Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Cancer Gene Ther ; 19(7): 476-88, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22595794

RESUMO

Adenovirus serotype 5 remains one of the most promising vectors for delivering genetic material to cancer cells for imaging or therapy, but optimization of these agents to selectively promote tumor cell infection is needed to further their clinical development. Peptide sequences that bind to specific cell surface receptors have been inserted into adenoviral capsid proteins to improve tumor targeting, often in the background of mutations designed to ablate normal ligand:receptor interactions and thereby reduce off target effects and toxicities in non-target tissues. Different tumor types also express highly variable complements of cell surface receptors, so a customized targeting strategy using a particular peptide in the context of specific adenoviral mutations may be needed to achieve optimal efficacy. To further investigate peptide targeting strategies in adenoviral vectors, we used a set of peptide motifs originally isolated using phage display technology that evince tumor specificity in vivo. To demonstrate their abilities as targeting motifs, we genetically incorporated these peptides into a surface loop of the fiber capsid protein to construct targeted adenovirus vectors. We then systematically evaluated the ability of these peptide targeted vectors to infect several tumor cell types, both in vitro and in vivo, in a variety of mutational backgrounds designed to reduce CAR and/or HSG-mediated binding. Results from this study support previous observations that peptide insertions in the HI loop of the fiber knob domain are generally ineffective when used in combination with HSG detargeting mutations. The evidence also suggests that this strategy can attenuate other fiber knob interactions, such as CAR-mediated binding, and reduce overall viral infectivity. The insertion of peptides into fiber proved more effective for targeting tumor cell types expressing low levels of CAR receptor, as this strategy can partially compensate for the very low infectivity of wild-type adenovirus in those cells. Nevertheless, the incorporation of relatively low affinity peptide ligands into the fiber knob, while effective in vitro, has only minimal targeting efficacy in vivo and highlights the importance of high affinity ligand:receptor interactions to achieve tumor targeting.


Assuntos
Adenoviridae/genética , Neoplasias/metabolismo , Peptídeos/genética , Receptores Virais/metabolismo , Adenoviridae/metabolismo , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Estudos de Viabilidade , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Mutação , Transplante de Neoplasias , Neoplasias/genética , Peptídeos/metabolismo , Receptores Virais/genética
2.
Cancer Gene Ther ; 19(7): 451-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22555510

RESUMO

Adenoviral technology has been thoroughly evaluated for delivering genetic material to tumor tissue and the surrounding microenvironment. Almost any gene can be cloned into an adenovirus (Ad) vector, which when combined with strong, constitutively active promoters permit up to a million-fold amplification of the transgene in a single adenoviral particle, thus facilitating their use in cancer therapy and imaging. However, widespread infection of the liver and other non-targeted tissues by Ad vectors is a substantial problem that often results in significant liver inflammation and hepatotoxicity at doses required to achieve efficient tumor transduction. miR-122 is a highly expressed liver-specific microRNA (miRNA) that provides a unique opportunity for downregulating adenoviral transgene expression in liver tissue. The binding of endogenous miRNAs to complementary miRNA-targeting elements (miRTs) incorporated into the 3' untranslated region of adenoviral transgenes interferes with message stability and/or protein translation, and miRT elements against miR-122 (miRT-122) can selectively reduce adenoviral transgene expression in the liver. Previous studies using miR-122-based regulation, with and without other types of transcriptional targeting, have yielded promising preliminary results. However, investigations to date evaluating miRT-122 elements for improving tumor specificity have used either non-tumor-bearing animals or direct intratumoral injection as the mode of delivery. In the present study, we confirmed the ability of miRT-122 sequences to selectively downregulate adenoviral luciferase expression in the liver in vitro and in vivo, and show that this strategy can improve tumor-specific transgene expression in a HT1080 human fibrosarcoma model. Rapid growth and the inefficient flow of blood through tumor neovasculature often results in profound hypoxia, which provides additional opportunities for targeting solid tumors and their microenvironment using vectors incorporating hypoxia-responsive promoters to drive transgene expression. We therefore used a combinatorial approach using miRT-122 elements with hypoxia-responsive transcriptional targeting to further improve the tumor-specific expression of an adenoviral reporter gene. Results from this investigation reveal that miRT122 elements alone decrease off-target liver expression and improve tumor specificity of adenoviral vectors. Furthermore, increased tumor specificity can be achieved by combining miRT-122 elements with hypoxia-responsive promoters.


Assuntos
Adenoviridae/genética , Fibrossarcoma/terapia , Inativação Gênica , MicroRNAs/genética , Regiões 3' não Traduzidas , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Anidrase Carbônica IX , Anidrases Carbônicas/genética , Anidrases Carbônicas/metabolismo , Hipóxia Celular , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Terapia Genética/métodos , Vetores Genéticos , Humanos , Fígado/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Transplante de Neoplasias , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transgenes , Transplante Heterólogo
3.
Cancer Gene Ther ; 18(4): 275-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21183946

RESUMO

Gene targeting to tumors using adenoviral (Ad) vectors holds great potential for cancer imaging and therapy, but the limited efficacy of current methods used to improve delivery to target tissues and reduce unwanted interactions remain substantial barriers to further development. Progress in characterizing the set of molecular interactions used by Ad vectors to infect particular tissues has aided the development of novel strategies for retargeting vectors to tumor cells. One method is chemical retargeting of adenovirus using bispecific antibodies (bsAbs) against both viral capsid proteins and tumor-specific cell surface molecules. This approach can be combined either with competitive inhibitors designed to reduce viral tropism in undesired tissues, or with traditional therapeutics to increase the expression of surface molecules for improved tumor targeting. Ablating liver cell-specific interactions through mutation of capsid proteins or chemical means are promising strategies for reducing adenovirus-induced liver toxicity. The nature of tumor neovasculature also influences Ad delivery, and the use of vascular disrupting agents (VDAs) such as combretastatin can help elucidate these contributions. In this investigation, we evaluate a variety of these methods for retargeting Ad vectors to tumor cells in vitro and in vivo, and assess the contributions of specific molecular and tissue interactions that affect Ad transgene delivery.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Neoplasias/terapia , Adenoviridae/metabolismo , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/metabolismo , Linhagem Celular Tumoral , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Neoplasias/genética , Neoplasias/patologia , Transdução Genética , Transgenes/genética
4.
Cancer Gene Ther ; 17(6): 375-86, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20139924

RESUMO

Using biochemical, imaging and histological methods, we employed transcriptional targeting to increase the specificity of tumor gene expression in vivo for intravenously administered recombinant adenovirus vectors. Surprisingly, the relative specificity of tumor expression in comparison with other tissues was increased for a constitutively expressing recombinant adenovirus, AdCMVLuc, by simply reducing the viral dose. Even at lower doses, however, the high frequency of viral infection and transgene expression in the liver using constitutive promoters still represents a substantial problem. To further augment tumor specificity, we constructed a series of adenoviruses expressing luciferase from several other promoters and tested their ability to selectively transcribe genes in tumor cells, both in vitro and in vivo. Constitutively active viral promoters (RSV, SRalpha) varied widely in their tumor selectivity, but hypoxia-responsive promoters (carbonic anhydrase 9, PAI-1, SOD2 and several chimeric constructs) showed the most tumor-selective expression. Our results show that tumor targeting to HT1080 fibrosarcomas was readily achieved using transcriptional targeting mechanisms. We attribute the relatively high level of gene transfer and expression in HT1080 tumors in vivo to increased viral access to the tumor, presumably due to discontinuities in tumor vasculature and augmented expression from stress-responsive promoters in the hypoxic and inflammatory tumor microenvironment.


Assuntos
Adenoviridae/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Transgenes/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Injeções Intravenosas , Luciferases/genética , Luciferases/metabolismo , Camundongos , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transplante Heterólogo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
5.
J Mol Endocrinol ; 27(3): 321-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11719284

RESUMO

The androgen receptor (AR) and closely related members of the steroid receptor family have proven difficult to obtain in native form in large quantities. In the case of the human AR (hAR), high-level expression in prokaryotic or non-mammalian cells leads to the synthesis of a high proportion of non-binding, insoluble, or degraded forms of the receptor protein. To circumvent these difficulties, we have constructed a recombinant adenovirus that directs the expression of hAR under the control of a potent, constitutive promoter. Infection of eukaryotic cells with this recombinant virus leads to the synthesis of large quantities of the intact AR. In contrast to expression methods designed to direct the full-length AR in bacteria, yeast, and insect cells, AR expressed in mammalian cells using this adenoviral vector accumulates at high levels, retains many properties of the native AR, and is not rapidly proteolyzed. This method will prove useful for large-scale preparations of hAR for use in functional and structural studies.


Assuntos
Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Adenoviridae/genética , Animais , Linhagem Celular , Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Expressão Gênica , Genes Reporter , Vetores Genéticos , Humanos , Hidrocortisona/metabolismo , Ligantes , Luciferases/genética , Nandrolona/análogos & derivados , Nandrolona/metabolismo , Progesterona/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidade
6.
J Steroid Biochem Mol Biol ; 78(4): 319-28, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11717002

RESUMO

The lacrimal gland secretes most of the water and many proteins present in tear fluid. The composition of the tear fluid is affected dramatically by androgens, an observation which has been linked to the fact that more than 90% of the patients with Sjögren syndrome are female. Although the presence of androgen receptors in the lacrimal gland has been established, the molecular biology of the protective effects of androgens remains largely unknown. Here, we report the use of primary cultures of the lacrimal gland which express endogenous proteins under androgen control, as a more homologous test system for tissue-specific transcription studies. Infection with recombinant adenoviral vectors was the most efficient method to introduce foreign gene constructs in these cultures. A thus introduced mouse mammary tumor virus promoter was inducible with androgens and this effect was independent of the sexual genotype of the infected cells. By use of two recombinant adenoviral vectors containing genomic fragments of the SC gene, which is androgen responsive in the lacrimal gland, we could demonstrate the functionality of the sc promoter as well as its androgen regulation in this culture system.


Assuntos
Adenoviridae/genética , Androgênios/fisiologia , Técnicas de Transferência de Genes , Aparelho Lacrimal/fisiologia , Animais , Células Cultivadas , Células Epiteliais/fisiologia , Feminino , Vetores Genéticos , Aparelho Lacrimal/citologia , Luciferases/genética , Masculino , Ratos , Ratos Wistar , beta-Galactosidase/genética
7.
J Virol ; 75(11): 5215-21, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333903

RESUMO

Although many recombinant adenovirus vectors (rAd) have been developed, especially by using group C adenoviruses, to transfer and express genes, such rAd do not readily infect B-cell lines due to the lack of the coxsackievirus-adenovirus receptor. Bispecific antibodies have been used in different cell systems to facilitate entry of rAd into otherwise nonpermissive cells. Bispecific antibody is synthesized by covalently linking two monoclonal antibodies with distinct specificities. It has been shown that lymphoproliferative tumors commonly express the cell surface protein CD70, while this receptor is normally expressed on only a small subset of highly activated B cells and T cells. We therefore investigated whether a bispecific antibody with specificities for the adenovirus fiber protein and CD70 can facilitate rAd entry and subsequent expression of rAd-encoded genes in CD70-positive B cells. We found high CD70 expression on Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines (LCLs), as well as some, but not all, Burkitt lymphoma (BL) lines. We show here that rAd encoding green fluorescent protein (Ad-GFP) infects EBV-transformed LCLs and a CD70-positive BL line 10- to 20-fold more efficiently in the presence of the CD70-fiber bispecific antibody. In contrast, the bispecific antibody does not enhance Ad-GFP infection in CD70-deficient BL cells. Using the CD70-fiber bispecific antibody, we increased the ability of rAd vectors encoding the EBV immediate-early proteins BZLF1 and BRLF1 to induce the lytic form of EBV infection in LCLs. These results indicate that the CD70-fiber bispecific antibody can enhance rAd infection of CD70-positive B cells and suggest the use of this vector to explore EBV-positive LCLs.


Assuntos
Adenoviridae/genética , Anticorpos Biespecíficos , Antígenos CD , Linfócitos B/imunologia , Proteínas do Capsídeo , Capsídeo/imunologia , Vetores Genéticos , Proteínas de Membrana/imunologia , Transfecção/métodos , Anticorpos Facilitadores , Antígenos Virais/imunologia , Linfócitos B/virologia , Ligante CD27 , Linhagem Celular Transformada , Genes Virais , Humanos , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética
8.
J Cell Biol ; 152(4): 777-84, 2001 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11266468

RESUMO

The plasminogen (Plg)/plasminogen activator (PA) system plays a key role in cancer progression, presumably via mediating extracellular matrix degradation and tumor cell migration. Consequently, urokinase-type PA (uPA)/plasmin antagonists are currently being developed for suppression of tumor growth and angiogenesis. Paradoxically, however, high levels of PA inhibitor 1 (PAI-1) are predictive of a poor prognosis for survival of patients with cancer. We demonstrated previously that PAI-1 promoted tumor angiogenesis, but by an unresolved mechanism. We anticipated that PAI-1 facilitated endothelial cell migration via its known interaction with vitronectin (VN) and integrins. However, using adenoviral gene transfer of PAI-1 mutants, we observed that PAI-1 promoted tumor angiogenesis, not by interacting with VN, but rather by inhibiting proteolytic activity, suggesting that excessive plasmin proteolysis prevents assembly of tumor vessels. Single deficiency of uPA, tissue-type PA (tPA), uPA receptor, or VN, as well as combined deficiencies of uPA and tPA did not impair tumor angiogenesis, whereas lack of Plg reduced it. Overall, these data indicate that plasmin proteolysis, even though essential, must be tightly controlled during tumor angiogenesis, probably to allow vessel stabilization and maturation. These data provide insights into the clinical paradox whereby PAI-1 promotes tumor progression and warrant against the uncontrolled use of uPA/plasmin antagonists as tumor angiogenesis inhibitors.


Assuntos
Inibidores da Angiogênese/farmacologia , Endopeptidases/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Vitronectina/metabolismo , Animais , Endotélio Vascular/efeitos dos fármacos , Fibrinolisina/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Mutantes , Neoplasias Musculares/irrigação sanguínea , Invasividade Neoplásica , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/etiologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Vitronectina/genética
9.
Hum Gene Ther ; 11(9): 1329-39, 2000 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-10890742

RESUMO

Local intracoronary delivery of recombinant adenoviruses expressing anti-migratory or anti-proliferative proteins including human constitutive endothelial nitric oxide synthase (NOS3), plasminogen activator inhibitor 1 (PAI-1), or herpesvirus thymidine kinase (TK) (combined with ganciclovir) was used to prevent neointimal formation in porcine coronary arteries. After balloon injury of the left anterior descending (LAD) coronary artery, animals received an intramural injection of adenovirus (1.5 X 10(9) PFU) carrying either the NOS3 cDNA (AdCMVNOS3, n = 12), the PAI-1 cDNA (AdCMVPAI-1, n = 12), the TK cDNA (AdMLPItk, n = 12), or no cDNA (AdpL+, n = 12). After 28 days, morphometric analysis was performed on coronary sections from all segments demonstrating injury. The internal elastic lamina (IEL) fracture length normalized to the IEL perimeter (initial injury) and the neointimal area normalized to the vessel area (response to injury) were used to generate linear regression lines and calculate an index of stenosis for the respective treatment groups. The response to injury was significantly smaller in AdCMVNOS3- and AdMLPItk-infected animals than in AdpL+-infected animals (slopes = 0.86 +/- 0.05 and 0.69 +/- 0.07 versus 1.11 +/- 0.06, p < 0.005 and p < 0.0001, respectively) but not in AdCMVPAI-1-infected animals (slope = 1.26 +/- 0.04, p = 0.04). No viral shedding was observed and there was no acute systemic toxicity after gene transfer. An increase in neutralizing antibody titers against Ad vectors was observed without any detectable response to the transgene products (NOS3, PAI-1). Local gene transfer of NOS3 and TK may hold promise as a safe and effective adjunctive treatment to reduce neointimal formation after percutaneous coronary intervention in humans.


Assuntos
Arteriopatias Oclusivas/terapia , Vasos Coronários/lesões , Terapia Genética , Óxido Nítrico Sintase/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Timidina Quinase/genética , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Adenovírus Humanos/isolamento & purificação , Angioplastia Coronária com Balão/efeitos adversos , Animais , Anticorpos Antivirais/análise , Arteriopatias Oclusivas/patologia , Vasos Coronários/patologia , Elastina/análise , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Herpesvirus Humano 1/enzimologia , Óxido Nítrico Sintase/imunologia , Óxido Nítrico Sintase Tipo III , Inibidor 1 de Ativador de Plasminogênio/imunologia , Suínos , Timidina Quinase/imunologia
10.
Gene Ther ; 6(2): 227-36, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10435107

RESUMO

Recombinant adenoviral vectors expressing u-PA, t-PA, PAI-1 and PAI-2 were employed to correlate the expression of components of the fibrinolytic system with the invasiveness of HT 1080 tumor cells. Migration through Transwell inserts in vitro in the presence of plasminogen was increased up to 22% by overexpression of u-PA, whereas t-PA had no effect. Gene transfer of PAI-1 or PAI-2 both reduced migration in a dose-dependent manner by up to 43% with PAI-1 and 29% with PAI-2. Two routes of gene transfer were used to alter metastasis of subcutaneously implanted HT 1080 cells expressing firefly luciferase in nude mice. Infection of cultured tumor cells with adenovirus expressing either PAI-1 or PAI-2 before implantation significantly reduced the incidence of lung metastasis by 60% compared with control virus. However, only PAI-2 reduced the incidence of lung and brain metastasis following liver gene transfer. Although PAI gene transfer by either route reduced primary tumor size, it had little effect on tumor vascularization or host survival. The migratory and metastatic phenotype of HT 1080 tumor cells is thus directly dependent on u-PA expression levels and can be altered by gene transfer of u-PA or plasminogen activator inhibitors.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Metástase Neoplásica/terapia , Inativadores de Plasminogênio/genética , Adenoviridae/genética , Análise de Variância , Animais , Movimento Celular , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Fígado/metabolismo , Camundongos , Camundongos Nus , Estatísticas não Paramétricas , Células Tumorais Cultivadas
11.
Arterioscler Thromb Vasc Biol ; 19(7): 1796-803, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10397700

RESUMO

Tissue-type plasminogen activator (t-PA) is acutely released by endothelial cells. Although its endothelial storage compartment is still not well defined, t-PA release is often accompanied by release of von Willebrand factor (vWf), a protein stored in Weibel-Palade bodies. We investigated, therefore, whether t-PA is stored in these secretory organelles. Under basal culture conditions, a minority of human umbilical vein endothelial cells (HUVEC) exhibited immunofluorescent staining for t-PA, which was observed only in Weibel-Palade bodies. To increase t-PA expression, HUVEC were infected with a t-PA recombinant adenovirus (AdCMVt-PA). Overexpressed t-PA was detected in Weibel-Palade bodies and acutely released together with endogenous vWf by thrombin or calcium ionophore stimulation. In contrast, plasminogen activator inhibitor type 1 and urokinase were not detected in Weibel-Palade bodies after adenovirus-mediated overexpression. Infection of HUVEC with proinsulin recombinant adenovirus resulted in the storage of insulin in Weibel-Palade bodies, indicating that these organelles can also store nonendothelial proteins that show regulated secretion. Infection of AtT-20 pituitary cells, a cell type with regulated secretion, with AdCMVt-PA resulted in the localization of t-PA in adrenocorticotropic hormone-containing granules, indicating that t-PA can be diverted to secretory granules independently of vWf. Coinfection of AtT-20 cells with AdCMVt-PA and proinsulin recombinant adenovirus resulted in the colocalization of t-PA and insulin in the same granules. Taken together, these results suggest that HUVEC have protein sorting mechanisms similar to those of other regulated secretory cells. Although the results did not exclude an alternative storage site for t-PA in HUVEC, they established that t-PA can be stored in Weibel-Palade bodies. This finding may explain the acute coordinate secretion of t-PA and vWf.


Assuntos
Grânulos Citoplasmáticos/química , Endotélio Vascular/química , Ativador de Plasminogênio Tecidual/análise , Células Cultivadas , Endotélio Vascular/ultraestrutura , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Insulina/análise , Ativador de Plasminogênio Tipo Uroquinase/análise
12.
Hum Gene Ther ; 10(7): 1105-15, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10340543

RESUMO

Recombinant adenoviral (Ad) vectors represent an efficient gene transfer system for targeting the cardiovascular system. Phenotypic modulation of coronary vascular cells in vivo is, however, critically dependent on the efficacy of local delivery devices. Four local drug delivery catheters were tested for intracoronary gene transfer efficiency: the Infiltrator (INF, n = 10), the Crescendo (CRE, n = 10), the Infusasleeve (SLE, n = 8), and the Remedy balloon (channel balloon [CHA], n = 8). After balloon injury of the LAD, Ad vector containing the firefly luciferase cDNA (AdCMVluc, 1.5 x 10(10) plaque-forming units) was administered at the site of injury. On day 4, tissue samples from different regions in the heart and from the liver were assayed for luciferase activity to evaluate local and systemic gene transfer. INF, CRE, and SLE catheters showed higher transduction levels of the target LAD segment than did the CHA catheter (median luciferase activity = 4.2 x 10(6), 11 x 10(6), and 1.3 x 10(6) light units [LU]/vessel versus 0.09 x 10(6) LU/vessel, respectively, p < 0.05). Luciferase activity was occasionally observed in nontarget tissues (right and left ventricular free wall, distal LAD, and liver) and was not significantly different between groups. The viral circulatory half-life was similar for the four groups (<1 min). Gene transfer efficiency was positively correlated with the degree of injury for the intralumenal catheters (CRE, SLE, and CHA) but was independent of the vessel wall injury for the intramural INF. Local drug delivery catheters enable efficient vascular gene transfer in balloon-injured coronary arteries, a prerequisite for further development of intracoronary gene therapy for restenosis.


Assuntos
Adenoviridae/genética , Cateterismo , Doença das Coronárias/terapia , Técnicas de Transferência de Genes , Administração Cutânea , Angioplastia com Balão , Animais , Vasos Coronários/anatomia & histologia , Vasos Coronários/lesões , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Terapia Genética , Vetores Genéticos/administração & dosagem , Luciferases/metabolismo , Músculo Liso , Suínos
13.
Circulation ; 98(9): 919-26, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9738648

RESUMO

BACKGROUND: Nitric oxide, synthesized from L-arginine by nitric oxide synthase (NOS), is a vasodilator and inhibits vascular smooth muscle cell (SMC) proliferation and migration. The effects of local NOS gene transfer on restenosis after experimental balloon angioplasty were investigated. METHODS AND RESULTS: Left anterior descending coronary artery angioplasty was performed in 25 pigs. Animals received an intramural injection of adenovirus (1.5 x 10(9) pfu) carrying either the NOS cDNA (AdCMVceNOS) or no cDNA (AdRR5) via the Infiltrator. Local gene transfer efficiency and bioactivity of recombinant protein were assessed after 4 days. Indices of restenosis were evaluated by computerized planimetry on coronary artery sections prepared 28 days after angioplasty. Adenoviral vectors permitted efficient gene delivery to medial SMCs and adventitial cells of coronary arteries. Vascular cGMP levels were depressed after angioplasty from 1.30+/-0.42 to 0.33+/-0.20 pmol/mg protein (P<0.05) but were restored after constitutive endothelial (ce) NOS gene transfer to 1.82+/-0.98 pmol/mg (P<0.05 versus injured group and P=NS versus control). The ratio of the neointimal area to the internal elastic lamina fracture length, maximal neointimal thickness, and percent stenosis were all reduced in AdCMVceNOS- versus AdRR5-transduced pigs (0.59+/-0.14 versus 0.80+/-0.19 mm, P=0.02; 0.75+/-0.21 versus 1.04+/-0.25 mm, P=0.019; and 53+/-15% versus 75+/-11%, P=0.006, respectively). Lumen area was significantly larger (0.70+/-0.35 mm2 in AdCMVceNOS versus 0.32+/-0.18 mm2 in AdRR5, P=0.007). CONCLUSIONS: Percutaneous adenovirus-mediated NOS gene transfer resulted in efficient local overexpression of functional NOS after angioplasty in coronary arteries. Restored NO production in injured coronary arteries significantly reduced luminal narrowing, most likely through a combined effect on neointima formation and on vessel remodeling after angioplasty.


Assuntos
Adenoviridae , Angioplastia Coronária com Balão/efeitos adversos , Vasos Coronários/lesões , Técnicas de Transferência de Genes , Óxido Nítrico Sintase/genética , Animais , Vasos Coronários/enzimologia , Vasos Coronários/fisiopatologia , GMP Cíclico/metabolismo , Expressão Gênica , Genes Reporter , Músculo Liso Vascular/enzimologia , Isquemia Miocárdica/terapia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III , Recidiva , Suínos , Transgenes/genética , Túnica Íntima/enzimologia , beta-Galactosidase/genética
14.
Nat Med ; 4(8): 923-8, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9701244

RESUMO

Acquisition of invasive/metastatic potential through protease expression is an essential event in tumor progression. High levels of components of the plasminogen activation system, including urokinase, but paradoxically also its inhibitor, plasminogen activator inhibitor 1 (PAI1), have been correlated with a poor prognosis for some cancers. We report here that deficient PAI1 expression in host mice prevented local invasion and tumor vascularization of transplanted malignant keratinocytes. When this PAI1 deficiency was circumvented by intravenous injection of a replication-defective adenoviral vector expressing human PAI1, invasion and associated angiogenesis were restored. This experimental evidence demonstrates that host-produced PAI is essential for cancer cell invasion and angiogenesis.


Assuntos
Invasividade Neoplásica/prevenção & controle , Neovascularização Patológica/prevenção & controle , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Inibidor 1 de Ativador de Plasminogênio/deficiência , Neoplasias Cutâneas/patologia , Adenoviridae , Animais , Transformação Celular Neoplásica , Células Cultivadas , Progressão da Doença , Feminino , Vetores Genéticos , Genótipo , Humanos , Queratinócitos/patologia , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica , Inibidor 1 de Ativador de Plasminogênio/genética , Neoplasias Cutâneas/irrigação sanguínea , Transfecção
15.
Circulation ; 96(9): 3180-91, 1997 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-9386191

RESUMO

BACKGROUND: Plasminogen-deficient mice display impaired vascular wound healing and reduced arterial neointima formation after arterial injury, suggesting that inhibition of plasmin generation might reduce arterial neointima formation. Therefore, we studied the consequences of plasminogen activator inhibitor-1 (PAI-1) gene inactivation and adenoviral PAI-1 gene transfer on arterial neointima formation. METHODS AND RESULTS: Neointima formation was evaluated in PAI-1-deficient (PAI-1(-/-)) mice with perivascular electric or transluminal mechanical injury. PAI-1 deficiency improved vascular wound healing in both models: the cross-sectional neointimal area was 0.001+/-0.001 mm2 in PAI-1(+/+) and 0.016+/-0.008 mm2 in PAI-1(-/-) mice within 1 week after electric injury (P<.02) and 0.055+/-0.008 mm2 in PAI-1(+/+) and 0.126+/-0.006 mm2 in PAI-1(-/-) mice within 3 weeks after mechanical injury (P<.001). Proliferation of smooth muscle cells was not affected by PAI-1 deficiency. Topographic analysis of arterial wound healing after electric injury revealed that PAI-1(-/-) smooth muscle cells, originating from the uninjured borders, more rapidly migrated into the necrotic center of the arterial wound than wild-type smooth muscle cells. On the basis of immunostaining, PAI-1 expression was markedly upregulated during vascular wound healing. There were no genotypic differences in reendothelialization of the vascular wound. When PAI-1(-/-) mice were intravenously injected with replication-defective adenovirus expressing human PAI-1 (AdCMVPAI-1), plasma PAI-1 antigen levels increased in a dose-dependent fashion up to to 61+/-8 microg/mL with 2x10(9) plaque-forming units (pfu) virus. Luminal stenosis was 35+/-13% in control AdRR5-treated (2x10(9) pfu) and suppressed to 5+/-5% in AdCMVPAI-1-treated (6x10(8) pfu) PAI-1(-/-) mice (P<.002). CONCLUSIONS: By affecting cellular migration, PAI-1 plays an inhibitory role in vascular wound healing and arterial neointima formation after injury, and adenoviral PAI-1 gene transfer reduces arterial neointima formation in mice.


Assuntos
Marcação de Genes , Técnicas de Transferência de Genes , Músculo Liso Vascular/patologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Cicatrização , Adenoviridae/genética , Animais , Divisão Celular , Movimento Celular , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidor 1 de Ativador de Plasminogênio/análise , Inibidor 1 de Ativador de Plasminogênio/genética , Trombose/prevenção & controle , Doenças Vasculares/prevenção & controle
16.
Blood ; 90(7): 2738-46, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9326241

RESUMO

Uveal melanoma is the most common intraocular malignancy in adults and results in the death of 50% of the patients. Plasminogen activators (PA) are believed to facilitate tumor metastasis by promoting invasion of tissue barriers. The present study explored the possibility of preventing the metastasis of intraocular melanomas by disrupting plasminogen activator function through gene transfer. A replication-deficient adenovirus vector was used for the in vivo transfer of plasminogen activator inhibitor type 1 (PAI-1) cDNA. Intraocular injection of an adenovirus vector (AdCMV-PAI-1) expressing plasminogen activator inhibitor-1 resulted in: (1) the transduction of more than 95% of human and murine uveal melanoma cells in the eyes of nude mice; (2) a 50% reduction in the number of animals developing liver metastases; and (3) a 78% reduction in the metastatic tumor burden in animals that eventually developed metastases. In other experiments intravenous injections of AdCMV-PAI-1 resulted in transduction of normal liver cells and culminated in a sharp reduction in the incidence of metastases and a significant prolongation of host survival. The results support the feasibility of disruption of PA function through gene transfer as a therapeutic strategy for preventing metastases and prolonging host survival.


Assuntos
Adenovírus Humanos/genética , Vírus Defeituosos/genética , Terapia Genética , Vetores Genéticos/genética , Melanoma Experimental/terapia , Melanoma/terapia , Metástase Neoplásica/prevenção & controle , Inibidor 1 de Ativador de Plasminogênio/uso terapêutico , Neoplasias Uveais/terapia , Adulto , Animais , Estudos de Avaliação como Assunto , Humanos , Neoplasias Hepáticas Experimentais/prevenção & controle , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Melanoma/patologia , Melanoma Experimental/patologia , Melanoma Experimental/secundário , Camundongos , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Transplante de Neoplasias , Inibidor 1 de Ativador de Plasminogênio/administração & dosagem , Inibidor 1 de Ativador de Plasminogênio/genética , Ativadores de Plasminogênio/antagonistas & inibidores , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Transfecção , Transplante Heterólogo , Neoplasias Uveais/patologia
17.
Blood ; 90(4): 1527-34, 1997 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9269770

RESUMO

Impaired fibrinolysis, resulting from increased plasminogen activator inhibitor-1 (PAI-1) or reduced tissue-type plasminogen activator (t-PA) plasma levels, may predispose the individual to subacute thrombosis in sepsis and inflammation. The objective of these studies was to show that adenovirus-mediated gene transfer could increase systemic plasma t-PA levels and thrombolytic capacity in animal model systems. Recombinant adenovirus vectors were constructed that express either human wild type or PAI-1-resistant t-PA from the cytomegalovirus (CMV) promoter. Both t-PA-deficient (t-PA(-/-)) and PAI-1-overexpressing transgenic mice were infected by intravenous injection of these viruses. Intravenous injection of recombinant adenovirus resulted in liver gene transfer, t-PA synthesis, and secretion into the plasma. Virus dose, human t-PA antigen, and activity concentrations in plasma and extent of lysis of a 125I-fibrin-labeled pulmonary embolism were all closely correlated. Plasma t-PA antigen and activity were increased approximately 1,000-fold above normal levels. Clot lysis was significantly increased in mice injected with a t-PA-expressing virus, but not in mice injected with saline or an irrelevant adenovirus. Comparable levels of enzyme activity and clot lysis were obtained with wild type and inhibitor-resistant t-PA viruses. Adenovirus-mediated t-PA gene transfer was found to augment clot lysis as early as 4 hours after infection, but expression levels subsided within 7 days. Adenovirus-mediated transfer of a t-PA gene can effectively increase plasma fibrinolytic activity and either restore (in t-PA-deficient mice) or augment (in PAI-1-overexpressing mice) the thrombolytic capacity in simple animal models of defective fibrinolysis.


Assuntos
Fibrinólise , Terapia Genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Trombose/terapia , Ativador de Plasminogênio Tecidual/metabolismo , Adenoviridae , Alelos , Animais , Células Cultivadas , Fibrinólise/efeitos dos fármacos , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Camundongos Transgênicos , Inibidor 1 de Ativador de Plasminogênio/genética , Proteínas Recombinantes/farmacologia , Ativador de Plasminogênio Tecidual/deficiência , Ativador de Plasminogênio Tecidual/genética
19.
J Clin Invest ; 98(10): 2364-72, 1996 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-8941655

RESUMO

Endothelin-1 (ET-1) has been implicated in the regulation of vascular tone in various pathological conditions. To examine the effect of in vivo overexpression of the peptide in rats, we prepared recombinant adenovirus stocks encoding the human preproET-1 cDNA (Ad.ET-1) or Escherichia coli lacZ (Ad.betaGal), each driven by cytomegalovirus early promoter. Ad.ET-1 or Ad.betaGal was injected into the caudal vein of rats and the animals were studied under anesthesia 96 h later. Hepatic overexpression of the virus-derived human ET-1 mRNA was accompanied by a 13-fold elevation of liver ET-1 content in the Ad.ET-1 group. Circulating plasma ET-1 levels in the Ad.ET-1 group were sixfold higher than those in the Ad.betaGal group. Mean arterial blood pressure was increased by 28 mmHg in the Ad.ET-1 group as compared with the Ad.betaGal group. In the Ad.ET-1 group, intravenous infusion of the ET(A) receptor antagonist FR 139317 reduced the blood pressure to levels seen in the Ad.betaGal group, whereas the same antagonist did not significantly alter the blood pressure in the Ad.betaGal group. Intravenous infusion of the ET(B) receptor antagonist BQ-788 caused a small but significant increase in blood pressure in both groups. These findings demonstrate that endogenous overexpression of preproET-1, accompanied by an elevation of plasma ET-1 concentrations to the levels seen in pathophysiological states, can cause systemic hypertension through the activation of the ETA receptor.


Assuntos
Endotelina-1/genética , Endotelina-1/fisiologia , Regulação da Expressão Gênica , Hipertensão/genética , Hipertensão/metabolismo , Adenovírus Humanos/genética , Animais , Azepinas/farmacologia , Pressão Sanguínea/genética , Northern Blotting , Citomegalovirus/genética , DNA Complementar/genética , Endotelina-1/antagonistas & inibidores , Endotelina-1/sangue , Escherichia coli/genética , Vetores Genéticos , Humanos , Indóis/farmacologia , Rim/fisiologia , Óperon Lac , Fígado/metabolismo , Fígado/fisiologia , Masculino , Oligopeptídeos/farmacologia , Piperidinas/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Recombinação Genética
20.
J Clin Invest ; 98(2): 317-24, 1996 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8755640

RESUMO

Nitric oxide (NO), a vasodilator involved in the regulation of pulmonary vascular tone, is synthesized by a family of enzymes, nitric oxide synthases (NOS). To investigate whether adenoviral-mediated overexpression of constitutive endothelial NOS (ceNOS) would attenuate hypoxic pulmonary vasoconstriction, we aerosolized 3 X 10(9) plaque forming units of a recombinant adenovirus containing the ceNOS gene (AdCMVceNOS) into rat lungs. Four days after infection, transgene expression was confirmed using immunoblot techniques. Diffuse ceNOS immunostaining was detected in alveoli and medium-sized and small pulmonary vessels of AdCMVceNOS-transduced lungs. AdCMVceNOS-transduction was associated with an 86% increase in [3H]arginine to [3H]citrulline conversion and a rise in pulmonary cGMP levels from 7 +/- 1 to 59 +/- 9 pmol/mg protein in lungs from AdCMVceNOS versus control rats, (P < 0.05). During acute hypoxia (FIO2 = 0.10) for 25 min, mean pulmonary artery pressure (PAP) increased significantly from 17 +/- 1 to 27 +/- 1 mmHg in rats aerosolized with saline (n = 4) and from 18 +/- 1 to 28 +/- 1 mmHg in rats given an adenoviral vector expressing a nuclear-targeted beta-galactosidase gene (AdCMV beta gal, n = 8). In contrast, in AdCMVceNOS-transduced rats (n = 8) the hypoxia-induced increase in PAP was significantly attenuated (18 +/- 1 to 23 +/- 2 mmHg). Systemic blood pressure was not affected by aerosol gene transfer. Thus, adenoviral-mediated ceNOS gene transfer to rat lungs increases ceNOS expression and activity, and reduces acute hypoxic pulmonary vasoconstriction. Aerosolized recombinant adenovirus overexpressing vasodilatory proteins can act as a selective pulmonary vasodilator and may hold promise as a future therapeutic strategy for pulmonary hypertension.


Assuntos
Endotélio Vascular/enzimologia , Técnicas de Transferência de Genes , Pulmão/fisiologia , Óxido Nítrico Sintase/biossíntese , Circulação Pulmonar/fisiologia , Vasoconstrição , Adenoviridae , Análise de Variância , Animais , Sequência de Bases , Linhagem Celular , Citomegalovirus , Primers do DNA , Vetores Genéticos , Humanos , Hipóxia , Imuno-Histoquímica , Cinética , Pulmão/irrigação sanguínea , Dados de Sequência Molecular , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/genética , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Mapeamento por Restrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...