Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(36): E3425-34, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959876

RESUMEN

Vision loss from ischemic retinopathies commonly results from the accumulation of fluid in the inner retina [macular edema (ME)]. Although the precise events that lead to the development of ME remain under debate, growing evidence supports a role for an ischemia-induced hyperpermeability state regulated, in part, by VEGF. Monthly treatment with anti-VEGF therapies is effective for the treatment of ME but results in a major improvement in vision in a minority of patients, underscoring the need to identify additional therapeutic targets. Using the oxygen-induced retinopathy mouse model for ischemic retinopathy, we provide evidence showing that hypoxic Müller cells promote vascular permeability by stabilizing hypoxia-inducible factor-1α (HIF-1α) and secreting angiogenic cytokines. Blocking HIF-1α translation with digoxin inhibits the promotion of endothelial cell permeability in vitro and retinal edema in vivo. Interestingly, Müller cells require HIF--but not VEGF--to promote vascular permeability, suggesting that other HIF-dependent factors may contribute to the development of ME. Using gene expression analysis, we identify angiopoietin-like 4 (ANGPTL4) as a cytokine up-regulated by HIF-1 in hypoxic Müller cells in vitro and the ischemic inner retina in vivo. ANGPTL4 is critical and sufficient to promote vessel permeability by hypoxic Müller cells. Immunohistochemical analysis of retinal tissue from patients with diabetic eye disease shows that HIF-1α and ANGPTL4 localize to ischemic Müller cells. Our results suggest that ANGPTL4 may play an important role in promoting vessel permeability in ischemic retinopathies and could be an important target for the treatment of ME.


Asunto(s)
Angiopoyetinas/metabolismo , Permeabilidad Capilar , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuronas Retinianas/metabolismo , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Animales , Western Blotting , Hipoxia de la Célula , Células Cultivadas , Retinopatía Diabética/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunohistoquímica , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Neuronas Retinianas/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cancer Cell ; 10(2): 133-43, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16904612

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV), the infectious causative agent of Kaposi's sarcoma (KS), encodes a G protein-coupled receptor (vGPCR) implicated in the initiation of KS. Here we demonstrate that Kaposi's sarcomagenesis involves stimulation of tuberin (TSC2) phosphorylation by vGPCR, promoting the activation of mTOR through both direct and paracrine mechanisms. Pharmacologic inhibition of mTOR with rapamycin prevented vGPCR sarcomagenesis, while overactivation of this pathway was sufficient to render endothelial cells oncogenic. Moreover, mice haploinsufficient for TSC2 are predisposed to vascular sarcomas remarkably similar to KS. Collectively, these results implicate mTOR in KS initiation and suggest that the sarcomagenic potential of KSHV may be a direct consequence of the profound sensitivity of endothelial cells to vGPCR dysregulation of the TSC2/mTOR pathway.


Asunto(s)
Transformación Celular Neoplásica , Células Endoteliales/patología , Herpesvirus Humano 8/genética , Proteínas Quinasas/metabolismo , Receptores de Quimiocina/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Virales/fisiología , Animales , Línea Celular , Proliferación Celular , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/metabolismo , Comunicación Paracrina , Fosforilación , Proteínas Quinasas/genética , Receptores de Quimiocina/genética , Sarcoma de Kaposi/patología , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Virales/genética
3.
Proc Natl Acad Sci U S A ; 107(32): 14363-8, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660728

RESUMEN

Kaposi's sarcoma (KS) is an enigmatic vascular tumor thought to be a consequence of dysregulated expression of the human herpesvirus-8 (HHV-8 or KSHV)-encoded G protein-coupled receptor (vGPCR). Indeed, transgenic animals expressing vGPCR manifest vascular tumors histologically identical to human KS, with expression of the viral receptor limited to a few cells, suggestive of a paracrine mechanism for vGPCR tumorigenesis. Both human and vGPCR experimental KS lesions are characterized by prominent angiogenesis and vascular permeability attributed to the release of angiogenic molecules, most notably vascular endothelial growth factor. However, the relative contribution of these paracrine mediators to the angiogenic and exudative phenotype of KS lesions remains unclear. Here we show that vGPCR up-regulation of Angiopoietin-like 4 (ANGPTL4) plays a prominent role in promoting the angiogenesis and vessel permeability observed in KS. Indeed, ANGPTL4 expression is a hallmark of vGPCR experimental and human KS lesions. Inhibition of ANGPTL4 effectively blocks vGPCR promotion of the angiogenic switch and vascular leakage in vitro and tumorigenesis in vivo. These observations suggest that ANGPTL4 is a previously unrecognized target for the treatment of patients with KS. As angiogenesis and increased vessel permeability are common themes in all solid tumors, these findings may have a broad impact on our understanding and treatment of cancer.


Asunto(s)
Angiopoyetinas/biosíntesis , Permeabilidad Capilar , Neovascularización Patológica , Receptores de Quimiocina/fisiología , Sarcoma de Kaposi/fisiopatología , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Comunicación Paracrina , Factor A de Crecimiento Endotelial Vascular
4.
Cancer Immunol Immunother ; 61(11): 1977-87, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22527247

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most aggressive carcinomas. Limited therapeutic options, mainly due to a fragmented genetic understanding of HCC, and major HCC resistance to conventional chemotherapy are the key reasons for a poor prognosis. Thus, new effective treatments are urgent and gene therapy may be a novel option. Signal transducer and activator of transcription 3 (Stat3) is a highly studied member of the STAT family. Inhibition of Stat3 signaling has been found to suppress tumor growth and improve survival, providing a molecular target for cancer therapy. Furthermore, HCC is a hypervascular tumor and angiogenesis plays a crucial role in tumor growth and metastasis. Thus, anti-angiogenic therapy, combined with inhibition of Stat3, may be an effective approach to combat HCC. We tested the effect that the combination therapy consisting of endostatin (a powerful angiogenesis inhibitor) and Stat3-specific small interfering RNA, using a DNA vector delivered by attenuated S. typhimurium, on an orthotopic HCC model in C57BL/6 mice. Although antitumor effects were observed with either single therapeutic treatment, the combination therapy provided superior antitumor effects. Correlated with this finding, the combination treatment resulted in significant alteration of Stat3 and endostatin levels and that of the downstream gene VEGF, decreased cell proliferation, induced cell apoptosis and inhibited angiogenesis. Importantly, combined treatment also elicited immune system regulation of various immune cells and cytokines. This study has provided a novel cancer gene therapeutic approach.


Asunto(s)
Carcinoma Hepatocelular/terapia , Endostatinas/genética , Terapia Genética/métodos , Neoplasias Hepáticas Experimentales/terapia , Neoplasias Hepáticas/terapia , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Salmonella typhimurium , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Silenciador del Gen , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/terapia , Factor de Transcripción STAT3/genética , Factor A de Crecimiento Endotelial Vascular/análisis
5.
J Pharmacol Exp Ther ; 338(1): 173-83, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21444629

RESUMEN

Our objective was to evaluate cell growth and death effects by inhibiting Murine Double Minute 2 (MDM2) expression in human prostate cancer cells overexpressing the wild-type (WT) p53 gene. Prostate PC-3 tumor cells were transfected with a plasmid containing either mdm2 small interfering (Si-mdm2) or the WT p53 gene (Pp53) alone, or both (Pmp53), using Lipofectamine in vitro and attenuated Salmonella enterica serovar Typhi vaccine strain Ty21a (Salmonella Typhi Ty21a) in vivo. Cell growth, apoptosis, and the expression of related genes and proteins were examined in vitro and in vivo by flow cytometry and Western blot assays. We demonstrated that human prostate tumors had increased expression of MDM2 and mutant p53 proteins. Transfection of the PC-3 cells with the Pmp53 plasmid in vitro offered significant inhibition of cell growth and an increase in apoptotic cell death compared with that of the Si-mdm2 or Pp53 group. These effects were associated with up-regulation of p21 and down-regulation of hypoxia-inducible factor 1α expression in Pmp53-transfected cells. To validate the in vitro findings, the nude mice implanted with PC-3 cells were treated with attenuated Salmonella Typhi Ty21a carrying the plasmids, which showed that the Pmp53 plasmid significantly inhibited the tumor growth rate in vivo compared with that of the Si-mdm2 or Pp53 plasmid alone. Tumor tissues from mice treated with the Pmp53 plasmid showed increased expression of p21 and decreased expression of hypoxia-inducible factor 1α proteins, with an increased apoptotic effect. These results suggest that knockdown of mdm2 expression by its specific small interfering RNA with overexpression of the WT p53 gene offers synergistic inhibition of prostate cancer cell growth in vitro and in vivo.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes p53/fisiología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Clin Cancer Res ; 14(2): 559-68, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18223232

RESUMEN

PURPOSE: Persistent activation of signal transducers and activators of transcription 3 (Stat3) and its overexpression contribute to the progression and metastasis of several different tumor types. For this reason, Stat3 is a reasonable target for RNA interference-mediated growth inhibition. Blockade of Stat3 using specific short hairpin RNAs (shRNA) can significantly reduce prostate tumor growth in mice. However, RNA interference does not fully ablate target gene expression in vivo, owing to the idiosyncrasies associated with shRNAs and their targets. To enhance the therapeutic efficacy of Stat3-specific shRNA, we applied a combination treatment involving gene associated with retinoid-IFN-induced mortality 19 (GRIM-19), another inhibitor of STAT3, along with shRNA. EXPERIMENTAL DESIGN: The coding sequences for GRIM-19, a cellular STAT3-specific inhibitor, and Stat3-specific shRNAs were used to create a dual expression plasmid vector and used for prostate cancer therapy in vitro and in mouse xenograft models in vivo. RESULTS: The coexpressed Stat3-specific shRNA and GRIM-19 synergistically and more effectively suppressed prostate tumor growth and metastases when compared with treatment with either single agent alone. CONCLUSION: The simultaneous use of two specific, but mechanistically different, inhibitors of STAT3 activity exerts enhanced antitumor effects.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , NADH NADPH Oxidorreductasas/genética , Plásmidos , Próstata/metabolismo , Neoplasias de la Próstata/genética , Factor de Transcripción STAT3/genética , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 67(13): 6212-20, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616678

RESUMEN

Signal transducers and activators of transcription 3 (STAT3) was originally identified as a transcription factor that mediates cytokine-induced responses. In these pathways, Janus-activated kinase (JAK)-induced transient tyrosine phosphorylation of STAT3 promotes gene expression in response to a number of cytokines, which is inhibited by feedback mechanisms. A number of studies have shown that STAT3 is constitutively activated in human cancer cells, leading to cell proliferation. It is unclear, apart from a chronic tyrosyl phosphorylation of STAT3, what mechanisms contribute to the STAT3 deregulation in tumors. Earlier, we have isolated a novel growth inhibitory gene product, gene associated with retinoid-IFN-induced mortality 19 (GRIM-19), using a genetic approach. GRIM-19 is an IFN/retinoic acid-regulated growth suppressor. Subsequent analyses have shown that GRIM-19 binds to STAT3 and prevents interleukin-6-induced transcription of cellular genes. However, its effects on a constitutively active STAT3 and cellular transformation are unknown. In this study, we show that GRIM-19 suppresses constitutive STAT3-induced cellular transformation in vitro and in vivo by down-regulating the expression of a number of cellular genes involved in cell proliferation and apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Regulación Neoplásica de la Expresión Génica , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/fisiología , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Muerte Celular , Proliferación Celular , Transformación Celular Neoplásica , Humanos , Interleucina-6/metabolismo , Ratones , Fosforilación , Ratas , Transcripción Genética , Tretinoina/metabolismo
8.
Cancer Res ; 67(12): 5859-64, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575154

RESUMEN

The facultative anaerobic, invasive Salmonella enterica serovar typhimurium (S. typhimurium) has been shown to retard the growth of established tumors. We wondered if a more effective antitumor response could be achieved in vivo if these bacteria were used as tools for delivering specific molecular antitumor therapeutics. Constitutively activated transcription factor signal transducer and activator of transcription 3 (STAT3) promotes the survival of a number of human tumors. In this study, we investigated the relative efficacies of attenuated S. typhimurium alone or combined with Stat3-specific small interfering RNA (siRNA) in terms of tumor growth and metastasis. The bacteria preferentially homed into tumors over normal liver and spleen tissues in vivo. S. typhimurium expressing plasmid-based Stat3-specific siRNAs significantly inhibited tumor growth, reduced the number of metastastic organs, and extended the life time for C57BL6 mice bearing an implanted prostate tumor, versus bacterial treatment alone. These results suggest that attenuated S. typhimurium combined with an RNA interference approach might be more effective for the treatment of primary as well as metastatic cancer.


Asunto(s)
Neoplasias de la Próstata/terapia , ARN Interferente Pequeño , Factor de Transcripción STAT3/genética , Salmonella typhimurium , Animales , Apoptosis , Northern Blotting , Western Blotting , Proliferación Celular , Terapia Genética/métodos , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos , Neoplasias de la Próstata/microbiología , Interferencia de ARN
9.
Cancer Res ; 66(1): 168-74, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16397229

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a G protein-coupled receptor (vGPCR) that has been implicated in the initiation of Kaposi's sarcoma, identifying vGPCR as an attractive target for preventing Kaposi's sarcoma. However, as only a fraction of cells in advanced Kaposi's sarcoma lesions express vGPCR, it is unclear whether this unique viral oncogene contributes to Kaposi's sarcoma progression. We therefore set out to determine whether the few cells that express vGPCR in established tumors represent an appropriate therapeutic target for the treatment of patients with preexisting Kaposi's sarcoma. To this end, we generated endothelial cell lines stably expressing vGPCR or key KSHV latently expressed proteins (vCyclin, vFlip, and LANA1). The endothelial cell line expressing vGPCR was rendered sensitive to treatment with the nucleoside analogue ganciclovir by using a bicistronic construct coexpressing the herpes simplex virus 1 thymidine kinase. S.c. injection into nude mice with mixed-cell populations formed tumors that approximate the ratio of vGPCR-expressing and KSHV latent gene-expressing cells. These mice were then treated with ganciclovir to specifically target only the vGPCR-expressing cells. Surprisingly, despite the expression of KSHV latent genes in the vast majority of tumor cells, specifically targeting only the few vGPCR-expressing cells in established tumors resulted in tumor regression. Moreover, we observed an increase in apoptosis of latent gene-expressing cells after the pharmacologic deletion of the vGPCR-expressing cells. These findings indicate that vGPCR may play a key role in Kaposi's sarcoma progression and provide experimental justification for developing molecular-based therapies specifically targeting vGPCR and its effectors for the treatment of Kaposi's sarcoma patients.


Asunto(s)
Herpesvirus Humano 8/fisiología , Receptores Acoplados a Proteínas G/fisiología , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/terapia , Animales , Transformación Celular Viral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Femenino , Ganciclovir/farmacología , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Ratones , Ratones Desnudos , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virología , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Transfección
10.
J Interferon Cytokine Res ; 38(9): 378-387, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30230984

RESUMEN

Melanoma is one of the most lethal skin malignancies in the world. Interferons (IFNs) have been also demonstrated in response to tumor cell and IFNs such as IFN-α have been used for melanoma treatment. The long chain double-stranded RNA (dsRNA) (from a variety of nonviral sources) is a potent activator of the IFN system and an inducer of cell apoptosis. Panaxadiolsaponins (PDS) is a major Panax ginseng-derived active component with known antitumor activity and immune modulation. Here, we investigated a hypothesis that the combination of PDS and total natural dsRNA (as opposed to the synthetic dsRNA) will suppress tumor growth better than the individual agents. We have evaluated the antitumor and immunostimulatory effects of the combination of natural long chain dsRNA (derived from yeast) and PDS on melanoma cell line B16 and mice xenograft model. The underlying mechanisms of growth suppression were investigated by analyzing dsRNA-activated pathways, apoptosis, and cell cycle. Natural dsRNA and PDS exert superior anticancer effects than either agent alone. Natural dsRNA and PDS combination might be a promising strategy for treating malignancies, including melanoma.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Melanoma Experimental/tratamiento farmacológico , ARN Bicatenario/farmacología , Sapogeninas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Melanoma Experimental/patología , Ratones , Panax/química , Células Tumorales Cultivadas
11.
Clin Cancer Res ; 11(17): 6333-41, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16144938

RESUMEN

PURPOSE: Signal transducer and activator of transcription 3 (Stat3) is constitutively activated in a variety of cancers and it is a common feature of prostate cancer. Thus, Stat3 represents a promising molecular target for tumor therapy. We applied a DNA vector-based Stat3-specific RNA interference approach to block Stat3 signaling and to evaluate the biological consequences of Stat3 down-modulation on tumor growth using a mouse model. EXPERIMENTAL DESIGN: To investigate the therapeutic potential of blocking Stat3 in cancer cells, three small interfering RNAs (siRNA; Stat3-1, Stat3-2, and Stat3-3) specific for different target sites on Stat3 mRNA were designed and used with a DNA vector-based RNA interference approach expressing short hairpin RNAs to knockdown Stat3 expression in human prostate cancer cells in vitro as well as in vivo. RESULTS: Of the three equivalently expressed siRNAs, only Stat3-3 and Stat3-2, which target the region coding for the SH2 domain and the coiled-coil domain, respectively, strongly suppressed the expression of Stat3 in PC3 and LNCaP cells. The Stat3-1 siRNA, which targeted the DNA-binding domain, exerted no effect on Stat3 expression, indicating that the gene silencing efficiency of siRNA may be dependent on the local structure of Stat3 mRNA. The Stat3 siRNAs down-regulated the expression of Bcl-2 (an anti-apoptotic protein), and cyclin D1 and c-Myc (cell growth activators) in prostate cancer cells. Inhibition of Stat3 and its related genes was accompanied by growth suppression and induction of apoptosis in cancer cells in vitro and in tumors implanted in nude mice. CONCLUSIONS: These data indicate that Stat3 signaling is a promising molecular target for prostate cancer therapy and that vector-based Stat3 siRNA may be useful as a therapeutic agent for treatment of prostate cancer.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Vectores Genéticos , Neoplasias de la Próstata/prevención & control , ARN Interferente Pequeño/farmacología , Transactivadores/metabolismo , Animales , Apoptosis , Emparejamiento Base , Secuencia de Bases , Ciclo Celular , Proliferación Celular , Ciclina D1/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3 , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Células Tumorales Cultivadas
12.
Oncogene ; 21(12): 1882-9, 2002 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-11896621

RESUMEN

We recently identified inositol hexakisphosphate kinase 2 (IP6K2) as a positive regulator of apoptosis. Overexpression of IP6K2 enhances apoptosis induced by interferon-beta (IFN-beta) and cytotoxic agents in NIH-OVCAR-3 ovarian carcinoma cells. In this study, we contrast and compare IFN-beta and radiation-induced death, and show that IP6K2 expression sensitizes tumor cells. Unirradiated NIH-OVCAR-3 cells transfected with IP6K2 formed fewer colonies compared to unirradiated vector-expressing cells. IP6K2 overexpression caused increased radiosensitivity, evidenced by decreased colony forming units (CFU). Both IFN-beta and radiation induced caspase 8. IFN-beta, but not gamma-irradiation, induced TRAIL in NIH-OVCAR-3 cells. Gamma irradiation, but not IFN-beta, induced DR4 mRNA. Apoptotic effects of IFN-beta or gamma-irradiation were blocked by expression of a dominant negative mutant death receptor 5 (DR5Delta) or by Bcl-2. Caspase-8 mRNA induction was more pronounced in IP6K2-expressing cells compared to vector-expressing cells. These data suggest that overexpression of IP6K2 enhances sensitivity of some ovarian carcinomas to radiation and IFN-beta. IP6K2 may function to enhance the expression and/or function of caspase 8 and DR4 following cell injury. Both IFN-beta and gamma-irradiation induce apoptosis through the extrinsic, receptor-mediated pathway, IFN-beta through TRAIL, radiation through DR4, and both through caspase 8. The function of both death inducers is positively regulated by IP6K2.


Asunto(s)
Neoplasias Ováricas/enzimología , Fosfotransferasas (Aceptor del Grupo Fosfato)/fisiología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Caspasa 8 , Caspasa 9 , Caspasas/genética , Caspasas/metabolismo , Femenino , Rayos gamma , Regulación Enzimológica de la Expresión Génica , Humanos , Interferón beta/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/radioterapia , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , ARN Mensajero/metabolismo , Fármacos Sensibilizantes a Radiaciones , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Ribonucleasas/genética , Ribonucleasas/metabolismo , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación , Ensayo de Tumor de Célula Madre
13.
J Interferon Cytokine Res ; 25(11): 666-73, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16318580

RESUMEN

Interferon-gamma (IFN-gamma) regulates a number of cellular genes using a variety of cellular signaling pathways. Previously, we identified a novel IFN-regulated element, IFN-gamma-activated transcriptional element (GATE), in the promoter of the murine IFN regulatory factor-9 (IRF-9) gene. This element binds to novel factors. We have recently characterized a novel regulatory factor, GATE binding factor 1 (GBF1), which promoted IFN-gamma-induced transcription. Although GBF1 was a potent inducer of transcription, it did not bind to DNA well in vitro. To understand its role in IFN-gamma-induced actions, we raised monoclonal antibodies (mAb) against GBF1. These antibodies are highly useful in Western, immunoprecipitation, and immunocytochemical analyses. Employing these antibodies, we show that GBF1 is recruited to the endogenous IRF-9 promoter. We also show GBF1 interacts with CAAAT/enhancer binding protein-beta (C/EBP-beta), the other GATE binding factor. Furthermore, other cytokines, such as interleukin-1 (IL-1) and IL-6, induced the expression of GBF1. These antibodies may be useful tools for investigating the role of GBF1 in cytokine-induced responses.


Asunto(s)
Anticuerpos Monoclonales/química , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Factores de Intercambio de Guanina Nucleótido/química , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Interferón gamma/metabolismo , Interleucina-1/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Citocinas/metabolismo , ADN/química , Ensayo de Inmunoadsorción Enzimática , Factores de Intercambio de Guanina Nucleótido/inmunología , Células HeLa , Humanos , Inmunohistoquímica , Inmunoprecipitación , Interferones/metabolismo , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Recombinantes/química , Transducción de Señal , Factores de Tiempo , Transcripción Genética
14.
Clin Cancer Res ; 8(10): 3210-8, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12374691

RESUMEN

The IFN and retinoic acid (RA) combination suppresses cell growth by inducing apoptosis in the cultured tumor cells. Using a genetic technique, we have isolated several "genes associated with retinoid-IFN-induced mortality" (GRIM) that participate in this death pathway. One such gene, GRIM-12, encodes the redox enzyme thioredoxin reductase (TR). Antisense-mediated inhibition of TR abrogates cell death. To test the in vivo relevance of TR for growth suppression, we have conducted the following study. A wild-type TR or a catalytically defective mutant were expressed in MCF-7 breast carcinoma cells and transplanted into athymic nude mice. These mice were treated with IFN-beta and all-trans RA combination. Tumors expressing the vector or wild-type TR were readily suppressed by the IFN/RA combination. In contrast, the tumors bearing a mutant TR were resistant to regression. We further show that markers of apoptosis are stimulated in the regressing tumors. These studies show a prominent role for TR in tumor-growth suppression.


Asunto(s)
Antineoplásicos/farmacología , Interferón beta/farmacología , Neoplasias Mamarias Experimentales/enzimología , Reductasa de Tiorredoxina-Disulfuro/fisiología , Tretinoina/farmacología , Animales , Anexina A5/metabolismo , Apoptosis , Western Blotting , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Regulación hacia Abajo , Activación Enzimática , Femenino , Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Trasplante de Neoplasias , Transfección , Células Tumorales Cultivadas/efectos de los fármacos
15.
J Interferon Cytokine Res ; 22(10): 1017-26, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12433281

RESUMEN

A combination of interferon-beta (IFN-beta) and all-trans retinoic acid (IFN/RA) induces tumor cell apoptosis via some unknown mechanisms. Apoptosis is a gene-directed process that limits the proliferation of undesired cells. Several genes are required to regulate cell death in the higher-order animals. Earlier, we employed a gene expression knockout technique to isolate cell death-related genes. A novel gene, the gene associated with retinoid-interferon-induced mortality-19 (GRIM-19), was found to be essential for tumor cell death induced by IFN/RA. Here, we describe the development and characterization of three monoclonal antibodies (mAbs) against GRIM-19. GRIM-19 is present in the nucleus and cytoplasm. Its expression is induced by the IFN/RA combination. We also show that GRIM-19 inhibits the cell-transforming property of viral oncogenic protein viral IFN regulatory factor-1 (vIRF-1) via a physical interaction. mAbs developed in this study should be useful for studying the other physiologic roles of GRIM-19 and serve as a potent tool for studying tumor responses to IFN/RA therapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Apoptosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Interferón beta/farmacología , NADH NADPH Oxidorreductasas/inmunología , Células 3T3 , Animales , Proteínas Reguladoras de la Apoptosis , Humanos , Factores Inmunológicos/farmacología , Ratones , Ratones Mutantes , NADH NADPH Oxidorreductasas/metabolismo , Proteínas Nucleares/inmunología , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Tretinoina/farmacología , Células Tumorales Cultivadas
16.
Cancer Lett ; 335(1): 242-50, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23435374

RESUMEN

The E6 protein of the oncogenic HPV-16 functions by interfering with the normal cell cycle control mechanisms, particularly those controlled by p53. In this study, we developed a dual expression plasmid that coexpressed-E6-specific siRNA and wild type p53, and to evaluate its effects on cervical cancer growth. We found that simultaneous expression of pSi-E6-P53 caused a robust suppression of tumor growth when compared to the controls either E6-specific siRNA or p53 alone. In conclusion, our findings demonstrate that a combined strategy of co-expressed E6-specific siRNA and p53 synergistically and more effectively suppressed cervical tumor growth when compared with single treatment.


Asunto(s)
Proliferación Celular , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/terapia , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/terapia , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Plásmidos/genética , Proteínas Represoras/metabolismo , Salmonella typhimurium/genética , Transformación Bacteriana , Carga Tumoral , Proteína p53 Supresora de Tumor/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
17.
J Cancer Res Clin Oncol ; 139(6): 971-80, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23463096

RESUMEN

OBJECTIVES: To investigate the therapeutic utility of an attenuated bacterium carrying a plasmid that co-expresses Endostatin, an inhibitor of tumor neovasculogenesis, and a shRNA that targets Stat3 to suppress prostate cancer growth. METHODS: Plasmid pEndo-Si-Stat3 was constructed and introduced into an attenuated strain of Salmonella enterica serovar typhimurium. The resultant recombinant bacterium was used as a vector to deliver the plasmid to tumor cells growing in vivo. Tumor-associated gene and protein expression changes were measured by using RT-PCR and Western blot analyses. Expression of Endostatin in tumor tissue was detected by ELISA. The presence of vector bacteria in tissues was monitored and tumor destruction was assessed by using TUNEL and H&E staining assays. RESULTS: Bacterially delivered pEndo-Si-Stat3 decreased Stat3 levels and increased Endostatin expression in mouse tumors, resulting in a significant suppression of tumor growth (P < 0.01). Expression of Bcl-2 and PCNA was down-regulated and Caspase3 expression was up-regulated to promote apoptosis of tumor cells. CONCLUSIONS: Successful delivery by attenuated Salmonella of the combination therapeutic plasmid simultaneously knocked down the expression of Stat3 and resulted in over-expression of Endostatin, which synergistically inhibited prostate cancer growth.


Asunto(s)
Endostatinas/genética , Técnicas de Transferencia de Gen , Neoplasias de la Próstata/terapia , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Animales , Antígenos CD34/metabolismo , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Terapia Combinada , Endostatinas/metabolismo , Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Salmonella typhimurium/genética , Factores de Tiempo , Carga Tumoral/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
PLoS One ; 6(4): e19103, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559457

RESUMEN

BACKGROUND: Kaposi's sarcoma (KS) is a vascular neoplasm characterized by the dysregulated expression of angiogenic and inflammatory cytokines. The driving force of the KS lesion, the KSHV-infected spindle cell, secretes elevated levels of vascular endothelial growth factor (VEGF), essential for KS development. However, the origin of VEGF in this tumor remains unclear. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that the KSHV G protein-coupled receptor (vGPCR) upregulates VEGF in KS through an intricate paracrine mechanism. The cytokines secreted by the few vGPCR-expressing tumor cells activate in neighboring cells multiple pathways (including AKT, ERK, p38 and IKKß) that, in turn, converge on TSC1/2, promoting mTOR activation, HIF upregulation, and VEGF secretion. Conditioned media from vGPCR-expressing cells lead to an mTOR-dependent increase in HIF-1α and HIF-2α protein levels and VEGF upregulation. In a mouse allograft model for KS, specific inhibition of the paracrine activation of mTOR in non-vGPCR-expressing cells was sufficient to inhibit HIF upregulation in these cells, and abolished the ability of the vGPCR-expressing cells to promote tumor formation in vivo. Similarly, pharmacologic inhibition of HIF in this model blocked VEGF secretion and also lead to tumor regression. CONCLUSIONS/SIGNIFICANCE: Our findings provide a compelling explanation for how the few tumor cells expressing vGPCR can contribute to the dramatic amplification of VEGF secretion in KS, and further provide a molecular mechanism for how cytokine dysregulation in KS fuels angiogenesis and tumor development. These data further suggest that activation of HIF by vGPCR may be a vulnerable target for the treatment of patients with KS.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica , Receptores Acoplados a Proteínas G/metabolismo , Sarcoma de Kaposi/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Citocinas/metabolismo , Células Endoteliales/citología , Femenino , Humanos , Inflamación , Ratones , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Oral Oncol ; 47(5): 371-5, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21421336

RESUMEN

Kaposi's sarcoma (KS) remains among the most common causes of oral cancer in HIV-infected individuals. Infection with the KS-associated herpesvirus (KSHV/HHV8) is a necessary event for disease development. Emerging evidence suggests that KSHV infects vascular endothelial (or endothelial progenitor) cells promoting the formation of the KS tumor (or spindle) cell. These cells elaborate angiogenic growth factors and cytokines that promote the dysregulated angiogenesis and profuse edema that characterizes this unusual vascular tumor. Central among these secreted factors is the potent endothelial cell mitogen, vascular endothelial growth factor (VEGF). Indeed, VEGF has proven to be a key player in KSHV pathogenesis and is a molecular hallmark of KS lesions. We have recently shown that a second angiogenic factor, Angiopoietin-like 4 (ANGPTL4), may also play a critical role in KS development. Here we demonstrate that ANGPTL4 is upregulated both directly and indirectly by the KSHV oncogene, vGPCR. We further show that ANGPTL4 is a molecular hallmark of oral KS lesions. Indeed, expression of this protein was observed in more tumor cells and in more biopsies specimens than expression of VEGF (23/25 or 92% vs. 19/25 or 76%, respectively) in oral KS. These surprising results support a key role for ANGPTL4 in Kaposi's sarcomagenesis and further suggest that this angiogenic factor may provide a novel diagnostic and therapeutic marker for oral KS patients.


Asunto(s)
Angiopoyetinas/metabolismo , Herpesvirus Humano 8/metabolismo , Neoplasias de la Boca/metabolismo , Sarcoma de Kaposi/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Inductores de la Angiogénesis , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Línea Celular Tumoral , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/virología , Femenino , Herpesvirus Humano 8/genética , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/patología , Neoplasias de la Boca/virología , Proteínas de Neoplasias/metabolismo , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética
20.
Cancer Res ; 68(20): 8361-8, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922908

RESUMEN

Rapamycin (or sirolimus), the prototypical inhibitor of the mammalian target of rapamycin (mTOR) and an immunosuppressant used for the prevention of renal transplant rejection, has recently emerged as an effective treatment for Kaposi's sarcoma (KS), an enigmatic vascular tumor and a model for pathologic angiogenesis. Indeed, recent work supports a role for mTOR as a central player in the transformation of endothelial cells by the KS-associated herpesvirus-encoded G protein-coupled receptor (vGPCR), the viral oncogene believed to be responsible for causing KS. However, emerging evidence that rapamycin may transiently promote the activation of Akt may limit its use as an anti-KS therapy. Here, we show that activation of Akt in endothelial cells expressing vGPCR is augmented by treatment with rapamycin, resulting in the up-regulation of several Akt proliferative and survival pathways. However, use of a novel dual phosphatidylinositol 3-kinase alpha (PI3Kalpha)/mTOR inhibitor, PI-103, effectively and independently blocked activation of both PI3K and mTOR in vGPCR-expressing endothelial cells. This resulted in more effective inhibition of endothelial cell proliferation and survival in vitro and tumor growth in vivo. Our results suggest that PI-103 may be an effective therapeutic option for the treatment of patients with KS. Moreover, as KS may serve as a model for pathologic angiogenesis, our results further provide the basis for the early assessment of PI-103 as an antiangiogenic chemotherapeutic.


Asunto(s)
Antineoplásicos/farmacología , Furanos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Quinasas/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Sarcoma de Kaposi/tratamiento farmacológico , Sirolimus/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Furanos/uso terapéutico , Inmunosupresores/farmacología , Ratones , Trasplante de Neoplasias , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Piridinas/uso terapéutico , Pirimidinas/uso terapéutico , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA