Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters










Publication year range
1.
Genom Data ; 5: 36-37, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26090325

ABSTRACT

TCF21 is a basic helix-loop-helix transcription factor that has recently been implicated as contributing to susceptibility to coronary heart disease based on genome wide association studies. In order to identify transcriptionally regulated target genes in a major disease relevant cell type, we performed siRNA knockdown of TCF21 in in vitro cultured human coronary artery smooth muscle cells and compared the transcriptome of siTCF21 versus siCONTROL treated cells. The raw (FASTQ) as well as processed (BED) data from 3 technical replicates per treatment has been deposited with Gene Expression Omnibus (GSE44461).

2.
Handb Exp Pharmacol ; (178): 441-68, 2007.
Article in English | MEDLINE | ID: mdl-17203666

ABSTRACT

This chapter will review conditional mouse model systems that have been developed to study gene function in skeletal, cardiac, and vascular smooth muscle cells in vivo with an emphasis on the utility of these models for investigating developmental and pathophysiological gene function in muscle. In general, these systems have utilized muscle-specific/selective promoter-enhancers in conjunction with site-specific DNA recombinases, e.g., Cre-loxP, and fusion proteins with these recombinases that confer temporal control, such as tamoxifen-inducible CreER systems. A major focus of this chapter will be to discuss unique challenges of studying Cre-mediated mutagenesis/gene targeting in these muscle types during development and in the adult animal, some of which are inherent of the muscle cell type being studied. For example, unlike cardiac and skeletal muscle cells, the vascular SMC is extremely plastic and able to undergo rapid phenotypic modulation to various environmental cues in vivo. Thus, employing SMC marker gene promoter enhancers for conditional gene targeting in SMCs must take into account the possibility and/or certainty that the particular SMC promoter enhancers used may or may not be transcriptionally active in SMCs of a vessel wall under normal and some pathophysiological conditions. Moreover, individual floxed loci within the same muscle cell type and tissue have different degrees of sensitivity to Cre, most likely dependent on the chromatin state of that particular gene, i.e., closed/condensed state or open/active state. Thus, Cre recombination may be ineffective for specific floxed gene DNA. Lastly, rigorous efforts must be made to confirm the degree of recombination in a tissue, taking into full account the multicellularity of the tissue, to understand the extent of the physiological effect in that organ.


Subject(s)
Muscles/embryology , Muscles/metabolism , Animals , Gene Targeting , Mice , Models, Animal , Mutagenesis , Recombinases/metabolism
3.
Proc Natl Acad Sci U S A ; 103(7): 2440-5, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16461919

ABSTRACT

Cyclic nucleotides can relax smooth muscle without a change in [Ca2+]i, a phenomenon termed Ca2+ desensitization, contributing to vasodilation, gastrointestinal motility, and airway resistance. The physiological importance of telokin, a 17-kDa smooth muscle-specific protein and target for cyclic nucleotide-induced Ca2+ desensitization, was determined in telokin null mice bred to a congenic background. Telokin null ileal smooth muscle homogenates compared to wild type exhibited an approximately 30% decrease in myosin light-chain phosphatase (MLCP) activity, which was reflected in a significant leftward shift (up to 2-fold at pCa 6.3) of the Ca2+ force relationship accompanied by an increase in myosin light-chain phosphorylation. No difference in the Ca2+ force relationship occurred in telokin WT and knockout (KO) aortas, presumably reflecting the normally approximately 5-fold lower telokin content in aorta vs. ileum smooth muscle. Ca2+ desensitization of contractile force by 8-Br-cGMP was attenuated by 50% in telokin KO intestinal smooth muscle. The rate of force relaxation reflecting MLCP activity, in the presence of 50 microM 8-Br-cGMP, was also significantly slowed in telokin KO vs. WT ileum and was rescued by recombinant telokin. Normal thick filaments in telokin KO smooth muscles indicate that telokin is not required for filament formation or stability. Results indicate that a primary role of telokin is to modulate force through increasing MLCP activity and that this effect is further potentiated through phosphorylation by cGMP in telokin-rich smooth tissues.


Subject(s)
Calcium/pharmacology , Cyclic GMP/pharmacology , Muscle Relaxation , Muscle, Smooth/drug effects , Myosin-Light-Chain Kinase/physiology , Peptides/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Aorta/drug effects , Ileum/drug effects , Mice , Mice, Knockout , Muscle Relaxation/genetics , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Muscle, Smooth/ultrastructure , Myosin-Light-Chain Kinase/deficiency , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase/metabolism , Peptide Fragments , Peptides/deficiency , Peptides/genetics
4.
Circ Res ; 98(3): 378-85, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16397143

ABSTRACT

Lipoma preferred partner (LPP) has been identified as a protein highly expressed in smooth muscle (SM) tissues. The aim of the present study was to determine mechanisms that regulate LPP expression in an in vitro model of SM cell (SMC) differentiation and in stent-induced pig coronary vessel injury. All trans-retinoic acid treatment of A404 cells induced a strong increase in LPP, as well as SM alpha-actin, SM myosin heavy chain, and smoothelin mRNA levels, in a Rho kinase (ROK)-dependent manner. Adenovirus mediated overexpression of myocardin in A404 cells significantly increased LPP mRNA expression. Interestingly, inactivation of RhoA with C3-exoenzyme or treatment with ROK inhibitors strongly inhibited myocardin mRNA expression in retinoic acid-treated A404 cells or human iliac vein SMCs. LPP silencing with short interfering RNA significantly decreased SMC migration. LPP expression was also markedly decreased in focal adhesion kinase (FAK)-null cells known to have impaired migration but rescued with inducible expression of FAK. LPP expression in FAK-null fibroblasts enhanced cell spreading. In stented pig coronary vessels, LPP was expressed in the neointima of cells lacking smoothelin and showed expression patterns identical to those of SM alpha-actin. In conclusion, LPP appears to be a myocardin-, RhoA/ROK-dependent SMC differentiation marker that plays a role in regulating SMC migration.


Subject(s)
Carrier Proteins/physiology , Muscle Proteins/physiology , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Coronary Vessels/physiology , Humans , Iliac Vein , Leucine Zippers , Mice , Models, Animal , Muscle Proteins/genetics , Muscle, Smooth, Vascular/physiology , Stents , Swine
6.
Circ Res ; 95(10): 981-8, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15486317

ABSTRACT

A hallmark of smooth muscle cell (SMC) phenotypic switching in atherosclerotic lesions is suppression of SMC differentiation marker gene expression. Yet little is known regarding the molecular mechanisms that control this process. Here we show that transcription of the SMC differentiation marker gene SM22alpha is reduced in atherosclerotic lesions and identify a cis regulatory element in the SM22alpha promoter required for this process. Transgenic mice carrying the SM22alpha promoter-beta-galactosidase (beta-gal) reporter transgene were crossed to apolipoprotein E (ApoE)-/- mice. Cells of the fibrous cap, intima, and underlying media showed complete loss of beta-gal activity in advanced atherosclerotic lesions. Of major significance, mutation of a G/C-rich cis element in the SM22alpha promoter prevented the decrease in SM22alpha promoter-beta-gal reporter transgene expression, including in cells that compose the fibrous cap of the lesion and in medial cells in proximity to the lesion. To begin to assess mechanisms whereby the G/C repressor element mediates suppression of SM22alpha in atherosclerosis, we tested the hypothesis that effects may be mediated by platelet-derived growth factor (PDGF)-BB-induced increases in the G/C binding transcription factor Sp1. Consistent with this hypothesis, results of studies in cultured SMCs showed that: (1) PDGF-BB increased expression of Sp1; (2) PDGF-BB and Sp1 profoundly suppressed SM22alpha promoter activity as well as smooth muscle myosin heavy chain promoter activity through mechanisms that were at least partially dependent on the G/C cis element; and (3) a short interfering RNA to Sp1 increased basal expression and attenuated PDGF-BB induced suppression of SM22alpha. Together, these results support a model whereby a G/C repressor element within the SM22alpha promoter mediates transcriptional repression of this gene within phenotypically modulated SMCs in experimental atherosclerosis and provide indirect evidence implicating PDGF-BB and Sp1 as possible mediators of these effects.


Subject(s)
Arteriosclerosis/genetics , Gene Silencing/physiology , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid , Animals , Aorta/cytology , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Becaplermin , Cell Differentiation/genetics , Cells, Cultured/metabolism , Cells, Cultured/pathology , Crosses, Genetic , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Genes, Reporter , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Lac Operon , Mice , Mice, Inbred CBA , Mice, Transgenic , Muscle Proteins/biosynthesis , Myocytes, Smooth Muscle/pathology , Phenotype , Platelet-Derived Growth Factor/pharmacology , Platelet-Derived Growth Factor/physiology , Protein Binding , Proto-Oncogene Proteins c-sis , Rats , Recombinant Fusion Proteins/physiology , Serum Response Element , Sp1 Transcription Factor/physiology
7.
Circ Res ; 95(4): 406-14, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15256479

ABSTRACT

Vascular smooth muscle cell (SMC) contraction is mediated in part by calcium influx through L-type voltage-gated Ca2+ channels (VGCC) and activation of the RhoA/Rho kinase (ROK) signaling cascade. We tested the hypothesis that Ca2+ influx through VGCCs regulates SMC differentiation marker expression and that these effects are dependent on RhoA/ROK signaling. Depolarization-induced activation of VGCCs resulted in a nifedipine-sensitive increase in endogenous smooth muscle myosin heavy chain (SMMHC) and SM alpha-actin expression and CArG-dependent promoter activity, as well as c-fos promoter activity. The ROK inhibitor, Y-27632, prevented depolarization-induced increase in SMMHC/SM alpha-actin but had no effect on c-fos expression. Conversely, the Ca2+/calmodulin-dependent kinase inhibitor, KN93, prevented depolarization-induced increases in c-fos expression with no effect on SMMHC/SM alpha-actin. Depolarization increased expression of myocardin, a coactivator of SRF that mediates CArG-dependent transcription of SMC marker gene promoters containing paired CArG cis regulatory elements (SMMHC/SM alpha-actin). Both nifedipine and Y-27632 prevented the depolarization-induced increase in myocardin expression. Moreover, short interfering RNA (siRNA) specific for myocardin attenuated depolarization-induced SMMHC/SM alpha-actin transcription. Chromatin immunoprecipitation (ChIP) assays revealed that depolarization increased SRF enrichment of the CArG regions in the SMMHC, SM alpha-actin, and c-fos promoters in intact chromatin. Whereas Y-27632 decreased basal and depolarization-induced SRF enrichment in the SMMHC/SM alpha-actin promoter regions, it had no effect of SRF enrichment of c-fos. Taken together, these results provide evidence for a novel mechanism whereby Ca2+ influx via VGCCs stimulates expression of SMC differentiation marker genes through mechanisms that are dependent on ROK, myocardin, and increased binding of SRF to CArG cis regulatory elements.


Subject(s)
Calcium Channels, L-Type/physiology , Gene Expression Regulation, Developmental , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Protein Serine-Threonine Kinases/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Actins/physiology , Animals , Aorta , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Cell Differentiation/physiology , Cells, Cultured/cytology , Cells, Cultured/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Genes, fos , Intracellular Signaling Peptides and Proteins , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin Heavy Chains/physiology , Nifedipine/pharmacology , Nuclear Proteins/physiology , Organoids/cytology , Patch-Clamp Techniques , Potassium Chloride/pharmacology , Protein Transport , RNA, Small Interfering/pharmacology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Element/genetics , Serum Response Factor/physiology , Trans-Activators/physiology , Transfection , rho-Associated Kinases , rhoA GTP-Binding Protein/physiology
8.
J Biol Chem ; 276(42): 39076-87, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11489897

ABSTRACT

Previous studies in our laboratory demonstrated that the transgene consisting of the -4.2 to +11.6 kilobase (kb) region of the smooth muscle (SM) myosin heavy chain (MHC) gene was expressed in virtually all SM tissue types in vivo in transgenic mice and that the multiple CArG elements within this region were differentially required in SMC subtypes, implying that the SM-MHC gene was controlled by multiple transcriptional regulatory modules. To investigate this hypothesis, we analyzed specific regulatory regions within the SM-MHC -4.2 to +11.6 kb region by a combination of deletion analyses of various SM-MHC transgenes as well as by DNaseI hypersensitivity assays and in vivo footprinting in intact SMC tissues. The results showed that SM-MHC transgene expression depended on a large number of required regulatory modules that were widely spread over the -4.2 to +11.6 region. Moreover, the results revealed several unexpected novel features of regulation of the SM-MHC gene including: 1) unique combinations of regulatory modules were required for SM-MHC expression in different SMC-subtypes; 2) repressor modules as well as activator modules were both critical for SMC specificity of the gene; 3) certain modules were required in certain contexts but were dispensable in others within a given SMC-subtype (i.e. the net activity of the module was determined by interaction between modules not simply by the sum of module activities); and 4) we identified a highly conserved 200-base pair transcriptional regulatory module at +8 kb that was required in the large arteries but dispensable in the coronary arteries and airways in transgenic mice and contained multiple potential cis-elements that were occupied by nuclear proteins in the intact aorta based on in vivo footprinting. Taken together, the results suggest a model of complex modular control of expression of the SM-MHC gene that varies between SMC subtypes. Moreover, the studies establish the possibility of designing derivatives of the SM-MHC promoter that might be used for targeting gene expression to specific SMC subtypes in vivo.


Subject(s)
Muscle, Smooth/metabolism , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cell Nucleus/metabolism , Deoxyribonuclease I/metabolism , Gene Deletion , Genes, Reporter , Introns , Lac Operon , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Transcription, Genetic , Transfection
9.
Circ Res ; 88(11): 1127-34, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11397778

ABSTRACT

Little is known regarding transcriptional regulatory mechanisms that control the sequential and coordinate expression of genes during smooth muscle cell (SMC) differentiation. To facilitate mechanistic studies of SMC differentiation, we established a novel P19-derived clonal cell line (designated A404) harboring a smooth muscle (SM) alpha-actin promoter/intron-driven puromycin resistance gene. Retinoic acid plus puromycin treatment stimulated rapid differentiation of multipotential A404 cells into SMCs that expressed multiple SMC differentiation marker genes, including the definitive SM-lineage marker SM myosin heavy chain. Using this system, we demonstrated that various transcription factors were upregulated coincidentally with the expression of SMC differentiation marker genes. Of interest, the expression of serum response factor (SRF), whose function is critical for SMC-specific transcription, was high in undifferentiated A404 cells, and it did not increase over the course of differentiation. However, chromatin immunoprecipitation analyses showed that SRF did not bind the target sites of endogenous SMC marker genes in chromatin in undifferentiated cells, but it did in differentiated A404 cells, and it was associated with hyperacetylation of histones H3 and H4. The present studies define a novel cell system for studies of transcriptional regulation during the early stages of SMC differentiation, and using this system, we obtained evidence for the involvement of chromatin remodeling and selective recruitment of SRF to CArG elements in the induction of cell-selective marker genes during SMC differentiation.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Histones/metabolism , Muscle, Smooth, Vascular/metabolism , Nuclear Proteins/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Acetylation/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Differentiation/biosynthesis , Cell Differentiation/drug effects , Cell Line , Chromatin/metabolism , Gene Expression Regulation/drug effects , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myosin Heavy Chains/biosynthesis , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Precipitin Tests , Puromycin/pharmacology , Regulatory Sequences, Nucleic Acid/drug effects , Serum Response Factor , Transcription Factors/metabolism , Tretinoin/pharmacology , Up-Regulation/drug effects
10.
J Clin Invest ; 107(7): 823-34, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285301

ABSTRACT

Expression of smooth muscle myosin heavy chain (SM-MHC) is tightly controlled depending on the differentiated state of smooth muscle cells (SMCs). To better understand the mechanisms that regulate transcription of the SM-MHC gene in vivo, we tested the function of several conserved CArG elements contained within the -4200 to +11,600 region of this gene that we had previously shown to drive SMC-specific expression in transgenic mice. CArG1 in the 5'-flanking sequence was required for all SMCs, while CArG2 and a novel intronic CArG element were differentially required in SMC subtypes. Of particular note, mutation of the intronic CArG selectively abolished expression in large arteries. A promoter construct containing three repeats of a conserved 227-bp intronic CArG-containing region was sufficient to direct transcription in vascular SMCs in transgenic mice, although this construct was also expressed in skeletal and cardiac muscle. These results support a model in which transcriptional regulation of SM-MHC is controlled by multiple positive and negative modular control regions that differ between SMCs and non-SMCs and among SMC subtypes. We also demonstrated that the CArG elements of the endogenous SM-MHC gene were bound by SRF in chromatin.


Subject(s)
Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , Myosin Heavy Chains/genetics , Transcription, Genetic , Animals , Base Sequence , Cells, Cultured , Chromatin , DNA-Binding Proteins/metabolism , Humans , Introns , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Mutagenesis , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Rats , Serum Response Factor
11.
Mol Cell Biol ; 21(5): 1565-72, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238893

ABSTRACT

Extracellular matrix signaling via integrin receptors is important for smooth muscle cell (SMC) differentiation during vasculogenesis and for phenotypic modulation of SMCs during atherosclerosis. We previously reported that the noncatalytic carboxyl-terminal protein binding domain of focal adhesion kinase (FAK) is expressed as a separate protein termed FAK-related nonkinase (FRNK) and that ectopic expression of FRNK can attenuate FAK activity and integrin-dependent signaling (A. Richardson and J. T. Parsons, Nature 380:538-540, 1996). Herein we report that in contrast to FAK, which is expressed ubiquitously, FRNK is expressed selectively in SMCs, with particularly high levels observed in conduit blood vessels. FRNK expression was low during embryonic development, was significantly upregulated in the postnatal period, and returned to low but detectable levels in adult tissues. FRNK expression was also dramatically upregulated following balloon-induced carotid artery injury. In cultured rat aortic smooth muscle cells, overexpression of FRNK attenuated platelet-derived growth factor (PDGF)-BB-induced migration and also dramatically inhibited [(3)H]thymidine incorporation upon stimulation with PDGF-BB or 10% serum. These effects were concomitant with a reduction in SMC proliferation. Taken together, these data indicate that FRNK acts as an endogenous inhibitor of FAK signaling in SMCs. Furthermore, increased FRNK expression following vascular injury or during development may alter the SMC phenotype by negatively regulating proliferative and migratory signals.


Subject(s)
Muscle, Smooth, Vascular/cytology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Adenoviridae/metabolism , Animals , Blotting, Northern , Blotting, Western , Carotid Arteries/metabolism , Cell Adhesion , Cell Cycle , Cell Division , Cell Movement , Cells, Cultured , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins/metabolism , Phenotype , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Precipitin Tests , Rats , Time Factors , Tissue Distribution , Up-Regulation
12.
J Biol Chem ; 276(1): 341-7, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11035001

ABSTRACT

Smooth muscle cell (SMC) differentiation is regulated by a complex array of local environmental cues, but the intracellular signaling pathways and the transcription mechanisms that regulate this process are largely unknown. We and others have shown that serum response factor (SRF) contributes to SMC-specific gene transcription, and because the small GTPase RhoA has been shown to regulate SRF, the goal of the present study was to test the hypothesis that RhoA signaling is a critical mechanism for regulating SMC differentiation. Coexpression of constitutively active RhoA in rat aortic SMC cultures significantly increased the activity of the SMC-specific promoters, SM22 and SM alpha-actin, whereas coexpression of C3 transferase abolished the activity of these promoters. Inhibition of either stress fiber formation with the Rho kinase inhibitor Y-27632 (10 microm) or actin polymerization with latrunculin B (0.5 microm) significantly decreased the activity of SM22 and SM alpha-actin promoters. In contrast, increasing actin polymerization with jasplakinolide (0.5 microm) increased SM22 and SM alpha-actin promoter activity by 22-fold and 13-fold, respectively. The above interventions had little or no effect on the transcription of an SRF-dependent c-fos promoter or on a minimal thymidine kinase promoter that is not SRF-dependent. Taken together, the results of these studies indicate that in SMC, RhoA-dependent regulation of the actin cytoskeleton selectively regulates SMC differentiation marker gene expression by modulating SRF-dependent transcription. The results also suggest that RhoA signaling may serve as a convergence point for the multiple signaling pathways that regulate SMC differentiation.


Subject(s)
Actins/metabolism , Cell Differentiation , Depsipeptides , Gene Expression Regulation , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Animals , Aorta , Biomarkers , Biopolymers/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Cytochalasin D/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Genes, Reporter , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides, Cyclic/pharmacology , Promoter Regions, Genetic/genetics , Pyridines/pharmacology , Rats , Serum Response Factor , Signal Transduction , Stress Fibers/drug effects , Stress Fibers/metabolism , Thiazoles/pharmacology , Thiazolidines , Transcription, Genetic/drug effects , Transfection , rhoA GTP-Binding Protein/antagonists & inhibitors
13.
J Clin Invest ; 106(9): 1139-47, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11067866

ABSTRACT

While it is well established that phenotypic modulation of vascular smooth muscle cells (VSMCs) contributes to the development and progression of vascular lesions, little is known regarding the molecular mechanisms of phenotypic modulation in vivo. Here we show that vascular injury reduces transcription of VSMC differentiation marker genes, and we identify cis regulatory elements that may mediate this decrease. Using a carotid wire-injury model in mice carrying transgenes for smooth muscle alpha-actin, smooth muscle myosin heavy chain, or a SM22alpha promoter-beta-gal reporter, we collected arteries 7 and 14 days after injury and assessed changes in endogenous protein and mRNA levels and in beta-gal activity. Endogenous levels for all markers were decreased 7 days after injury and returned to nearly control levels by 14 days. beta-gal staining in all lines followed a similar pattern, suggesting that transcriptional downregulation contributed to the injury-induced decreases. To begin to dissect this response, we mutated a putative G/C-rich repressor in the SM22alpha promoter transgene and found that this mutation significantly attenuated injury-induced downregulation. Hence, transcriptional downregulation contributes to injury-induced decreases in VSMC differentiation markers, an effect that may be partially mediated through a G/C-rich repressor element.


Subject(s)
Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/metabolism , Actins/genetics , Actins/metabolism , Animals , Base Sequence , Biomarkers , Cell Differentiation , DNA Primers/genetics , Gene Expression , Genes, Reporter , In Situ Hybridization , Lac Operon , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , beta-Galactosidase/genetics
14.
Circ Res ; 87(5): 363-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969033

ABSTRACT

The use of genetically modified mice has been an important model system to study gene function in cardiovascular development and under pathophysiological conditions. Although conventional gene knockout studies have provided important insights into gene function in the cardiovascular system, they may be limited by upregulation of compensatory pathways and the inability to differentiate direct versus indirect functions in vivo. As a first step in developing systems that can target gene activation or inactivation specifically to smooth muscle cells (SMCs), we coupled the smooth muscle myosin heavy chain (SMMHC) promoter to the cre recombinase gene and generated transgenic mice that express cre in SMCs. In addition, we used these mice to address whether the heterogeneous staining observed in SMMHC-LacZ mice was due to subsets of SMCs that required different regulatory cassettes of the promoter or if it reflected episodic expression of the transgene. To address both the feasibility of SMC targeting and the apparent heterogeneous expression, we bred SMMHC-cre mice to indicator mice containing a cre-activated LacZ gene. Results showed high-level expression in SMCs at various embryonic time points and in adult tissues. Because breeding of SMMHC-cre mice to an indicator line provided an integration of cre activity over time, results of this study revealed that expression of the SMMHC promoter fragment more closely resembled the expression of the endogenous gene, both with respect to the onset of activation during development and uniformity of staining among individual cells within tissues. Overall, these mice will provide a powerful tool to researchers to study gene function in vascular development/disease by using cre/lox technology to direct smooth muscle-specific gene activation or inactivation in vivo.


Subject(s)
Integrases/genetics , Muscle, Smooth, Vascular/metabolism , Myosin Heavy Chains/genetics , Viral Proteins , beta-Galactosidase/biosynthesis , Animals , Aorta, Thoracic/embryology , Carotid Arteries/embryology , Crosses, Genetic , Embryo, Mammalian/metabolism , Gene Expression Regulation , Gene Targeting , Gestational Age , Heart/embryology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/enzymology , Promoter Regions, Genetic , Recombination, Genetic , Transcriptional Activation , Transgenes , beta-Galactosidase/analysis , beta-Galactosidase/genetics
15.
J Biol Chem ; 275(48): 37798-806, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10954723

ABSTRACT

Transforming growth factor beta (TGF-beta) is implicated in the regulation of smooth muscle cell (SMC) differentiation. We previously identified a novel TGF-beta control element (TCE) in the promoters of SMC differentiation marker genes, including alpha-smooth muscle actin and SM22alpha. In this study, the importance of the TCE in regulation of SM22alpha gene expression in vivo was investigated by mutating it within the context of a mouse SM22alpha promoter-lacZ transgenic construct. Mutation of the TCE completely abolished SM22alpha promoter activity in arterial SMCs as well as in developing heart and skeletal muscle. To identify the transcription factor(s) binding to the TCE, we performed yeast one-hybrid cloning analysis and identified gut-enriched Krüppel-like factor (GKLF). However, cotransfection studies in cultured cells showed that GKLF repressed the TGF-beta-dependent increases in SM22alpha and alpha-smooth muscle actin promoter activities. Furthermore, GKLF was not highly expressed in differentiated SMCs in vivo, and TGF-beta down-regulated GKLF expression in dedifferentiated cultured SMCs. In contrast, overexpression of a related factor (BTEB2) transactivated SM22alpha promoter activity. Thus, our findings suggest a reciprocal role for related Krüppel-like transcription factors in the regulation of SMC differentiation through a TCE-dependent mechanism.


Subject(s)
Biomarkers , Cell Differentiation , DNA-Binding Proteins/metabolism , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth/cytology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Aorta, Thoracic/cytology , Base Sequence , DNA , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Mice , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Rats , Recombinant Proteins/metabolism , Sequence Homology, Nucleic Acid
16.
Circ Res ; 86(2): 221-32, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10666419

ABSTRACT

Previous studies have shown that multiple serum response factor (SRF)-binding CArG elements were required for smooth muscle cell (SMC)-specific regulation of smooth muscle (SM) alpha-actin expression. However, a critical question remains as to the mechanisms whereby a ubiquitously expressed transcription factor such as SRF might contribute to SMC-specific expression. The goal of the present study was to investigate the hypothesis that SMC-selective expression of SM alpha-actin is due at least in part to (1) unique CArG flanking sequences that distinguish the SM alpha-actin CArGs from other ubiquitously expressed CArG-dependent genes such as c-fos, (2) cooperative interactions between CArG elements, and (3) SRF-dependent binding of SMC-selective proteins to the CArG-containing regions of the promoter. Results demonstrated that specific sequences flanking CArG B were important for promoter activity in SMCs but not in bovine aortic endothelial cells. We also provided evidence indicating that the structural orientation between CArGs A and B was an important determinant of promoter function. Electrophoretic mobility shift assays and methylation interference footprinting demonstrated that a unique SRF-containing complex formed that was selective for SMCs and, furthermore, that this complex was probably stabilized by protein-protein interactions and not by specific interactions with CArG flanking sequences. Taken together, the results of these studies provide evidence that SM alpha-actin expression in SMCs is complex and may involve the formation of a unique multiprotein initiation complex that is coordinated by SRF complexes bound to multiple CArG elements.


Subject(s)
Actins/genetics , DNA-Binding Proteins/genetics , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Nuclear Proteins/genetics , Promoter Regions, Genetic/physiology , Actins/analysis , Actins/metabolism , Animals , Aorta/cytology , Cattle , Cells, Cultured , DNA Footprinting , DNA Methylation , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Erythroid-Specific DNA-Binding Factors , Gene Expression Regulation/physiology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Mutagenesis/physiology , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding/genetics , Proto-Oncogene Proteins/analysis , Rats , Serum Response Factor , Transcription Factors/analysis , Transcription, Genetic/physiology , ets-Domain Protein Elk-1 , ets-Domain Protein Elk-4
17.
Arterioscler Thromb Vasc Biol ; 20(1): 89-95, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634804

ABSTRACT

All-trans-retinoic acid (atRA) has potent in vitro effects on a number of processes involved in vascular injury and repair, such as modulating smooth muscle cell (SMC) proliferation and inducing SMC differentiation, and may play an important role in the in vivo response to vascular injury. We hypothesized that atRA would limit restenosis after balloon angioplasty through SMC-modulated changes in plaque size and vessel geometry. Balloon angioplasty was performed on rabbits with focal femoral atherosclerosis randomized to treatment with atRA or saline. At 28 days after balloon angioplasty, minimal luminal diameter was significantly larger in the atRA group (1.24+/-0.17 versus 1.12+/-0.22 mm, P=0.02). Histomorphometry confirmed a larger lumen area (0.51+/-0.20 versus 0. 34+/-0.13 mm(2), P=0.004) in the atRA group, with no difference in absolute plaque area. Internal elastic lamina and external elastic lamina areas were significantly larger in the atRA group (0.89+/-0. 27 versus 0.66+/-0.24 mm(2), P=0.001, and 1.29+/-0.38 versus 0. 98+/-0.32 mm(2), P=0.001, respectively). Vessel sections exhibited significantly more alpha-actin and desmin immunostaining (P=0.01) in the atRA-treated group. No differences in early cellular proliferation and collagen content were detected with the use of bromodeoxyuridine. In this atherosclerotic model of vascular injury, atRA limits restenosis after balloon angioplasty by effects secondary to overall vessel segment enlargement at the angioplasty site rather than by effects on plaque size or cellular proliferation. Increased alpha-actin and desmin immunostaining suggest a possible role for phenotypic modulation of SMCs in this favorable remodeling effect.


Subject(s)
Angioplasty, Balloon/adverse effects , Arteriosclerosis/drug therapy , Arteriosclerosis/therapy , Tretinoin/pharmacology , Actins/metabolism , Animals , Arteriosclerosis/pathology , Cell Division/drug effects , Collagen/metabolism , Desmin/metabolism , Immunohistochemistry , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Rabbits , Recurrence , Time Factors
18.
Arterioscler Thromb Vasc Biol ; 19(11): 2591-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10559000

ABSTRACT

Identification of the regulators of smooth muscle specific gene expression is critical for understanding smooth muscle cell (SMC) differentiation and the alterations in SMC phenotype seen in vascular diseases. Previous studies have identified that a 2-bp mutation in a conserved cis-acting element (TGTTTATC) in the promoter of the chicken smooth muscle (SM) alpha-actin gene abolished nuclear factor binding and decreased transcriptional activity of a 271-bp SM alpha-actin promoter fragment when transfected into rat aortic SMC. However, the promoter region containing this conserved sequence has negative cis regulatory activity when studied in homologous systems. The goal of the present studies was to further characterize the transcriptional activity of the rat SM alpha-actin promoter region between -224 and -236 that is conserved across mammals. DNAse I analysis and electrophoretic mobility shift assays demonstrated that SMC nuclear proteins bound an extended sequence (TGTTTATCCCCATAA). Transient transfection experiments of wild-type and mutant rat SM alpha-actin promoter-luciferase constructs into rat aortic SMC revealed that promoter activity was enhanced by mutations of specific nucleotides in the TGTTTATCCCCA region. Interestingly, the TGTTTATCCCCA element in the rat SM alpha-actin promoter is centered between 2 canonical E-boxes. Mutations of the flanking E-boxes abolished the enhancement in promoter activity seen with mutation of the TGTTTATCCCCA element alone. Thus studies provide evidence for a regulatory cassette in the rat SM alpha-actin promoter that regulates gene expression via combinatorial interactions between 2 E-boxes and a newly described TGTTTATCCCCA element.


Subject(s)
Actins/genetics , Conserved Sequence , Muscle, Smooth, Vascular/physiology , Promoter Regions, Genetic/physiology , Animals , Aorta/cytology , Binding Sites/genetics , Cell Differentiation/physiology , Cells, Cultured , DNA Footprinting , Deoxyribonuclease I , Gene Expression Regulation/physiology , Genetic Complementation Test , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Mutagenesis/physiology , Rats , Sequence Homology, Nucleic Acid , Transcription, Genetic/physiology
19.
J Vasc Res ; 36(5): 343-52; discussion 430-3, 1999.
Article in English | MEDLINE | ID: mdl-10559674

ABSTRACT

UNLABELLED: Intravascular delivery of an E1/E3 deleted adenovirus encoding the hirudin protein reduces neointimal formation in the rat arterial injury model. Given the interspecies variability in response to adenoviral vectors, we tested this same construct in the hirudin-sensitive cholesterol-fed rabbit arterial balloon injury model. We hypothesized that local delivery of an E1/E3-deleted adenovirus encoding hirudin (Ad-Hir) in addition to early hirudin infusion would limit neointimal formation compared to early hirudin alone. METHODS AND RESULTS: Local delivery of Ad-Hir, 2.5 x 10(10) PFU/ml, using a double balloon catheter [n = 6 vessels (v)] produced a 79% reduction in vessel wall thrombin activity at 48 h after balloon angioplasty (BA) compared with vehicle (Veh, n = 6v; p = 0. 05). In chronic experiments, hypercholesterolemic rabbits underwent femoral BA, and received either early hirudin alone (n = 9v) or early hirudin plus locally delivered Ad-Hir (early hirudin + Ad-Hir; n = 9v), an E1/E3-deleted adenovirus encoding beta-galactosidase (early hirudin + AdGal; n = 7v), or Veh (early hirudin + Veh; n = 10v). Early hirudin + Ad-Hir did not limit the arterial response to injury versus the other groups at 4 weeks after BA. Plaque area, cross-sectional luminal area narrowing by plaque, and T cell infiltration were significantly increased in the adenovirus- versus non-adenovirus-treated arteries. Plaque area correlated with T cell density. CONCLUSION: Following BA in cholesterol-fed rabbits, local transduction with A-Hir produced a marked reduction in vessel wall-associated thrombin activity. However, this strategy increased rather than decreased the arterial response to BA injury. Our results suggest that the lack of therapeutic effect resulted from adenovirus-stimulated plaque formation, possibly resulting from a T cell-mediated inflammatory response.


Subject(s)
Adenoviruses, Human , Angioplasty, Balloon/adverse effects , Antithrombins/genetics , Femoral Artery/injuries , Gene Transfer Techniques , Genetic Vectors , Hirudins/genetics , Adenoviruses, Human/immunology , Animals , Antithrombins/therapeutic use , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Arteriosclerosis/therapy , Disease Models, Animal , Genetic Vectors/immunology , Hirudin Therapy , Humans , Rabbits , Thrombin/metabolism
20.
Arterioscler Thromb Vasc Biol ; 19(9): 2049-58, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479645

ABSTRACT

Transforming growth factor-beta (TGF-beta) has been shown to stimulate smooth muscle (SM) alpha-actin expression in smooth muscle cells (SMCs) and non-SMCs. We previously demonstrated that the 2 CArG boxes A and B and a novel TGF-beta control element (TCE) located within the first 125 bp of the SM alpha-actin promoter were required for TGF-beta inducibility of SM alpha-actin in SMCs. The aims of the present study were (1) to determine whether the TCE exhibits SMC specificity or contributes to TGF-beta induction of SM alpha-actin expression in non-SMCs (ie, endothelial cells and fibroblasts) and (2) to determine whether TGF-beta can induce expression of multiple TCE-containing SMC differentiation marker genes, such as SM22alpha, h(1) calponin, and SM myosin heavy chain (SM MHC) in non-SMCs. Results of transient transfection assays demonstrated that mutation of CArG A, CArG B, or the TCE within a 125-bp promoter context completely abolished TGF-beta inducibility of SM alpha-actin in endothelial cells and fibroblasts. However, in contrast to observations in SMCs, inclusion of regions upstream from (-155) completely repressed TGF-beta responsiveness in non-SMCs. Electrophoretic mobility shift assays showed that TGF-beta enhanced binding of a serum response factor to the CArG elements and the binding of an as-yet-unidentified factor to the TCE in endothelial cells and fibroblasts, but to a much lesser extent compared with SMCs. TGF-beta also stimulated expression of the SMC differentiation marker SM22alpha in non-SMCs. However, in contrast to SMCs, TGF-beta did not induce expression of h(1) calponin and SM MHC in non-SMCs. In summary, these results suggest a conserved role for CArG A, CArG B, and the TCE in TGF-beta-induced expression of SM alpha-actin in SMCs and non-SMCs that is modified by a complex interplay of positive- and negative-acting cis elements in a cell-specific manner. Furthermore, observations that TGF-beta stimulated expression of several early but not late differentiation markers in non-SMCs indicate that TGF-beta alone is not sufficient to induce transdifferentiation of non-SMCs into SMCs.


Subject(s)
Actins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Transforming Growth Factor beta/pharmacology , Actins/genetics , Amino Acid Sequence/genetics , Animals , Calcium-Binding Proteins/genetics , Cattle , Cells, Cultured , Conserved Sequence , DNA-Binding Proteins/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Myosin Heavy Chains/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats , Serum Response Factor , Calponins
SELECTION OF CITATIONS
SEARCH DETAIL
...