Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Res ; 66(24): 11869-77, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17178884

RESUMEN

Biochemical and genetic mutation-based analyses confirm that the MDA-7/IL-24 protein can induce transformed cell-specific apoptosis through a mechanism involving endoplasmic reticulum (ER) stress-associated pathways. Covalent modifications by N-linked glycans in the ER contribute to the conformational maturation and biological functions of many proteins. Because MDA-7/IL-24 is a glycosylated protein, we investigated the role of glycosylation in mediating the specific biological and "bystander" antitumor activities of this cytokine. An adenovirus vector expressing a nonsecreted and nonglycosylated version of MDA-7/IL-24 protein was generated via deletion of its signal peptide and point mutations of its three N-glycosylated sites. In this study, we showed that this intracellular nonglycosylated protein was as effective as wild-type MDA-7/IL-24 protein in inducing apoptosis in multiple tumor cell lines. Both constructs (a) displayed transformed cell specificity and localization to the ER compartment, (b) mediated apoptosis through JAK/STAT-independent and p38(MAPK)-dependent pathways, (c) induced sustained ER stress as evidenced by expression of ER stress markers (BiP/GRP78, GRP94, XBP-1, and eIF2alpha), and (d) generated proteins that physically interacted with BiP/GRP78. Additionally, an expression construct containing the mda-7/IL-24 signal peptide linked to the mutated nonglycosylated mda-7/IL-24 gene retained the ability to induce bystander antitumor activity. These studies reveal that MDA-7/IL-24 glycosylation is not mandatory for inducing cell death or bystander activities in different cancer cells, providing new insights into the mechanism by which MDA-7/IL-24 induces apoptosis and ER stress.


Asunto(s)
Apoptosis/fisiología , Interleucinas/genética , Interleucinas/metabolismo , Neoplasias de la Mama , Efecto Espectador , Muerte Celular , Línea Celular Tumoral , ADN Complementario/genética , Chaperón BiP del Retículo Endoplásmico , Femenino , Citometría de Flujo , Ingeniería Genética , Glicosilación , Hemo-Oxigenasa 1/metabolismo , Humanos
2.
Cancer Res ; 66(4): 2403-13, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16489047

RESUMEN

Pancreatic cancer is genetically complex, and without effective therapy. Mutations in the Kirsten-ras (K-ras) oncogene occur early and frequently (approximately 90%) during pancreatic cancer development and progression. In this context, K-ras represents a potential molecular target for the therapy of this highly aggressive cancer. We now show that a bipartite adenovirus expressing a novel cancer-specific apoptosis-inducing cytokine gene, mda-7/interleukin-24 (IL-24), and a K-ras AS gene, but not either gene alone, promotes growth suppression, induction of apoptosis, and suppression of tumor development mediated by K-ras mutant pancreatic cancer cells. Equally, the combination of an adenovirus expressing mda-7/IL-24 and pharmacologic and genetic agents simultaneously blocking K-ras or downstream extracellular regulated kinase 1/2 signaling also promotes similar inhibitory effects on the growth and survival of K-ras mutant pancreatic carcinoma cells. This activity correlates with the reversal of a translational block in mda-7/IL-24 mRNA in pancreatic cancer cells that limits message association with polysomes, thereby impeding translation into protein. Our study provides support for a "dual molecular targeted therapy" involving oncogene inhibition and selective cancer apoptosis-inducing gene expression with potential for effectively treating an invariably fatal cancer.


Asunto(s)
Genes ras , Terapia Genética/métodos , Interleucinas/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animales , Apoptosis/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cytokine Growth Factor Rev ; 14(1): 35-51, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12485618

RESUMEN

The melanoma differentiation-associated gene-7 (mda-7) was cloned by subtraction hybridization as a molecule whose expression is elevated in terminally differentiated human melanoma cells. Current information based on structural and sequence homology, has led to the recognition of MDA-7 as an IL-10 family cytokine member and its renaming as IL-24. Northern blot analysis revealed mda-7/IL-24 expression in human tissues associated with the immune system such as spleen, thymus, peripheral blood leukocytes and normal melanocytes. The MDA-7/IL-24 mouse counterpart, FISP, appears to be a Th2-specific protein and the rat counterpart, C49A/MOB-5, is associated with wound healing and is also induced as a consequence of ras-transformation. A notable property of MDA-7/IL-24 is its ability to induce apoptosis in a large spectrum of human cancer derived cell lines, in mouse xenografts and upon intratumoral injection in human tumors (phase I clinical trials). Various aspects of this intriguing molecule including its cytokine and anti-tumoral effects are described and discussed.


Asunto(s)
Antineoplásicos/metabolismo , Apoptosis/fisiología , Genes Supresores de Tumor , Interleucinas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucinas/genética , Melanoma/metabolismo , Melanoma/patología , Datos de Secuencia Molecular , Alineación de Secuencia
4.
Oncogene ; 24(4): 585-96, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15580305

RESUMEN

Pancreatic cancer is exceptionally aggressive with no long-term effective therapy. Current interventional approaches, including surgery, radiation and/or chemotherapy, have done little to quell the mortality associated with this malignancy. Subtraction hybridization identified a cancer-specific apoptosis-inducing cytokine gene, melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), with a broad range of selective antitumor activity in diverse cancers both in vitro and in vivo in nude mice and recently in patients with advanced carcinomas and melanomas. Unlike most neoplasms, pancreatic cancers display innate resistance to mda-7/IL-24-induced apoptosis, which correlates with a diminished capacity to convert mda-7/IL-24 mRNA into protein. We presently demonstrate that this translational block can be reversed by treatment with agents that elevate reactive oxygen species (ROS). Induction of apoptosis in vitro and suppression of tumorigenesis in vivo in nude mice are induced in pancreatic cancers, irrespective of the status of their K-ras gene, only when tumor cells simultaneously express mda-7/IL-24 and are treated with a ROS-inducer, such as arsenic trioxide (ARS), N-(4-hydroxyphenyl) retinamide (HPR) or dithiophene (NSC656240 (NSC)). In pancreatic cancer cells constitutively expressing mda-7/IL-24 mRNA, a single treatment with arsenic trioxide, HPR or NSC656240 induces apoptosis, which correlates with production of MDA-7/IL-24 protein. The specificity of this action is documented by the ability of ROS inhibitors, including N-acetyl-L-cysteine and Tiron, to block this killing effect. Of potential clinical significance, similar treatment of normal cells does not elicit significant changes in growth nor does it induce apoptosis. Analysis of signal transduction changes in pancreatic carcinoma cells infected with Ad.mda-7 in combination with a ROS-inducer indicate that cell death correlates with modulation of discrete cassettes of multiple signaling pathways in a pancreatic cancer cell-specific manner, supporting global signaling dysregulation as a potential mediator of apoptosis induction. These findings suggest a promising combinatorial approach for safely promoting cell death in pancreatic tumors that provides a rational framework for developing a selective and effective therapy for this invariably fatal cancer.


Asunto(s)
Apoptosis , Interleucinas/metabolismo , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Arseniatos/farmacología , Línea Celular Tumoral , Genes Supresores de Tumor , Humanos , Interleucinas/genética , Ratones , Ratones Desnudos , Mutación/genética , Trasplante de Neoplasias , Neoplasias Pancreáticas/genética , ARN Mensajero/genética , Transducción de Señal
5.
Cancer Res ; 64(9): 2988-93, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126330

RESUMEN

Melanoma differentiation associated gene-7/interleukin-24 (Mda-7/IL-24), a novel member of the IL-10 family of cytokines, uniquely displays cancer-specific apoptosis-inducing activity. Positive results in ongoing phase I/II clinical trials have strengthened the possibility of its utilization as a cancer gene therapeutic. Previous studies document that signaling events leading to Ad.mda-7-induced transformed cell apoptosis are tyrosine kinase-independent. These results suggest that mda-7/IL-24 cancer cell-specific activity could occur through mechanisms independent of binding to its currently recognized cognate receptors and might even occur independent of receptor function. An adenovirus vector expressing a nonsecreted version of MDA-7/IL-24 protein was generated via deletion of its signal peptide. This nonsecreted protein was as effective as wild-type secreted MDA-7/IL-24 in inducing apoptosis in prostate carcinoma cell lines and displayed transformed cell specificity and localization of MDA-7/IL-24 in the Golgi/endoplasmic reticulum compartments. Our results indicate that mda-7/IL-24-mediated apoptosis can be triggered through a combination of intracellular as well as secretory mechanisms and can occur efficiently in the absence of protein secretion.


Asunto(s)
Apoptosis/fisiología , Interleucinas/fisiología , Adenoviridae/genética , Apoptosis/genética , División Celular/genética , División Celular/fisiología , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Genes Supresores de Tumor , Terapia Genética/métodos , Vectores Genéticos/genética , Aparato de Golgi/metabolismo , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
6.
Oncogene ; 23(9): 1789-800, 2004 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-14676839

RESUMEN

Melanoma differentiation associated gene-5 (mda-5) was identified by subtraction hybridization as a novel upregulated gene in HO-1 human melanoma cells induced to terminally differentiate by treatment with IFN-beta+MEZ. Considering its unique structure, consisting of a caspase recruitment domain (CARD) and an RNA helicase domain, it was hypothesized that mda-5 contributes to apoptosis occurring during terminal differentiation. We have currently examined the expression pattern of mda-5 in normal tissues, during induction of terminal differentiation and after treatment with type I IFNs. In addition, we have defined its genomic structure and chromosomal location. IFN-beta, a type I IFN, induces mda-5 expression in a biphasic and dose-dependent manner. Based on its temporal kinetics of induction and lack of requirement for prior protein synthesis mda-5 is an early type I IFN-responsive gene. The level of mda-5 mRNA is in low abundance in normal tissues, whereas expression is induced in a spectrum of normal and cancer cells by IFN-beta. Expression of mda-5 by means of a replication incompetent adenovirus, Ad.mda-5, induces apoptosis in HO-1 cells as confirmed by morphologic, biochemical and molecular assays. Additionally, the combination of Ad.mda-5+MEZ further augments apoptosis as observed in Ad.null or uninfected HO-1 cells induced to terminally differentiate by treatment with IFN-beta+MEZ. The mda-5 gene is located on human chromosome 2q24 and consists of 16 exons, without pseudogenes, and is conserved in the mouse genome. Present data documents that mda-5 is a novel type I IFN-inducible gene, which may contribute to apoptosis induction during terminal differentiation and during IFN treatment. The conserved genomic and protein structure of mda-5 in human and mouse will permit analysis of the evolution and developmental aspects of this gene.


Asunto(s)
Apoptosis/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Melanoma/genética , Melanoma/patología , ARN Helicasas/genética , ARN Helicasas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , División Celular , Línea Celular Tumoral , Supervivencia Celular , Cromosomas Humanos Par 2/genética , ARN Helicasas DEAD-box , Inducción Enzimática/efectos de los fármacos , Exones/genética , Perfilación de la Expresión Génica , Genómica , Humanos , Helicasa Inducida por Interferón IFIH1 , Melanoma/metabolismo , Ratones , Estructura Terciaria de Proteína , ARN Helicasas/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia/genética , Transducción de Señal/efectos de los fármacos
7.
Oncogene ; 22(8): 1164-80, 2003 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-12606943

RESUMEN

Malignant gliomas are extremely aggressive cancers currently lacking effective treatment modalities. Gene therapy represents a promising approach for this disease. A requisite component for improving gene-based therapies of brain cancer includes tumor suppressor genes that exhibit cancer constrained inhibitory activity. Subtraction hybridization identified melanoma differentiation associated gene-7 (mda-7) as a gene associated with melanoma cell growth, differentiation and progression. Ectopic expression of mda-7 by means of a replication-incompetent adenovirus (Ad), Ad.mda-7, induces growth suppression and apoptosis selectively in diverse human cancers, without producing any apparent harmful effect in normal cells. We presently demonstrate that Ad.mda-7 induces growth inhibition and apoptosis in malignant human gliomas expressing both mutant and wild-type p53, and these effects correlate with an elevation in expression of members of the growth arrest and DNA damage (GADD) gene family. In contrast, infection with a recombinant Ad expressing wild-type p53, Ad.wtp53, specifically affects mutant p53 expressing gliomas. When tested in early passage normal and immortal human fetal astrocytes, growth inhibition resulting from infection with Ad.mda-7 or Ad.wtp53 is significantly less than in malignant gliomas and no toxicity is evident in these normal cells. Moreover, infection of gliomas with Ad.mda-7 or treatment with purified GST-MDA-7 protein sensitizes both wild-type and mutant p53 expressing tumor cells to the growth inhibitory and antisurvival effects of ionizing radiation, and this response correlates with increased expression of specific members of the GADD gene family. Since heterogeneity in p53 expression is common in evolving gliomas, the present findings suggest that Ad.mda-7 may, in many instances, prove more beneficial for the gene-based therapy of malignant gliomas than administration of wild-type p53.


Asunto(s)
Apoptosis , Astrocitos/citología , Neoplasias Encefálicas/patología , Glioma/patología , Interleucinas/fisiología , Apoptosis/genética , Apoptosis/efectos de la radiación , Astrocitos/efectos de la radiación , Neoplasias Encefálicas/terapia , División Celular/efectos de la radiación , Línea Celular Transformada/citología , Línea Celular Transformada/efectos de la radiación , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Daño del ADN , Genes Supresores de Tumor , Genes p53 , Terapia Genética , Vectores Genéticos/farmacología , Glioma/terapia , Humanos , Interleucinas/genética , Tolerancia a Radiación/genética , Receptores Virales/análisis , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de la radiación , Proteína p53 Supresora de Tumor/fisiología
8.
Oncogene ; 23(46): 7679-90, 2004 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-15334067

RESUMEN

The human mda-7/IL-24 gene product is normally expressed in melanocytes and certain lymphocyte populations. Loss of expression, a distinctive feature of many tumor suppressor genes, has been documented at RNA and protein levels in association with melanoma progression both in vitro as well as in human tumor-derived material. The MDA-7/IL-24 protein undergoes post-translational processing, including removal of an amino-terminal 48-residue signal peptide and extensive glycosylation prior to its secretion by producing cells. Its inherent cytokine properties have been documented in multiple reports, which have identified and characterized its cognate receptors and activation of the JAK/STAT signaling pathway following ligand/receptor docking. A notable and incompletely understood property of MDA-7/IL-24 is its ability to induce apoptosis in transformed cells, while having marginal growth suppressive effects on normal primary or immortalized cell lines. MDA-7/IL-24 has been delivered to cells, tumor xenografts and patients in clinical trials via a nonreplicating adenovirus (Ad.mda-7). Studies utilizing eukaryotically expressed and purified MDA-7/IL-24 protein from several sources have recapitulated some of the molecule's reported properties including receptor binding and JAK/STAT activation. Here, we report the properties and characteristics of a bacterially expressed and purified GST-MDA-7 fusion protein. These studies reveal that GST-MDA-7 retains its cancer-selective apoptosis-inducing properties, thereby providing a new reagent that will assist in defining the mechanism of action of this novel cytokine. In addition, retention of tumor-specific activity of GST-MDA-7 suggests that this protein may also have therapeutic applications.


Asunto(s)
Apoptosis/fisiología , Interleucinas/genética , Interleucinas/metabolismo , Animales , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Genes Supresores de Tumor , Terapia Genética , Glutatión Transferasa/genética , Humanos , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Trasplante Heterólogo
9.
Clin Cancer Res ; 9(9): 3272-81, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12960112

RESUMEN

PURPOSE: Despite therapeutic interventions including surgery, chemotherapy, and radiotherapy, glioblastoma multiforme (GBM) has a very poor prognosis and novel therapies are required. EXPERIMENTAL DESIGN: Melanoma differentiation-associated 7 (mda-7) (interleukin 24), when expressed via a recombinant replication-defective adenovirus, adenovirus (Ad).mda-7, has profound antiproliferative and cytotoxic effects in a variety of tumor cells but not in nontransformed cells. The present studies examined the combined impact of Ad.mda-7 and ionizing radiation on the proliferation and survival of GBM cell lines. RESULTS: Ad.mda-7 caused a dose-dependent reduction in the proliferation of glioma cells in 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. The antiproliferative effects of Ad.mda-7 were enhanced by radiation in a greater than additive fashion. These effects were not observed in cultures of nontransformed primary astrocytes. Purified MDA-7 protein caused a similar dose-dependent reduction in GBM cell growth that was enhanced after radiation exposure. The enhanced reduction in growth correlated with increased necrosis and DNA degradation. These modifications in cell phenotype correlated with reduced expression of Bcl-(XL) and enhanced expression of BAX. Overexpression of Bcl-(XL) protected cells from the antiproliferative and cytotoxic effects of Ad.mda-7 + radiation. Incubation of cells with N-acetyl cysteine abolished the enhancing effects of radiation. In vitro, Ad.mda-7 and radiation reduced colony formation ability, which was significantly increased when the two treatments were combined. In vivo, Ad.mda-7 enhanced the survival of Fischer 344 rats implanted intracranially with glioma cells. Radiation did not alter survival in control infected animals, whereas it prolonged survival in those infected with Ad.mda-7. CONCLUSIONS: These findings demonstrate that mda-7 reduces the proliferation and enhances the radiosensitivity of GBM cells in vitro and in vivo.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Glioma/tratamiento farmacológico , Interleucinas/genética , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Acetilcisteína/metabolismo , Adenoviridae/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Células Cultivadas , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Regulación hacia Abajo , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Glioma/metabolismo , Glioma/radioterapia , Glutatión Transferasa/metabolismo , Humanos , Interleucinas/metabolismo , Necrosis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Ratas Endogámicas F344 , Sales de Tetrazolio/farmacología , Tiazoles/farmacología , Factores de Tiempo , Proteína X Asociada a bcl-2 , Proteína bcl-X
10.
Mol Cancer Ther ; 2(7): 623-32, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12883035

RESUMEN

The median survival of metastatic renal cell carcinoma (RCC) is 12 months, and the majority of treatment options are palliative. MDA-7 (interleukin-24), when expressed via a recombinant replication defective adenovirus, Ad.mda-7, has profound antiproliferative and cytotoxic effects in a wide variety of tumor cells but not in nontransformed cells. The studies in this study examined the impact of MDA-7 on RCC proliferation and survival. RCC lines (A498 and UOK121N), but not primary renal epithelial cells, were resistant to adenoviral infection that correlated with a lack of coxsackievirus and adenovirus receptor expression. Additional studies were performed using purified preparations of bacterially synthesized glutathione S-transferase (GST)-MDA-7 protein. GST-MDA-7, but not GST, caused a dose-dependent inhibition of RCC proliferation but not of primary renal epithelial cells. Clinically achievable concentrations of the novel therapeutic agent arsenic trioxide (0.5-1 micro M) were found to have little effect on RCC growth. However, the combination of GST-MDA-7 and arsenic trioxide resulted in a greater than additive reduction in cell growth that correlated with a large increase in tumor cell death. The free radical scavenger N-acetyl cysteine abolished the potentiating effect of arsenic trioxide. Although pro-caspase 3, poly(ADP-ribose) polymerase, and Bcl-(XL) levels, as well as nucleosomal DNA integrity, were reduced by combined treatment, cell killing was predominantly nonapoptotic. Combined treatment of RCC lines with GST-MDA-7 and arsenic trioxide also resulted in a substantial reduction in clonogenic survival compared with either treatment individually. Collectively, these findings demonstrate that MDA-7 protein, in combination with agents that generate free radicals, may have potential in the treatment of RCC.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , Radicales Libres/metabolismo , Interleucinas/farmacología , Neoplasias Renales/tratamiento farmacológico , Acetilcisteína/farmacología , Adenoviridae/genética , Trióxido de Arsénico , Arsenicales/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Caspasas/metabolismo , División Celular/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Genes Supresores de Tumor , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Glutatión Transferasa/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Óxidos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Virales/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas , Proteína bcl-X
11.
Cancer Biol Ther ; 2(4 Suppl 1): S23-37, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14508078

RESUMEN

An obstacle to effective gene-based cancer therapies is the limited number of cancer-specific growth suppressing and apoptosis-inducing genes. Using a differentiation induction subtraction hybridization (DISH) approach with human melanoma cells, melanoma differentiation associated (mda) genes were isolated that display elevated expression as a function of irreversible growth arrest, cancer reversion and terminal differentiation. This screening paradigm resulted in the cloning of mda-7 in the context of terminal differentiation of human melanoma cells. Based on its structure, chromosomal location, sequence homology and cytokine-like properties, mda-7 has now been renamed IL-24 and classified as a member of the expanding IL-10 cytokine gene family. Expression of mda-7/IL-24 inversely correlates with melanoma progression and administration of mda-7/IL-24 by means of a replication incompetent adenovirus, Ad.mda-7, results in growth suppression and apoptosis in melanoma cells as well as in a broad-spectrum of additional cancer cell types. In contrast, Ad.mda-7 does not elicit deleterious effects in normal cells, including those of epithelial, fibroblast, astrocyte, melanocyte or endothelial origin. Based on these distinctive properties and anti-tumor and anti-angiogenic activities in human tumor xenograft animal models, mda-7/IL-24 has now entered the clinical arena. A Phase I/II clinical trial in patients with advanced carcinomas involving intratumoral administration of mda-7/IL-24 [using a replication incompetent adenovirus; ING241 (Ad.mda-7)] has documented that this gene is safe and well tolerated by patients and a single virus injection elicits apoptosis in a majority of the tumor. Current data suggests that mda-7/IL-24 may function as a dual-acting cytokine in which its normal physiological functions may be related to specific aspects of the immune system and over-expression culminates in cancer-specific apoptosis. This review will provide a prospectus of our current understanding of mda-7/IL-24.


Asunto(s)
Apoptosis , Interleucinas/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Adenoviridae/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Citocinas/metabolismo , Progresión de la Enfermedad , Genes Supresores de Tumor , Terapia Genética , Humanos , Melanoma/metabolismo , Melanoma/patología , Modelos Biológicos , Trasplante de Neoplasias , Neovascularización Patológica , Hibridación de Ácido Nucleico
12.
Cancer Biol Ther ; 2(4): 347-53, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14508103

RESUMEN

Despite therapeutic interventions including surgery, chemotherapy and radiotherapy, glioblastoma multiforme (GBM) has a very poor prognosis and novel therapies are required. MDA-7 (IL-24), when expressed via a recombinant replication defective adenovirus, Ad.mda-7, has profound anti-proliferative and cytotoxic effects in a variety of tumor cells, but not in non-transformed cells. The present studies examined the combined impact of Ad.mda-7 and ionizing radiation on the proliferation and survival of GBM cells. Ad.mda-7 reduced the proliferation of rodent and human glioma cells in MTT assays and in colony formation assays. The anti-proliferative effects of Admda-7 were enhanced by radiation in a greater than additive fashion. In vitro, this cellular change correlated with enhanced cell numbers in G1/G0 and G2/M phases of the cell cycle, implying Ad.mda-7 radiosensitizes tumor cells in a cell cycle-independent manner. The radiosensitizing effects were not observed in cultures of non-transformed primary astrocytes. The enhanced reduction in growth correlated with increased necrosis and DNA degradation. Ad.mda-7 enhanced p38 and ERK1/2 activity but did not alter JNK or Akt activity. Irradiation of cells expressing MDA-7 suppressed ERK1/2 activity and dramatically enhanced JNK1/2 activity without altering either Akt or p38 activity. Inhibition of JNK1/2, but not p38, signaling abolished the radiosensitizing properties of MDA-7. Inhibition of neither ERK1/2 nor PI3K signaling enhanced the anti-proliferative effects of Ad.mda-7, whereas combined inhibition of both pathways enhanced cell killing, suggesting that ERK and PI3K signaling can be protective against MDA-7 lethality.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias Encefálicas/radioterapia , Glioma/radioterapia , Interleucinas/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Transducción de Señal , Adenoviridae/genética , Animales , Apoptosis/fisiología , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Relación Dosis-Respuesta en la Radiación , Genes Supresores de Tumor , Glioma/metabolismo , Glioma/patología , Glutatión Transferasa/metabolismo , Interleucinas/genética , MAP Quinasa Quinasa 4 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Radiación Ionizante , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
13.
Cancer Biol Ther ; 3(8): 739-51, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15197348

RESUMEN

We examined the impact of purified bacterially synthesized GST-MDA-7 (IL-24) and ionizing radiation on the proliferation and survival of nonestablished human glioblastoma multiforme (GBM) cells. Glioma cell types expressing mutated PTEN and p53 molecules, activated ERBB1VIII, overexpressing wild type ERBB1 or without receptor overexpression were selected. In MTT assays, GST-MDA-7 caused a dose-dependent reduction in the proliferation of nonestablished glioma cells; however only at higher concentrations did GST-MDA-7 reduce cell viability. The anti-proliferative and cytotoxic effects of GST-MDA-7 were enhanced by radiation in a greater than additive fashion that correlated with JNK1/2/3 activation. The reduction in cell growth and enhancement in cell killing by the combination of GST-MDA-7 and radiation were blocked by an ROS scavenger, N-acetyl cysteine (NAC), a JNK1/2/3 inhibitor SP600125, a pan-caspase inhibitor (zVAD) and by an inhibitor of caspase 9 (LEHD), but not by an inhibitor of caspase 8 (IETD). Low concentrations of either GST-MDA-7 or radiation reduced clonogenic survival, however colony formation ability was significantly further decreased when the two treatments were combined, which was also blocked by inhibition of caspase 9 function. In general agreement with activation of the intrinsic caspase pathway, cell death correlated with reduced BCL-XL expression and with increased levels of the pro-apoptotic proteins BAD and BAX. Inhibition of caspase 9 after combination treatment blunted neither JNK1/2/3 activation nor the enhanced expression of BAD and BAX, but did block caspase 3 cleavage, reduced expression of BCL-XL and inhibition of ERK1/2 activity. In contrast, incubation with NAC blocked JNK1/2/3 activation and cell killing, but not the increases in BAD and BAX expression. These findings argue that after combination treatment JNK1/2/3 activation is a primary pro-apoptotic event and loss of BCL-XL expression and ERK1/2 activity are secondary caspase-dependent processes. This data also argues that GST- MDA-7 induces two parallel pro-apoptotic pathways via ROS-dependent and -independent mechanisms. Infection of primary human astrocytes with a recombinant adenovirus to express MDA-7, Ad.mda-7, but not infection with either Ad.cmv or Ad.mda-7SP- lacking MDA-7 secretion, resulted in the suppression of GBM cell colony formation in soft agar overlay assays, an effect that was enhanced in a greater than additive fashion by radiation. Collectively, our findings demonstrate that MDA-7 reduces proliferation and enhances the radiosensitivity of nonestablished human GBM cells in vitro, and when grown in 3 dimensions, and that sensitization occurs independently of basal EGFR/ERK1/2/AKT activity or the functions of PTEN and p53.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias Encefálicas/terapia , Proliferación Celular/efectos de la radiación , Glioblastoma/terapia , Interleucinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Acetilcisteína/farmacología , Adenoviridae/genética , Adyuvantes Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/efectos de la radiación , Neoplasias Encefálicas/metabolismo , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Genes Supresores de Tumor , Genes erbB-1/fisiología , Glioblastoma/metabolismo , Humanos , Interleucinas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tolerancia a Radiación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína X Asociada a bcl-2 , Proteína bcl-X
14.
Biotechniques ; Suppl: 30-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12395925

RESUMEN

One hallmark of neoplasia is abnormal differentiation. Induction of differentiation, by chemical or biological methods, provides a possible therapeutic intervention. "Differentiation therapy" is well documented in several model systems. These include melanoma, in which treatment with interferon-beta and the protein kinase C activator mezerein induces irreversible growth arrest and terminal differentiation culminating in programmed cell death. Subtraction hybridization between terminally differentiated and untreated melanoma cells identified melanoma differentiation-associated gene-7 (mda-7), which is selectively induced during the process of melanoma terminal differentiation. Since its identification seven years ago, mda-7 has been the object of intense focus because of its unique biological properties. Firstly, mda-7 is a secreted protein having cytokine-like properties and belonging to the IL-10 cytokine family. Based on this consideration, mda-7 was renamed IL-24. Secondly if delivered by means of an adenoviral vector, mda-7 induces selective apoptosis in cancer cells of diverse origin, while sparing their normal cellular counterparts. As such, mda-7 has become a novel tool for cancer gene therapy and is currently undergoing phase II clinical trials to determine its clinical efficacy in patients. The present review examines the biological properties of mda-7 and the signaling pathways that contribute to its unique cancer-specific apoptosis-inducing properties.


Asunto(s)
Interleucinas/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Apoptosis , Diferenciación Celular , Cromosomas Humanos Par 1/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Terapia Genética , Humanos , Interleucinas/química , Interleucinas/genética , Interleucinas/uso terapéutico , Mamíferos/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Ratas , Proteínas Recombinantes de Fusión/fisiología , Seguridad , Transducción de Señal , Células Tumorales Cultivadas/patología
15.
Int Immunopharmacol ; 4(5): 635-47, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15120649

RESUMEN

Melanoma Differentiation Associated gene-7 (mda-7)/IL-24 has shown potent tumor cell apoptosis inducing capacity in multiple cancers, making it a strong candidate for use as a human cancer gene therapeutic. Several independent studies have currently documented and confirmed mda-7/IL-24's cytokine nature including presence of a canonical secretory signal peptide, processing and secretion of the molecule by cells and it's binding to specific interleukin receptors on the cell surface. Receptor binding has been shown to activate the JAK/STAT signal transduction pathway with concomitant stimulation of STAT 1 and 3 transactivators. The physiological role(s) of this molecule in modulating immune responses, as a member of the IL-10 family of cytokines, is not well documented and most current information pertains to its apparently restricted expression patterns in specific cell types with immunomodulatory activity. On the other hand, several additional signal transduction pathways were modulated when cells overexpress mda-7/IL-24, not all of which are necessarily downstream of mda-7/IL-24 induced JAK/STAT activation. A summary of the current status of information is presented to provide a perspective for the cytokine-related properties of mda-7/IL-24 in correlation to its tumor cell apoptosis inducing activity. Moreover, new evidence has surfaced pointing toward apoptosis induction via mechanisms independent of cytokine activity-related JAK/STAT activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citocinas/farmacología , Interleucinas/farmacología , Neoplasias/patología , Animales , Apoptosis/inmunología , Ensayos Clínicos Fase I como Asunto , Citocinas/clasificación , Citocinas/inmunología , Genes Supresores de Tumor/fisiología , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , Interleucinas/genética , Interleucinas/inmunología , Interleucinas/fisiología , Neoplasias/fisiopatología
16.
Mol Ther ; 11(1): 4-18, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15585401

RESUMEN

The mda-7/IL-24 cDNA was isolated almost a decade ago in a screen for genes differentially upregulated following growth arrest and terminal differentiation of a human melanoma cell line employed as an in vitro cell differentiation model. The underlying rationale for the screen was that oncogenesis arises from a cellular dedifferentiation process culminating in uncontrolled proliferation and acquisition of invasive and metastatic potential. Identification of genes upregulated during the process of reactivation of faulty or inoperational differentiation maintenance programs was postulated to have cancer gene therapeutic potential. In this context, it is heartening to note that mda-7/IL-24 has made a methodical and progressive journey, from an unidentified novel sequence with little homology to known genes at its time of isolation to currently having the status of a molecule belonging to the IL-10-related family of cytokines, with considerable cancer gene therapeutic potential. Extensive in vitro and in vivo human tumor xenograft studies have established its transformed cell apoptosis-inducing capacity in various model systems. It has recently taken an important step for a candidate cancer gene therapeutic molecule, in the ultimate goal of benchtop to clinic, by being currently utilized in human Phase I/II clinical trials. This review provides a current perspective of our understanding of mda-7/IL-24, including established and more recent information about the molecular properties, specificity of anti-tumor-cell apoptosis-inducing activity, and underlying mechanisms of this action relative to its cancer gene therapeutic potential.


Asunto(s)
Terapia Genética , Interleucinas/metabolismo , Neoplasias/genética , Neoplasias/terapia , Animales , Apoptosis/genética , Ensayos Clínicos Fase I como Asunto , Genes Supresores de Tumor , Humanos , Interleucinas/genética , Neoplasias/metabolismo , Neoplasias/patología
17.
J Cell Physiol ; 192(1): 34-44, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12115734

RESUMEN

Genomic instability is a fundamental component of cancer progression. Subtraction hybridization identified a novel rodent gene, progression elevated gene-3 (PEG-3) whose expression directly correlates with cancer aggressiveness and progression. Moreover, ectopic expression of PEG-3 in rodent or human tumor cells produces an aggressive transformed phenotype. We demonstrate that PEG-3 expression in rodent tumor cells correlates directly with genomic instability as characterized by alterations in chromosome composition and structure. Additionally, elevated endogenous or ectopic expression of PEG-3 in rodent and human tumor cells, respectively, enhances gene amplification, as monitored by resistance to methothrexate (MTX) and amplification of the dihydrofolate reductase (dhfr) gene. Stable expression of PEG-3 in normal cloned rat embryo fibroblast (CREF) cells marginally elevates MTX resistance, but morphology remains unaltered and anchorage independence is not induced, suggesting that these phenotypes are separable in immortal cells and gene amplification may precede the acquisition of morphological and oncogenic transformation. The present studies document that stable, inducible, and transient expression of PEG-3 in cancer cells augments genomic instability. In these contexts, one mechanism by which PEG-3 influences cancer progression may be by preferentially facilitating the development of genomic changes in evolving cancer cells.


Asunto(s)
Antígenos de Diferenciación , Genoma , Proteínas de Neoplasias/fisiología , Neoplasias/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Carcinoma/patología , Carcinoma/fisiopatología , Proteínas de Ciclo Celular , División Celular/efectos de los fármacos , Células Cultivadas , Aberraciones Cromosómicas , Resistencia a Antineoplásicos , Embrión de Mamíferos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Amplificación de Genes , Humanos , Masculino , Metotrexato/farmacología , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Proteína Fosfatasa 1 , Proteínas Proto-Oncogénicas , ARN Mensajero/metabolismo , Ratas , Tetrahidrofolato Deshidrogenasa/genética , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/fisiopatología
18.
Proc Natl Acad Sci U S A ; 99(2): 637-42, 2002 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-11805321

RESUMEN

Human melanoma cells can be reprogrammed to terminally differentiate and irreversibly lose proliferative capacity by appropriate pharmacological manipulation. Subtraction hybridization identified melanoma differentiation-associated gene-5 (mda-5) as a gene induced during differentiation, cancer reversion, and programmed cell death (apoptosis). This gene contains both a caspase recruitment domain and putative DExH group RNA helicase domains. Atypical helicase motifs of MDA-5 deviate from consensus sequences but are well conserved in a potentially new group of cloned and hypothetical proteins. mda-5 is an early response gene inducible by IFN and tumor necrosis factor-alpha, responding predominantly to IFN-beta. Protein kinase C activation by mezerein further augments mda-5 expression induced by IFN-beta. Expression of mda-5 is controlled transcriptionally by IFN-beta, and the MDA-5 protein localizes in the cytoplasm. mda-5 displays RNA-dependent ATPase activity, and ectopic expression of mda-5 in human melanoma cells inhibits colony formation. In these contexts, mda-5 may function as a mediator of IFN-induced growth inhibition and/or apoptosis. MDA-5 is a double-stranded RNA-dependent ATPase that contains both a caspase recruitment domain and RNA helicase motifs, with a confirmed association with growth and differentiation in human melanoma cells.


Asunto(s)
Melanoma/enzimología , ARN Helicasas/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Diferenciación Celular , División Celular , Clonación Molecular , ARN Helicasas DEAD-box , ADN Complementario/genética , Inhibidores de Crecimiento/metabolismo , Humanos , Interferón Tipo I/farmacología , Helicasa Inducida por Interferón IFIH1 , Melanoma/genética , Melanoma/patología , Datos de Secuencia Molecular , ARN Helicasas/genética , ARN Bicatenario/metabolismo , Proteínas Recombinantes , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
19.
J Cell Physiol ; 196(2): 334-45, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12811827

RESUMEN

Experimental evidence documents that the MDA-7/IL-24 protein (an IL-10 family cytokine) binds to IL-20 and IL-22 receptor complexes resulting in the activation of JAK/STAT signaling pathways. Recent published reports utilizing human blood derived primary lymphocytes have provided additional confirmatory evidence relating to the cytokine properties of this molecule. A notable attribute of mda-7/IL-24 is its cancer cell-specific apoptosis inducing capacity, which currently remains incompletely understood. Treatment with distinctive tyrosine kinase inhibitors (Genistein and AG18) or a JAK-selective inhibitor (AG490) did not prevent Ad.mda-7 induced apoptosis in diverse cell lines. In addition, there is no apparent correlation between patterns of expression of IL-20R1, IL-20R2, and IL-22R mRNA and susceptibility to Ad.mda-7 in different cell lines. Furthermore, Ad.mda-7 is able to induce killing in STAT/JAK deficient cells. In contrast, treatment with the p38(MAPK) selective inhibitor SB203580, partially inhibited apoptosis induced by Ad.mda-7 in different cell lines. These results demonstrate for the first time that signaling events leading to susceptibility to Ad.mda-7 induced apoptosis, might be tyrosine kinase independent and can thus be distinguished from its cytokine function related properties mediated by the IL-20/IL-22 receptor complexes that require JAK/STAT kinase activity.


Asunto(s)
Apoptosis , Interleucinas/metabolismo , Neoplasias/fisiopatología , Proteínas Tirosina Quinasas/metabolismo , Transactivadores/metabolismo , Adenoviridae/genética , Línea Celular Transformada , Resistencia a Medicamentos , Inhibidores Enzimáticos/farmacología , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Vectores Genéticos , Células HeLa , Humanos , Interleucinas/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptores de Interleucina/metabolismo , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Proc Natl Acad Sci U S A ; 99(15): 10054-9, 2002 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-12114539

RESUMEN

Subtraction hybridization identified melanoma differentiation-associated gene-7 (mda-7) as a gene induced during terminal differentiation in human melanoma cells. On the basis of structure, chromosomal localization and cytokine-like properties, mda-7 is classified as IL-24. Administration of mda-7/IL-24 by means of a replication-incompetent adenovirus (Ad.mda-7) induces apoptosis selectively in diverse human cancer cells without inducing harmful effects in normal fibroblast or epithelial cells. The present studies investigated the mechanism underlying this differential apoptotic effect. Infection of melanoma cells, but not normal immortal melanocytes, with Ad.mda-7 induced a time- and dose-dependent increase in expression, mRNA and protein, of a family of growth arrest and DNA damage (GADD)-inducible genes, which correlated with induction of apoptosis. Among the members of the GADD family of genes, GADD153, GADD45 alpha, and GADD34 displayed marked, and GADD45 gamma showed minimal induction. Treatment of melanoma cells with SB203580, a selective inhibitor of the p38 mitogen-activated protein kinase (MAPK) pathway, effectively inhibited Ad.mda-7-induced apoptosis. Additional support for an involvement of the p38 MAPK pathway in Ad.mda-7-mediated apoptosis was documented by using an adenovirus expressing a dominant negative mutant of p38 MAPK. Infection with Ad.mda-7 increased the phosphorylation of p38 MAPK and heat shock protein 27 in melanoma cells but not in normal immortal melanocytes. In addition, SB203580 effectively inhibited Ad.mda-7-mediated induction of the GADD family of genes in a time- and dose-dependent manner, and it effectively blocked Ad.mda-7-mediated down-regulation of the antiapoptotic protein BCL-2. Inhibition of GADD genes by an antisense approach either alone or in combination also effectively blocked Ad.mda-7-induced apoptosis in melanoma cells. These results support the hypothesis that Ad.mda-7 mediates induction of the GADD family of genes by means of the p38 MAPK pathway, thereby resulting in the selective induction of apoptosis in human melanoma cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Ciclo Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico , Interleucinas/farmacología , Melanoma/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares/genética , Proteínas/genética , Factores de Transcripción/genética , Antígenos de Diferenciación , Apoptosis/fisiología , Ciclo Celular , Supervivencia Celular , Genes Supresores de Tumor , Proteínas de Choque Térmico HSP27 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas Activadas por Mitógenos/genética , Modelos Biológicos , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Fosforilación , Proteína Fosfatasa 1 , Factor de Transcripción CHOP , Transfección , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos , Proteinas GADD45
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA