase reporter gene assays identified upstream stimulatory factors as the transcription factors bound to the E-boxes in the 591-bp promoter. Site-directed mutagenesis of the E-boxes abolished the binding of upstream stimulatory factor proteins and decreased the activity of the cGMP-dependent protein kinase I 591-bp promoter, thus confirming the involvement of these transcription factors in mediating gene expression. Cotransfection experiments demonstrated that overexpression of upstream stimulatory factors 1 and 2 increased cGMP-dependent protein kinase I promoter activity. Collectively, these data suggest that the human proximal cGMP-dependent protein kinase I promoter is regulated by tandem E-boxes that bind upstream stimulatory factors.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/biosynthesis , DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic/physiology , Muscle, Smooth, Vascular/enzymology , Transcription Factors/physiology , Animals , Base Sequence , Cattle , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , DNA-Binding Proteins/genetics , Humans , Molecular Sequence Data , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic , Protein Binding/physiology , Rats , Transcription Factors/genetics , Upstream Stimulatory Factors
20.
J Cardiovasc Pharmacol ; 45(5): 404-13, 2005 May.
Article in English | MEDLINE | ID: mdl-15821435

ABSTRACT

We have previously shown that type I cGMP-dependent protein kinase (PKG) can alter the phenotype of cultured vascular smooth muscle cells (VSMCs). Although the expression of contractile proteins in VSMCs has been shown to be modulated with the induction of PKG, experiments in which PKG inhibition brings about reduced expression of contractile markers have not been performed. To more thoroughly examine the role of PKG in the expression of contractile proteins, recombinant adenovirus containing the PKG coding sequence (AD-PKG) was used to induce gene expression and morphologic changes in adult rat aortic VSMCs. Cells expressing PKG, but not control adenovirus-infected cells, began to express a specific marker protein for the contractile phenotype, smooth muscle myosin heavy chain (SMMHC), within 48 hours of PKG induction. The morphology of the AD-PKG-infected cells began to change from a fibroblastic phenotype to a spindle-shaped phenotype within 72 hours after PKG induction. The specific cell-permeable PKG inhibitory peptide DT-2, but not control peptides, reversed the biochemical and morphologic changes associated with PKG expression. These results suggest that PKG expression and activity in cultured VSMCs is capable of altering the VSMC phenotype. These data also verify the intracellular action of DT-2 and reveal uptake and dynamic properties of this PKG-inhibiting peptide.


Subject(s)
Biomarkers/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Muscle, Smooth, Vascular/drug effects , Phenotype , Adenoviridae/genetics , Animals , Aorta, Abdominal/cytology , Blotting, Western , Cell Culture Techniques , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/pharmacology , Enzyme Inhibitors/metabolism , Fluorescein , Fluorescent Dyes , Gene Expression , Male , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL