Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell Death Differ ; 23(1): 64-75, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26024393

RESUMEN

The long terminal repeat (LTR) of human endogenous retrovirus type 9 (ERV9) acts as a germline-specific promoter that induces the expression of a proapoptotic isoform of the tumor suppressor homologue p63, GTAp63, in male germline cells. Testicular cancer cells silence this promoter, but inhibitors of histone deacetylases (HDACs) restore GTAp63 expression and give rise to apoptosis. We show here that numerous additional transcripts throughout the genome are driven by related ERV9-LTRs. 3' Rapid amplification of cDNA ends (3'RACE) was combined with next-generation sequencing to establish a large set of such mRNAs. HDAC inhibitors induce these ERV9-LTR-driven genes but not the LTRs from other ERVs. In particular, a transcript encoding the death receptor DR5 originates from an ERV9-LTR inserted upstream of the protein coding regions of the TNFRSF10B gene, and it shows an expression pattern similar to GTAp63. When treating testicular cancer cells with HDAC inhibitors as well as the death ligand TNF-related apoptosis-inducing ligand (TRAIL), rapid cell death was observed, which depended on TNFRSF10B expression. HDAC inhibitors also cooperate with cisplatin (cDDP) to promote apoptosis in testicular cancer cells. ERV9-LTRs not only drive a large set of human transcripts, but a subset of them acts in a proapoptotic manner. We propose that this avoids the survival of damaged germ cells. HDAC inhibition represents a strategy of restoring the expression of a class of ERV9-LTR-mediated genes in testicular cancer cells, thereby re-enabling tumor suppression.


Asunto(s)
Apoptosis/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Secuencias Repetidas Terminales/genética , Neoplasias Testiculares/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/administración & dosificación , Retrovirus Endógenos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Germinativas , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Humanos , Masculino , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , ARN Mensajero/biosíntesis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Receptores del Factor de Necrosis Tumoral/genética , Neoplasias Testiculares/patología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
3.
Nature ; 523(7560): 352-6, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26009011

RESUMEN

Missense mutations in p53 generate aberrant proteins with abrogated tumour suppressor functions that can also acquire oncogenic gain-of-function activities that promote malignant progression, invasion, metastasis and chemoresistance. Mutant p53 (mutp53) proteins undergo massive constitutive stabilization specifically in tumours, which is the key requisite for the acquisition of gain-of-functions activities. Although currently 11 million patients worldwide live with tumours expressing highly stabilized mutp53, it is unknown whether mutp53 is a therapeutic target in vivo. Here we use a novel mutp53 mouse model expressing an inactivatable R248Q hotspot mutation (floxQ) to show that tumours depend on sustained mutp53 expression. Upon tamoxifen-induced mutp53 ablation, allotransplanted and autochthonous tumours curb their growth, thus extending animal survival by 37%, and advanced tumours undergo apoptosis and tumour regression or stagnation. The HSP90/HDAC6 chaperone machinery, which is significantly upregulated in cancer compared with normal tissues, is a major determinant of mutp53 stabilization. We show that long-term HSP90 inhibition significantly extends the survival of mutp53 Q/- (R248Q allele) and H/H (R172H allele) mice by 59% and 48%, respectively, but not their corresponding p53(-/-) littermates. This mutp53-dependent drug effect occurs in H/H mice treated with 17DMAG+SAHA and in H/H and Q/- mice treated with the potent Hsp90 inhibitor ganetespib. Notably, drug activity correlates with induction of mutp53 degradation, tumour apoptosis and prevention of T-cell lymphomagenesis. These proof-of-principle data identify mutp53 as an actionable cancer-specific drug target.


Asunto(s)
Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Terapia Molecular Dirigida/métodos , Proteínas Mutantes/antagonistas & inhibidores , Estabilidad Proteica , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Aloinjertos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Histona Desacetilasa 6 , Histona Desacetilasas/metabolismo , Humanos , Linfoma/genética , Linfoma/patología , Masculino , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Trasplante de Neoplasias , Estabilidad Proteica/efectos de los fármacos , Tasa de Supervivencia , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Triazoles/farmacología , Triazoles/uso terapéutico , Proteína p53 Supresora de Tumor/genética
4.
Cell Death Dis ; 6: e1634, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25675294

RESUMEN

The p53 family and its cofactors are potent inducers of apoptosis and form a barrier to cancer. Here, we investigated the impact of the supposedly inhibitory member of the apoptosis-stimulating protein of p53, iASPP, on the activity of the p53 homolog TAp73, and its cofactors p300 and CBP. We found that iASPP interacted with and stabilized the histone acetyltransferase p300 and its homolog CBP upon cisplatin treatment. Vice versa, iASPP depletion by shRNA resulted in decreased amounts of p300 and CBP, impaired binding of p300 and TAp73 to target site promoters, reduced induction of pro-apoptotic TAp73 target genes, and impaired apoptosis. Mechanistically, we observed that the p300-regulatory E3 ubiquitin ligase BRMS1 could rescue the degradation of p300 and CBP in cisplatin-treated, iASPP-depleted cells. This argues that iASPP stabilizes p300 and CBP by interfering with their BRMS1-mediated ubiquitination, thereby contributing to apoptotic susceptibility. In line, iASPP overexpression partially abolished the interaction of BRMS1 and CBP upon DNA damage. Reduced levels of iASPP mRNA and protein as well as CBP protein were observed in human melanoma compared with normal skin tissue and benign melanocytic nevi. In line with our findings, iASPP overexpression or knockdown of BRMS1 each augmented p300/CBP levels in melanoma cell lines, thereby enhancing apoptosis upon DNA damage. Taken together, destabilization of p300/CBP by downregulation of iASPP expression levels appears to represent a molecular mechanism that contributes to chemoresistance in melanoma cells.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Represoras/metabolismo , Apoptosis , Proteína de Unión a CREB/genética , Ciclo Celular , Inmunoprecipitación de Cromatina , Proteína p300 Asociada a E1A/genética , Células HEK293 , Humanos , Inmunoprecipitación , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/genética , Melanoma/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Represoras/genética
5.
Cell Death Dis ; 5: e1411, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25210794

RESUMEN

HSP90 inhibition represents a promising route to cancer therapy, taking advantage of cancer cell-inherent proteotoxic stress. The HSP90-inhibitor ganetespib showed benefit in advanced clinical trials. This raises the need to identify the molecular determinants of treatment response. We tested the efficacy of ganetespib on a series of colorectal cancer (CRC)-derived cell lines and correlated their sensitivities with comprehensive gene expression analysis. Notably, the drug concentration required for 50% growth inhibition (IC50) varied up to 70-fold (from 36 to 2500 nM) between different cell lines. Correlating cell line-specific IC50s with the corresponding gene expression patterns revealed a strong association between ganetespib resistance (IC50>500 nM) and high expression of the UDP glucuronosyltransferase 1A (UGT1A) gene cluster. Moreover, CRC tumor samples showed a comparable distribution of UGT1A expression levels. The members of the UGT1A gene family are known as drug-conjugating liver enzymes involved in drug excretion, but their function in tumor cells is hardly understood. Chemically unrelated HSP90 inhibitors, for example, 17-N-allylamino-17-demethoxygeldanamycin (17-AAG), did not show correlation of drug sensitivities with UGT1A levels, whereas the ganetespib-related compound NVP-AUY922 did. When the most ganetespib-resistant cell line, HT29, was treated with ganetespib, the levels of HSP90 clients were unaffected. However, HT29 cells became sensitized to the drug, and HSP90 client proteins were destabilized by ganetespib upon siRNA-mediated UGT1A knockdown. Conversely, the most ganetespib-sensitive cell lines HCT116 and SW480 became more tolerant toward ganetespib upon UGT1A overexpression. Mechanistically, ganetespib was rapidly glucuronidated and excreted in resistant but not in sensitive CRC lines. We conclude that CRC cell-expressed UGT1A inactivates ganetespib and other resorcinolic Hsp90 inhibitors by glucuronidation, which renders the drugs unable to inhibit Hsp90 and thereby abrogates their biological activity. UGT1A levels in tumor tissues may be a suitable predictive biomarker to stratify CRC patients for ganetespib treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/enzimología , Glucuronosiltransferasa/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Triazoles/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/fisiopatología , Glucuronosiltransferasa/genética , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos
6.
Cell Death Dis ; 5: e980, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24384723

RESUMEN

Overexpression of the human epidermal growth factor receptor-2 (HER2) in breast cancer strongly correlates with aggressive tumors and poor prognosis. Recently, a positive correlation between HER2 and MIF (macrophage migration inhibitory factor, a tumor-promoting protein and heat-shock protein 90 (HSP90) client) protein levels was shown in cancer cells. However, the underlying mechanistic link remained unknown. Here we show that overexpressed HER2 constitutively activates heat-shock factor 1 (HSF1), the master transcriptional regulator of the inducible proteotoxic stress response of heat-shock chaperones, including HSP90, and a crucial factor in initiation and maintenance of the malignant state. Inhibiting HER2 pharmacologically by Lapatinib (a dual HER2/epidermal growth factor receptor inhibitor) or CP724.714 (a specific HER2 inhibitor), or by knockdown via siRNA leads to inhibition of phosphoactivated Ser326 HSF1, and subsequently blocks the activity of the HSP90 chaperone machinery in HER2-overexpressing breast cancer lines. Consequently, HSP90 clients, including MIF, AKT, mutant p53 and HSF1 itself, become destabilized, which in turn inhibits tumor proliferation. Mechanistically, HER2 signals via the phosphoinositide-3-kinase (PI3K)-AKT- mammalian target of rapamycin (mTOR) axis to induce activated pSer326 HSF1. Heat-shock stress experiments confirm this functional link between HER2 and HSF1, as HER2 (and PI3K) inhibition attenuate the HSF1-mediated heat-shock response. Importantly, we confirmed this axis in vivo. In the mouse model of HER2-driven breast cancer, ErbB2 inhibition by Lapatinib strongly suppresses tumor progression, and this is associated with inactivation of the HSF1 pathway. Moreover, ErbB2-overexpressing cancer cells derived from a primary mouse ErbB2 tumor also show HSF1 inactivation and HSP90 client destabilization in response to ErbB2 inhibition. Furthermore, in HER2-positive human breast cancers HER2 levels strongly correlate with pSer326 HSF1 activity. Our results show for the first time that HER2/ErbB2 overexpression controls HSF1 activity, with subsequent stabilization of numerous tumor-promoting HSP90 clients such as MIF, AKT and HSF1 itself, thereby causing a robust promotion in tumor growth in HER2-positive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptor ErbB-2/metabolismo , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Femenino , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Humanos , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/genética , Factores de Transcripción/genética
7.
Oncogene ; 30(33): 3612-24, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21441950

RESUMEN

Mutant p53 frequently accumulates in cancer cells and promotes tumor cell invasion, as part of its gain of function. Its accumulation is partially due to enhanced stability, but little is known about how the mRNA levels of mutant p53 can be regulated. Likewise, the impact of cancer therapy on the levels of mutant p53 is poorly understood. We show here that the anthracyclines doxorubicin, daunorubicin and epirubicin further increase the amounts of mutant p53 mRNA and protein in cancer cells. Moreover, we show for the first time that the transcription factor E2F1 associates with the promoter DNA of TP53. Upon genotoxic treatment, E2F1 contributed to the expression of mutant p53, both directly and through induction of TAp73. In contrast, the anthracycline idarubicin and also another topoisomerase inhibitor, etoposide, failed to increase the levels of p53 mRNA, despite their ability to induce the synthesis of TAp73 mRNA. Instead, a natural antisense transcript of TP53, WRAP53, was strongly augmented by idarubicin and etoposide, but only less so by the other anthracyclines under study. RNA corresponding to the first exon of WRAP53 was mainly found in cell nuclei and it reduced the levels of mutant p53. Taken together, this suggests a reciprocal activation pattern of TP53 and WRAP53 by different chemotherapeutics. Reducing the levels of mutant p53 by small-interfering RNA increased chemosensitivity, and idarubicin prevented cell survival more efficiently than the mutant p53-inducing doxorubicin. We conclude that even closely related anthracyclines induce the synthesis of different, opposing transcripts from the TP53 locus. When using these drugs for cancer therapy, the increased levels of mutant p53 may augment its gain of function and thus favor unwanted chemoresistance and tumor progression.


Asunto(s)
Antraciclinas/farmacología , Factor de Transcripción E2F1/fisiología , Genes p53 , Mutación , Neoplasias/genética , Proteína p53 Supresora de Tumor/metabolismo , Células Cultivadas , Progresión de la Enfermedad , Doxorrubicina/farmacología , Humanos , Chaperonas Moleculares , Neoplasias/tratamiento farmacológico , Regiones Promotoras Genéticas , Telomerasa/biosíntesis , Inhibidores de Topoisomerasa/farmacología
8.
Oncogene ; 30(7): 865-75, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-20935676

RESUMEN

Wild-type adenovirus type 5 eliminates p53 through the E1B-55kDa and E4-34kDa gene products. Deletion or mutation of E1B-55kDa has long been thought to confer p53-selective replication of oncolytic viruses. We show here that infection with E1B-defective adenovirus mutants induces massive accumulation of p53, without obvious defects in p53 localization, phosphorylation, conformation and oligomerization. Nonetheless, p53 completely failed to induce its target genes in this scenario, for example, p21/CDKN1A, Mdm2 and PUMA. Two regions of the E1A gene products independently contributed to the suppression of p21 transcription. Depending on the E1A conserved region 3, E1B-defective adenovirus impaired the ability of the transcription factor Sp1 to bind the p21 promoter. Moreover, the amino terminal region of E1A, binding the acetyl transferases p300 and CREB-binding protein, blocked p53 K382 acetylation in infected cells. Mutating either of these E1A regions, in addition to E1B, partially restored p21 mRNA levels. Our findings argue that adenovirus attenuates p53-mediated p21 induction, through at least two E1B-independent mechanisms. Other virus species and cancer cells may employ analogous strategies to impair p53 activity.


Asunto(s)
Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Infecciones por Adenovirus Humanos/genética , Infecciones por Adenovirus Humanos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Acetiltransferasas/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína p300 Asociada a E1A , Regulación Viral de la Expresión Génica , Humanos , Mutación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Transcripción Sp1/metabolismo
9.
Cell Death Differ ; 17(12): 1816-29, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21076477

RESUMEN

The p53 family member p73 is essential for brain development, but its precise role and scope remain unclear. Global p73 deficiency determines an overt and highly penetrant brain phenotype marked by cortical hypoplasia with ensuing hydrocephalus and hippocampal dysgenesis. The ΔNp73 isoform is known to function as a prosurvival factor of mature postmitotic neurons. In this study, we define a novel essential role of p73 in the regulation of the neural stem cell compartment. In both embryonic and adult neurogenesis, p73 has a critical role in maintaining an adequate neurogenic pool by promoting self-renewal and proliferation and inhibiting premature senescence of neural stem and early progenitor cells. Thus, products of the p73 gene locus are essential maintenance factors in the central nervous system, whose broad action stretches across the entire differentiation arch from stem cells to mature postmitotic neurons.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Proteínas de Unión al ADN/fisiología , Células-Madre Neurales/citología , Neurogénesis , Neuronas/citología , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Senescencia Celular , Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Hidrocefalia/patología , Ratones , Ratones Noqueados , Mitosis , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Notch/metabolismo , Fase S , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Cell Death Differ ; 17(3): 452-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19960022

RESUMEN

E2F1 is a positive regulator of cell cycle progression and also a potent inducer of apoptosis, especially when activated by DNA damage. We identified E2F1-inducible microRNAs (miRNAs) by microarray hybridization and found that the levels of miRNAs 449a and 449b, as well as their host gene CDC20B, are strongly upregulated by E2F1. High miR-449 levels were found in testes, lung, and trachea, but not in testicular and other cancer cells. MiR-449a/b structurally resemble the p53-inducible miRNA 34 family. In agreement with a putative tumor-suppressive role, miR-449a as well as miR-34a reduced proliferation and strongly promoted apoptosis by at least partially p53-independent mechanisms. Both miRNAs reduced the levels of CDK6, implying miR-449 in a negative feedback mechanism for E2F1. Moreover, miR-449a and miR-34a diminished the deacetylase Sirt1 and augmented p53 acetylation. We propose that both miRNAs provide a twofold safety mechanism to avoid excessive E2F1-induced proliferation by cell cycle arrest and by apoptosis. While responding to different transactivators, miRNAs 449 and 34 each repress E2F1, but promote p53 activity, allowing efficient cross-talk between two major DNA damage-responsive gene regulators.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular , Factor de Transcripción E2F1/metabolismo , MicroARNs/metabolismo , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Análisis por Micromatrices , Sirtuinas/genética , Sirtuinas/metabolismo
11.
Oncogene ; 29(12): 1773-86, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20023703

RESUMEN

The adenovirus type 5 E1B-55 kDa oncoprotein forms a complex with the tumor suppressor p53 and inactivates it. E1B-55 kDa and p53 are each capable of forming oligomers. We mapped the oligomerization domain of E1B-55 kDa to the central portion of the protein. Disturbing E1B-55 kDa self-association by point mutations at residues 285/286 or 307 not only impairs its intracellular localization to the cytoplasmic clusters, but in addition, its association with p53. Strikingly, tetramerization of p53 is also required for efficient association with E1B-55 kDa. Moreover, two different E1B-55 kDa mutants defective for p53 binding but proficient for oligomerization can trans-complement each other for p53 relocalization. We propose that the homo-oligomerization of each component enables efficient interaction between E1B-55 kDa and p53 through increased avidity.


Asunto(s)
Adenoviridae/fisiología , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/metabolismo , Proteínas E1B de Adenovirus/química , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/fisiología , Apoptosis , Ciclo Celular , Codón de Terminación/genética , Humanos , Peso Molecular , Mutación Puntual , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Eliminación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo
12.
Arch Virol ; 151(6): 1085-92, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16421637

RESUMEN

Adenoviruses are an extensively studied system for modeling oncogenesis and for experimental cancer therapy. The most commonly analyzed virus types are 2 and 5, and little distinction has been made between them in past studies. Adenoviruses used for therapeutic purposes are frequently hybrids between these types, including the prototype dl1520/Onyx015. We tested the replication of the wild-type viruses WtD (a hybrid of the type 2 E1 region and type 5) and dl309 (type 5) in comparison with the mutants dl1520 (hybrid) and dl338 (type 5), the latter two lacking part of the E1B-55 kDa coding region. We found that the hybrid viruses replicated with considerably lower efficiency than their type 5 counterparts in H1299 cells (dl309:WtD = 3-4, dl338:dl1520 > 10). Moreover, adenovirus type 2 E1A expression from the hybrid viruses was strongly reduced in comparison to adenovirus type 5 E1A, as revealed by immunoblot analysis and RT-PCR, providing a potential explanation for the differences in virus yield. Differential E1A expression levels need to be taken into account for the construction of effective therapeutic viruses and when studying viral transformation.


Asunto(s)
Adenoviridae/fisiología , Proteínas E1A de Adenovirus/genética , Expresión Génica , Adenoviridae/genética , Proteínas E1A de Adenovirus/biosíntesis , Línea Celular Tumoral , Vectores Genéticos , Humanos , Immunoblotting , ARN Mensajero/análisis , ARN Viral/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral
13.
Curr Top Microbiol Immunol ; 273: 291-334, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14674605

RESUMEN

The potential use of adenoviruses in therapy against cancer has evoked a rapidly moving field of research. Unlike conventional gene therapy vectors, oncolytic adenoviruses retain the ability to replicate. However, replication is restricted as much as possible to tumor cells, with the aim of eliminating these cells through viral cytotoxicity. The two key issues are to improve the efficiency of virus replication and cell killing while ensuring the specificity of these activities for tumor cells. Wild-type adenoviruses as such may already be usable for cancer therapy. Strategies to further improve efficiency and specificity include the partial or complete removal of viral genes. The idea is that functions carried out by the corresponding gene products are not required for replication in tumor cells, but are needed in normal cells. Accordingly, the removal of genes encoding E1B-55 kDa or E1B-19 kDa, or the mutation of E1A may improve the selective killing of tumor cells. On the other hand, the overexpression of the adenovirus death protein (ADP) may enhance viral spread and oncolytic efficiency. Other strategies to improve the specific oncolytic activity of replicating adenoviruses have been pursued. For instance, some promoters are active specifically in tumor cells, and these promoters were introduced into the viral genome, to regulate essential viral genes. Moreover, replicating viruses were engineered to express toxic proteins or drug converters. A number of these viruses have been tested successfully using tumor xenografts in nude mice as a model system. An oncolytic adenovirus lacking the E1B-55 kDa gene product, termed dl1520 or ONYX015, was injected into squamous cell carcinomas of head and neck in phase II clinical trials, and the results were encouraging when chemotherapy was applied in parallel. In the future, further progress might be achieved on the level of virus constructs, but also by refining and adjusting simultaneous conventional therapies, and by standardizing the assessment of the clinical outcome. Recent progress has been made towards the use of replicating virus constructs in cancer therapy. The goal of these developments is to remove cancerous cells from patients with the help of viruses that selectively replicate in these cells. These viruses are generally termed oncolytic viruses. Some convenient properties of adenovirus make this virus particularly useful for this purpose. It infects a large number of human cell types, especially epithelial cells, which give rise to the vast majority of human malignancies. It can be grown easily and to high titers, and the creation of virus recombinants is well established. Finally, a large body of basic research has already been carried out on this virus, facilitating its manipulation. Various approaches to use adenovirus as a cancer drug have been reviewed (Alemany et al. 1999a, 2000; Curiel 2000; Galanis et al. 2001b; Gromeier 2001; Heise and Kirn 2000; Kirn 2000a; Kirn et al. 2001; Kirn and McCormick 1996; Smith and Chiocca 2000; Sunamura 2000; Wells 2000; Wodarz 2001). The aim of this chapter is to provide an integrated overview of these strategies.


Asunto(s)
Adenovirus Humanos/genética , Terapia Genética , Neoplasias/terapia , Replicación Viral , Adenovirus Humanos/fisiología , Animales , Vectores Genéticos/genética , Humanos
14.
Cancer Res ; 61(15): 5941-7, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11479237

RESUMEN

The adenoviral oncoproteins E1B-55 kDa and E4orf6 inactivate and destabilize the tumor suppressor protein p53, thereby contributing to malignant transformation. However, it is unclear whether the elimination of p53 also contributes to the efficiency of viral replication. Furthermore, it is controversial whether adenoviruses with a deletion in the E1B-55 kDa-coding region might selectively replicate in cells with a mutation or deletion of the p53 gene and, therefore, represent a tool in cancer therapy. To address the role of p53 in virus replication, amino acid substitutions were introduced into the NH(2)-terminal portion of p53, replacing residues 24-28 with the corresponding sequence of the human p53-homologue p73. This replacement leaves p53 transcriptionally active but renders the modified protein, termed p53mt24-28, completely resistant to inhibition and degradation by adenoviral oncoproteins. Surprisingly, even strong overexpression of p53 or p53mt24-28 allowed the virus to replicate as efficiently as in the absence of p53 proteins, both in tumor cells and in primary endothelial cells. Also, p53 or p53mt24-28 did not reduce the amount of virus released from infected cells. These observations were made in primary cells or in cell lines that were capable of expressing the p53-agonist p14ARF. Thus, active p53 does not inhibit the growth of adenovirus. Alternative strategies should be used to improve the utility of adenoviruses in cancer therapy.


Asunto(s)
Adenoviridae/fisiología , Proteína p53 Supresora de Tumor/fisiología , Replicación Viral , Adenoviridae/genética , Proteínas E1B de Adenovirus/fisiología , Línea Celular Transformada , Transformación Celular Viral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/metabolismo , Endotelio Vascular/virología , Regulación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Biosíntesis de Proteínas , Proteínas Recombinantes de Fusión , Transfección , Proteína Tumoral p73 , Proteína p14ARF Supresora de Tumor , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor
15.
J Virol ; 75(11): 5391-7, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11333923

RESUMEN

Promyelocytic leukemia (PML) oncogenic domains (PODs) accumulate the transcriptional cofactor named CREB binding protein (CBP) and have been suggested to function as centers of transcription. Transcriptional activation by nuclear hormones, such as glucocorticoids, is augmented by the key constituent of PODs, the PML protein, and decreased by the POD-associated Tax protein of human T-cell leukemia virus type 1 (HTLV-1). This led to the hypothesis that intact PODs might play a positive role in the activation of these promoters. We report here that transiently expressed E4orf3 protein of adenovirus type 5, immediate-early protein 1 of human cytomegalovirus, and the PML-retinoic acid receptor fusion protein from leukemia cells each redistribute CBP within the nucleus. However, unlike the Tax protein of HTLV-1, these factors did not inhibit a glucocorticoid-inducible promoter but strongly enhanced its activity. Thus, at least glucocorticoid-induced transcription does not depend on POD integrity.


Asunto(s)
Proteínas E4 de Adenovirus/fisiología , Glucocorticoides , Proteínas Inmediatas-Precoces/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Regiones Promotoras Genéticas/fisiología , Adenoviridae/química , Proteínas E4 de Adenovirus/genética , Citomegalovirus/química , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas de Neoplasias/genética , Proteína de la Leucemia Promielocítica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Proteínas Supresoras de Tumor
16.
Oncogene ; 19(14): 1834-42, 2000 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-10777217

RESUMEN

The p53 tumor suppressor protein induces apoptosis through a mechanism that may involve the transcriptional activation of cellular genes, including the PIG3 gene. A p53 protein lacking the proline-rich region (p53delta62-91) induces many p53-responsive genes but not PIG3. In parallel, this mutant induces growth arrest but not apoptosis. We show here that the replacement of the N-terminal (amino acids 1-80) or C-terminal (amino acids 344-393) domains of p53 with heterologous domains does not interfere with transcription from the PIG3 promoter, but these chimeras still require the proline-rich region for PIG3 activation. The p53-homolog p73beta also activated the PIG3 promoter, but in contrast to p53, the proline-rich domain of p73beta (residues 81-113) was dispensable to induce the PIG3 promoter. Some tumor-derived p53-mutants, especially M246I, retained the ability to activate transcription of mdm2 but specifically failed to induce the PIG3 promoter, thus resembling p53delta62-91. Further, p53delta62-91 and p53M246I were defective for induction of apoptosis. Finally, p53delta62-91 and p53M246I both showed reduced binding to the DNA of the PIG3 promoter and also to the DNA of the mdm2 and p21 promoters in vitro. Correspondingly, at low expression levels, p53delta62-91 and p53M246I poorly activated the mdm2 promoter when compared to wild type p53. Our results suggest that the proline-rich domain of p53 affects the ability of the central domain to bind DNA. Moreover, some tumor-derived mutations within the central DNA binding domain of p53 mimic the loss of the proline-rich domain.


Asunto(s)
Prolina/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Sitios de Unión , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes Supresores de Tumor , Humanos , Proteínas Nucleares/metabolismo , Fenotipo , Fosforilación , Prolina/genética , Regiones Promotoras Genéticas , Eliminación de Secuencia , Activación Transcripcional , Células Tumorales Cultivadas , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor
17.
J Virol ; 74(2): 764-72, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10623738

RESUMEN

During the late phase of adenovirus infection, viral mRNA is efficiently transported from the nucleus to the cytoplasm while most cellular mRNA species are retained in the nucleus. Two viral proteins, E1B-55 kDa and E4orf6, are both necessary for these effects. The E4orf6 protein of adenovirus type 5 binds and relocalizes E1B-55 kDa, and the complex of the two proteins was previously shown to shuttle continuously between the nucleus and cytoplasm. Nucleocytoplasmic transport of the complex is achieved by a nuclear export signal (NES) within E4orf6. Mutation of this signal sequence severely reduces the ability of the E1B-55 kDa-E4orf6 complex to leave the nucleus. Here, we examined the role of functional domains within E4orf6 during virus infection. E4orf6 or mutants derived from it were transiently expressed, followed by infection with recombinant adenovirus lacking the E4 region and determination of virus yield. An arginine-rich putative alpha helix near the carboxy terminus of E4orf6 contributes to E1B-55 kDa binding and relocalization as well as to the synthesis of viral DNA, mRNA, and proteins. Further mutational analysis revealed that mutation of the NES within E4orf6 considerably reduces its ability to support virus production. The same effect was observed when nuclear export was blocked with a competitor. Further, a functional NES within E4orf6 contributed to the efficiency of late virus protein synthesis and viral DNA replication, as well as total and cytoplasmic accumulation of viral late mRNA. Our data support the view that NES-mediated nucleocytoplasmic shuttling strongly enhances most, if not all, intracellular activities of E4orf6 during the late phase of adenovirus infection.


Asunto(s)
Proteínas E4 de Adenovirus/metabolismo , Adenovirus Humanos/fisiología , Proteínas de la Cápside , Señales de Clasificación de Proteína , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Replicación Viral , Proteínas E1A de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/genética , Adenovirus Humanos/genética , Adenovirus Humanos/crecimiento & desarrollo , Sitios de Unión , Transporte Biológico , Cápside/biosíntesis , Núcleo Celular , Citoplasma/metabolismo , Citoplasma/virología , Replicación del ADN , ADN Viral/biosíntesis , Células HeLa , Humanos , Mutagénesis
18.
J Virol ; 74(1): 193-202, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10590106

RESUMEN

The p53 tumor suppressor protein represents a target for viral and cellular oncoproteins, including adenovirus gene products. Recently, it was discovered that several proteins with structural and functional homologies to p53 exist in human cells. Two of them were termed p51 and p73. We have shown previously that the E1B 55-kDa protein (E1B-55 kDa) of adenovirus type 5 (Ad5) binds and inactivates p53 but not p73. Further, p53 is rapidly degraded in the presence of E1B-55 kDa and the E4orf6 protein of this virus. Here, it is demonstrated that p51 does not detectably associate with E1B-55 kDa. While p53 is relocalized to the cytoplasm by E1B-55 kDa, p51's location is unaffected. Finally, p51 retains its full transcriptional activity in the presence of E1B-55 kDa. Apparently, p51 does not represent a target of Ad5 E1B-55 kDa, suggesting that the functions of p51 are distinct from p53-like tumor suppression. E1B-55 kDa from highly oncogenic adenovirus type 12 (Ad12) was previously shown to surpass the oncogenic activity of Ad5 E1B-55 kDa in various assay systems, raising the possibility that Ad12 E1B-55 kDa might target a broader range of p53-like proteins. However, we show here that Ad12 E1B-55 kDa also inhibits p53's transcriptional activity without measurably affecting p73 or p51. Moderate inhibition of p51's transcriptional activity was observed in the presence of the E4orf6 proteins from Ad5 and Ad12. p53 and Ad12-E1B-55 kDa colocalize in the nucleus and also in cytoplasmic clusters when transiently coexpressed. Finally, E1B-55 kDa and E4orf6 of Ad12 mediate rapid degradation of p53 with an efficiency comparable to that of the Ad5 proteins in human and rodent cells. Our results suggest that E1B-55 kDa of either virus type has similar effects on p53 but does not affect p73 and p51.


Asunto(s)
Proteínas E1B de Adenovirus/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas , Transactivadores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Secuencia de Bases , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Cartilla de ADN , Genes Supresores de Tumor , Humanos , Sistemas de Lectura Abierta , Factores de Transcripción , Transcripción Genética , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
19.
Oncogene ; 18(12): 2101-6, 1999 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-10321734

RESUMEN

The p73beta protein shares structural and functional similarities with the tumor suppressor gene product p53. Both proteins activate transcription from p53-responsive promoters. p53's activity is antagonized by the mdm2 protein (also termed hdm2 in human cells). Complex formation between p53 and mdm2 results in p53's transcriptional inactivation and destabilization. Here we show that overexpression of mdm2 reduces p73beta's ability to activate transcription, too. The mdm2 protein forms a specific complex with p73beta in vitro with an efficiency comparable to p53-binding. Further, both p73beta and p53 relocalize a transport-defective mutant of mdm2 from the cytoplasm to the nucleus, arguing that complex formation occurs in vivo as well. Mutational analysis suggests that the interaction between p73beta and mdm2 follows structural principles analogous to the p53-mdm2-complex. Whereas p53 is destabilized in the presence of mdm2, the amount of intracellular p73beta was not detectably reduced by mdm2. The carboxyterminal RING finger domain of mdm2 was found to be required to reduce the intracellular abundance of p53, but it was dispensable for transcriptionally inactivating either p53 or p73beta. Our results suggest that the autoregulatory feedback loop between p53 and mdm2 also controls p73's activity, but that mdm2-mediated protein degradation is unique to p53.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Activación Transcripcional , Transporte Biológico , Compartimento Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Genes Supresores de Tumor , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
20.
J Virol ; 73(3): 2253-62, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9971808

RESUMEN

The E1B-55-kDa protein of adenovirus type 5 and the p53 tumor suppressor gene product form a complex that localizes to the cytoplasm, thereby downregulating p53's transcriptional activity. The E4orf6 protein binds and relocalizes E1B-55-kDa, and the proteins act synergistically to inactivate p53. We show that another adenovirus E4 gene product, E4orf3, is also sufficient to relocalize E1B-55-kDa from the cytoplasm to the nucleus. Both proteins are then found in discrete nuclear structures (tracks) that are known to contain components of the promyelocytic leukemia-associated nuclear structure. Simultaneously, p53 is dissociated from E1B-55-kDa and is found evenly distributed over the nucleoplasm. In the presence of E4orf3, p53-dependent transcriptional activity is no longer repressed by E1B-55-kDa. When E1B-55-kDa is coexpressed with E4orf3 and E4orf6, E1B-55-kDa is found to colocalize with E4orf6 rather than E4orf3. In parallel, p53 is inhibited and degraded by the combination of E1B-55-kDa and E4orf6, regardless of coexpressed E4orf3. This suggests that the effects of E4orf6 on E1B-55-kDa overrule the actions of E4orf3. When cells are infected with virus expressing E4orf3 but not E4orf6, E1B is found in the cell nucleus and p53 enters the virus replication centers. After infection with wild-type adenovirus, E4orf3 is expressed before E4orf6 and E1B temporarily colocalizes with E4orf3 in nuclear tracks before associating with E4orf6. We propose that during adenovirus infection, the E4orf3 protein transiently liberates p53 from its association with E1B-55-kDa. Subsequently, p53 is inactivated and degraded by the combination of E1B-55-kDa and E4orf6.


Asunto(s)
Proteínas E1B de Adenovirus/fisiología , Proteínas E4 de Adenovirus/fisiología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Núcleo Celular/química , Células Cultivadas , Replicación del ADN , Peso Molecular , Proteína p53 Supresora de Tumor/análisis , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA