Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vaccine ; 29(7): 1463-71, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21184856

RESUMEN

The efficacy of recombinant adenoviruses (Ads) vaccine vectors is diminished by the high prevalence of anti-Ad antibodies (Abs) that hampers gene transfer. Epitope display on Ad capsid constitutes an alternative approach to bypass anti-Ad Ab capacity from blocking antigen expression. To understand the role of the epitope insertion site, an ovalbumin-derived epitope was genetically inserted into either Ad hexon or fiber proteins. Hexon-modified Ads triggered higher anti-ovalbumin Ab responses after one injection but surprisingly fiber-modified Ads were by far more potent after two or several administrations. Our data unravel a role for anti-Ad humoral immunity in controlling anti-epitope humoral responses.


Asunto(s)
Proteínas de la Cápside/genética , Cápside/inmunología , Epítopos de Linfocito B/inmunología , Inmunidad Humoral , Vacunas Sintéticas/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Epítopos de Linfocito B/genética , Femenino , Vectores Genéticos , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Vacunas Sintéticas/genética
2.
Int J Oncol ; 38(1): 179-88, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21109939

RESUMEN

We previously demonstrated, using the glioblastoma cell line U87MG as an experimental model, that the adenoviral mediated overexpression of the truncated protein HARPΔ111-136 inhibits the proliferation of these cells in vitro as well as tumor growth and angiogenesis in vivo. This study focused on identifying the underlying mechanisms for the observed antitumoral effect. The present study demonstrated that HARPΔ111-136 induced the ATF4/ATF3/CHOP cascade resulting in a strong expression of the proapoptotic protein CHOP, leading to tumor cell apoptosis as demonstrated by PARP cleavage and FACS analysis. siRNA-mediated CHOP gene silencing abolished Ad-HARPΔ111-136 induced apoptosis. Moreover, Ad-HARPΔ111-136 increased the expression of the death receptor DR5 and enhanced U87MG cells sensitivity in vitro to TRAIL a DR5 ligand with subsequent activation of caspase 8. Infection of U87MG cells with Ad-HARPΔ111-136 also enhanced radiation-induced apoptosis. In vivo, the combination of Ad-HARPΔ111-136 and radiation therapy resulted in a striking inhibition (92%) of the growth of U87MG xenografts, resulting from the potent effect on tumor angiogenesis and tumor cell apoptosis as determined by TUNEL analysis. Taken together, our results indicated that the inhibitor HARPΔ111-136 sensitized U87MG cells to apoptosis.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , ADN Helicasas/biosíntesis , Glioblastoma/patología , Glioblastoma/radioterapia , Fragmentos de Péptidos/biosíntesis , Animales , Apoptosis/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , ADN Helicasas/genética , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Fragmentos de Péptidos/genética , Factor de Transcripción CHOP , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Cancer ; 127(5): 1038-51, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20013808

RESUMEN

Glioblastoma is the most common primary brain tumor in human adults. Since existing treatments are not effective enough, novel therapeutic targets must be sought. The heparin-binding growth factor, heparin affin regulatory peptide (HARP), also known as pleiotrophin (PTN), could potentially represent such a target. We have previously shown that a mutant protein, HARPDelta111-136, which lacks HARP's C-terminal 26 amino acids, acts as a dominant negative HARP effector by heterodimerizing with the wild-type growth factor. The aim of our study was to evaluate the potential inhibitory activity of HARPDelta111-136 on the U87 MG human glioblastoma cell line. By overexpressing the truncated form of HARP in stably established clones of U87 MG cells, we observed an inhibition of proliferation under both anchorage-dependent and anchorage-independent conditions. We confirmed these results in an in vivo subcutaneous tumor xenograft model. In addition, we found that HARPDelta111-136 inhibited cell proliferation in a paracrine manner. Analysis of key cellular pathways revealed a decrease of cell adhesion in U87 MG cells that overexpressed the mutant protein, which could explain this inhibitory effect. A replication-defective adenovirus model that encoded HARPDelta111-136 supported a putative antiproliferative role for the truncated protein in vitro and in vivo. Interestingly, HARPDelta111-136 was also able to abolish angiogenic activity in HUVEC proliferation and in a Matrigel plug assay. These results demonstrate that considering its antiproliferative and angiostatic effects, HARPDelta111-136 could be of great interest when used in conjunction with standard treatments.


Asunto(s)
Neoplasias Encefálicas/patología , Proteínas Portadoras/genética , Citocinas/genética , Glioblastoma/patología , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Animales , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Células CHO , Adhesión Celular , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Cricetinae , Cricetulus , Combinación de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Laminina/metabolismo , Ratones , Ratones Desnudos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Proteoglicanos/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 7(9): 2817-27, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790762

RESUMEN

Pleiotrophin (PTN) is a 136-amino acid secreted heparin-binding protein that is considered as a rate-limiting growth and an angiogenic factor in the onset, invasion, and metastatic process of many tumors. Its mitogenic and tumorigenic activities are mediated by the COOH-terminal residues 111 to 136 of PTN, allowing it to bind to cell surface tyrosine kinase-linked receptors. We investigated a new strategy consisting in evaluating the antitumor effect of a truncated PTN, lacking the COOH-terminal 111 to 136 portion of the molecule (PTNDelta111-136), which may act as a dominant-negative effector for its mitogenic, angiogenic, and tumorigenic activities by heterodimerizing with the wild-type protein. In vitro studies showed that PTNDelta111-136 selectively inhibited a PTN-dependent MDA-MB-231 breast tumor and endothelial cell proliferation and that, in MDA-MB-231 cells expressing PTNDelta111-136, the vascular endothelial growth factor-A and hypoxia-inducible factor-1alpha mRNA levels were significantly decreased by 59% and 71%, respectively, compared with levels in wild-type cells. In vivo, intramuscular electrotransfer of a plasmid encoding a secretable form of PTNDelta111-136 was shown to inhibit MDA-MB-231 tumor growth by 81%. This antitumor effect was associated with the detection of the PTNDelta111-136 molecule in the muscle and tumor extracts, the suppression of neovascularization within the tumors, and a decline in the Ki-67 proliferative index. Because PTN is rarely found in normal tissue, our data show that targeted PTN may represent an attractive and new therapeutic approach to the fight against cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/farmacología , Citocinas/farmacología , Células Endoteliales/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Desnudos , Peso Molecular , Proteínas Mutantes/metabolismo , Neovascularización Patológica/patología , Receptores de Superficie Celular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Stem Cells ; 26(11): 2981-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18757301

RESUMEN

Prostate cancer metastasis to bone results in mixed osteolytic and osteoblastic lesions associated with high morbidity, and there is mounting evidence that the urokinase-type plasminogen system is causatively involved in the progression of prostate cancer. Adult mesenchymal stem cells (MSCs) are promising tools for cell-mediated gene therapy with the advantage of osteogenic potential, a critical issue in the case of osteolytic metastases. In this study, we evaluated the therapeutic use of engineered murine MSCs for in vivo delivery of the urokinase-type plasminogen antagonist amino-terminal fragment (hATF) to impair osteolytic prostate cancer cell progression in bone and to repair bone lesions. Bioluminescence imaging revealed that both primary MSCs and the MSC line C3H10T1/2 (C3) expressing hATF (MSC-hATF) significantly inhibited intratibial PC-3 Luciferase (Luc) growth following coinjection in SCID mice. Furthermore, microcomputed tomography imaging of vascular network clearly demonstrated a significant decrease in tumor-associated angiogenesis and a protection from tumor-induced osteolysis in MSC-hATF-treated mice. Importantly, the osteogenic potential of MSC-hATF cells was unaffected, and an area of new bone formation was evidenced in 60% of animals. Together, these data support the concept of MSC-based therapy of tumor osteolysis disease, indicating that MSCs may combine properties of vehicle for angiostatic agent with osteogenic potential. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Neoplasias Óseas/terapia , Células Madre Mesenquimatosas/citología , Fragmentos de Péptidos/biosíntesis , Neoplasias de la Próstata/terapia , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Adenoviridae/genética , Animales , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/secundario , Comunicación Celular , Línea Celular Tumoral , Estudios de Factibilidad , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Neovascularización Patológica/terapia , Osteogénesis , Osteólisis/patología , Osteólisis/terapia , Fragmentos de Péptidos/genética , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
6.
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
7.
Endocr Relat Cancer ; 14(3): 691-702, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17914099

RESUMEN

KiSS1 is a putative metastasis suppressor gene in melanoma and breast cancer-encoding kisspeptins, which are also described as neuroendocrine regulators of the gonadotropic axis. Negative as well as positive regulation of KiSS1 gene expression by estradiol (E(2)) has been reported in the hypothalamus. Estrogen receptor alpha (ERalpha level is recognized as a marker of breast cancer, raising the question of whether expression of KiSS1 and its G-protein-coupled receptor (GPR54) is down- or upregulated by estrogens in breast cancer cells. KiSS1 was found to be expressed in MDA-MB-231, MCF7, and T47D cell lines, but not in ZR75-1, L56Br, and MDA-MB-435 cells. KiSS1 mRNA levels decreased significantly in ERalpha-negative MDA-MB-231 cells expressing recombinant ERalpha. In contrast, tamoxifen (TAM) treatment of ERalpha-positive MCF7 and T47D cells increased KiSS1 and GPR54 levels. The clinical relevance of this negative regulation of KiSS1 and GPR54 by E(2) was then studied in postmenopausal breast cancers. KiSS1 mRNA increased with the grade of the breast tumors. ERalpha-positive invasive primary tumors expressed sevenfold lower KiSS1 levels than ERalpha-negative tumors. Among ERalpha-positive breast tumors from postmenopausal women treated with TAM, high KiSS1 combined with high GPR54 mRNA tumoral levels was unexpectedly associated with shorter relapse-free survival (RFS) relative to tumors expressing low tumoral mRNA levels of both genes. The contradictory observation of putative metastasis inhibitor role of kisspeptins and RFS to TAM treatment suggests that evaluation of KiSS1 and its receptor tumoral mRNA levels could be new interesting markers of the tumoral resistance to anti-estrogen treatment.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Neoplasias Hormono-Dependientes/diagnóstico , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/terapia , Supervivencia sin Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Estrógenos/farmacología , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Kisspeptinas , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/terapia , Pronóstico , Receptores de Kisspeptina-1 , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Células Tumorales Cultivadas
8.
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
9.
Exp Hematol ; 35(1): 64-74, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17198875

RESUMEN

Myelofibrosis is characterized by excessive deposits of extracellular matrix proteins, which occur as a marrow microenvironment reactive response to cytokines released from the clonal malignant myeloproliferation. The observation that mice exposed to high systemic levels of thrombopoietin (TPO) invariably developing myelofibrosis has allowed demonstration of the crucial role of transforming growth factor (TGF)-beta1 released by hematopoietic cells in the onset of myelofibrosis. The purpose of this study was to investigate whether TGF-beta1 inhibition could directly inhibit fibrosis development in a curative approach of this mice model. An adenovirus encoding for TGF-beta1 soluble receptor (TGF-beta-RII-Fc) was injected either shortly after transplantation (preventive) or 30 days post-transplantation (curative). Mice were transplanted with syngenic bone marrow cells transduced with a retrovirus encoding for murine TPO. All mice developed a myeloproliferative syndrome. TGF-beta-RII-Fc was detected in the blood of all treated mice, leading to a dramatic decrease in TGF-beta1 level. Histological analysis show that the two approaches (curative or preventive) were successful enough to inhibit bone marrow and spleen fibrosis development in this model. However, lethality of TPO overexpression was not decreased after treatment, indicating that in this mice model, myeloproliferation rather than fibrosis was probably responsible for the lethality induced by the disorder.


Asunto(s)
Terapia Genética/métodos , Mielofibrosis Primaria/terapia , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Adenoviridae , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Ratones , Ratones SCID , Mielofibrosis Primaria/prevención & control , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Enfermedades del Bazo/terapia , Análisis de Supervivencia , Trombopoyetina/administración & dosificación , Trombopoyetina/genética , Transducción Genética , Trasplante Isogénico
10.
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
11.
J Cancer Res Clin Oncol ; 132(9): 561-71, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16763806

RESUMEN

PURPOSE: To identify the characteristics and function of the truncated cadherin cDNA which encodes a soluble molecule containing the sequence of VE-cadherin extracellular domain repeats from repeat 1 to 4 (designated as CED1-4) and a secreting signal peptide at N terminal. METHODS: A pMSCV/CED1-4 vector was constructed. Recombinant retrovirus ReCED1-4 and ReEmpty were produced by 293 package cells and transfected into MDA-MB435 human breast cancer cells. The expression of CED1-4 in transfectants and their supernatant was analyzed by RT-PCR and Western blot, respectively. MDA-MB435 cell proliferation assays were performed in vitro and in vivo. CED-14-induced apoptosis was demonstrated using Annexin V binding, TUNEL and caspase 3 assays. The expression of integrin beta1 and c-fos mRNA was detected by RT-PCR. RESULTS: The constructed soluble CED1-4 encoded 484 amino acids and a secreting signal peptide (27 amino acids). CED1-4 was expressed by MDA-MB435/CED1-4 cells, and detected in the supernatant of CED1-4 tranfectants. CED1-4 transfection significantly inhibited the growth of MDA-MB435 cells in vitro and in vivo. About 22-fold increase in the early apoptotic cells in MDA-MB435/CED1-4 cells was observed as compared with MDA-MB435/empty cells. Increased activity of caspase 3 in MDA-MB435/CED1-4 cells was more than two times as compared with that of the control cells. Interestingly, integrin beta1 transcriptional level in MDA-MB435/CED1-4 cells was down-regulated as compared with control cells. The resistance of fibronectin to CED1-4 apoptotic inducibility was confirmed by detection of caspase 3. The blockage of c-fos transcriptional expression was detected in MDA-MB435/CED1-4 cells. CONCLUSIONS: The soluble truncated cadherin may be considered an apoptotic inducer and growth inhibitor in the MDA-MB435 breast carcinoma cell line. Down-regulation of integrin beta1 and blockage of c-fos expression may be related to CED1-4-induced apoptosis and growth inhibition.


Asunto(s)
Apoptosis/genética , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Carcinoma/metabolismo , Proteínas Recombinantes/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Neoplasias de la Mama/genética , Cadherinas/biosíntesis , Carcinoma/genética , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Clonación Molecular , Regulación hacia Abajo/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/genética , Proteínas Recombinantes/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Solubilidad , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
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
13.
Anticancer Drugs ; 17(2): 195-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428938

RESUMEN

The aim of the present study was to examine modifications of anti-tumor activity and toxicity of paclitaxel (PLX) when given p.o. after recombinant interleukin-2 (rIL-2) to Lewis lung carcinoma-bearing mice. PLX was given orally to mice at the dose of 15 mg/kg on day 8 and 30 mg/kg on day 15, either alone or after 16.5 microg of rIL-2 given i.p. twice a day either 1 or 3 days before. The anti-tumor activity was higher and PLX hematological toxicity not increased if orally administered PLX was given after a 3-day rIL-2 pre-treatment rather than if given alone. Lung metastasis was significantly lower and s.c. tumors were smaller in the PLX+rIL-2 group than in the PLX or rIL-2 or non-treated groups. In addition, a decrease in lung P-glycoprotein expression (investigated by Western blot analysis) was observed 1 h after the last administration of rIL-2 on day 7.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos Fitogénicos/administración & dosificación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Interleucina-2/uso terapéutico , Pulmón/efectos de los fármacos , Paclitaxel/administración & dosificación , Administración Oral , Animales , Western Blotting , Carcinoma Pulmonar de Lewis/metabolismo , Terapia Combinada , Femenino , Inyecciones Subcutáneas , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/uso terapéutico
14.
Hum Gene Ther ; 16(10): 1157-67, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16218777

RESUMEN

The urokinase plasminogen activator (uPA) is implicated in both cancer cell invasion and angiogenesis. It can interact with a specific receptor (uPAR) via the epidermal growth factor (EGF)-like domain in the urokinase amino-terminal fragment (ATF) in a species-specific manner. Our previous studies showed that adenovirusmediated delivery of murine ATF (AdmATF) suppressed human tumor growth in mouse models, by inhibiting murine angiogenesis. However, we cannot exclude its putative inhibitory action on human cancer cell invasion through a uPAR-independent pathway. To further investigate the mechanisms of ATF, we constructed another adenovirus, AdhmATF, expressing humanized murine ATF (hmATF). hmATF binds to human uPAR but not to murine uPAR. We compared the antagonist effect of both AdmATF and AdhmATF on human and murine cancer cells. In vitro, the supernatant from AdhmATF-infected cells repressed 79% of membrane-associated uPA activity on human MDA-MB-231 cells, whereas that from AdmATF-infected cells repressed 35% of membrane-associated uPA activity. On murine LLC cells, the supernatant from AdhmATF-infected cells inhibited 29% of cell surface uPA activity, whereas that from AdmATF-infected cells inhibited 74% of cell surface uPA activity. Similar results were obtained in a cell invasion assay. In vivo, intratumoral injection of the adenoviruses into LLC tumors on day 24 postinjection induced lower but significant tumor growth suppression by AdhmATF (tumor volume was 1185 +/- 128 mm3), whereas suppression by AdmATF was greater (407 +/- 147 mm3). In the MDA-MB-231 tumor model, on day 52 postinjection, tumor size was 187 +/- 47 mm3 in the AdhmATF-treated group and 468 +/- 65 mm3 in the AdmATF-treated group. The LLC and MDA-MB- 231 cell lines transfected by mATF or hmATF genes showed growth inhibition In vivo equivalent to the results obtained by adenovirus treatment. These results demonstrate the strong anticancer activity of ATF even when its uPAR-binding affinity has been suppressed, and indicate that ATF exerts an antitumor effect via dual mechanisms: essentially through targeting the uPA-uPAR system via the EGF-like domain and partially through targeting a uPAR-independent interaction via the kringle domain.


Asunto(s)
Adenoviridae , Terapia Genética , Vectores Genéticos , Neoplasias Experimentales/terapia , Receptores de Superficie Celular/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa , Animales , Línea Celular Tumoral , Femenino , Terapia Genética/métodos , Humanos , Ratones , Ratones Mutantes , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activador de Plasminógeno de Tipo Uroquinasa/administración & dosificación , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/genética
15.
Invest Radiol ; 40(8): 536-44, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16024992

RESUMEN

OBJECTIVES: This work includes (1) the characterization of a reproducible poly[lactide-coglycolide] (PLGA) microparticle preparation with an optimial mean diameter and size distribution and (2) the preliminary in vivo ultrasonographic investigation of PLGA microparticles. METHODS: A first series of PLGA microparticle preparations (1 to 15 mum) was acoustically characterized on a hydrodynamic device to select the most appropriate for ultrasound contrast agent application. Preparations of 3-microm microparticles were selected, characterized at different doses, and then injected into 20 melanoma grafted mice for contrast-enhanced power Doppler ultrasonography evaluation. RESULTS: The 3-microm microparticles (3.26-microm mean diameter with 0.41-microm standard deviation) led to in vitro enhancement of 18.3 dB at 0.62 mg/mL. In vivo experiments showed 47% enhancement of intratumoral vascularization detection after PLGA injection, significantly correlated (P < 0.0001) with preinjection intravascularization and tumoral volume. No toxicity was histologically observed. CONCLUSION: The 3-microm PLGA microparticles provided significant enhancement in vitro and in vivo without any toxicity.


Asunto(s)
Ácido Láctico , Melanoma/diagnóstico por imagen , Microesferas , Ácido Poliglicólico , Polímeros , Ultrasonografía Doppler , Animales , Medios de Contraste , Técnicas In Vitro , Ratones , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polisacáridos , Reproducibilidad de los Resultados
16.
Cancer Res ; 65(10): 4353-61, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15899827

RESUMEN

Canstatin, the noncollagenous domain of collagen type IV alpha-chains, belongs to a series of collagen-derived angiogenic inhibitors. We have elucidated the functional receptors and intracellular signaling induced by canstatin that explain its strong antitumor efficacy in vivo. For this purpose, we generated a canstatin-human serum albumin (CanHSA) fusion protein, employing the HSA moiety as an expression tag. We show that CanHSA triggers a crucial mitochondrial apoptotic mechanism through procaspase-9 cleavage in both endothelial and tumor cells, which is mediated through cross-talk between alphavbeta3- and alphavbeta5-integrin receptors. As a point of reference, we employed the first three kringle domains of angiostatin (K1-3), fused with HSA, which, in contrast to CanHSA, act only on endothelial cells through alphavbeta3-integrin receptor-mediated activation of caspase-8 alone, without ensuing mitochondrial damage. Taken together, these results provide insights into how canstatin might exert its strong anticancer effect.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Colágeno Tipo IV/farmacología , Células Endoteliales/efectos de los fármacos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Mitocondrias/efectos de los fármacos , Receptores de Vitronectina/metabolismo , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Caspasas/metabolismo , Línea Celular Tumoral , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Células Endoteliales/enzimología , Humanos , Isoenzimas , Ratones , Mitocondrias/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Albúmina Sérica/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 64(21): 8045-51, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520214

RESUMEN

Radioiodine therapy of nonthyroid cancers after sodium iodide symporter (NIS) gene delivery has been proposed as a potential application of gene therapy. However, it seems to be precluded by the rapid efflux of taken up iodine from most transduced xenografted tumors. We present an in vivo kinetic study of NIS-related hepatic iodine uptake in an aggressive model of hepatocarcinoma induced by diethylnitrosamine in immunocompetent Wistar rats. We followed the whole-body iodine distribution by repeated imaging of live animals. We constructed a rat NIS (rNIS) adenoviral vector, Ad-CMV-rNIS, using the cytomegalovirus (CMV) as a promoter. Injected in the portal vein in 5 healthy and 25 hepatocarcinoma-bearing rats and liver tumors in 9 hepatocarcinoma-bearing rats, Ad-CMV-rNIS drove expression of a functional NIS protein by hepatocytes and allowed marked (from 20 to 30% of the injected dose) and sustained (>11 days) iodine uptake. This contrasts with the massive iodine efflux found in vitro in human hepatic tumor cell lines. In vivo specific inhibition of NIS by sodium perchlorate led to a rapid iodine efflux from the liver, indicating that the sustained uptake was not attributable to an active retention mechanism but to permanent recycling of the effluent radioiodine via the high hepatic blood flow. Radioiodine therapy after Ad-CMV-rNIS administration achieved a strong inhibition of tumor growth, the complete regression of small nodules, and prolonged survival of hepatocarcinoma-bearing rats. This demonstrates for the first time the efficacy of NIS-based radiotherapy in a relevant preclinical model of nonthyroid human carcinogenesis.


Asunto(s)
Terapia Genética , Radioisótopos de Yodo/uso terapéutico , Neoplasias Hepáticas Experimentales/radioterapia , Simportadores/genética , Animales , Radioisótopos de Yodo/farmacocinética , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratas , Ratas Wistar
18.
Invest Radiol ; 39(6): 350-6, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15167101

RESUMEN

RATIONALE AND OBJECTIVES: At present, the gold standard to evaluate tumor necrosis is histology. We described here a new method to quantify the degree of tumor necrosis by ultrasonography. This technique combines ultrasound exploration of tissue and post-treatment of the numerical sequences using a dedicated software to evaluate backscattered power within the tumor. MATERIALS AND METHODS: In order to establish that the backscattered power could be considered as a relevant marker of tumor necrosis, we performed (1) intra- and interoperator reproducibility in estimation of tumor dimensions obtained on sonographic scans; and (2) intra- and interoperator reproducibility in quantification of backscattered power in postprocessing using the HDILab software. The third part of the study consisted of correlating the degree of tumor necrosis estimated by histology and the ultrasound backscattered power, both obtained on xenografted melanomas at different days after tumor transplantation. RESULTS: Results concerning tumor size estimations and quantification of echogenicity were reproducible (coefficient of variation < 4.33%). The degree of necrosis measured in histology and echogenicity were significantly negatively correlated (P < 0.003). CONCLUSION: In conclusion, backscattered power could be considered as a relevant parameter to quantify tumor necrosis in vivo.


Asunto(s)
Necrosis , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/patología , Animales , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Neuroblastoma/diagnóstico por imagen , Neuroblastoma/patología , Reproducibilidad de los Resultados , Programas Informáticos , Ultrasonografía
19.
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
20.
Stem Cells ; 22(1): 74-85, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14688393

RESUMEN

Currently available murine models to evaluate mesenchymal stem cell (MSC) differentiation are based on cell injection at ectopic sites such as muscle or skin. Due to the importance of environmental factors on the differentiation capacities of stem cells in vivo, we investigated whether the peculiar synovial/cartilaginous environment may influence the lineage specificity of bone morphogenetic protein (BMP)-2-engineered MSCs. To this aim, we used the C3H10T1/2-derived C9 MSCs that express BMP-2 under control of the doxycycline (Dox)-repressible promoter, Tet-Off, and showed in vitro, using the micropellet culture system that C9 MSCs kept their potential to differentiate toward chondrocytes. Implantation of C9 cells, either into the tibialis anterior muscles or into the joints of CB17-severe combined immunodeficient bg mice led to the formation of cartilage and bone filled with bone marrow as soon as day 10. However, no differentiation was observed after injection of naïve MSCs or C9 cells that were repressed to secrete BMP-2 by Dox addition. The BMP-2-induced differentiation of adult MSCs is thus independent of soluble factors present in the local environment of the synovial/cartilaginous tissues. Importantly, we demonstrated that a short-term expression of the BMP-2 growth factor is necessary and sufficient to irreversibly induce bone formation, suggesting that a stable genetic modification of MSCs is not required for stem cell-based bone/cartilage engineering.


Asunto(s)
Proteínas Morfogenéticas Óseas/biosíntesis , Cartílago/citología , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Factor de Crecimiento Transformador beta , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/genética , Huesos/citología , Huesos/embriología , Huesos/metabolismo , Cartílago/embriología , Cartílago/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular/genética , Linaje de la Célula/genética , Condrocitos/citología , Condrocitos/metabolismo , Líquido Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Sustancias de Crecimiento/metabolismo , Articulaciones/citología , Articulaciones/crecimiento & desarrollo , Articulaciones/cirugía , Ratones , Ratones Endogámicos C3H , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/cirugía , Células 3T3 NIH , Osteogénesis/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA