Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 369
Filtrar
1.
2.
Handb Exp Pharmacol ; (176 Pt 2): 335-50, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16999232

RESUMO

Heart failure associated with coronary artery disease is a major cause of morbidity and mortality. Recent developments in the understanding of the molecular mechanisms of heart failure have led to the identification of novel therapeutic targets which, combined with the availability of efficient gene delivery vectors, offer the opportunity for the design of gene therapies for protection of the myocardium. Viral-based therapies have been developed to treat polygenic and complex diseases such as myocardial ischaemia, hypertension, atherosclerosis and restenosis. Some of these experimental therapies are now undergoing clinical evaluation in patients with cardiovascular diseases. In this review we will focus on the latest advances in the field of gene therapy for treatment of heart failure and their clinical application.


Assuntos
Terapia Genética/tendências , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Cardiomegalia/metabolismo , DNA Antissenso/genética , DNA Antissenso/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Humanos , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
3.
Gene Ther ; 11(12): 962-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15029230

RESUMO

In high-risk patients, the ideal cardiovascular gene therapy requires a strategy that provides long-term protection of myocardium against episodes of ischemic/reperfusion injury. We report the development of an efficient, long-lasting pre-emptive gene therapy strategy in a rat model of ischemic-reperfusion (I/R) injury of heart. At 6 weeks prior to myocardial injury, the human extracellular superoxide dismutase (Ec-SOD) gene was delivered by direct intramyocardial injections, using a recombinant adeno-associated virus vector. Significant myocardial protection was documented by the decrease in infarct size at 24 h post I/R, improved left ventricular function at 7 weeks postinjury, and enhanced long-term survival in the SOD treated group. This concept of preinjury delivery and 'pre-emptive' gene therapy via the expression of a secreted protein that renders paracrine therapeutic action can be an effective strategy for organ protection against future injury.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Superóxido Dismutase/genética , Transdução Genética/métodos , Animais , Líquido Extracelular/enzimologia , Masculino , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Traumatismo por Reperfusão Miocárdica/mortalidade , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/enzimologia , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Fatores de Tempo , Função Ventricular
4.
Gene Ther ; 10(18): 1600-7, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12907952

RESUMO

The development of accurate, safe, and efficient gene delivery remains a major challenge towards the realization of gene therapeutic prevention and treatment of cardiovascular diseases. In this study, we investigated the ability of high-intensity focused ultrasound (HIFU), a form of mechanical wave transmission, to act as a noninvasive tool for the enhancement of in vivo gene transfer into rabbit carotid arteries. Segments of the common carotid arteries of New Zealand white rabbits were isolated and infused with plasmid DNA encoding the reporter beta-galactosidase either with or without the addition of ultrasound contrast agent consisting of small (approximately 2-5 microm) gas-filled human albumin microspheres to augment cavitation. Infused arteries were exposed to pulsed ultrasound for 1 min (frequency 0.85 MHz, burst length 50 ms, repetition frequency 1 Hz, duration 60 s, peak pressure amplitude of 15 MPa). At 6.3 MPa, HIFU enhanced gene expression eight-fold, and 17.5-fold in the presence of contrast. We found increasing amounts of beta-galactosidase expression in the carotid vessel with increasing pressure amplitude. This dose-response relation was present with and without contrast. Without contrast, no vessel damage was detected up to 15 MPa, while the addition of contrast induced side effects above a threshold of 6.3 MPa peak pressure. The entire procedure was feasible and safe for the animals, and the results suggest that HIFU has the potential to assist in the noninvasive spatial regulation of gene transfer into the vascular system.


Assuntos
Artérias Carótidas/enzimologia , Doenças das Artérias Carótidas/terapia , DNA/administração & dosagem , Terapia Genética/métodos , Transfecção/métodos , Terapia por Ultrassom/métodos , Animais , Expressão Gênica , Técnicas In Vitro , Óperon Lac , Microesferas , Modelos Animais , Coelhos
5.
J Endocrinol ; 175(2): 475-85, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12429045

RESUMO

Atherosclerotic cardiovascular disease results from complex interactions among multiple genetic and environmental factors. Thus, it is important to elucidate the influence of each factor on cholesterol metabolism. For this purpose, transgenic/gene-targeting technology is a powerful tool for studying gene functions. However, this technology has several disadvantages such as being time consuming and expensive. Accordingly, we established new animal models using in vivo gene transfer technology. In this study, we examined the feasibility of the creation of a new animal model for the study of atherosclerosis. We hypothesized that apolipoprotein (apo) E-deficient mice can be created by systemic administration of antisense apo E oligodeoxynucleotides (ODN) coupled to the HVJ-liposome complex. Initially, we examined the localization and cellular fate of FITC-labeled antisense ODN administered intravenously. FITC-labeled ODN transfection by the HVJ-liposome method resulted in fluorescence in the liver, spleen and kidney, but not in other organs such as brain. Moreover, fluorescence with the HVJ-liposome method was sustained for up to 2 weeks after transfection, which resulted in a striking difference from transfection of ODN alone or ODN in liposomes without HVJ, which showed rapid disappearance of fluorescence (within 1 day). Given these unique characteristics of the HVJ-liposome method, we next examined transfection of antisense apo E ODN by intravenous administration. Transfection of antisense apo E ODN resulted in a marked reduction of apo E mRNA levels in the liver, but no change in apo B and beta-actin mRNA levels. In mice fed a normal diet, a transient increase in cholesterol and triglyceride levels was observed in the antisense apo E-treated group, but they returned to normal levels by 6 days after transfection. Similar findings were also found in mice fed a high cholesterol diet. Neither scrambled nor mismatched ODN resulted in any increase in cholesterol. To make chronic hypercholesterolemic mice, we therefore performed repeated injections of apo E antisense ODN. Whenever antisense apo E ODN were injected, mice showed a transient increase in cholesterol and triglyceride. Cumulative administration of antisense apo E ODN resulted in a sustained increase in cholesterol for up to 3 weeks after the last transfection. Finally, mice treated with repeated injections of antisense apo E every week developed sustained hypercholesterolemia and hypertriglyceridemia until withdrawal of injections. Apolipoprotein-deficient mice created by intravenous administration of antisense ODN are a promising new animal model to help understand the role of apolipoprotein in vivo and develop a new drug therapy targeting apolipoprotein.


Assuntos
Apolipoproteínas E/deficiência , Arteriosclerose/metabolismo , Modelos Animais de Doenças , Lipoproteínas/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Animais , Northern Blotting , Colesterol/análise , Colesterol na Dieta/administração & dosagem , Dieta , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Oligonucleotídeos Antissenso/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Triglicerídeos/análise
6.
Proc Natl Acad Sci U S A ; 98(23): 13219-24, 2001 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-11687612

RESUMO

The mineralocorticoid hormone, aldosterone, is known to play a role in sodium homeostasis. We serendipitously found, however, highly significant association between single-nucleotide polymorphisms in the aldosterone synthase gene and plasma glucose levels in a large population of Chinese and Japanese origin. Two polymorphisms--one in the putative promoter (T-344C) and another resulting in a lysine/arginine substitution at amino acid 173, which are in complete linkage disequilibrium in this population--were associated with fasting plasma glucose levels (P = 0.000017) and those 60 (P = 0.017) and 120 (P = 0.0019) min after an oral glucose challenge. A C/T variant in intron 1, between these polymorphisms, was not associated with glucose levels. Arg-173 and -344C homozygotes were most likely to be diabetic [odds ratio 2.51; 95% confidence interval (C.I.) 1.39-3.92; P = 0.0015] and have impaired fasting glucose levels (odds ratio 3.53; 95% C.I. 2.02-5.5; P = 0.0000036). These results suggest a new role for aldosterone in glucose homeostasis.


Assuntos
Glicemia/análise , Citocromo P-450 CYP11B2/genética , Variação Genética , Adulto , Sequência de Bases , Primers do DNA , Diabetes Mellitus/sangue , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Feminino , Genótipo , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade
7.
Am J Cardiol ; 88(9A): 1L-20L, 2001 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-11694220

RESUMO

Angiotensin-converting enzyme (ACE) is primarily localized (>90%) in various tissues and organs, most notably on the endothelium but also within parenchyma and inflammatory cells. Tissue ACE is now recognized as a key factor in cardiovascular and renal diseases. Endothelial dysfunction, in response to a number of risk factors or injury such as hypertension, diabetes mellitus, hypercholesteremia, and cigarette smoking, disrupts the balance of vasodilation and vasoconstriction, vascular smooth muscle cell growth, the inflammatory and oxidative state of the vessel wall, and is associated with activation of tissue ACE. Pathologic activation of local ACE can have deleterious effects on the heart, vasculature, and the kidneys. The imbalance resulting from increased local formation of angiotensin II and increased bradykinin degradation favors cardiovascular disease. Indeed, ACE inhibitors effectively reduce high blood pressure and exert cardio- and renoprotective actions. Recent evidence suggests that a principal target of ACE inhibitor action is at the tissue sites. Pharmacokinetic properties of various ACE inhibitors indicate that there are differences in their binding characteristics for tissue ACE. Clinical studies comparing the effects of antihypertensives (especially ACE inhibitors) on endothelial function suggest differences. More comparative experimental and clinical studies should address the significance of these drug differences and their impact on clinical events.


Assuntos
Peptidil Dipeptidase A/fisiologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Sistema Cardiovascular/enzimologia , Sistema Cardiovascular/fisiopatologia , Doença das Coronárias/tratamento farmacológico , Doença das Coronárias/enzimologia , Doença das Coronárias/fisiopatologia , Endotélio Vascular/fisiopatologia , Coração/fisiopatologia , Humanos , Rim/enzimologia , Rim/fisiopatologia , Nefropatias/enzimologia , Nefropatias/fisiopatologia , Miocárdio/enzimologia , Peptidil Dipeptidase A/genética , Disfunção Ventricular Esquerda/fisiopatologia
8.
J Mol Cell Cardiol ; 33(10): 1879-86, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11603929

RESUMO

The number of genes encoded by the human genome has long sought to be determined. With the recent publications of the complete sequence of the human genome, the number of genes encoded by the human genome has now been estimated to be approximately 32,000-38,000. Now the next step will be to determine which of these genes are expressed in a given cell, tissue or organ. Using three unique approaches taking advantage of our current cardiovascular EST database and the complete nucleotide sequence of human chromosomes 21 and 22 as well as cDNA microarray hybridization, we estimate that between 20,930-27,160 genes are expressed in the cardiovascular system.


Assuntos
Sistema Cardiovascular , Genoma Humano , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 22 , Análise por Conglomerados , DNA Complementar/metabolismo , Etiquetas de Sequências Expressas , Humanos , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos
9.
Life Sci ; 69(5): 509-16, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11510946

RESUMO

The recently cloned angiotensin II type 2 (AT2) receptor is a member of the seven transmembrane G-protein coupled receptor superfamily with a relatively low sequence homology with the angiotensin II type 1 (AT1) receptor subtype and counteracts the growth action of AT1 receptor. Intracellular third loops are known to be involved in interactions with various G proteins. We hypothesized that the intracellular third loop plays critical roles in determining the specificity of opposite functions of AT1 and AT2 receptor subtypes and examined this possibility using chimeric AT1 receptor, of which intracellular third loop is replaced with that of AT2 receptor. We transfected this chimeric receptor into PC 12 cells and observed that stimulation of this receptor inhibited extracellular signal-regulated kinase (ERK) activation and induces apoptosis, whereas the binding characteristics of this receptor remained those of ATI receptor. Taken together, these results support the notion that intracellular third loop is the critical determinant for mutually antagonistic AT1 and AT2 receptors' signaling pathways.


Assuntos
Apoptose , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Angiotensina/metabolismo , Animais , Técnicas de Transferência de Genes , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Células PC12 , Estrutura Terciária de Proteína , Ratos , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
10.
Genome Res ; 11(7): 1262-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11435409

RESUMO

To make large-scale association studies a reality, automated high-throughput methods for genotyping with single-nucleotide polymorphisms (SNPs) are needed. We describe PCR conditions that permit the use of the TaqMan or 5' nuclease allelic discrimination assay for typing large numbers of individuals with any SNP and computational methods that allow genotypes to be assigned automatically. To demonstrate the utility of these methods, we typed >1600 individuals for a G-to-T transversion that results in a glutamate-to-aspartate substitution at position 298 in the endothelial nitric oxide synthase gene, and a G/C polymorphism (newly identified in our laboratory) in intron 8 of the 11-beta hydroxylase gene. The genotyping method is accurate-we estimate an error rate of fewer than 1 in 2000 genotypes, rapid-with five 96-well PCR machines, one fluorescent reader, and no automated pipetting, over one thousand genotypes can be generated by one person in one day, and flexible-a new SNP can be tested for association in less than one week. Indeed, large-scale genotyping has been accomplished for 23 other SNPs in 13 different genes using this method. In addition, we identified three "pseudo-SNPs" (WIAF1161, WIAF2566, and WIAF335) that are probably a result of duplication.


Assuntos
Polimorfismo de Nucleotídeo Único/genética , Alelos , Pareamento Incorreto de Bases/genética , Genótipo , Humanos , Reação em Cadeia da Polimerase/métodos , Mapeamento de Híbridos Radioativos , Taq Polimerase/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 21(7): 1152-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11451744

RESUMO

Cell cycle progression represents a key event in vascular proliferative diseases, one that depends on an increased rate of protein synthesis. An increase in phosphatidylinositol 3-kinase (PI 3-kinase) activity is associated with vascular smooth muscle cell proliferation, and rapamycin, which blocks the activity of the mammalian target of rapamycin, inhibits this proliferation in vitro and in vivo. We hypothesized that these 2 molecules converge on a critical pathway of translational regulation that is essential for successful upregulation of cell cycle-regulatory proteins in activated smooth muscle cells. p70(S6) kinase, a target of PI 3-kinase and the mammalian target of rapamycin, was rapidly activated on growth factor stimulation of quiescent coronary artery smooth muscle cells and after balloon injury of rat carotid arteries. The translational repressor protein 4E-binding protein 1 was similarly hyperphosphorylated under these conditions. These events were associated with increases in the protein levels of cyclin B1, cyclin D1, cyclin E, cyclin-dependent kinase 1, cyclin-dependent kinase 2, proliferating cell nuclear antigen, and p21(Cip1) in vivo and in vitro, whereas inhibition of the PI 3-kinase signaling pathway with either rapamycin or wortmannin blocked the upregulation of these cell cycle proteins, but not mRNA, and arrested the cells in vitro before S phase. In contrast to findings in other cell types, growth factor- or balloon injury-induced downregulation of the cell cycle inhibitor p27(Kip1) was not affected by rapamycin treatment. These data suggest that cell cycle progression in vascular cells in vitro and in vivo depends on the integrity of the PI 3-kinase signaling pathway in allowing posttranscriptional accumulation of cell cycle proteins.


Assuntos
Arteriopatias Oclusivas/metabolismo , Proteínas de Ciclo Celular/biossíntese , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Sirolimo/farmacologia , Angioplastia com Balão/efeitos adversos , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Divisão Celular , Células Cultivadas , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais
12.
Circ Res ; 89(2): 168-73, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11463724

RESUMO

Heme oxygenase (HO)-1 degrades the pro-oxidant heme and generates carbon monoxide and antioxidant bilirubin. We have previously shown that in response to hypoxia, HO-1-null mice develop infarcts in the right ventricle of their hearts and that their cardiomyocytes are damaged by oxidative stress. To test whether HO-1 protects against oxidative injury in the heart, we generated cardiac-specific transgenic mice overexpressing different levels of HO-1. By use of a Langendorff preparation, hearts from transgenic mice showed improved recovery of contractile performance during reperfusion after ischemia in an HO-1 dose-dependent manner. In vivo, myocardial ischemia and reperfusion experiments showed that infarct size was only 14.7% of the area at risk in transgenic mice compared with 56.5% in wild-type mice. Hearts from these transgenic animals had reduced inflammatory cell infiltration and oxidative damage. Our data demonstrate that overexpression of HO-1 in the cardiomyocyte protects against ischemia and reperfusion injury, thus improving the recovery of cardiac function.


Assuntos
Heme Oxigenase (Desciclizante)/genética , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Animais , Regulação Enzimológica da Expressão Gênica/fisiologia , Genótipo , Coração/fisiopatologia , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Humanos , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia
15.
Ann Med ; 33(3): 153-5, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11370767

RESUMO

Autologous saphenous vein is the conduit of choice for the bypass of arterial occlusive disease, be it in the peripheral arterial tree or in the coronary system. This technique is limited by primary graft failure rates approaching 20% in the first year for peripheral arterial disease and 50% at 10 years for coronary artery bypass grafting. The PREVENT trial describes a novel, safe and effective means of ex vivo transfection of harvested vein grafts with an E2F decoy oligonucleotide, with 70-74% decreases in the level of proliferating cell nuclear antigen (PCNA) and c-myc mRNA expressed by the smooth muscle cells in the vein. This translated into a statistically significant reduction in primary graft failure when used to bypass peripheral arterial occlusions in a high-risk human patient population.


Assuntos
Ponte de Artéria Coronária , Doença das Coronárias/cirurgia , Terapia Genética , Animais , Feminino , Humanos , Técnicas In Vitro , Masculino , Oligonucleotídeos/uso terapêutico
16.
Hypertension ; 37(4): 1047-52, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11304501

RESUMO

There is increasing evidence that direct pathobiological events in the vessel wall play an important role in vascular disease. An important mechanism involves the perturbation of the homeostatic balance between NO and reactive oxygen species. Increased reactive oxygen species can inactivate NO and produce peroxynitrite. Angiotensin II is a potent mediator of oxidative stress and stimulates the release of cytokines and the expression of leukocyte adhesion molecules that mediate vessel wall inflammation. Inflammatory cells release enzymes (including ACE) that generate angiotensin II. Thus, a local positive-feedback mechanism could be established in the vessel wall for oxidative stress, inflammation, and endothelial dysfunction. Angiotensin II also acts as a direct growth factor for vascular smooth muscle cells and can stimulate the local production of metalloproteinases and plasminogen activator inhibitor. Taken together, angiotensin II can promote vasoconstriction, inflammation, thrombosis, and vascular remodeling. In this article, we propose a model that unifies the interrelationship among cardiovascular risk factors, angiotensin II, and the pathobiological mechanisms contributing to cardiovascular disease. This model may also explain the beneficial effects of ACE inhibitors on cardiovascular events beyond blood pressure reduction.


Assuntos
Angiotensina II/metabolismo , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Angiotensina II/biossíntese , Moléculas de Adesão Celular/metabolismo , Comunicação Celular , Citocinas/metabolismo , Ativação Enzimática , Homeostase , Humanos , Inflamação , Modelos Biológicos , Óxido Nítrico/metabolismo , Peptidil Dipeptidase A/metabolismo , Espécies Reativas de Oxigênio , Fatores de Risco
17.
Curr Cardiol Rep ; 3(3): 198-207, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11305973

RESUMO

Heart failure is not a single disease entity, but a syndrome with various causes, including hypertension, ischemic and congenital heart disease, cardiomyopathy, and myocarditis. Because of the multiple etiologies and secondary adaptations contributing to heart failure, the study of the cellular and molecular mechanisms underlying the development and progression of this syndrome has been rather challenging. Much has been learned about the remodeling processes in heart failure, which involve complex interactions among numerous mediators in signaling and regulatory pathways. The Human Genome Project and related projects have provided a preliminary database for a genome-wide analysis of complex polygenic disorders such as heart failure. With the aid of expressed sequence tag technology and microarray applications, both known and previously uncharacterized genes involved in the induction and regression of cardiac hypertrophy and its progression to heart failure can be analyzed simultaneously. Deciphering the complexity of sequence-structure-function relationships in heart failure is a goal for the future, and will require advances in structural biology, proteomics, and computational technology. In this review, we summarize the cellular and molecular aspects of heart failure, and how recent applications of genomic technologies have been successful in achieving a more complete portrait of gene expression in this pathologic state.


Assuntos
Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Animais , Humanos , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/fisiologia
18.
J Thorac Cardiovasc Surg ; 121(4): 714-22, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11279413

RESUMO

OBJECTIVE: We tested the hypothesis that a single intraoperative transfection of rabbit vein grafts with a decoy oligonucleotide that blocks cell-cycle gene transactivation by the transcription factor E2F induces long-term stable adaptation that involves medial hypertrophy and a resistance to neointimal hyperplasia and atherosclerosis. METHODS: Jugular vein to carotid artery interposition vein grafts in hypercholesterolemic rabbits were treated, using pressure-mediated delivery, with either E2F decoy oligonucleotide, scrambled oligonucleotide, or vehicle alone. E2F decoy inhibition of cell-cycle gene expression was determined by measuring proliferating cell nuclear antigen upregulation and bromodeoxyuridine incorporation in vascular smooth muscle cells. Neointimal hyperplasia and atherosclerosis were compared between groups at 6 months after operation. Wall stress was derived from the ratio of luminal radius to wall thickness. Normal rabbits exposed to 6 weeks of diet-induced hypercholesterolemia starting 6 months after operation were analyzed in the same manner. RESULTS: The E2F decoy oligonucleotide, but not scrambled oligonucleotide or vehicle alone, inhibited proliferating cell nuclear antigen expression and smooth muscle cell proliferation. Furthermore, this manipulation of cell-cycle gene expression yielded an inhibition of neointimal hyperplasia and atherosclerotic plaque formation throughout the 6 months of cholesterol feeding. In normocholesterolemic rabbits, vehicle-treated and scrambled oligonucleotide-treated vein grafts remain susceptible to diet-induced atherosclerosis as well, whereas resistance to this disease induction remained stable in genetically engineered grafts. CONCLUSION: A single intraoperative pressure-mediated delivery of E2F decoy effectively provides vein grafts with long-term resistance to neointimal hyperplasia and atherosclerosis. These findings suggest that long-term reduction in human vein graft failure rates may be feasible with this ex vivo gene therapy approach.


Assuntos
Arteriosclerose/terapia , Doenças das Artérias Carótidas/prevenção & controle , Artéria Carótida Primitiva/cirurgia , Proteínas de Transporte , Proteínas de Ciclo Celular/uso terapêutico , Proteínas de Ligação a DNA , Terapia Genética/métodos , Veias Jugulares/transplante , Fatores de Transcrição/uso terapêutico , Anastomose Cirúrgica , Animais , Antimetabólitos/farmacocinética , Antimetabólitos/uso terapêutico , Arteriosclerose/induzido quimicamente , Arteriosclerose/genética , Arteriosclerose/metabolismo , Bromodesoxiuridina/farmacocinética , Bromodesoxiuridina/uso terapêutico , Doenças das Artérias Carótidas/induzido quimicamente , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Colesterol na Dieta/toxicidade , Sondas de DNA/química , Progressão da Doença , Fatores de Transcrição E2F , Hipertrofia , Veias Jugulares/metabolismo , Veias Jugulares/patologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Antígeno Nuclear de Célula em Proliferação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Coelhos , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Transfecção , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/patologia , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...