Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biol ; 132(5): 849-59, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8603917

RESUMO

SM22alpha is a putative calcium-binding protein that is expressed in cardiac, smooth, and skeletal muscle lineages during mouse embryogenesis and in adult smooth muscle cells (SMC). To define the mechanisms that regulate smooth muscle-specific gene transcription, we isolated the SM22alpha gene and analyzed its 5'-flanking region for elements that direct smooth muscle expression in transgenic mice. Using a series of promoter deletions, a region of the SM22alpha promoter containing 445 base pairs of 5'-flanking sequence was found to be sufficient to direct the specific expression of a lacZ transgene in mouse embryos in the vascular smooth, cardiac, and skeletal muscle lineages in a temporospatial pattern similar to the endogenous SM22alpha gene. However, in contrast to the endogenous gene, transgene expression was not detected in venous, nor visceral SMCs. This SM22alpha-lacZ transgene was therefore able to distinguish between the transcriptional regulatory programs that control gene expression in vascular and visceral SMCs and revealed heretofore unrecognized differences between these SMC types. These results suggest that distinct transcriptional regulation programs control muscle gene expression in vascular and visceral SMCs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas dos Microfilamentos , Proteínas Musculares/biossíntese , Músculo Liso/embriologia , Regiões Promotoras Genéticas , Transcrição Gênica , Animais , Artérias/embriologia , Biomarcadores , Células Cultivadas , Genes Reporter , Coração/embriologia , Óperon Lac , Camundongos , Camundongos Transgênicos , Morfogênese , Proteínas Musculares/genética , Músculo Liso Vascular/embriologia , Veias/embriologia , Vísceras/embriologia
2.
Mol Cell Biol ; 14(3): 1647-56, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8114702

RESUMO

Members of the myocyte-specific enhancer-binding factor 2 (MEF2) family of transcription factors bind a conserved A/T-rich sequence in the control regions of numerous muscle-specific genes. Mammalian MEF2 proteins have been shown previously to be encoded by three genes, Mef2, xMef2, and Mef2c, each of which gives rise to multiple alternatively spliced transcripts. We describe the cloning of a new member of the MEF2 family from mice, termed MEF2D, which shares extensive homology with other MEF2 proteins but is the product of a separate gene. MEF2D binds to and activates transcription through the MEF2 site and forms heterodimers with other members of the MEF2 family. Deletion mutations show that the carboxyl terminus of MEF2D is required for efficient transactivation. MEF2D transcripts are widely expressed, but alternative splicing of MEF2D transcripts gives rise to a muscle-specific isoform which is induced during myoblast differentiation. The mouse Mef2, Mef2c, and Mef2d genes map to chromosomes 7, 13, and 3, respectively. The complexity of the MEF2 family of regulatory proteins provides the potential for fine-tuning of transcriptional responses as a consequence of combinatorial interactions among multiple MEF2 isoforms encoded by the four Mef2 genes.


Assuntos
Proteínas de Ligação a DNA/genética , Músculos/metabolismo , Fatores de Transcrição/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Clonagem Molecular , Primers do DNA/química , DNA Complementar/genética , Expressão Gênica , Fatores de Transcrição MEF2 , Substâncias Macromoleculares , Camundongos , Dados de Sequência Molecular , Fatores de Regulação Miogênica , RNA Mensageiro/genética , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Transcrição Gênica , Ativação Transcricional
3.
Circ Res ; 87(10): 881-7, 2000 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-11073883

RESUMO

Retinoids exert antiproliferative and prodifferentiating effects in vascular smooth muscle cells (SMCs) and reduce neointimal mass in balloon-injured blood vessels. The mechanisms through which retinoids carry out these effects are unknown but likely involve retinoid receptor-mediated changes in gene expression. Here we report the cloning, chromosomal mapping, and biological activity of the retinoid-response gene rat tissue transglutaminase (tTG). Northern blotting studies showed that tTG is rapidly and dose-dependently induced in a protein synthesis-independent manner after stimulation with the natural retinoid all-trans retinoic acid (atRA). The induction of tTG was selective for atRA and its stereoisomers 9-cis and 13-cis RA, because little or no elevation in mRNA expression was observed with a panel of growth factors. Western blotting and immunofluorescence confocal microscopy showed an accumulation of cytosolic tTG protein after atRA stimulation. Radiolabeled cross-linking studies revealed a corresponding elevation in in vitro tTG activity. The increase in tTG activity was reduced in the presence of 2 distinct inhibitors of tTG (monodansylcadaverine and cystamine). atRA-induced tTG mRNA and protein expression were followed by a significant elevation in SMC apoptosis. Such retinoid-induced programmed cell death could be partially inhibited with each tTG inhibitor and was completely blocked when both inhibitors were used simultaneously. These results establish a role for atRA in the sequential stimulation of tTG and apoptosis in cultured SMCs. atRA-mediated apoptosis in SMCs seems to require the participation of active tTG, suggesting a potential mechanistic link between this retinoid-inducible gene and programmed cell death.


Assuntos
Apoptose , Cadaverina/análogos & derivados , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Músculo Liso Vascular/metabolismo , Transglutaminases/genética , Transglutaminases/metabolismo , Tretinoína/metabolismo , Animais , Northern Blotting , Western Blotting , Cadaverina/farmacologia , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , Cistamina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/farmacologia , Masculino , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transcrição Gênica/efeitos dos fármacos , Transglutaminases/antagonistas & inibidores , Tretinoína/farmacologia
4.
Cardiovasc Res ; 36(1): 118-26, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9415280

RESUMO

OBJECTIVES: The A10 cell line was derived from the thoracic aorta of embryonic rat and is a commonly used model of vascular smooth muscle cells (VSMC). Despite its wide use this cell line has not been well characterized. This is especially important in light of recent evidence of phenotypically distinct cell populations isolated from rat vascular tissue. Therefore, the present study was undertaken to confirm the VSMC nature of A10 cells and to investigate whether these cells particularly resemble adult, neonatal, or neointimal rat VSMC. METHODS: A variety of defining characteristics were used that included immunofluorescent analysis for smooth muscle alpha-actin, smooth and non-muscle myosin heavy chains, desmin and vimentin; Western analysis for smooth muscle and non-muscle myosin heavy chains; mRNA analysis for smooth muscle myosin heavy chain, calponin, SM22 alpha, tropoelastin and PDGF-B peptide; and functional assays of cell migration, proliferation and agonist induced intracellular Ca transients. RESULTS: A10 cells expressed smooth muscle alpha-actin, SM22 alpha, smooth muscle calponin and vimentin, characteristic of in vivo rat VSMCs; however they also resembled de-differentiated smooth muscle cells in that they expressed non-muscle myosin rather than smooth muscle myosin heavy chain. A10 cells resembled cultured rat neonatal smooth muscle cells ("pup cells") in that they had an epithelioid shape and lacked functional PDGF-alpha receptors: however they did not express PDGF-B mRNA or proliferate in low serum containing medium as do neonatal cells. A10 cells had several characteristics in common with neointimal cells including the expression of alpha-actin, vimentin, and non-muscle myosin and the lack of expression of PDGF-B mRNA as well as the ability to migrate in response to PDGF-BB. CONCLUSION: In conclusion, A10 cells are nondifferentiated VSMC that differ from neonatal but bear significant resemblance to neointimal cells.


Assuntos
Linhagem Celular , Músculo Liso Vascular/citologia , Actinas/análise , Animais , Animais Recém-Nascidos , Aorta Torácica , Northern Blotting , Western Blotting , Proteínas de Ligação ao Cálcio/análise , Diferenciação Celular , Movimento Celular , Desmina/análise , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Proteínas dos Microfilamentos , Modelos Biológicos , Cadeias Pesadas de Miosina/análise , Miosinas/análise , Fator de Crescimento Derivado de Plaquetas/análise , Ratos , Tropoelastina/análise , Túnica Íntima/citologia , Vimentina/análise , Calponinas
5.
Cell Death Dis ; 6: e2011, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26633717

RESUMO

Serum response factor (SRF) is a transcription factor known to mediate phenotypic plasticity in smooth muscle cells (SMCs). Despite the critical role of this protein in mediating intestinal injury response, little is known about the mechanism through which SRF alters SMC behavior. Here, we provide compelling evidence for the involvement of SRF-dependent microRNAs (miRNAs) in the regulation of SMC apoptosis. We generated SMC-restricted Srf inducible knockout (KO) mice and observed both severe degeneration of SMCs and a significant decrease in the expression of apoptosis-associated miRNAs. The absence of these miRNAs was associated with overexpression of apoptotic proteins, and we observed a high level of SMC death and myopathy in the intestinal muscle layers. These data provide a compelling new model that implicates SMC degeneration via anti-apoptotic miRNA deficiency caused by lack of SRF in gastrointestinal motility disorders.


Assuntos
Mucosa Intestinal/metabolismo , MicroRNAs/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Humanos , Intestinos/citologia , Intestinos/patologia , Camundongos , Miócitos de Músculo Liso , Transdução de Sinais
6.
Gene ; 197(1-2): 215-24, 1997 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9332369

RESUMO

Smooth muscle cells (SMC) express a battery of cell-restricted differentiation genes, many of which are down-regulated during the course of vascular disease. Here, we present the mRNA expression, genomic structure and chromosomal mapping of the gene encoding human smooth muscle cell calponin (SMCC). Human SMCC transcripts are restricted to tissues and cells of SMC origin and, in the latter case, appear to be uniquely controlled in two distinct human SMC lines of uterine and aortic origin. Restriction mapping. Southern blot and PCR analysis of a 70-kb human bacterial artificial chromosome (BAC) revealed a genomic structure (seven exons spanning > 11 kb) very similar to that reported for the mouse SMCC gene. Using a variety of human-rodent somatic cell hybrid and radiation hybrid mapping panels, the human SMCC gene was mapped to a genomic interval of less than 1.32 Mb in 19p13.2. These results provide new information concerning the regulation of SMCC gene expression and demonstrate the utility of two human SMC lines for the further characterization of this gene's expression control. The identification of a BAC harboring the entire human SMCC locus represents an important reagent for future analysis of SMCC regulatory sequences. Finally, the localization of SMCC to a defined genomic interval will facilitate an analysis of its potential as a candidate gene for disease phenotypes mapping to 19p13.2.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 19/genética , Regulação da Expressão Gênica/fisiologia , Proteínas Musculares/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Clonagem Molecular/métodos , Éxons/genética , Genes/genética , Humanos , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Músculo Liso/citologia , Especificidade de Órgãos , RNA Mensageiro/análise , Homologia de Sequência do Ácido Nucleico , Calponinas
7.
Methods Mol Med ; 30: 25-35, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-21341013

RESUMO

Recent molecular genetic studies in cardiac and skeletal muscle have revealed mutations in a battery of sarcomeric muscle-restricted genes that appear to be associated with various myopathies (1,2). In sharp contrast, no mutations in smooth muscle cell (SMC)-restricted genes have been linked to a SMC disease phenotype, although a review of the literature indicates that many SMC diseases with a presumed genetic basis are present in human populations (3-13). An important first step in linking a disease phenotype to a mutation within a specific gene is the accurate physical mapping of the candidate gene to a specific chromosomal region within the context of other genetic markers, such as highly polymorphic microsatellite markers now routinely used for recombination-based linkage analysis of families segregating a particular disease phenotype. Several methods exist for the physical mapping of genes, including fluorescent in situ hybridization (FISH) (14) and interspecific mouse back-crossing (15). FISH analysis is relatively fast, but often requires large genomic clones and does not afford the high-resolution mapping required to link a gene locus to a disease phenotype. Interspecific mouse back-crossing can be quite powerful with respect to resolution, but studies are necessarily limited to the mouse genome. Thus, a broadly applicable, fast and simple method of gene mapping would be desirable to aid investigators in localizing potential candidate disease genes, especially those pertaining to SMC-associated diseases.

8.
In Vitro Cell Dev Biol Anim ; 34(3): 217-26, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9557939

RESUMO

Transcriptional regulation of smooth muscle cell (SMC) differentiation is a rapidly growing area of interest that has relevance for understanding intimal disease. Despite the wealth of data accumulating in vitro, however, no study has compared the cell-specific marker profile, transfectability, promoter activity, and growth characteristics among several SMC culture systems. Accordingly, we performed a comprehensive analysis of the marker profile, growth properties, transfectability, and SMC promoter activity in four rat SMC lines (A7r5, adult and pup aortic, and PAC1). Despite alterations in chromosomal number and structure, A7r5, adult aortic, and PAC1 cells express all SMC markers studied including SM alpha-actin, SM calponin, SM22, tropoelastin, and to a lesser extent, SM myosin heavy chain (SMMHC). In contrast, pup aortic cells express very low or undetectable levels of all the above markers except tropoelastin. Adult aortic, pup, and PAC1 cells display similar growth curves and levels of proto-oncogene transcripts, whereas those in the A7r5 line are comparatively less. All cell lines studied except pup cells show expression of SMC differentiation genes during active growth, indicating that growth and differentiation are not mutually exclusive in cultured smooth muscle. Transfection studies reveal dramatic differences in DNA uptake and SMC-restricted promoter activity between cell lines. Collectively, these results provide detailed information relating to SMC molecular biology in culture that should facilitate the selection of a cell line for studying the transcriptional regulatory mechanisms underlying SMC differentiation.


Assuntos
Músculo Liso Vascular/metabolismo , Actinas/análise , Animais , Biomarcadores , Proteínas de Ligação ao Cálcio/análise , Divisão Celular , Linhagem Celular , Proteínas dos Microfilamentos , Músculo Liso Vascular/citologia , Cadeias Pesadas de Miosina/análise , Regiões Promotoras Genéticas , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Soroalbumina Bovina/farmacologia , Transfecção , Calponinas
12.
J Biol Chem ; 271(12): 7095-103, 1996 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-8636144

RESUMO

Although several genes are considered markers for vascular smooth muscle cell (SMC) differentiation, few have been rigorously tested for SMC specificity in mammals, particularly during development where considerable overlap exists between different muscle gene programs. Here we describe the temporospatial expression pattern of the SMC calponin gene (formerly h1 or basic calponin) during mouse embryogenesis and in adult mouse tissues and cell lines. Whereas SMC calponin mRNA expression is restricted exclusively to SMCs in adult tissues, during early embryogenesis, SMC calponin transcripts are expressed throughout the developing cardiac tube as well as in differentiating SMCs. Transcription of the SMC calponin gene initiates at two closely juxtaposed sites in the absence of a consensus TATAA or initiator element. Transient transfection assays in cultured SMC demonstrated that high level SMC calponin promoter activity required no more than 549 nucleotides of 5 sequence. In contrast to the strict cell type-specificity of SMC calponin mRNA expression, the SMC calponin promoter showed activity in several cell lines that do not express the endogenous SMC calponin gene. These results demonstrate that SMC calponin responds to cardiac and smooth muscle gene regulatory programs and suggest that the cardiac and smooth muscle cell lineages may share a common gene regulatory program early in embryogenesis, which diverges as the heart matures. The finding that the isolated SMC calponin promoter is active in a wider range of cells than the endogenous SMC calponin gene also suggests that long-range repression or higher order regulatory mechanism(s) are involved in cell-specific regulation of SMC calponin expression.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso/metabolismo , Miocárdio/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Coração/embriologia , Camundongos , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Músculo Liso/citologia , Músculo Liso/embriologia , Miocárdio/citologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , TATA Box , Transcrição Gênica , Calponinas
13.
Am J Hosp Pharm ; 51(6): 772-7, 1994 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8010315

RESUMO

A medication-review service at a managed care facility was studied. The service, developed in 1991, provides in-depth analysis of medication use and patient consultation by a pharmacist. Patients are seen by referral. During a visit, the pharmacist reviews each medication for patterns of use, clinical response, and adverse effects and if necessary teaches the patient how to use the drugs more appropriately. The pharmacist may change some aspect of the prescription and may schedule follow-up visits. The investigators reviewed data on (1) medication-related problems for all new patients seen by the pharmacist during the first 12 months of the service, (2) patient demographics for the first 23 months, (3) utilization of the service and pharmacist productivity for the first 23 months, (4) utilization of medications and health care services in a representative subset of patients 12 months before and after they used the service, and (5) costs. A total of 2720 medications were reviewed during months 1-12. On average, 64.9% of the drugs reviewed each month were problematic. A total of 836 patients were seen during the first 23 months; most of the referrals were from physicians. The patients averaged 2.6 diagnoses and 4.7 drugs each and were not dominated by any one age group, gender, diagnosis, or drug therapy. A representative subgroup showed reductions in the number of unscheduled physician visits, urgent care visits, emergency room visits, and hospital days; a savings of $644 per patient per year was calculated. Patients used fewer health services during the year after they began participating in a pharmacist-managed medication-review program.


Assuntos
Revisão de Uso de Medicamentos/organização & administração , Programas de Assistência Gerenciada/normas , Educação de Pacientes como Assunto/organização & administração , Assistência Farmacêutica/organização & administração , Adolescente , Adulto , Idoso , California , Redução de Custos , Eficiência , Feminino , Humanos , Masculino , Programas de Assistência Gerenciada/economia , Pessoa de Meia-Idade , Assistência Farmacêutica/economia , Encaminhamento e Consulta
14.
Biochem Biophys Res Commun ; 281(2): 475-82, 2001 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-11181072

RESUMO

A modified suppression subtractive hybridization assay was performed to uncover genes induced by all-trans retinoic acid in cultured smooth muscle cells (SMC). Northern blotting studies confirmed the induction of 14 genes, many of which have heretofore been unrecognized as retinoid-inducible. Temporal expression and cycloheximide studies allowed us to categorize these genes as either immediate-early (LOX-1, endolyn, Stoned B/TFIIA alpha/beta-like factor, Src Suppressed C Kinase Substrate, and tissue transglutaminase) or delayed (cathepsin-L, ceruloplasmin, epithelin, importin alpha, alpha(8)-integrin, lactate dehydrogenase B, retinol dehydrogenase, spermidine/spermine N(1)-acetyltransferase, and VCAM-1) retinoid-response genes. A survey of rat tissues showed two of the genes (tissue transglutaminase and alpha(8)-integrin) to be highly restricted to vascular tissue. In situ hybridization verified expression of both tissue transglutaminase and alpha(8)-integrin to SMC in balloon-injured rat carotid artery. These findings unveil a new retinoid-response gene set that should be exploited to define molecular pathways involved in the antagonistic effects of retinoids on SMC growth and neointimal formation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Retinoides/farmacologia , Animais , Northern Blotting , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Células Cultivadas , Cicloeximida/farmacologia , Hibridização In Situ , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Hibridização de Ácido Nucleico/métodos , Inibidores da Síntese de Proteínas/farmacologia , RNA/efeitos dos fármacos , RNA/genética , RNA/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Distribuição Tecidual , Tretinoína/farmacologia
15.
Mamm Genome ; 12(3): 187-91, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11252166

RESUMO

Smooth muscle calponin (Cnn1) is a multifunctional protein whose expression is tightly restricted to differentiated smooth muscle cell (SMC) lineages during embryonic and post-natal life. As such, Cnn1 represents an ideal locus from which to dissect out regulatory elements that control its expression and hence the mature SMC phenotype. Previous work has focused on the expression and chromosomal mapping of the rat and human Cnn1 orthologs. In this report, we describe a unique pattern of Cnn1 expression during the growth and differentiation of BC3H1 cells, a mouse cell line that has transcriptional characteristics of both smooth and skeletal muscle lineages. Actively growing BC3H1 cells exhibit Cnn1 mRNA expression, which is extinguished when these cells are induced to differentiate upon serum withdrawal. Replating differentiated BC3H1 cells restores steady-state Cnn1 mRNA levels. The down-regulation of Cnn1 mRNA during BC3H1 differentiation coincides with the induction of myogenin, a skeletal muscle transcription factor that is not present in SMC lineages. Results from cycloheximide and actinomycin D studies suggest the existence of a labile repressor protein(s) that destabilizes the pool of Cnn1 mRNA and/or silences transcription of the Cnn1 locus. Mapping of the mouse Cnn1 locus to Chr 9, which is homologous to human Cnn1 on 19p13.2 and rat Cnn1 on 8q, suggests no gross rearrangement of this locus in the BC3H1 cell line. These results are the first to show reversible expression of Cnn1 and demonstrate the utility of the BC3H1 muscle cell line as a model system for the further characterization of Cnn1 gene regulation.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas Musculares/genética , Músculo Liso , Animais , Linhagem Celular , Mapeamento Cromossômico , Expressão Gênica , Cariotipagem , Camundongos , Proteínas dos Microfilamentos , Músculo Esquelético/fisiologia , RNA Mensageiro , Calponinas
16.
Am J Pathol ; 137(4): 761-5, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2221010

RESUMO

To study the mechanism(s) of vascular smooth muscle cell proliferation in vivo, mRNA levels of c-fos, c-jun, and c-myc were determined by Northern blot analysis following vascular balloon de-endothelialization (BDE). Medial smooth muscle cells (SMC) were separated and studied by enzymatic digestion of the vessel wall. mRNA levels of c-fos and c-jun from aortic smooth muscle cells (SMC) were simultaneously induced within 30 minutes of BDE and declined to baseline by 1.5 hours, c-myc mRNA did not begin to increase until 1 hour after vascular injury. Levels of c-myc peaked at 2 hours and were sustained for an additional 4 hours before gradually declining. Smooth muscle cells derived from enzyme-treated control aortae that did not undergo BDE expressed c-fos and c-jun, but showed no evidence of c-myc message. In contrast, nonenzymatically treated, non-BDE whole aortae (containing both media and adventitia) demonstrated a prominent c-myc signal, but failed to express c-fos and c-jun. Corresponding examination of adventitia derived from enzyme-treated aortae showed this tissue to be a source of all three proto-oncogenes. The results of this study demonstrate the earliest in vivo molecular markers of vascular injury reported to date and implicate SMC proto-oncogene expression in the initiation of SMC proliferation. Furthermore these findings suggest two avenues for proto-oncogene induction, that are due to (1) vessel wall manipulation and (2) humoral stimulation.


Assuntos
Endotélio Vascular/fisiologia , Expressão Gênica , Músculo Liso Vascular/metabolismo , Proto-Oncogenes/fisiologia , Animais , Aorta , Divisão Celular , Sondas de DNA , Masculino , Modelos Biológicos , Músculo Liso Vascular/citologia , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos
17.
Circ Res ; 75(5): 803-12, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7923625

RESUMO

We cloned a portion of the mouse smooth muscle myosin heavy chain (SM-MHC) cDNA and analyzed its mRNA expression in adult tissues, several cell lines, and developing mouse embryos to determine the suitability of the SM-MHC promoter as a tool for identifying smooth muscle-specific transcription factors and to define the spatial and temporal pattern of smooth muscle differentiation during mouse development. RNase protection assays showed SM-MHC mRNA in adult aorta, intestine, lung, stomach, and uterus, with little or no signal in brain, heart, kidney, liver, skeletal muscle, spleen, and testes. From an analysis of 14 different cell lines, including endothelial cells, fibroblasts, and rhabdomyosarcomas, we failed to detect any SM-MHC mRNA; all of the cell lines induced to differentiate also showed no detectable SM-MHC. In situ hybridization of staged mouse embryos first revealed SM-MHC transcripts in the early developing aorta at 10.5 days post coitum (dpc). No hybridization signal was demonstrated beyond the aorta and its arches until 12.5 to 13.5 dpc, when SM-MHC mRNA appeared in smooth muscle cells (SMCs) of the developing gut and lungs as well as peripheral blood vessels. By 17.5 dpc, SM-MHC transcripts had accumulated in esophagus, bladder, and ureters. Except for blood vessels, no SM-MHC transcripts were ever observed in developing brain, heart, or skeletal muscle. These results indicate that smooth muscle myogenesis begins by 10.5 days of embryonic development in the mouse and establish SM-MHC as a highly specific marker for the SMC lineage. The SM-MHC promoter should therefore serve as a useful model for defining the mechanisms that govern SMC transcription during development and disease.


Assuntos
Músculo Liso/embriologia , Miosinas/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Cultivadas , DNA Complementar/isolamento & purificação , Feminino , Expressão Gênica , Marcadores Genéticos , Hibridização In Situ , Masculino , Camundongos , Dados de Sequência Molecular , Músculo Liso/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Transcrição Gênica
18.
J Biol Chem ; 276(36): 34175-81, 2001 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-11447226

RESUMO

Retinoids block smooth muscle cell (SMC) proliferation and attenuate neointimal formation after vascular injury, presumably through retinoid receptor-mediated changes in gene expression. To identify target genes in SMC whose encoded proteins could contribute to such favorable biological effects, we performed a subtractive screen for retinoid-inducible genes in cultured SMC. Here, we report on the cloning and initial characterization of a novel retinoid-inducible serine carboxypeptidase (RISC). Expression of RISC is low in cultured SMC but progressively increases over a 5-day time-course treatment with all-trans-retinoic acid. A near full-length rat RISC cDNA was cloned and found to have a 452-amino acid open reading frame containing an amino-terminal signal sequence, followed by several conserved domains comprising the catalytic triad common to members of the serine carboxypeptidase family. In vitro transcription and translation experiments showed that the rat RISC cDNA generates an approximately 51-kDa protein. Confocal immunofluorescence microscopy of COS-7 cells transiently transfected with a RISC-His tag plasmid revealed cytosolic localization of the fusion protein. Western blotting studies using conditioned medium from transfected COS-7 cells suggest that RISC is a secreted protein. Tissue Northern blotting studies demonstrated robust expression of RISC in rat aorta, bladder, and kidney with much lower levels in all other tissues analyzed; high level RISC expression was also observed in human kidney. In situ hybridization verified the localization of RISC to medial SMC of the adult rat aorta. Interestingly, expression in kidney was restricted to proximal convoluted tubules; little or no expression was observed in glomerular cells, distal convoluted and collecting tubules, or medullary cells. Radiation hybrid mapping studies placed the rat RISC locus on chromosome 10q. These studies reveal a novel retinoid-inducible protease whose activity may be involved in vascular wall and kidney homeostasis.


Assuntos
Carboxiliases/genética , Carboxipeptidases/química , Carboxipeptidases/genética , Músculo Liso Vascular/metabolismo , Sequência de Aminoácidos , Animais , Aorta/metabolismo , Sequência de Bases , Northern Blotting , Western Blotting , Células COS , Carboxiliases/química , Carboxipeptidases/biossíntese , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar/metabolismo , Hibridização In Situ , Rim/metabolismo , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Fases de Leitura Aberta , Fenótipo , Plasmídeos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Mapeamento de Híbridos Radioativos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual , Transcrição Gênica , Transfecção
19.
Microcirculation ; 8(6): 403-13, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781813

RESUMO

OBJECTIVE: Our purpose was to develop a method for adenovirus delivery to the hamster cheek pouch to experimentally target gene transfer in tissue used for microvascular studies. METHODS: Separate constructs were tested with transgenes for lacZ or green fluorescent protein (GFP) driven by three promoters: RSV, CMV, and SM22. With university approval, adenovirus was delivered in anesthetized (pentobarbital, 70 mg/kg) hamsters (n = 28) by using either a vascular systemic injection or tissue infiltration (interstitial space behind the pouch). During 3 days, animals receiving infiltration gained the expected weight, whereas those receiving vascular injection lost weight; no other behavior changes were noted. RESULTS: On day 3 postadenoviral delivery (infiltration), expression of lacZ (histology, beta-galactosidase) or GFP (fluorescence microscopy) was confirmed across the tissue (CMV and RSV promoters) and exclusively in vascular smooth muscle cells (specific SM22 promoter), without evidence of tissue inflammation. In vitro microvascular experiments verified normal responses in the cheek pouch of day 3 postadenoviral delivery animals. We tested local dilation to methacholine, adenosine, remote dilation to methacholine, adenosine, nitroprusside, and LM609 (alpha(v)beta3 integrin agonist), flow-dependent dilation, and flow recruitment. CONCLUSIONS: Thus, this method enables targeted, cell-specific gene transfer to one tissue important for microvascular studies, without significant systemic exposure and without adverse inflammation.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Liso Vascular/metabolismo , Animais , Bochecha , Cricetinae , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Vetores Genéticos/toxicidade , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/farmacocinética , Masculino , Mesocricetus , Microcirculação , Regiões Promotoras Genéticas , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , beta-Galactosidase/farmacocinética
20.
Circ Res ; 82(5): 566-75, 1998 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-9529161

RESUMO

In recent years, significant progress has been made toward understanding skeletal muscle development. However, the mechanisms that regulate smooth muscle development and differentiation are presently unknown. To better understand smooth muscle-specific gene expression, we have focused our studies on the smooth muscle myosin heavy chain (SMHC) gene, a highly specific marker of differentiated smooth muscle cells. The goal of the present study was to isolate and characterize the mouse SMHC gene promoter, since the mouse promoter would be particularly suited for in vivo promoter analyses in transgenic mice and would serve as a tool for targeting genes of interest into smooth muscle cells. We report here the isolation and characterization of the mouse SMHC promoter and its 5' flanking region. DNA sequence analysis of a 2.6-kb portion of the promoter identified several potential binding sites for known transcription factors. Transient transfection analysis of promoter deletion constructs in primary cultures of smooth muscle cells showed that the region between -1208 and -1050 bp is critical for maximal SMHC promoter activity. A comparison of SMHC promoter sequences from mouse, rat, and rabbit revealed the presence of a highly conserved region located between -967 and -1208 bp. This region includes three CArG/CArG*-like elements, two SP-1 binding sites, a NF-1-like element, an Nkx2-5 binding site, and an Elk-1 binding site. Gel mobility shift assay and DNase I footprinting analyses show that all three CArG/CArG*-like elements can form DNA-protein complexes with nuclear extract from vascular smooth muscle cells. Protein binding to the CArG* elements can be competed out by either serum response element or by an authentic CArG element from the cardiac alpha-actin gene. Using a serum response factor (SRF) antibody, we demonstrate that SRF is part of the protein complex. In addition, we show that cotransfection with the SRF dominant-negative mutant expression vector abolishes SMHC promoter activity, suggesting that SRF protein plays a critical role in SMHC gene regulation.


Assuntos
Sequência Conservada , Músculo Liso Vascular/enzimologia , Cadeias Pesadas de Miosina/genética , Regiões Promotoras Genéticas/genética , Animais , Aorta Torácica/citologia , Sequência de Bases , Núcleo Celular/química , Células Cultivadas , Pegada de DNA , Desoxirribonuclease I , Regulação da Expressão Gênica , Camundongos , Dados de Sequência Molecular , Coelhos , Ratos , Análise de Sequência de DNA , Fatores de Transcrição/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA