Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Clin Invest ; 117(7): 1844-55, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17557121

RESUMEN

Tumor radioresponsiveness depends on endothelial cell death, which leads in turn to tumor hypoxia. Radiation-induced hypoxia was recently shown to trigger tumor radioresistance by activating angiogenesis through hypoxia-inducible factor 1-regulated (HIF-1-regulated) cytokines. We show here that combining targeted radioiodide therapy with angiogenic inhibitors, such as canstatin, enhances direct tumor cell apoptosis, thereby overcoming radio-induced HIF-1-dependent tumor survival pathways in vitro and in vivo. We found that following dual therapy, HIF-1alpha increases the activity of the canstatin-induced alpha(v)beta(5) signaling tumor apoptotic pathway and concomitantly abrogates mitotic checkpoint and tetraploidy triggered by radiation. Apoptosis in conjunction with mitotic catastrophe leads to lethal tumor damage. We discovered that HIF-1 displays a radiosensitizing activity that is highly dependent on treatment modalities by regulating key apoptotic molecular pathways. Our findings therefore support a crucial role for angiogenesis inhibitors in shifting the fate of radiation-induced HIF-1alpha activity from hypoxia-induced tumor radioresistance to hypoxia-induced tumor apoptosis. This study provides a basis for developing new biology-based clinically relevant strategies to improve the efficacy of radiation oncology, using HIF-1 as an ally for cancer therapy.


Asunto(s)
Apoptosis/efectos de la radiación , Colágeno Tipo IV/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fragmentos de Péptidos/metabolismo , Adenoviridae/genética , Animales , Línea Celular , Colágeno Tipo IV/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Integrinas/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Fragmentos de Péptidos/genética , Transducción de Señal , Simportadores/genética , Simportadores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Ther ; 16(8): 1474-80, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18560416

RESUMEN

Liver tropism potentially leading to massive hepatocyte transduction and hepatotoxicity still represents a major drawback to adenovirus (Ad)-based gene therapy. We previously demonstrated that substitution of the hexon hypervariable region 5 (HVR5), the most abundant capsid protein, constituted a valuable platform for efficient Ad retargeting. The use of different mouse strains revealed that HVR5 substitution also led to dramatically less adenovirus liver transduction and associated toxicity, whereas HVR5-modified Ad were still able to transduce different cell lines efficiently, including primary hepatocytes. We showed that HVR5 modification did not significantly change Ad blood clearance or liver uptake at early times. However, we were able to link the lower liver transduction to enhanced HVR5-modified Ad liver clearance and impaired use of blood factors. Most importantly, HVR5-modified vectors continued to transduce tumors in vivo as efficiently as their wild-type counterparts. Taken together, our data provide a rationale for future design of retargeted vectors with a safer profile.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside/genética , Hígado/metabolismo , Alanina Transaminasa/sangre , Animales , Carcinoma Pulmonar de Lewis/sangre , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Vectores Genéticos/genética , Interleucina-6/sangre , Recuento de Leucocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Desnudos , Recuento de Plaquetas , Reacción en Cadena de la Polimerasa , Transducción Genética/métodos
3.
Biomacromolecules ; 9(10): 2881-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18788777

RESUMEN

Specific siRNAs that target estrogen receptor alpha (ERalpha) were encapsulated in nanocapsules (NCs). We produced small (approximately 100-200 nm) ERalpha-siRNA NCs with a water core by incorporating two mixed duplexes of specific ERalpha-siRNAs (ERalpha-mix-siRNA) into NCs. The encapsulation yield that was obtained with poly(iso-butylcyanoacrylate) (PIBCA) NCs was low, whereas no release of trapped siRNA was observed for poly(ethylene)glycol-poly(D,L-lactide-co-glycolide) (PEG-PLGA) NCs. High levels of ERalpha-siRNA incorporation into PEG-epsilon-caprolactone-malic acid (PEG-PCL/MA) NCs (3.3 microM in a polymer solution at 16 mg/mL) were observed (72% yield). No difference in size or zeta potential was observed between siRNA NCs that were based on PEG-PCL/MA and empty NCs. Fluorescence quenching assays confirmed the incorporation of siRNA into the NC core. A persistent loss of ERalpha (90% over 5 days) was observed in MCF-7 human breast cancer cells that were exposed to PEG-PCL/MA NCs that were loaded with ERalpha-siRNA. The intravenous injection of these NCs into estradiol-stimulated MCF-7 cell xenografts led to a significant decrease in tumor growth and a decrease in ERalpha expression in tumor cells. These data indicate that a novel strategy, based on ERalpha-siRNA delivery, could be developed for the treatment of hormone-dependent breast cancers.


Asunto(s)
Materiales Biocompatibles/química , Química Física/métodos , Receptor alfa de Estrógeno/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Emulsiones , Femenino , Humanos , Ensayo de Materiales , Ratones , Ratones Desnudos , Nanocápsulas/química , Trasplante de Neoplasias
4.
J Steroid Biochem Mol Biol ; 102(1-5): 114-27, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17056251

RESUMEN

Anti-oestrogens (AEs) are currently used for treating hormone-dependent breast cancers. They specifically bind to oestrogen receptors (ERs) and inhibit their transactivation capacity. However, ERs are present in various other tissues in which AEs may have either a beneficial or detrimental action. AE administration via systems targeting breast tumours may be an important therapeutic improvement. Thus, several biodegradable drug delivery systems containing either "mixed" (4-hydroxytamoxifen - 4-HT) or "pure" (RU 58668 - RU) AEs were prepared. Liposomes and nanospheres (NS, composed of non-toxic and biodegradable lipids and poly(d,l-lactic acid) incorporated up to 1 and 0.5 mM AE, respectively. Nanocapsules (NCs) in which an oily core solubilises the AE incorporated no more than 0.02 mM of the drug. PEG-functionalised nanoparticles survived longer in plasma and had better controlled release of the drug. The small size of the vectors (100-250 nm) was compatible with their extravasation through the discontinuous endothelium of tumour vasculature, allowing their accumulation in MCF-7 cell xenografts and leading to a prolonged exposure of the tumour to AEs. In these tumours and in MCF-7/ras xenografts, RU-NS and RU-NC (6.5mg/kg/week and 0.27 mg/kg/week, respectively, doses at which free RU had a very weak effect), both inhibited tumour growth. Entrapped RU significantly induced involution of tumours and strongly induced apoptosis in tumour cells, concomitantly with inhibiting tumour angiogenesis. 4-HT-nanoparticles also arrest oestradiol-induced tumour growth, inducing apoptosis and inhibiting angiogenesis. However, unlike RU-nanoparticles, they did not promote ERalpha subtype loss in tumour cells. Subcutaneous administration of both RU- and 4-HT-NS in MCF-7 xenografts strongly arrested tumour growth for prolonged periods and RUNS decreased the number of tumour epithelial cells. Analysis of the proteins involved in cell cycle proliferation and apoptosis confirmed that RU-nanoparticles were more efficient than 4-HT-nanoparticles. Their lack of toxicity and high anti-tumour potency that affects only tumour cells in the xenograft models mean these AE-loaded colloidal systems are a breakthrough in hormone-dependent breast cancer treatment.


Asunto(s)
Coloides , Sistemas de Liberación de Medicamentos , Moduladores de los Receptores de Estrógeno/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanocápsulas , Receptores de Estrógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Nanopartículas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Steroid Biochem Mol Biol ; 100(1-3): 67-78, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16753295

RESUMEN

In most of multiple myeloma (MM) cells, the "pure" antiestrogen (AE) RU 58668 (RU) induced either a G1-arrest (LP-1, OPM-2, NCI-H929, U266 cells) or apoptosis (RPMI 8226 cells). In RPMI 8226 cells, RU activates a caspase-dependent cell death pathway leading to the release of cytochrome c, the decrease of the essential MM survival factor Mcl-1, the cleavage of Bid and the activation of caspases-3 and -8. Incorporation of RU in pegylated cholesterol-containing liposomes allowed a controlled RU release, improving its anti-proliferative and apoptotic effects in cells. In RPMI 8226 xenografts, i.v. injected RU-liposomes but not free RU, exhibited antitumor activity. In vivo, RU-liposomes triggered the mitochondrial death pathway, concomitantly with a down-regulation of Mcl-1 and Bid cleavage. The decrease of CD34 immunoreactivity indicated a reduction of angiogenesis. The decrease of VEGF secretion in vitro supported a direct effect of RU on angiogenesis. These pro-apoptotic and antiangiogenic effects were explained by a prolonged exposure to the drug and to the endocytosis capacity of liposomes which might increase RU uptake and bypass a membrane export of free RU. Thus, these combined enhanced activities of RU-liposomes support that such a delivery of an AE may constitute a strategy of benefit for MM treatment.


Asunto(s)
Antineoplásicos/farmacología , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Liposomas , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos/métodos , Estradiol/aislamiento & purificación , Estradiol/farmacología , Antagonistas de Estrógenos/aislamiento & purificación , Moduladores de los Receptores de Estrógeno/administración & dosificación , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Modelos Biológicos , Mieloma Múltiple/patología , Tonsila Palatina/citología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Res ; 64(6): 2062-9, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15026344

RESUMEN

Metargidin, a transmembrane protein of the adamalysin family, and integrins, e.g., alpha5beta1 and alphav, are preferentially expressed on endothelial cells on angiogenesis. Furthermore, metargidin interacts with these integrins via its disintegrin domain. In this study, recombinant human disintegrin domain (RDD) was produced in Escherichia coli by subcloning its cDNA into the pGEX-2T vector, and the effect of purified RDD on different steps of angiogenesis was evaluated. At concentrations of 2-10 micro g/ml, RDD exhibited inhibitory activities in a variety of in vitro functional assays, including endothelial cell proliferation and adhesion on the integrin substrates fibronectin, vitronectin, and fibrinogen. RDD (10 micro g/ml) totally abrogated endothelial cell migration and blocked most capillary formation in a three-dimensional fibrin gel. To test RDD efficacy in vivo, the RDD gene inserted into pBi vector containing a tetracycline-inducible promoter was electrotransferred into nude mouse muscle. RDD was successfully synthesized by muscle cells in vivo as shown by immunolabeling and Western blotting. In addition, 78% less MDA-MB-231 tumor growth, associated with strong inhibition of tumor angiogenesis, was observed in athymic mice bearing electrotransferred RDD. Moreover, in the presence of RDD, 74% fewer B16F10 melanoma lung metastases were found in C57BL/6 mice. Taken together, these results identified this RDD as a potent intrinsic inhibitor of angiogenesis, tumor growth, and metastasis, making it a promising tool for use in anticancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Desintegrinas/uso terapéutico , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/prevención & control , Proteínas de la Membrana/uso terapéutico , Metaloendopeptidasas/uso terapéutico , Neovascularización Patológica/prevención & control , Proteínas ADAM , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Escherichia coli/genética , Femenino , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Músculo Esquelético/patología , Proteínas Recombinantes/uso terapéutico , Células Tumorales Cultivadas
7.
Drug Metab Dispos ; 35(2): 306-14, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17132762

RESUMEN

Busulfan is an example of a drug eliminated through glutathione S-transferase (GST)-catalyzed conjugation with reduced glutathione (GSH). We studied the pharmacokinetics and toxicity of busulfan in C57BL6 mice in correlation with liver GST activity and GSH synthesis by accurate determination of precursors, namely, gamma-glutamyl-cysteine and cysteine. A significantly lower incidence of acute toxicity was observed in mice receiving busulfan 16.5 mg/kg twice a day compared with animals receiving 33 mg/kg once a day. In both cases, a total dose of 132 mg/kg was administered over 4 days. The difference in toxicity was explained by pharmacokinetics since a strong induction of clearance was observed only in animals treated twice daily. Induction of metabolism was correlated with an increase in liver cysteine content and enhanced glutathione synthesis rate, whereas GST activity was unchanged. To our knowledge, this is the first time that in vivo flux of GSH synthesis has been shown to be closely related to a drug plasma clearance and toxicity. These results allow hypothesizing that GSH liver synthesis may directly influence busulfan clearance in humans with possible implications in the occurrence of hepatic veno-occlusive disease.


Asunto(s)
Busulfano/toxicidad , Glutatión/biosíntesis , Animales , Trasplante de Médula Ósea , Busulfano/farmacocinética , Interacciones Farmacológicas , Glutatión Transferasa/metabolismo , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL
8.
PLoS One ; 2(11): e1135, 2007 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17989772

RESUMEN

We report the first successful use of irreversible electroporation for the minimally invasive treatment of aggressive cutaneous tumors implanted in mice. Irreversible electroporation is a newly developed non-thermal tissue ablation technique in which certain short duration electrical fields are used to permanently permeabilize the cell membrane, presumably through the formation of nanoscale defects in the cell membrane. Mathematical models of the electrical and thermal fields that develop during the application of the pulses were used to design an efficient treatment protocol with minimal heating of the tissue. Tumor regression was confirmed by histological studies which also revealed that it occurred as a direct result of irreversible cell membrane permeabilization. Parametric studies show that the successful outcome of the procedure is related to the applied electric field strength, the total pulse duration as well as the temporal mode of delivery of the pulses. Our best results were obtained using plate electrodes to deliver across the tumor 80 pulses of 100 micros at 0.3 Hz with an electrical field magnitude of 2500 V/cm. These conditions induced complete regression in 12 out of 13 treated tumors, (92%), in the absence of tissue heating. Irreversible electroporation is thus a new effective modality for non-thermal tumor ablation.


Asunto(s)
Electroporación , Sarcoma Experimental/terapia , Animales , Línea Celular Tumoral , Daño del ADN , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Temperatura
9.
Hepatology ; 44(2): 399-409, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16871589

RESUMEN

Fas and tumor necrosis factor receptor 1 (TNFR1) are death receptors involved in various diseases such as hepatitis, sepsis, or graft rejection. Neutralizing antibodies to death ligands or soluble death receptors can inhibit cell death; however, they induce side effects because of their systemic actions. To specifically block death signaling to target cells, we created death domain-deficient (DeltaDD) membrane-anchored receptors, delivered to the liver by either recombinant adenovirus or hydrodynamic pressure of nonviral recombinant plasmids. In anti-Fas antibody-induced fulminant hepatitis, mice expressing recombinant Fas-decoy receptors (FasDeltaDD) in their livers were completely protected against apoptosis and survived fulminant hepatitis. In T-cell-dependent concanavalin A-induced autoimmune hepatitis, FasDeltaDD antagonist expression prevented hepatocyte damage and mouse death. Finally, TNFR1DeltaDD effectively protected mice against LPS-induced septic shock. In conclusion, such DeltaDD-decoy receptors act as dominant-negative receptors exerting local inhibition, while avoiding systemic neutralization of apoptosis ligands, and might have therapeutic potential in hepatitis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/fisiología , Expresión Génica , Hepatitis/metabolismo , ARN/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Western Blotting , Modelos Animales de Enfermedad , Proteína de Dominio de Muerte Asociada a Fas , Femenino , Hepatitis/patología , Hepatitis/prevención & control , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Proc Natl Acad Sci U S A ; 103(49): 18721-6, 2006 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17130448

RESUMEN

Angiopoietin-like 4 (ANGPTL4), a secreted protein of the angiopoietin-like family, is induced by hypoxia in both tumor and endothelial cells as well as in hypoxic perinecrotic areas of numerous cancers. Here, we investigated whether ANGPTL4 might affect tumor growth as well as metastasis. Metastatic 3LL cells were therefore xenografted into control mice and mice in which ANGPTL4 was expressed by using in vivo DNA electrotransfer. Whereas primary tumors grew at a similar rate in both groups, 3LL cells metastasized less efficiently to the lungs of mice that expressed ANGPTL4. Fewer 3LL emboli were observed in primary tumors, suggesting that intravasation of 3LL cells was inhibited by ANGPTL4. Furthermore, melanoma B16F0 cells injected into the retro-orbital sinus also metastasized less efficiently in mice expressing ANGPTL4. Although B16F0 cells were observed in lung vessels, they rarely invaded the parenchyma, suggesting that ANGPTL4 affects extravasation. In addition, recombinant B16F0 cells that overexpress ANGPTL4 were generated, showing a lower capacity for in vitro migration, invasion, and adhesion than control cells. Expression of ANGPTL4 induced reorganization of the actin cytoskeleton through inhibition of actin stress fiber formation and vinculin localization at focal contacts. Together, these results show that ANGPTL4, through its action on both vascular and tumor compartments, prevents the metastatic process by inhibiting vascular activity as well as tumor cell motility and invasiveness.


Asunto(s)
Permeabilidad Capilar/fisiología , Movimiento Celular/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/prevención & control , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas , Animales , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma Pulmonar de Lewis/secundario , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Ganglios Linfáticos/patología , Metástasis Linfática/patología , Metástasis Linfática/prevención & control , Linfopoyesis/fisiología , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Neovascularización Fisiológica
11.
Mol Ther ; 14(2): 175-82, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16600689

RESUMEN

Angiogenesis is essential for tumor growth and metastatic dissemination. We have previously shown that human angiotensinogen (AGT) can in vitro inhibit endothelial cell proliferation and migration, capillary-like tube formation, and neovascularization. To determine whether AGT can exert an antitumoral effect through its antiangiogenic properties, we constructed a recombinant adenovirus carrying the human angiotensinogen gene under the control of the cytomegalovirus promoter (AdAGT). In vitro studies showed that AdAGT selectively inhibited endothelial cell proliferation. In vivo, injections of AdAGT into preestablished human MDA-MB-231 mammary carcinomas in nude mice inhibited tumor growth by 70% compared to controls, with 21% total regression. This effect was associated with the suppression of intratumoral vascularization and marked necrosis. Furthermore, in vitroAdAGT infection of MDA-MB-231 and murine melanoma B16F10 cells strongly blocked their in vivo tumorigenicity. Then, in mice expressing high levels of AGT (i.e., either iv injected with AdAGT or HuAGT transgenic mice), the number of B16F10 pulmonary metastases was 85% lower than in control C57BL/6 mice. Our data demonstrate that AGT is a very potent antiangiogenic factor in vivo, independent of angiotensin II generation. Its delivery by gene transfer represents a promising new strategy to block primary tumor growth and to prevent metastasis.


Asunto(s)
Adenoviridae/genética , Angiotensinógeno/genética , Terapia Genética/métodos , Metástasis de la Neoplasia/terapia , Neoplasias Experimentales/terapia , Neovascularización Patológica/terapia , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/citología , Endotelio Vascular/citología , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/secundario , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Trasplante Heterólogo
12.
J Gene Med ; 5(2): 109-19, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12539149

RESUMEN

BACKGROUND: The advent of cardiac gene therapy in clinical practice requires a more efficient and safer myocardial gene delivery in large animals. A new approach to adenovirus-mediated intracoronary gene transfer in the piglet, using a heterotopic heart transplantation model, was designed to maximize the duration of contact between the vector and the heart in noncoronary flow conditions. METHODS: Recombinant adenoviruses harboring a nucleus-localized beta-galactosidase gene under the control of a viral promoter were injected into the coronary vessels of the harvested hearts at a dose ranging from 10(10) to 2 x 10(11) pfu. The graft was maintained for 75 min in saline solution and then implanted in the abdomen of recipients. Gene transfer to allografts was evaluated 4 days after grafting by immunohistochemical and enzymatic analysis of beta-galactosidase expression. RESULTS: Transgene expression was detected in all cardiac areas and up to 64, 44, 32, and 15% of positive nuclei were estimated in the left ventricle wall in four animals out of eleven. In the remaining animals, transgene expression was focally distributed, mainly in the left ventricle wall. PCR analysis revealed the presence of adenoviral sequences, albeit minimal, in exposed organs such as the liver and lung. CONCLUSIONS: This procedure demonstrated that direct intracoronary gene transfer can be achieved using an ex vivo gene transfer strategy.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Trasplante de Corazón , Corazón/fisiología , Adenoviridae/metabolismo , Animales , Línea Celular , Vectores Genéticos , Porcinos , Distribución Tisular , Transgenes , Trasplante Heterotópico , beta-Galactosidasa/metabolismo
13.
Mol Ther ; 8(3): 425-33, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12946315

RESUMEN

We describe an approach employing intramuscular plasmid electrotransfer to deliver secretable forms of K1-5 and K1-3-HSA (a fusion of K1-3 with human serum albumin), which span, respectively, five and three of the five kringle domains of plasminogen. A tetracycline-inducible system (Tet-On) composed of three plasmids coding, respectively, for the transgene, the tetracycline transcriptional activator rtTA, and the silencer tTS was employed. K1-3-HSA and K1-5, produced from C2C12 muscle cells, were found to inhibit endothelial cell (HMEC-1) proliferation by 30 and 51%, respectively. In vivo, the expression of the transgene upon doxycycline stimulation was rapid, stable, and tightly regulated (no background expression) and could be maintained for at least 3 months. Blood half-lives of 2.1 and 3.7 days were found for K1-5 and K1-3-HSA, respectively. The K1-5 protein was secreted from muscle into blood at a level of 45 ng/ml, which was sufficient to inhibit MDA-MB-231 tumor growth by 81% in nude mice and B16-F10 melanoma cell lung invasion in C57BL/6 mice by 73%. PECAM-1 immunostaining studies revealed modest tumor vasculature in mice expressing K1-5. In contrast, K1-3-HSA, although secreted into blood at much higher level (250 ng/ml) than K1-5, had no effect on tumor growth.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Electroporación , Vectores Genéticos , Músculo Esquelético/metabolismo , Neoplasias/terapia , Inhibidores de la Angiogénesis/metabolismo , Regulación de la Expresión Génica , Metástasis de la Neoplasia/terapia , Péptidos/genética , Péptidos/metabolismo , Plásmidos , Plasminógeno/genética , Plasminógeno/metabolismo , Tetraciclina/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA