Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35511740

RESUMEN

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Asunto(s)
Inmunoconjugados , Proteínas ADAM , Línea Celular Tumoral , Xenoinjertos , Humanos , Inmunoconjugados/química , Proteínas de la Membrana/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Ther ; 19(11): 1999-2011, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21712814

RESUMEN

In mice, liver-restricted expression of lysosomal enzymes from adeno-associated viral serotype 8 (AAV8) vectors results in reduced antibodies to the expressed proteins. To ask whether this result might translate to patients, nonhuman primates (NHPs) were injected systemically with AAV8 encoding α-galactosidase A (α-gal). As in mice, sustained expression in monkeys attenuated antibody responses to α-gal. However, this effect was not robust, and sustained α-gal levels were 1-2 logs lower than those achieved in male mice at the same vector dose. Because our mouse studies had shown that antibody levels were directly related to expression levels, several strategies were evaluated to increase expression in monkeys. Unlike mice, expression in monkeys did not respond to androgens. Local delivery to the liver, immune suppression, a self-complementary vector and pharmacologic approaches similarly failed to increase expression. While equivalent vector copies reached mouse and primate liver and there were no apparent differences in vector form, methylation or deamination, transgene expression was limited at the mRNA level in monkeys. These results suggest that compared to mice, transcription from an AAV8 vector in monkeys can be significantly reduced. They also suggest some current limits on achieving clinically useful antibody reduction and therapeutic benefit for lysosomal storage diseases using a systemic AAV8-based approach.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Tolerancia Inmunológica , Inmunidad Humoral , Hígado/metabolismo , alfa-Galactosidasa/genética , Andrógenos/farmacología , Animales , Metilación de ADN , Desaminación , Dependovirus/inmunología , Dosificación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/inmunología , Humanos , Inyecciones , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Transcripción Genética , alfa-Galactosidasa/inmunología , alfa-Galactosidasa/metabolismo
3.
J Gene Med ; 11(5): 390-400, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19291676

RESUMEN

BACKGROUND: Diabetes mellitus is a common comorbidity of atherosclerosis. Hypoxia-inducible factor-1 (HIF-1) is the master regulator of the angiogenic response to hypoxia. METHODS: We studied the effects of adenoviral vectors expressing a constitutively active HIF-1 alpha hybrid (Ad2/HIF-1 alpha/VP16) or vascular endothelial growth factor (Ad2/VEGF) on collateral development and vascular leakiness in a diabetic rat model of hindlimb ischemia. RESULTS: After the removal of the right femoral artery, the mRNA levels of VEGF, angiopoietin-1 and angiopietin-4 in the calf muscles, as measured by Taqman reverse transcriptase-polymerase chain reaction, were transiently elevated in Zucker lean (ZL) but not Zucker diabetic fatty (ZDF) rats. The angiographic score, as determined by post-mortem angiography, was significantly lower in ZDF animals 35 days after surgery compared to their ZL counterparts. In separate animals, intramuscular injection of Ad2/HIF-1a/VP16 and Ad/2VEGF into the thigh muscles significantly increased the angiographic score and capillary density 21 and 35 days after the injection compared to Ad2/CMVEV (a vector expressing no transgene) or vehicle. After the injection of Ad2/CMVEV or vehicle, the Evans-blue dye content in the thigh muscles was significantly higher in ZDF rats than their ZL counterparts. Ad2/HIF-1 alpha/VP16 but not Ad2/VEGF reduced tissue Evans blue dye content. CONCLUSIONS: The endogenous angiogenic response to ischemia was impaired in ZDF rats, possibly due to down-regulation of angiogenic factors. Ad2/HIF-1 alpha/VP16 enhanced collateral development and reduced vascular leakage in the ischemic hindlimb of ZDF rats indicating that hybrid HIF-1 alpha angiogenic therapy may be efficacious for peripheral vascular disease with a diabetic comorbidity.


Asunto(s)
Vasos Sanguíneos/patología , Circulación Colateral/fisiología , Diabetes Mellitus Experimental/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Recombinantes/metabolismo , Adenoviridae/genética , Animales , Peso Corporal , ADN/genética , Arteria Femoral/cirugía , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hemoglobina Glucada/metabolismo , Miembro Posterior/irrigación sanguínea , Inmunohistoquímica , Inyecciones Intramusculares , Isquemia , Neovascularización Fisiológica/genética , Ratas , Ratas Zucker , Transgenes
4.
Biochem Biophys Res Commun ; 364(3): 567-72, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17963722

RESUMEN

In response to cellular hypoxia, cardiomyocytes adapt to consume less oxygen by shifting ATP production from mitochondrial fatty acid beta-oxidation to glycolysis. The transcriptional activation of glucose transporters and glycolytic enzymes by hypoxia is mediated by hypoxia-inducible factor 1 (HIF-1). In this study, we examined whether HIF-1 was involved in the suppression of mitochondrial fatty acid beta-oxidation in hypoxic cardiomyocytes. We showed that either hypoxia or adenovirus-mediated expression of a constitutively stable hybrid form (HIF-1alpha/VP16) suppressed mitochondrial fatty acid metabolism, as indicated by an accumulation of intracellular neutral lipid. Both treatments also reduced the mRNA levels of muscle carnitine palmitoyltransferase I which catalyzes the rate-limiting step in the mitochondrial import of fatty acids for beta-oxidation. Furthermore, adenovirus-mediated expression of HIF-1alpha/VP16 in cardiomyocytes under normoxic conditions also mimicked the reduction in the DNA binding activity of peroxisome proliferator-activated receptor alpha (PPARalpha)/retinoid X receptor (RXR), in the presence or absence of a PPARalpha ligand. These results suggest that HIF-1 may be involved in hypoxia-induced suppression of fatty acid metabolism in cardiomyocytes by reducing the DNA binding activity of PPARalpha/RXR.


Asunto(s)
ADN/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Metabolismo de los Lípidos/fisiología , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , PPAR alfa/metabolismo , Receptores X Retinoide/metabolismo , Animales , Animales Recién Nacidos , Hipoxia de la Célula/fisiología , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Ratas
5.
Mol Pharmacol ; 69(6): 1953-62, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16507742

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a primary regulator of the physiological response to hypoxia. A recombinant adenovirus expressing a constitutively active hybrid form of the HIF-1alpha subunit (Ad2/HIF-1alpha/VP16) is being evaluated as a gene therapy for the treatment of peripheral vascular disease. Ad2/HIF-1alpha/VP16 up-regulates known HIF-1-responsive genes, including those involved in angiogenesis. Expression profile analysis revealed that the brain natriuretic peptide (BNP) gene was significantly up-regulated in response to HIF-1alpha/VP16 in human fetal cardiac cells. Real-time reverse transcription-polymerase chain reaction analyses confirmed transcriptional activation of the BNP gene by HIF-1alpha/VP16 in human but not rat cardiac cells. Because hypoxia itself did not increase human BNP gene expression in these analyses, the mechanism of the HIF-1alpha/VP16 effect was determined. Analyses of promoter deletion mutants suggested that the cis-acting sequence in the human BNP promoter mediating activation by HIF-1alpha/VP16 was a putative HIF-1 responsive element (HRE) located at -466. An SV40 basal promoter-luciferase plasmid containing a minimal BNP HRE was up-regulated by HIF-1alpha/VP16, whereas a similar construct carrying a mutation within the HIF-1 binding site was not. Mutation of an E-box motif within the BNP HRE reduced HIF-1alpha/VP16-mediated transcriptional activation by 50%. Gel-shift analyses showed that both the native HIF-1alpha and HIF-1alpha/VP16 are able to bind to a probe containing the HIF-1 binding site. These experiments demonstrate the existence of a functional HRE in the BNP promoter and further define the scope and mechanism of action of Ad2/HIF-1alpha/VP16.


Asunto(s)
Terapia Genética , Péptido Natriurético Encefálico/genética , Enfermedades Vasculares Periféricas/terapia , Proteínas Recombinantes de Fusión/genética , Activación Transcripcional , Secuencia de Bases , Células Cultivadas , Análisis Mutacional de ADN , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia
6.
Circulation ; 111(13): 1679-84, 2005 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-15781730

RESUMEN

BACKGROUND: Hemodialysis vascular access dysfunction is the single most important cause of morbidity in kidney hemodialysis patients. Failure of an arteriovenous polytetrafluoroethylene (PTFE) graft, the most common form of hemodialysis access, is primarily due to intimal hyperplasia and thrombosis at the venous anastomosis. METHODS AND RESULTS: This study was aimed at evaluating the efficacy and safety of an adenoviral vector (Ad2/betaARKct) encoding the carboxyl terminus of beta-adrenergic receptor kinase (betaARKct) in a pig model of arteriovenous PTFE graft failure. Transduction of the external jugular vein with Ad2/betaARKct (5E9, 5E10, or 5E11 particles per vein) did not result in systemic toxicity, as measured by clinical and pathological assessments. Ad2/betaARKct significantly reduced neointimal hyperplasia in the graft/vein anastomosis. It also improved the graft patency rate and angiographic score, as measured histologically and angiographically, compared with vehicle or empty viral vector controls. CONCLUSIONS: Our results suggest that local administration of adenoviral vectors encoding betaARKct into the jugular vein represents a viable strategy to treat AV graft hemodialysis vascular access failure.


Asunto(s)
Oclusión de Injerto Vascular/terapia , Hiperplasia/terapia , Politetrafluoroetileno/uso terapéutico , Receptores Adrenérgicos beta/administración & dosificación , Túnica Íntima/patología , Adenoviridae/genética , Animales , Derivación Arteriovenosa Quirúrgica/efectos adversos , Catéteres de Permanencia/efectos adversos , Falla de Equipo , Regulación de la Expresión Génica , Diálisis Renal , Porcinos , Transducción Genética
7.
Am J Physiol Cell Physiol ; 288(2): C314-20, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15496478

RESUMEN

Preconditioning in cultured cardiomyocytes elevates the expression of several protective genes including Glut-4 and heat shock protein (HSP)70. Hypoxia-inducible factor-1 (HIF-1) is known to mediate the transcriptional activation of hypoxia-responsive genes. In this study, we examined the effect of adenovirus-mediated expression of constitutively stable hybrid forms of HIF-1alpha on cardiomyocyte viability and gene expression. Cultured neonatal rat cardiomyocytes were subjected to simulated ischemia-reperfusion with or without preinfection with recombinant adenoviral vectors [Ad2/HIF-1alpha/herpes simplex virus protein VP16 and Ad2/HIF-1alpha/nuclear factor-kappaB (NF-kappaB)]. Cellular viability and mRNA levels of several cardioprotective genes were measured. We demonstrated that infection with Ad2/HIF-1alpha/VP16 and Ad2/HIF-1alpha/NF-kappaB mimicked the upregulation of the mRNA levels of vascular endothelial growth factor (VEGF), Glut-1, Glut-4, HSP70, and inducible NO synthase (iNOS) and the protection of cultured neonatal rat cardiomyocytes by late-phase preconditioning against simulated ischemia-reperfusion. The same dose of a control viral vector expressing no transgene had no effect. Preconditioning also elevated HIF-1alpha protein levels. These results suggest that adenovirus-mediated expression of HIF-1alpha/VP16 or HIF-1alpha/NF-kappaB, a constitutively stable hybrid transcriptional factor, protected cultured neonatal cardiomyocytes against simulated ischemia-reperfusion injury by inducing multiple protective genes.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica/fisiología , Precondicionamiento Isquémico Miocárdico , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/biosíntesis , Daño por Reperfusión/fisiopatología , Factores de Transcripción/biosíntesis , Adenoviridae , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Proteína Vmw65 de Virus del Herpes Simple/biosíntesis , Proteína Vmw65 de Virus del Herpes Simple/genética , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Miocitos Cardíacos/efectos de los fármacos , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas Nucleares/genética , ARN Mensajero/análisis , Ratas , Factores de Transcripción/genética
8.
J Gene Med ; 6(10): 1061-8, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15386742

RESUMEN

BACKGROUND: Previous studies have shown that incubation of balloon-injured rat carotid arteries with adenoviral vectors encoding the carboxyl terminus of the beta-adrenergic receptor kinase (Ad2/betaARKct) for 30 min reduces neointima formation. However, it is unclear whether this beneficial effect of betaARKct could be achieved using a catheter-based vector delivery system and whether the observed inhibition of neointima formation translated into a reduction of vessel stenosis. METHODS: In this study, Ad2/betaARKct was infused into the balloon-injured site of rabbit iliac arteries using a porous infusion catheter over 2 min. Twenty-eight days after gene transfer, angiographic and histological assessments were performed. RESULTS: Angiographic and histological assessments indicate significant (p < 0.05) inhibition of iliac artery neointima formation and lumen stenosis by Ad2/betaARKct. Our studies demonstrate that an inhibitory effect of Ad2/betaARKct on neointima formation is achievable using a catheter-based vector delivery system and that the inhibition of neointima formation translates into a gain in the vessel minimal luminal diameter. The extent of inhibition (35%) was comparable to that observed with adenoviral-mediated expression of thymidine kinase plus ganciclovir treatment, a cytotoxic gene therapy approach for restenosis. CONCLUSIONS: These results suggest that adenoviral-mediated gene transfer of betaARKct is a clinically viable cytostatic gene therapy strategy for the treatment of restenosis.


Asunto(s)
Adenoviridae/genética , Constricción Patológica/terapia , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Hiperplasia/terapia , Arteria Ilíaca/patología , Túnica Íntima/patología , Angiografía , Animales , Arterias Carótidas/patología , Constricción Patológica/prevención & control , Cartilla de ADN/química , Hiperplasia/patología , Estructura Terciaria de Proteína , Conejos , Factores de Tiempo , Quinasas de Receptores Adrenérgicos beta , beta-Galactosidasa/metabolismo
9.
Am J Physiol Renal Physiol ; 287(4): F649-57, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15198927

RESUMEN

The hereditary von Hippel-Lindau (VHL) syndrome predisposes sufferers to highly vascularized tumors such as renal clear cell carcinoma (RCC) and central nervous system hemangioblastoma. In RCC4 and RCC786-0 VHL- cells with VHL mutations, the protein of hypoxia-inducible factor-1alpha (HIF-1alpha) is constitutively stabilized and the mRNA levels of HIF target genes, including vascular endothelial growth factor (VEGF), are elevated. However, the expression of angiopoietins in these cells and their involvement in angiogenesis are not well known. In this study, we compared the mRNA levels of angiopoietins in human kidney proximal tubule epithelial (RPTE) and RCC4 and RCC786-0 VHL- cells. In RPTE cells, angiopoietin-4 (Ang-4) expression was selectively induced by hypoxia or by expression of a hybrid form of HIF-1alpha. Under normoxic conditions, the mRNA levels of Ang-4 were higher in RCC4 and RCC786-0 VHL- than RPTE cells. Angiopoietin-1 expression was detectable in RCC4 and RCC786-0 VHL- cells but not RPTE cells. In RCC786-0 VHL+ cells, which were stably transfected with a wild-type copy of VHL, the mRNA levels of VEGF and Ang-4 were suppressed and the hypoxic response was restored. We also demonstrated that stimulation of endothelial tube formation by conditioned medium harvested from RCC4 cells was inhibited by a soluble Tie-2 receptor. These results suggest that the angiopoietin/Tie-2 system may participate in the angiogenic response to hypoxia in renal tissues and in tumor angiogenesis in renal carcinoma.


Asunto(s)
Adenocarcinoma de Células Claras , Angiopoyetina 1/genética , Angiopoyetina 2/genética , Hipoxia/fisiopatología , Neoplasias Renales , Neovascularización Patológica/fisiopatología , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Línea Celular Tumoral , Células Epiteliales/citología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Túbulos Renales Proximales/citología , ARN Mensajero/metabolismo , Receptor TIE-2/fisiología , Factores de Transcripción/genética , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Australas Radiol ; 48(1): 93-5, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15027932

RESUMEN

In August 2002, we performed MRI scans on a female juvenile Bengal tiger. We present the clinical course, imaging and autopsy findings, and some comparative anatomy of the tiger brain and skull. Magnetic resonance images of a tiger have not previously been published.


Asunto(s)
Animales de Zoológico , Encéfalo/patología , Carnívoros , Síndrome Hipereosinofílico/veterinaria , Hipoxia Encefálica/veterinaria , Imagen por Resonancia Magnética/veterinaria , Animales , Femenino , Síndrome Hipereosinofílico/diagnóstico por imagen , Hipoxia Encefálica/diagnóstico por imagen , Radiografía
11.
Mol Ther ; 9(2): 231-40, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14759807

RESUMEN

The successful application of gene therapy for the treatment of genetic diseases such as Fabry is reliant on the development of vectors that are safe and that facilitate sustained expression of therapeutic levels of the transgene product. Here, we report that intravenous administration of a recombinant AAV2 vector encoding human alpha-galactosidase A under the transcriptional control of a liver-restricted enhancer/promoter (AAV2/DC190-alphagal) generated significantly higher levels of expression in BALB/c and Fabry mice than could be realized using the ubiquitous CMV promoter (AAV2/CMVHI-alphagal). Moreover, AAV2/DC190-alphagal-mediated hepatic expression of alpha-galactosidase A was sustained for 12 months in BALB/c mice and was associated with a significantly reduced immune response to the expressed enzyme. Subsequent challenge of the AAV2/DC190-alphagal-treated animals with recombinant human alpha-galactosidase A at 6 months failed to elicit the production of anti-alpha-galactosidase A antibodies, suggesting the induction of immune tolerance in these animals. The levels of expression attained with AAV2/DC190-alphagal in the Fabry mice were sufficient to reduce the abnormal accumulation of globotriaosylceramide in the liver, spleen, and heart to basal levels and in the kidney by approximately 40% at 8 weeks. Together, these results demonstrate that AAV2-mediated gene transfer that limits the expression of alpha-galactosidase A to the liver may be a viable strategy for treating Fabry disease.


Asunto(s)
Dependovirus/genética , Enfermedad de Fabry/terapia , Terapia Genética , Tolerancia Inmunológica , Hígado/metabolismo , Regiones Promotoras Genéticas/genética , alfa-Galactosidasa/uso terapéutico , Animales , ADN Recombinante/genética , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Enfermedad de Fabry/genética , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
12.
Circ Res ; 93(7): 664-73, 2003 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-12958144

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) mediates transcriptional activation of vascular endothelial growth factor (VEGF) and other hypoxia-responsive genes. Transgenic expression of a constitutively stable HIF-1alpha mutant increases the number of vascular vessels without vascular leakage, tissue edema, or inflammation. This study aimed to investigate the molecular basis by which HIF-1 mediates the angiogenic response to hypoxia. In primary human endothelial cells, hypoxia, desferrioxamine, or infection with Ad2/HIF-1alpha/VP16, an adenoviral vector encoding a constitutively stable hybrid form of HIF-1alpha, increased the mRNA and protein levels of VEGF, angiopoietin-2 (Ang-2), and angiopoietin-4 (Ang-4). Infection with Ad2/CMVEV (a control vector expressing no transgene) had no effect. Angiopoietin-1 (Ang-1) expression was not detected in human endothelial cells. Ang-4 was also induced by hypoxia or Ad2/HIF-1alpha/VP16 in human cardiac cells, whereas Ang-1 expression remained unchanged. Recombinant Ang-4 protein protected endothelial cells against serum starvation-induced apoptosis and increased cultured endothelial cell migration and tube formation. Ad2/HIF-1alpha/VP16 stimulated endothelial cell proliferation and tube formation. Hypoxia- or Ad2/HIF-1alpha/VP16-induced tube formation was significantly reduced by a Tie-2 inhibitor. These results suggest that HIF-1 mediates the angiogenic response to hypoxia by upregulating the expression of multiple angiogenic factors. Ang-4 can function similarly as Ang-1 and substitute for Ang-1 to participate in hypoxia-induced angiogenesis. Activation of the angiopoietin/Tie-2 system may play a role in the ability of HIF-1 to induce hypervascularity without excessive permeability.


Asunto(s)
Inductores de la Angiogénesis/genética , Angiopoyetinas , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/metabolismo , Proteínas Nucleares/fisiología , Factores de Transcripción , Inductores de la Angiogénesis/metabolismo , Angiopoyetina 2 , Apoptosis/efectos de los fármacos , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Caspasa 3 , Caspasas/metabolismo , División Celular/efectos de los fármacos , Hipoxia de la Célula , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/genética , Deferoxamina/farmacología , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/genética , Linfocinas/metabolismo , Proteínas Nucleares/genética , Factor de Crecimiento Placentario , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor TIE-2 , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
13.
Hum Gene Ther ; 14(10): 947-57, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12869213

RESUMEN

The clinical use of doxorubicin, one of the most effective antitumor drugs, is limited by its cardiotoxicity, which results in irreversible cardiomyopathy and congestive heart failure. This study aimed to evaluate a gene therapy approach using adenovirus-mediated expression of p35, a baculoviral antiapoptotic gene, for alleviating doxorubicin-induced cardiomyopathy. In cultured neonatal rat cardiomyocytes, transduction with a recombinant adenoviral vector expressing p35 (Ad2/CMVp35) but not a control adenoviral vector expressing no transgene (Ad2/CMVEV) significantly inhibited doxorubicin-induced increase in cellular reactive oxygen species (ROS), the activity of caspases 8 and 3, cytochrome c release, and apoptosis. Direct injection of Ad2/CMVp35 into the left ventricular wall inhibited myocardial caspase 3 activity and apoptosis and improved left ventricular performance in rats treated with doxorubicin, whereas the same dose of Ad2/CMV beta gal encoding beta-galactosidase had no effect. These results suggest that adenovirus-mediated expression of p35 protects cardiomyocytes against doxorubicin cardiotoxicity, possibly by inhibiting caspase activity and by reducing cellular ROS levels. Localized delivery of gene transfer vectors expressing an antiapoptotic protein such as p35 to the myocardium may represent a therapeutic approach to alleviate doxorubicin-induced cardiomyopathy.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/toxicidad , Cardiomiopatías/terapia , Doxorrubicina/toxicidad , Proteínas Virales/genética , Animales , Apoptosis , Cardiomiopatías/inducido químicamente , Cardiomiopatías/patología , Caspasas/metabolismo , Células Cultivadas , Cromosomas Artificiales Bacterianos/genética , Terapia Genética , Vectores Genéticos , Proteínas Inhibidoras de la Apoptosis , Masculino , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Células Tumorales Cultivadas , Proteínas Virales/metabolismo
14.
J Mol Cell Cardiol ; 35(7): 811-21, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12818572

RESUMEN

Cardiomyocyte apoptosis by Fas ligand (FasL)/Fas signaling is associated with various pathophysiological conditions, such as ischemia/reperfusion injury and congestive heart failure. In this study, we tested the hypothesis that shedding of membrane FasL is a mechanism for downregulating FasL/Fas signaling and both membrane and soluble FasL are involved in cardiomyocyte hypoxia/reoxygenation (H/R) injury. We also examined the relative importance of mitochondrial damage and direct cleavage of the executioner caspases by activated initiator caspase 8 in the propagation of FasL/Fas signaling activated by either recombinant membrane FasL or H/R. We demonstrated that in neonatal rat cardiomyocytes maintained under normal culture conditions, recombinant human soluble FasL increased caspase 3 activation by twofold but did not reduce cell viability. In contrast, infection with a recombinant adenoviral vector expressing the non-cleavable human FasL (Ad2/nchFasL) resulted in cardiomyocyte death that was attenuated by soluble FasL. H/R increased the mRNA levels of both FasL and Fas and activated caspases 8, 9 and 3, indicating the activation of FasL/Fas signaling. Z-IETD.fmk and Z-LEHD.fmk, selective inhibitors for caspases 8 and 9, respectively, abolished caspase 3 activation induced by Ad2/nchFasL or H/R. Z-IETD.fmk also significantly reduced Ad2/nchFasL- or H/R-induced cardiomyocyte death. H/R potentiated membrane FasL-induced cell death. These results suggest that shedding of membrane FasL downregulates FasL/Fas signaling in cardiomyocytes and both membrane and soluble FasL contribute to H/R injury. Activation of FasL/Fas signaling by either recombinant membrane FasL under normal culture conditions or H/R causes cardiomyocyte death mainly through the mitochondrial damage/caspase 9 activation pathway.


Asunto(s)
Membrana Celular/metabolismo , Citoplasma/metabolismo , Glicoproteínas de Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Daño por Reperfusión/metabolismo , Animales , Caspasa 9 , Caspasas/metabolismo , Muerte Celular/fisiología , Proteína Ligando Fas , Humanos , Hipoxia/metabolismo , Mitocondrias/metabolismo , Ratas
15.
J Mol Cell Cardiol ; 34(7): 765-74, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12099716

RESUMEN

Peroxisome proliferator-activated receptors (PPAR), especially the PPARalpha and PPARgamma, are associated with an extraordinary diverse spectrum of cardiovascular diseases including hypertension, angiogenesis, cardiac hypertrophy, and atherosclerosis. PGAR (for PPAR gamma angiopoietin-related gene) is a recently identified PPAR target gene which is associated with adipose differentiation, systemic lipid metabolism, energy homeostasis, and possibly angiogenesis. We report here that WY-14643, a selective PPARalpha ligand up-regulated PGAR expression in neonatal rat cardiomyocytes. In parallel to activating the expression of vascular endothelial growth factor and glucose transporter-4, hypoxia increased PGAR mRNA levels. PGAR expression was also increased by desferrioxamine and CoCl(2), but not by sodium cyanide, results consistent with the pharmacological features of hypoxia-responsive genes. These studies are the first to demonstrate that hypoxia increases the mRNA levels of a PPAR target gene in cardiomyocytes. Furthermore, infection with adenoviral vectors encoding the wild-type or a hybrid form of HIF-1alpha highly increased PGAR mRNA levels. In contrast, neither hypoxia nor overexpression of HIF-1alpha affected the mRNA levels of PPARalpha, PPAR gamma, and muscle carnitine palmitoyltransferase, a known PPARalpha target gene. These results suggest that hypoxic activation of PGAR expression is likely mediated by HIF-1 but not the PPARalpha/RXR pathway.


Asunto(s)
Proteínas Sanguíneas , Proteínas de Unión al ADN/metabolismo , Glicoproteínas/metabolismo , Hipoxia/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción , Inductores de la Angiogénesis/metabolismo , Angiopoyetina 1 , Angiopoyetina 2 , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas , Animales , Regulación de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intercelular , Glicoproteínas de Membrana/metabolismo , Ratas , Regulación hacia Arriba
16.
Cardiovasc Res ; 55(2): 309-19, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12123770

RESUMEN

OBJECTIVE: This study aimed to examine the effects of adenovirus-mediated expression of p35, a baculovirus gene, on apoptosis induced by hypoxia/reoxygenation (H/R) in cardiomyocytes. METHODS: Neonatal rat cardiomyocytes were infected with recombinant adenoviral vectors expressing p35 (Ad2/CMVp35) or no transgene (Ad2/CMVEV) and were then subjected to H/R. Separate groups of non-infected cardiomyocytes were treated with pharmacological caspase inhibitors or antioxidants. Cell viability, apoptosis, caspase activity, and cellular reactive oxygen species (ROS) were measured using various assays. RESULTS: H/R decreased cell viability and increased cellular ROS levels, caspase activity, and cell apoptosis. Infection with Ad2/CMVp35 effectively inhibited the increase in cellular ROS levels, the activities of caspases 3 and 8, apoptosis, and cell death following H/R, whereas Ad2/CMVEV had no effect. Despite its ability to abolish the increase in caspase activity and partially inhibit apoptosis, the pan-caspase inhibitor ZVAD-fmk (100 microM) failed to significantly reduce cell death induced by H/R. N-acetyl-L-cysteine, an antioxidant, completely inhibited H/R-induced increase in cellular ROS levels, but reduced apoptosis and cell death by 30% only. CONCLUSIONS: Adenovirus-mediated expression of p35 effectively inhibits H/R-induced cardiomyocyte apoptosis by reducing cellular ROS levels and inhibiting caspase activity.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Caspasas/metabolismo , Hipoxia de la Célula/fisiología , Técnicas de Transferencia de Gen , Lipoproteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Virales , Adenoviridae/genética , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas de la Membrana Bacteriana Externa/genética , Inhibidores de Caspasas , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Inhibidores de Cisteína Proteinasa/farmacología , Vectores Genéticos , Lipoproteínas/genética , Ratas , Superóxido Dismutasa/metabolismo
17.
Physiol Genomics ; 8(1): 23-32, 2002 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-11842128

RESUMEN

The cellular response to hypoxia depends on rapid posttranslational modifications of proteins as well as regulation of gene expression. We performed serial analysis of gene expression (SAGE) on human cardiac cells under normoxia, subjected to hypoxia, or infected with Ad2/HIF-1alpha/VP16 (an adenoviral vector expressing a stable hybrid form of hypoxia-inducible factor 1alpha) or Ad2/CMVEV (an empty vector). Of the 97,646 SAGE tags that were sequenced, 27% matched GenBank entries, while an additional 32% matched expressed sequence tags (ESTs) in UniGene. We analyzed 161 characterized genes or ESTs with a putative identification. Expression of 35, 11, and 46 genes was increased by hypoxia, infection with Ad2/EVCMV, or infection with Ad2/HIF-1alpha/VP16, respectively, compared with normoxia; conversely, 20, 11, 38 genes, respectively, were expressed at lower levels. Genes regulated by hypoxia were associated with transcription, biosynthesis, extracellular matrix formation, glycolysis, energy production, cell survival, and cell stress. Changes following infection with Ad2/HIF-1alpha/VP16 mimicked the hypoxic response to a certain extent. Infection with Ad2/CMVEV affected expression of genes that were associated with extracellular matrix formation and membrane trafficking. Differential expression of select genes was confirmed using TaqMan in additional human cardiac cells and rat neonatal ventricular myocytes. These data provide insight into gene expression underlying the diverse and complex cellular response to hypoxia, expression of HIF-1alpha/VP16, or adenoviral infection.


Asunto(s)
Corazón Fetal/metabolismo , Corazón Fetal/fisiopatología , Perfilación de la Expresión Génica/métodos , Hipoxia/fisiopatología , Miocardio/metabolismo , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/genética , Adenoviridae/genética , Células Cultivadas , Regulación hacia Abajo/genética , Corazón Fetal/citología , Corazón Fetal/fisiología , Regulación de la Expresión Génica/fisiología , Vectores Genéticos/biosíntesis , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Miocardio/citología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología , Regulación hacia Arriba/genética
18.
Biochem Biophys Res Commun ; 291(4): 908-14, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11866451

RESUMEN

Hypoxia regulates expression of vascular endothelial growth factor (VEGF) by increasing its transcription and by stabilizing its mRNA. Despite the pivotal role of hypoxia-inducible factor 1 (HIF-1) in transcriptional activation of hypoxia-responsive genes, it is not known whether HIF-1 mediates hypoxia-induced stabilization of VEGF mRNA. We constructed adenoviral vectors expressing either the wild-type HIF-1 alpha (Ad2/HIF-1 alpha/FL), a constitutively stable hybrid form of HIF-1 alpha (Ad2/HIF-1 alpha/VP16), or no transgene (Ad2/CMVEV). In rat glioma (C6) cells and human cardiac, vascular smooth muscle, and endothelial cells, infection with Ad2/HIF-1 alpha/VP16 or Ad2/HIF-1 alpha/FL increased VEGF expression at both the mRNA and protein levels. Under normoxic conditions, the half-life of VEGF mRNA was 42 min in C6 cells. Hypoxia and Ad2/HIF-1 alpha/VP16 increased the half-life of VEGF mRNA to 3.3 and 2.7 h, respectively, while Ad2/CMVEV had no effect. These studies are the first to demonstrate that overexpression of HIF-1 alpha increases VEGF mRNA stability. Our results also suggest that stabilization of VEGF mRNA by hypoxia is mediated, at least in part, by HIF-1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Proteínas Nucleares/metabolismo , Estabilidad del ARN , Factores de Transcripción , Adenoviridae/genética , Animales , Hipoxia de la Célula , Células Cultivadas , Proteínas de Unión al ADN/genética , Factores de Crecimiento Endotelial/biosíntesis , Vectores Genéticos , Semivida , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cinética , Linfocinas/biosíntesis , Proteínas Nucleares/genética , ARN Mensajero/metabolismo , Ratas , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...