Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Br J Cancer ; 108(10): 2005-12, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23632480

RESUMEN

BACKGROUND: Current evidence indicates that a stem cell-like sub-population within malignant glioblastomas, that overexpress members of the adenosine triphosphate-binding cassette (ABC) family transporters, is responsible for multidrug resistance and tumour relapse. Eradication of the brain tumour stem cell (BTSC) compartment is therefore essential to achieve a stable and long-lasting remission. METHODS: Melatonin actions were analysed by viability cell assays, flow cytometry, quantitative PCR for mRNA expression, western blot for protein expression and quantitative and qualitative promoter methylation methods. RESULTS: Combinations of melatonin and chemotherapeutic drugs (including temozolomide, current treatment for malignant gliomas) have a synergistic toxic effect on BTSCs and A172 malignant glioma cells. This effect is correlated with a downregulation of the expression and function of the ABC transporter ABCG2/BCRP. Melatonin increased the methylation levels of the ABCG2/BCRP promoter and the effects on ABCG2/BCRP expression and function were prevented by preincubation with a DNA methyltransferase inhibitor. CONCLUSION: Our results point out a possible relationship between the downregulation of ABCG2/BCRP function and the synergistic toxic effect of melatonin and chemotherapeutic drugs. Melatonin could be a promising candidate to overcome multidrug resistance in the treatment of glioblastomas, and thus improve the efficiency of current therapies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Neoplasias Encefálicas/patología , Metilación de ADN/efectos de los fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/patología , Melatonina/farmacología , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/fisiología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Metilación de ADN/fisiología , Evaluación Preclínica de Medicamentos , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Humanos , Melatonina/administración & dosificación , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos
2.
Nat Med ; 4(6): 685-90, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9623977

RESUMEN

The transfer of apoptosis genes to tumors is one of the most promising strategies for cancer gene therapy. We have shown that massive apoptosis occurs when wild-type p53 expression is induced in glioma cells carrying a p53 gene mutation. However, adenovirus-mediated p53 gene transfer is ineffective in causing apoptosis in glioma cells that retain a wild-type p53 genotype. We evaluated the effect of E2F-1 overexpression on the growth of gliomas in vitro and in vivo. In the in vitro study, the adenovirus-mediated transfer of exogenous E2F-1 protein precipitated generalized apoptosis in gliomas. The treatment with Ad5CMV-E2F-1 of nude mice carrying subcutaneous gliomas arrested tumor growth. Our results indicate that E2F-1 has anti-glioma activity in vitro and in vivo.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras , Glioma/genética , Factores de Transcripción/genética , Adenovirus Humanos/genética , Animales , Apoptosis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Muerte Celular/genética , Muerte Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Expresión Génica/genética , Genes Supresores de Tumor , Terapia Genética , Vectores Genéticos/genética , Glioma/fisiopatología , Glioma/terapia , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteína de Retinoblastoma/metabolismo , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/fisiología , Transfección/genética , Células Tumorales Cultivadas
3.
Cancer Gene Ther ; 14(8): 756-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17557108

RESUMEN

Novel therapies are clearly needed for gliomas, and the combination of oncolytic vectors with chemotherapy possesses a significant hope for the treatment of this malignancy. In addition, combination with chemotherapy allows for lower virus doses to achieve anticancer effect, thus resulting in lower undesirable toxicities due to viral proteins. In this work, we sought to determine whether combination of an oncolytic adenovirus ICOVIR-5, with RAD001 or temozolomide (TMZ) could result in enhanced anti-glioma effect in vivo. We assessed the in vitro cytotoxic effect and replication properties of ICOVIR-5 in combination with RAD001 or TMZ in U87 MG glioma cell line by MTT and TCID(50), respectively. Our data showed that in vitro treatment with RAD001 or TMZ not only interfered with adenovirus replication but, in addition, enhanced its oncolytic properties. To evaluate the in vivo anticancer effect, athymic mice bearing glioma xenografts (5 x 10(5) U87 MG cells/animal) received a single intratumoral injection of ICOVIR-5 (10(7) PFU/animal). RAD001 was given as a regimen of 5 mg/kg 5 days per week until the end of the experiment and TMZ was administered for 5 days at 7.5 mg/kg/mice. Of significance, combination of ICOVIR-5 with RAD001 or TMZ showed a potent anti-glioma effect in vivo, resulting in a dramatic extension of the median animal survival and in 20-40% animals becoming free of disease beyond 90 days.


Asunto(s)
Adenoviridae , Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Viroterapia Oncolítica , Sirolimus/análogos & derivados , Animales , Línea Celular Tumoral , Dacarbazina/farmacología , Everolimus , Glioma/terapia , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Sirolimus/farmacología , Temozolomida
4.
J Natl Cancer Inst ; 89(14): 1036-44, 1997 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-9230885

RESUMEN

BACKGROUND: Alterations of the p53 (also called TP53) gene are one of the most common abnormalities in gliomas. We have previously reported that restoration of wild-type p53 protein function in glioma cells results in programmed cell death (apoptosis). Since p53 functions are mediated by genes that directly control the tumor suppressor effect of the p53 protein, understanding the relationship between p53 and p53-related genes in glioma cells will aid in the design of more rational treatment strategies for brain tumors. PURPOSE: We conducted this study to examine the timing of the p53-mediated events preceding apoptosis. More specifically, we undertook this work to characterize the genetic and cell cycle-related factors that may increase the resistance of glioma cells to p53-induced apoptosis. METHODS: Two human glioma cell lines (U-251 MG and U-373 MG) that express mutant p53 protein and two (U-87 MG and EFC-2) that express wild-type p53 protein were used. Replication-deficient adenovirus was utilized as an expression vector to transfer exogenous p53 and p21 complementary DNAs into the glioma cells; control cells were infected with the viral expression vector alone. To monitor gene transfer and the expression of exogenous genes (as well as the expression of endogenous genes), we used western blot analyses and immunohistochemistry analyses. Flow cytometry studies of cellular DNA content were performed to determine the cell cycle phenotype of the glioma cells before and after treatment. RESULTS: p53-mediated apoptosis was preceded by elevation in the levels of the p21 (cell cycle-related) and Bax (apoptosis-related) proteins. In addition, cell cycle analyses showed that glioma cells were arrested in the G2 phase before undergoing cell death. Transfer of p21 induced a G2 block but did not induce apoptosis. Moreover, coexpression of p21 and p53 prevented glioma cells from undergoing apoptosis. Expression of exogenous p53 in wild-type p53 cells did not induce elevation of Bax levels, arrest in G2 phase, or apoptosis. CONCLUSIONS AND IMPLICATIONS: Our data confirmed the ability of wild-type p53 to induce apoptosis in p53 mutant glioma cells. In addition, our results document that p21 plays a role in protecting cells from p53-mediated programmed cell death and suggest that p53-mediated apoptosis and p21 induction may represent, at least in certain cases, opposite signals. Finally, our data suggest that over expression of p21 in gliomas may be related to resistance to treatments that induce apoptosis.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Ciclinas/fisiología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/fisiopatología , Proteína p53 Supresora de Tumor/fisiología , Adenoviridae , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , ADN de Neoplasias/genética , Citometría de Flujo , Vectores Genéticos , Glioblastoma/genética , Humanos , Fenotipo , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba
5.
Cancer Res ; 56(4): 694-9, 1996 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8630997

RESUMEN

Wild-type p53 is involved in several aspects of cell cycle control and suppression of transformation, inducing either apoptosis or G1 block in cell cycle progression. Using a recombinant adenovirus containing the wild-type p53 cDNA, the biological effects of the newly expressed wild-type p53 protein were examined in six human glioma cell lines. Three cell lines (U-251 MG, U-373 MG, and A-172) expressed endogenous mutant p53, and the other three (U-87 MG, EFC-2, and D54 MG) expressed wild-type p53. The restoration of normal p53-encoded protein in the mutant cell lines induced apoptosis as assessed by morphological studies using nuclear staining, electron microscopy, and flow cytometric assays. In wild-type p53 cell lines, however, the overexpression of wild-type p53 did not result in apoptosis but inhibited cellular proliferation rather drastically and modified the neoplastic phenotype. Differential effects suggest two pathways for glioma oncogenesis and a possible therapeutic strategy.


Asunto(s)
Adenovirus Humanos , Apoptosis , Genes p53 , Transfección/métodos , Proteína p53 Supresora de Tumor/biosíntesis , Western Blotting , División Celular , Línea Celular , Citometría de Flujo , Expresión Génica , Vectores Genéticos , Glioma/genética , Homocigoto , Humanos , Cinética , Proteínas Recombinantes/biosíntesis , Factores de Tiempo , Células Tumorales Cultivadas
6.
Cancer Res ; 59(4): 895-900, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10029081

RESUMEN

Presently, there is no effective treatment for glioblastoma, the most malignant and common brain tumor. Angiogenic factors are potentially optimal targets for therapeutic strategies because they are essential for tumor growth and progression. In this study, we sought a strategy for efficiently delivering an antisense cDNA molecule of the vascular endothelial growth factor (VEGF) to glioma cells. The recombinant adenoviral vector Ad5CMV-alphaVEGF carried the coding sequence of wild-type VEGF165 cDNA in an antisense orientation. Infection of U-87 MG malignant glioma cells with the Ad5CMV-alphaVEGF resulted in reduction of the level of the endogenous VEGF mRNA and drastically decreased the production of the targeted secretory form of the VEGF protein. Treatment of s.c. human glioma tumors established in nude mice with intralesional injection of Ad5CMV-alphaVEGF inhibited tumor growth. Taken together, these findings indicate that the efficient down-regulation of the VEGF produced by tumoral cells using antisense strategies has an antitumor effect in vivo. This is the first time that an adenoviral vector is used to transfer antisense VEGF sequence into glioma cells in an animal model, and our results suggest that this system may have clinical and therapeutic utility.


Asunto(s)
ADN sin Sentido/farmacología , Factores de Crecimiento Endotelial/genética , Terapia Genética , Glioma/terapia , Linfocinas/genética , Neovascularización Patológica/terapia , Adenoviridae/genética , Animales , Regulación hacia Abajo , Glioma/irrigación sanguínea , Humanos , Ratones , Ratones Desnudos , ARN Mensajero/análisis , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
7.
Cancer Res ; 61(18): 6693-7, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11559537

RESUMEN

Strong evidence exists to support the tenet that activation of E2F transcription factors, via alterations in the p16-cyclin D-Rb pathway, is a key event in the malignant progression of most human malignant gliomas. The oncogenic ability of E2F has been related to the E2F-mediated up-regulation of several proteins that positively regulate cell proliferation. However, E2F may indirectly enhance proliferation by activating antiapoptotic molecules. In this work, we sought to ascertain whether E2F-1-mediated events involve the up-regulation of the antiapoptotic molecule Bcl-2. Western blot analyses showed up-regulation of Bcl-2 but not of Bcl-x(L) by 24 h after the transfer of E2F-1. Northern blot studies showed that transfer of E2F-1 also up-regulated Bcl-2 RNA. In support of these findings and the concept that E2F-1 has a direct effect in the induction of Bcl-2, we found a putative E2F binding site within the Bcl-2 sequence. Subsequent gel-mobility shift and supershift experiments involving the CTCCGCGC site in the bcl-2 promoter showed that E2F-1 bound Bcl-2. Transactivation experiments consistently showed that ectopic E2F-1 activated responsive elements located in the -1448/-1441 region in the P1 promoter region of the bcl-2 gene. As expected, other members of the E2F family of transcription factors such as E2F-2 and E2F-4 also transactivated the bcl-2 promoter. Our results demonstrate that E2F-1 modulates the expression of the antiapoptotic molecule Bcl-2 and suggest that up-regulation of Bcl-2 may favor the oncogenic role of E2F-1 and other members of the E2F family of transcription factors.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Glioma/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factores de Transcripción/genética , Activación Transcripcional , Sitios de Unión , Ciclo Celular/fisiología , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F2 , Factor de Transcripción E2F4 , Regulación Neoplásica de la Expresión Génica , Técnicas de Transferencia de Gen , Genes bcl-2/genética , Glioma/metabolismo , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Factores de Transcripción/biosíntesis , Regulación hacia Arriba
8.
Oncogene ; 12(1): 103-10, 1996 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-8552379

RESUMEN

The p16 (MTS1/CDKN2) gene localized at the 9p21 chromosomal region encodes for a cell cycle inhibitor protein and is altered in many human cancers. The frequency of p16 alterations in gliomas exceeds 50%. To restore the missing wild-type p16 gene efficiently in glioma cells an adenovirus vector carrying the full length coding sequence of the wild-type p16 cDNA, Ad5RSV-p16, was constructed. Three human glioma cell lines, U251 MG, U-87 MG and D54 MG, that did not express endogenous p16/CDKN2 gene and were easily infected with adenovirus vectors were selected for these experiments. Introduction of the Ad5RSV-p16 in these malignant glioma cell lines directed the biosynthesis of functional p16 protein in the majority of the exposed cells, significantly inhibited cell growth, influenced cell morphology and modified the transformed phenotype of cells including the ability to form colonies in soft agar. Flow cytometric studies revealed that the majority of the Ad5RSV-p16 infected glioma cells were arrested in the G0-G1 phases of the cell cycle. These results suggest that p16/CDKN2 inactivation is a significant factor in the genesis and progression of gliomas and that the restoration of the wild-type p16 protein could have clinical and therapeutic utility.


Asunto(s)
Adenoviridae/genética , Proteínas Portadoras/genética , Genes Supresores de Tumor , Terapia Genética , Glioma/terapia , División Celular , Transformación Celular Neoplásica , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Técnicas de Transferencia de Gen , Humanos , Células Tumorales Cultivadas
9.
Oncogene ; 13(8): 1615-9, 1996 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-8895506

RESUMEN

There is considerable evidence that lack of p16 protein expression is a frequent event in human gliomas. Nevertheless, the molecular mechanisms underlying this absence of p16 protein expression are not completely understood. In some gliomas, homozygous deletions are the main cause of p16/CDKN2 gene inactivation. However, other gliomas lacking p16 expression exhibit intact p16/CDKN2 gene, suggesting that p16/CDKN2 is down-regulated at the transcriptional level. In this study we investigated whether aberrant p16/CDKN2 gene methylation correlated with absence of p16 expression in the latter group of gliomas. In a series of 27 gliomas, 12 malignant tumors exhibited loss of p16/CDKN2 expression but not gene deletion. Methylation analysis of the CpG island in the 5' region of the p16/CDKN2 gene showed that exon 1 was extensively methylated in six and partially methylated in the other six of the 12 malignant gliomas. In contrast, no methylation was observed in four other malignant gliomas and two low-grade gliomas that expressed p16 protein. These results indicate that abnormal hypermethylation of the CpG island encompassing the 5' end of the p16/CDKN2 gene may be a mechanism of transcriptional silencing in gliomas without homozygous deletions.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas Portadoras/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Regulación hacia Abajo , Glioma/genética , Proteínas Portadoras/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Inmunohistoquímica , Metilación
10.
Oncogene ; 19(1): 2-12, 2000 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-10644974

RESUMEN

Effective anti cancer strategies necessitate the use of agents that target tumor cells rather than normal tissues. In this study, we constructed a tumor-selective adenovirus, Delta24, that carries a 24-bp deletion in the E1A region responsible for binding Rb protein. Immunoprecipitation analyses verified that this deletion rendered Delta24 unable to bind the Rb protein. However, titration experiments in 293 cells demonstrated that the Delta24 adenovirus could replicate in and lyse cancer cells with great efficiency. Lysis of most human glioma cells was observed within 10 - 14 days after infection with Delta24 at 10 PFU/cell. In vivo, a single dose of the Delta24 virus induced a 66.3% inhibition (P<0.005) and multiple injections, an 83.8% inhibition (P<0.01) of tumor growth in nude mice. However, normal fibroblasts or cancer cells with restored Rb activity were resistant to the Delta24 adenovirus. These results suggest that the E1A-mutant Delta24 adenovirus may be clinically and therapeutically useful against gliomas and possibly other cancers with disrupted Rb pathway.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Glioma/terapia , Proteína de Retinoblastoma/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Fase S , Células Tumorales Cultivadas , Replicación Viral
11.
Oncogene ; 15(17): 2049-57, 1997 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-9366522

RESUMEN

Malignant gliomas extensively infiltrate the surrounding normal brain, and their diffuse invasion is one of the most important barriers to successful therapy. Recent studies indicate that the progression of gliomas from low-grade to high-grade may depend on the acquisition of a new phenotype and the subsequent addition of genetic defects. One of the most frequent abnormalities in the progression of gliomas is the inactivation of tumor-suppressor gene p16, suggesting that loss of p16 is associated with acquisition of malignant characteristics. Consistent with this hypothesis, our previous studies showed that restoring wild-type p16 activity into p16-null malignant glioma cells modified their phenotype. In order to understand whether the biological consequences of p16 inactivation in high-grade gliomas included facilitating invasiveness, we used a recombinant replication-deficient adenovirus carrying the cDNA of the p16/CDKN2 gene to infect and express high levels of p16 protein in p16-null SNB19 glioma cells. Invasion of SNB19 glioma cells was tested into two models: invasion of glioma cells through Matrigel-coated transwell inserts and invasion of tumor-cell spheroids into fetal rat-brain aggregates in a co-culture system. Matrigel invasion assays showed that the SNB19 cells expressing exogenous p16 exhibited significantly reduced invasion. Similarly, invasion of p16-treated SNB19 cells into fetal rat-brain aggregates was reduced during a 72 h time period compared to invasion of the adenovirus-control and mock-infected cells. Expression of matrix metalloproteinase-2 (MMP-2), an enzyme involved in tumor-cell invasion, in SNB19 cells expressing p16 was significantly reduced compared to that of parental SNB19 and vector-infected cells. Our results show that restoring wild-type p16 activity into p16-null SNB19 glioma cells significantly inhibits tumor-cell invasion, thus suggesting a novel function of the p16 gene.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Gelatinasas/metabolismo , Técnicas de Transferencia de Gen , Genes Supresores de Tumor/genética , Glioma/genética , Glioma/secundario , Metaloendopeptidasas/metabolismo , Proteínas de Neoplasias/metabolismo , Adenoviridae/genética , Animales , Neoplasias Encefálicas/secundario , Colágeno , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Combinación de Medicamentos , Vectores Genéticos/genética , Glioma/metabolismo , Humanos , Laminina , Metaloproteinasa 2 de la Matriz , Invasividad Neoplásica , Proteoglicanos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína de Retinoblastoma/metabolismo , Esferoides Celulares/patología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
12.
Oncogene ; 34(41): 5295-301, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25619840

RESUMEN

Oncolytic adenoviruses, such as Delta-24-RGD (Δ24RGD), are replication-competent viruses that are genetically engineered to induce selective cancer cell lysis. In cancer cells, Δ24RGD induces massive autophagy, which is required for efficient cell lysis and adenoviral spread. Understanding the cellular mechanisms underlying the regulation of autophagy in cells treated with oncolytic adenoviruses may provide new avenues to improve the therapeutic effect. In this work, we showed that cancer cells infected with Δ24RGDundergo autophagy despite the concurrent activation of the AKT/mTOR pathway. Moreover, adenovirus replication induced sustained activation of JNK proteins in vitro. ERK1/2 phosphorylation remained unchanged during adenoviral infection, suggesting specificity of JNK activation. Using genetic ablation and pharmacological inactivation of JNK, we unequivocally demonstrated that cells infected with Δ24RGD required JNK activation. Thus, genetic co-ablation of JNK1 and JNK2 genes or inhibition of JNK kinase function rendered Δ24RGD-treated cells resistant to autophagy. Accordingly, JNK activation induced phosphorylation of Bcl-2 and prevented the formation of Bcl-2/Beclin 1 autophagy suppressor complexes. Using an orthotopic model of human glioma xenograft, we showed that treatment with Δ24RGD induced phosphorylation and nuclear translocation of JNK, as well as phosphorylation of Bcl-2. Collectively, our data identified JNK proteins as an essential mechanistic link between Δ24RGD infection and autophagy in cancer cells. Activation of JNK without inactivation of the AKT/mTOR pathway constitutes a distinct molecular signature of autophagy regulation that differentiates Δ24RGD adenovirus from the mechanism used by other oncolytic viruses to induce autophagy and provides a new rationale for the combination of oncolytic viruses and chemotherapy.


Asunto(s)
Adenoviridae/fisiología , Autofagia , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Virus Oncolíticos/fisiología , Línea Celular , Humanos , Viroterapia Oncolítica , Transducción de Señal
13.
Arch Neurol ; 56(4): 445-8, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10199333

RESUMEN

Cancer is a disease of a series of genes. Thus, theoretically, brain tumors could be treated by targeting their fundamental molecular defects. Currently, most of the approved clinical protocols for gene therapy involve cancer patients. Several of these protocols are designed to improve the treatment of brain tumors. In this brief report, we analyze the rationale, advantages, and disadvantages of a series of gene therapy approaches against brain tumors that include transfer of tumor suppressor genes and cell-cycle modulators; suicide or prodrug strategies; immunogene therapy; antiangiogenesis; and oncolytic virus therapy. In summary, in this review, we highlight the translational advances in molecular medicine that broaden our battery of therapies for patients with brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Terapia Genética/métodos , Glioma/genética , Glioma/terapia , Proteínas Supresoras de Tumor , Neoplasias Encefálicas/fisiopatología , Ciclo Celular/genética , Factores de Transcripción E2F , Genes Supresores de Tumor/genética , Genes p53 , Glioma/fisiopatología , Humanos , Neovascularización Patológica/genética , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/genética , Profármacos/uso terapéutico , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/genética , Virus/genética
14.
Neurology ; 51(5): 1250-5, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9818841

RESUMEN

The ability to transfer exogenous genes to cancer cells has yielded a wealth of information about the neoplastic processes that occur at molecular and cellular levels. Current research focuses on defining the biochemical factors that govern the interplay between cell growth and cell death in gliomas. The identification of tumor suppressor genes has greatly enhanced our understanding of the molecular mechanism of brain tumors. Accomplishing the transition from basic science to clinical practice is a major challenge for the future of brain tumor research. The concept of tumor suppressor genes is examined, with particular emphasis on the functional studies of the role of the p53, p16, Rb, and PTEN/MMAC1 genes in gliomas. Moreover, recent advances linking tumor suppressor genes, apoptosis, and cell-cycle control pathways in brain tumors are reviewed. The ability to detect mutations in tumor suppressor genes plays an important role in cancer diagnosis and prognosis. Perhaps of greatest significance has been the realization that tumor suppressor genes may provide novel targets for development of specific anticancer therapies for brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Genes Supresores de Tumor , Glioma/genética , Glioma/terapia , Proteínas Supresoras de Tumor , Apoptosis , Neoplasias Encefálicas/patología , Ciclo Celular , Cromosomas Humanos Par 10 , Genes de Retinoblastoma , Genes p16 , Genes p53 , Glioma/patología , Humanos , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/genética
15.
Neurology ; 50(5): 1307-15, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9595979

RESUMEN

OBJECTIVE: This study was conducted to obtain evidence that restoration of the retinoblastoma protein function may have therapeutic application for gliomas. BACKGROUND: The development of glioblastoma multiforme involves progressive inactivation of several tumor suppressor genes. Abnormalities of the retinoblastoma tumor suppressor gene are found in the majority of cancers, including at least 30% of malignant gliomas. No final evidence has been produced about the role of Rb in suppressing glioma growth. METHODS: To address this question, the Ad5CMV-Rb adenovirus carrying a 3.2-kb cDNA of the Rb gene was constructed. Expression of the exogenous protein was assessed by immunoblot and immunohistochemistry analyses. Growth curve assays were used to evaluate the effect of the Rb protein on glioma cell growth. Flow-cytometry analyses were used to analyze the phenotype of the cell cycle after the transfer of Rb. Human glioma xenografts implanted subcutaneously in nude mice were used for the tumorigenicity assay. RESULTS: After the transfer of Rb, 80% of the treated cells expressed high levels of the retinoblastoma protein for at least 7 days. Within 5 days of treatment, the cells lost the neoplastic morphology and showed marked growth suppression. The majority of the Rb-expressing cells were arrested in the G1 phase of the cell cycle. In addition, the restoration of the retinoblastoma activity rendered the human glioma cells unable to form tumors in nude mice. CONCLUSIONS: These findings provide direct evidence that inactivation of the retinoblastoma protein is a critical event in gliomas, and suggest that the restoration of wild-type retinoblastoma activity in these tumors may have therapeutic utility.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Genes de Retinoblastoma , Glioblastoma/terapia , Animales , Ciclo Celular/fisiología , División Celular/fisiología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/fisiología , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Fosforilación , Trasplante Heterólogo , Células Tumorales Cultivadas
16.
Int J Oncol ; 18(2): 343-7, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11172602

RESUMEN

Gliomas are highly resistant to conventional treatment. Improved knowledge of the molecular defects of glioma cells offers new avenues for the development of gene therapy strategies. Transfer of the p53 gene has proven effective in suppressing proliferation in human glioma cell lines. However, several human glioma cell lines are resistant to p53-induced cell death. The E2F family of transcription factors are pivotal for the regulation of cell-cycle and cell-death related genes in gliomas. In the present study, we sought a more effective strategy for glioma treatment by examining the therapeutic potential of the simultaneous transfer of p53 and E2F-2 to gliomas. Trypan blue cell viability assays and flow cytometric cell-cycle analysis demonstrated that the transfer of both p53 and E2F-2 induced cell death in D-54 MG, a p53-resistant glioma cell line. In addition, transfer of E2F-2 did not interfere with the apoptotic properties of exogenous wild-type p53 in U-251 MG cells. Finally, the expression of E2F-2 in D-54 MG cells suppressed the expression of the apoptotic molecule mdm-2 induced by exogenous p53 in these cells. These results show that co-expression of E2F-2 and p53 enhances the anti-cancer effect of p53 in gliomas.


Asunto(s)
Apoptosis/fisiología , Genes p53/fisiología , Glioblastoma/metabolismo , Glioma/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Actinas/metabolismo , Adenoviridae , Factor de Transcripción E2F2 , Glioblastoma/terapia , Glioma/terapia , Humanos , Proteínas Proto-Oncogénicas c-mdm2 , Factores de Transcripción/genética , Células Tumorales Cultivadas/metabolismo
17.
Int J Oncol ; 12(3): 665-9, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9472109

RESUMEN

Malignant gliomas are highly resistant to chemotherapy, in part because of the blood-brain barrier, which restricts the delivery of chemotherapy to certain areas of tumor and their cellular heterogeneity, which leads to the selection and propagation of resistant clones. However, the molecular basis of the drug resistance is poorly understood. In this study, we examined the effect of the cell cycle-inhibitory protein p16 on the chemosensitivity of human glioma cells. Treatment of the p16-null glioma cells, U-251 MG and D-54 MG, with paclitaxel and topotecan, resulted in cell death within 4 days. However, overexpression of exogenous wild-type p16 protein using an adenovirus vector resulted in G1 arrest of glioma cells and resistance to the anticancer effect of paclitaxel or topotecan. Specifically, the p16-expressing cells showed a 30-fold increase in the ID50 of topotecan and a more than 40-fold increase in the ID50 of paclitaxel. These observations indicate that overexpression of molecules that control cell-cycle progression may be partially responsible for causing the resistance of glioma cells to cytocidal drugs.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Glioma/tratamiento farmacológico , Paclitaxel/toxicidad , Topotecan/toxicidad , Adenoviridae , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Fase G1 , Humanos , Paclitaxel/uso terapéutico , Proteínas Recombinantes/biosíntesis , Factores de Tiempo , Topotecan/uso terapéutico , Transfección
18.
Int J Oncol ; 19(2): 359-65, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11445852

RESUMEN

The therapeutic efficacy of standard cancer treatments such as chemotherapy may be improved if they are combined with gene-therapy. Less than 30% of patients with glioblastoma multiforme respond to adjuvant chemotherapy. Actively dividing cells are generally more sensitive to chemotherapy than are non-dividing cells. To determine whether forced cell-cycle progression selectively sensitizes tumor cells to alkylating agents, we examined the effects of overexpressing the E2F-1 protein (a positive regulator of cell-cycle progression) on the sensitivity of two malignant human glioma cell lines, U-251 MG and D-54 MG, to BCNU and temozolomide. Treating these cells with 20-35 microM BCNU or 20-30 microM temozolomide resulted in 50% growth inhibition (IC50) within 4 or 6 days, respectively. By contrast, cells that were first induced to overexpress E2F-1 protein by infection with an adenoviral vector had IC50s that were 37-50% lower. Conversely, transferring the cyclin-dependent kinase inhibitors p16 and p21 to the cells, also by adenoviral infection, produced 3 to 4-fold increases in chemoresistance. Cell-cycle analyses showed that the combination of E2F-1 overexpression and treatment with BCNU or temozolomide increased the proportion of cells in S phase, but the combination of p16 or p21 overexpression and drug treatment reduced the proportion of cells in S phase. These observations suggest that overexpression of genes that positively control cell-cycle progression may be useful for increasing the sensitivity of glioma cells to alkylating agents.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Dacarbazina/farmacología , Glioma/patología , Factores de Transcripción/genética , Adenoviridae/genética , Antineoplásicos Alquilantes/farmacología , Carmustina/farmacología , Supervivencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Dacarbazina/análogos & derivados , Relación Dosis-Respuesta a Droga , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Técnicas de Transferencia de Gen , Glioma/metabolismo , Glioma/terapia , Humanos , Concentración 50 Inhibidora , Sensibilidad y Especificidad , Temozolomida , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
19.
Int J Mol Med ; 3(1): 81-5, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9864390

RESUMEN

Current therapy for glioma is suboptimal. The transfer of apoptosis genes to tumors constitutes one of the most promising strategies for cancer gene therapy. We have previously shown that massive apoptosis occurs when wild-type p53 or E2F-1 expression is induced in glioma. However, the mechanism of action and the efficiency in inducing apoptosis of these two proteins are not similar. Adenovirus-mediated p53 gene transfer is ineffective in causing apoptosis in glioma cells that retain wild-type p53 genotype or overexpress the p21 protein. The p16/Rb/E2F pathway is the most frequent target of genetic alterations in gliomas, and therefore constitutes a suitable target for gene therapy strategies. However, the transfer of either the p16 or Rb gene to glioma cells results in cytostatic effect. The E2F-1 protein is able to induce generalized apoptosis in gliomas independently of the p53, p16 or Rb status. In addition, p21- or p16-mediated growth arrest did not protect glioma cells from E2F-1-mediated apoptosis. The apoptotic molecule bax is induced in p53-mediated apoptosis, but bax is not induced in E2F-1-mediated apoptosis in glioma cells. Careful selection of patients may be necessary before designing therapeutic strategies using either p53 or E2F-1 as a therapeutic tools for glioma patients.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Terapia Genética , Glioma/genética , Apoptosis/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Regulación Neoplásica de la Expresión Génica , Glioma/terapia , Humanos , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
20.
Cancer Gene Ther ; 17(4): 289-97, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19876078

RESUMEN

The fiber-modified adenoviral vector Delta-24-RGD (D24RGD) offers vast therapeutic potential. Direct injection of D24RGD has been used to successfully target ovarian tumors in mice. However, systemic toxicity, especially in the liver, profoundly limits the efficacy of direct viral vector delivery. Mesenchymal stem cells (MSC) have the ability to function as a vector for targeted gene therapy because of their preferential engraftment into solid tumors and participation in tumor stroma formation. We show that MSC-guided delivery of D24RGD is specific and efficient and reduces the overall systemic toxicity in mice to negligible levels compared with D24RGD alone. In our model, we found efficient targeted delivery of MSC-D24RGD to both breast and ovarian cell lines. Furthermore, immunohistochemical staining for adenoviral hexon protein confirmed negligible levels of systemic toxicity in mice that were administered MSC-D24RGD compared with those that were administered D24RGD. These data suggest that delivery of D24RGD through MSC not only increases the targeted delivery efficiency, but also reduces the systemic exposure of the virus, thereby reducing overall systemic toxicity to the host and ultimately enhancing its value as an anti-tumor therapeutic candidate.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Viroterapia Oncolítica , Replicación Viral , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Neoplasias de la Mama/virología , Línea Celular Tumoral , Femenino , Humanos , Técnicas para Inmunoenzimas , Melanoma Experimental/genética , Melanoma Experimental/terapia , Melanoma Experimental/virología , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Neoplasias Ováricas/virología , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA