Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Acta Derm Venereol ; 99(1): 63-71, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30182136

RESUMEN

Deregulated Hedgehog signalling is a driver of basal cell carcinomas. One effector of the Hedgehog pathway is n-MYC. c/n-MYC proteins, NAMPT and DBC1 are linked to SIRT1 in a positive feedback loop that may contribute to tumorigenesis of basal cell carcinoma. In 5 basal cell carcinoma types immunohistochemistry revealed n-MYC, NAMPT and SIRT1 expression. DBC1 was homogenously expressed in all epithelial cells. NAMPT, SIRT1 and c-MYC were expressed in the stratum basale of human and murine skin. In hair follicles NAMPT and SIRT1 were expressed together with c-MYC and n-MYC, except for the matrix, where n-MYC was strongly positive, but c-MYC expression was absent. Therefore, a common pathway connecting n-MYC, NAMPT and SIRT1 may be active in basal cell carcinomas and in their cells of origin. This pathway may contribute to the development of basal cell carcinomas. Targeting factors in the feedback loop may offer novel therapeutic options.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma Basocelular/enzimología , Citocinas/análisis , Células Madre Neoplásicas/enzimología , Nicotinamida Fosforribosiltransferasa/análisis , Proteínas Proto-Oncogénicas c-myc/análisis , Sirtuina 1/análisis , Neoplasias Cutáneas/enzimología , Proteínas Adaptadoras Transductoras de Señales/análisis , Animales , Biopsia , Carcinoma Basocelular/patología , Proteínas de Ciclo Celular , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/análisis , Neoplasias Cutáneas/patología
2.
Proc Natl Acad Sci U S A ; 109(4): E187-96, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22190494

RESUMEN

Silent information regulator 1 (SIRT1) represents an NAD(+)-dependent deacetylase that inhibits proapoptotic factors including p53. Here we determined whether SIRT1 is downstream of the prototypic c-MYC oncogene, which is activated in the majority of tumors. Elevated expression of c-MYC in human colorectal cancer correlated with increased SIRT1 protein levels. Activation of a conditional c-MYC allele induced increased levels of SIRT1 protein, NAD(+), and nicotinamide-phosphoribosyltransferase (NAMPT) mRNA in several cell types. This increase in SIRT1 required the induction of the NAMPT gene by c-MYC. NAMPT is the rate-limiting enzyme of the NAD(+) salvage pathway and enhances SIRT1 activity by increasing the amount of NAD(+). c-MYC also contributed to SIRT1 activation by sequestering the SIRT1 inhibitor deleted in breast cancer 1 (DBC1) from the SIRT1 protein. In primary human fibroblasts previously immortalized by introduction of c-MYC, down-regulation of SIRT1 induced senescence and apoptosis. In various cell lines inactivation of SIRT1 by RNA interference, chemical inhibitors, or ectopic DBC1 enhanced c-MYC-induced apoptosis. Furthermore, SIRT1 directly bound to and deacetylated c-MYC. Enforced SIRT1 expression increased and depletion/inhibition of SIRT1 reduced c-MYC stability. Depletion/inhibition of SIRT1 correlated with reduced lysine 63-linked polyubiquitination of c-Myc, which presumably destabilizes c-MYC by supporting degradative lysine 48-linked polyubiquitination. Moreover, SIRT1 enhanced the transcriptional activity of c-MYC. Taken together, these results show that c-MYC activates SIRT1, which in turn promotes c-MYC function. Furthermore, SIRT1 suppressed cellular senescence in cells with deregulated c-MYC expression and also inhibited c-MYC-induced apoptosis. Constitutive activation of this positive feedback loop may contribute to the development and maintenance of tumors in the context of deregulated c-MYC.


Asunto(s)
Apoptosis/fisiología , Senescencia Celular/fisiología , Citocinas/metabolismo , Retroalimentación Fisiológica/fisiología , Nicotinamida Fosforribosiltransferasa/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cicloheximida , Cartilla de ADN/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Células HEK293 , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , NAD/metabolismo , Proteínas del Tejido Nervioso , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/aislamiento & purificación , Ubiquitinación
3.
Proc Natl Acad Sci U S A ; 105(39): 15046-51, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18818310

RESUMEN

In the majority of human tumors, expression of the c-MYC oncogene becomes constitutive. Here, we report that c-MYC directly regulates the expression of AP4 via CACGTG motifs in the first intron of the AP4 gene. Induction of AP4 was required for c-MYC-mediated cell cycle reentry of anti-estrogen arrested breast cancer cells and mitogen-mediated repression of the CDK inhibitor p21. AP4 directly repressed p21 by occupying four CAGCTG motifs in the p21 promoter via its basic region. AP4 levels declined after DNA damage, and ectopic AP4 interfered with p53-mediated cell cycle arrest and sensitized cells to apoptosis induced by DNA damaging agents. AP4 expression blocked induction of p21 by TGF-beta in human keratinocytes and interfered with up-regulation of p21 and cell cycle arrest during monoblast differentiation. Notably, AP4 is specifically expressed in colonic progenitor and colorectal carcinoma cells. In conclusion, our results indicate that c-MYC employs AP4 to maintain cells in a proliferative, progenitor-like state.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Humanos , Queratinocitos/metabolismo , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/farmacología
4.
Methods Mol Biol ; 2318: 119-160, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019289

RESUMEN

The c-MYC oncogene is activated in ~50% of all tumors and its product, the c-MYC transcription factor, regulates numerous processes, which contribute to tumor initiation and progression. Therefore, the genome-wide characterization of c-MYC targets and their role in different tumor entities is a recurrent theme in cancer research. Recently, next-generation sequencing (NGS) has become a powerful tool to analyze mRNA and miRNA expression, as well as DNA binding of proteins in a genome-wide manner with an extremely high resolution and coverage. Since the c-MYC transcription factor regulates mRNA and miRNA expression by binding to specific DNA elements in the vicinity of promoters, NGS can be used to generate integrated representations of c-MYC-mediated regulations of gene transcription and chromatin modifications. Here, we provide protocols and examples of NGS-based analyses of c-MYC-regulated mRNA and miRNA expression, as well as of DNA binding by c-MYC. Furthermore, we describe the validation of single c-MYC targets identified by NGS . Taken together, these approaches allow an accelerated and comprehensive analysis of c-MYC function in numerous cellular contexts. Ultimately, these analyses will further illuminate the role of this important oncogene.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Proteínas Proto-Oncogénicas c-myc/genética , ADN/genética , Proteínas de Unión al ADN/genética , Expresión Génica , Perfilación de la Expresión Génica/métodos , Genes myc , Humanos , MicroARNs/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN/genética , ARN Mensajero/genética , Análisis de Secuencia de ADN/métodos
5.
Neoplasia ; 21(10): 974-988, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31442917

RESUMEN

We recently described a positive feedback loop connecting c-MYC, NAMPT, DBC1 and SIRT1 that contributes to unrestricted cancer cell proliferation. Here we determine the relevance of the loop for serrated route intestinal tumorigenesis using genetically well-defined BrafV600E and K-rasG12D mouse models. In both models we show that c-MYC and SIRT1 protein expression increased through progression from hyperplasia to invasive carcinomas and metastases. It correlated with high NAMPT expression and was directly associated to activation of the oncogenic drivers. Assessing functional and molecular consequences of pharmacological interference with factors of the loop, we found that inhibition of NAMPT resulted in apoptosis and reduced clonogenic growth in human BRAF-mutant colorectal cancer cell lines and patient-derived tumoroids. Blocking SIRT1 activity was only effective when combined with a PI3K inhibitor, whereas the latter antagonized the effects of NAMPT inhibition. Interfering with the positive feedback loop was associated with down-regulation of c-MYC and temporary de-repression of TP53, explaining the anti-proliferative and pro-apoptotic effects. In conclusion we show that the c-MYC-NAMPT-DBC1-SIRT1 positive feedback loop contributes to murine serrated tumor progression. Targeting the feedback loop exerted a unique, dual therapeutic effect of oncoprotein inhibition and tumor suppressor activation. It may therefore represent a promissing target for serrated colorectal cancer, and presumably for other cancer types with deregulated c-MYC.


Asunto(s)
Transformación Celular Neoplásica , Citocinas/metabolismo , Neoplasias Intestinales/etiología , Neoplasias Intestinales/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Oncogenes , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 1/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Biopsia , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Inmunohistoquímica , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/patología , Ratones , Mutación , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Sirtuina 1/antagonistas & inhibidores
6.
Nucleic Acids Res ; 34(18): e119, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16998186

RESUMEN

Here we describe an episomal, one-vector system which allows the generation of cell populations displaying homogenous, inducible gene inactivation by RNA interference in a one step procedure. A dual tet-repressor/activator system tightly controls a bi-directional promoter, which simultaneously drives expression of microRNAs and a fluorescent marker protein. We demonstrate the effectiveness of this vector by knockdown of p53 expression in a human cell line which resulted in the expected loss of G1-arrest after DNA damage. The generation of a cell pool homogenously expressing the ectopic microRNAs was achieved in 1 week without the need for viral infections. Induction of microRNA expression did not elicit an interferon response. Furthermore, the vector was adapted for convenient ligation-free transfer of microRNA cassettes from public libraries. This conditional knockdown-system should prove useful for many research and gene therapeutic applications.


Asunto(s)
Regulación de la Expresión Génica , Vectores Genéticos , MicroARNs/genética , Plásmidos/genética , Interferencia de ARN , Línea Celular Tumoral , Humanos , MicroARNs/biosíntesis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
7.
Med Oncol ; 36(1): 5, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30460421

RESUMEN

We have recently identified a positive feedback loop in which c-MYC increases silent information regulator 1 (SIRT1) protein level and activity through transcriptional activation of nicotinamide phosphoribosyltransferase (NAMPT) and NAD+ increase. Here, we determined the relevance of the c-MYC-NAMPT-SIRT1 feedback loop, including the SIRT1 inhibitor deleted in breast cancer 1 (DBC1), for the development of conventional and serrated colorectal adenomas. Immunohistochemical analyses of 104 conventional adenomas with low- and high-grade dysplasia and of 157 serrated lesions revealed that elevated expression of c-MYC, NAMPT, and SIRT1 characterized all conventional and serrated adenomas, whereas DBC1 was not differentially regulated. Analyzing publicly available pharmacogenomic databases from 43 colorectal cancer cell lines demonstrated that responsiveness towards a NAMPT inhibitor was significantly associated with alterations in PTEN and TGFBR2, while features such as BRAF or RNF43 alterations, or microsatellite instability typical for serrated route colorectal cancer, showed increased sensitivities for inhibition of NAMPT and SIRT1. Our findings suggest an activation of the c-MYC-NAMPT-SIRT1 feedback loop that may crucially contribute to initiation and development of both routes to colorectal cancer. Targeting of NAMPT or SIRT1 may represent novel therapeutic strategies with putative higher sensitivity of the serrated route colorectal cancer subtype.


Asunto(s)
Adenoma/patología , Neoplasias Colorrectales/patología , Citocinas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 1/metabolismo , Adenoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/metabolismo , Retroalimentación Fisiológica/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Transducción de Señal/fisiología
8.
J Clin Invest ; 110(11): 1717-27, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12464677

RESUMEN

Using microarray analysis, we have detected downregulation of several components of the cGMP signaling pathway during replicative senescence of primary human diploid fibroblasts (HDFs). Therefore, the effect of pharmacological inhibition of cGMP synthesis was analyzed in HDFs. Treatment with 6-anilino-5,8-quinolinequinone (LY83583, referred to as LY hereafter), a previously described inhibitor of guanylate cyclase, induced cellular senescence. Microarray analysis revealed that LY treatment induced the Cdk inhibitor p21(WAF1/SDI/CIP1). In colorectal cancer cells, transcription of p21 was induced by LY in a p53-independent manner. Furthermore, p21, but not p53, was required for inhibition of proliferation by LY. The lack of p53 involvement suggests that LY does not induce DNA damage. Growth inhibition was also observed in malignant melanoma and breast cancer cell lines. Functional inactivation of the retinoblastoma tumor-suppressor protein, an effector of p21-mediated cell-cycle inhibition, converted LY-induced growth arrest to apoptosis. These results suggest that LY, or derivatives, may be useful therapeutic agents for the treatment of tumors.


Asunto(s)
Aminoquinolinas/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/genética , Inhibidores Enzimáticos/farmacología , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , GMP Cíclico/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Recién Nacido , Cinética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
9.
Cancer Res ; 65(10): 4218-27, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15899813

RESUMEN

In many cases, silencing of gene expression by CpG methylation is causally involved in carcinogenesis. Furthermore, cancer-specific CpG methylation may serve as a tumor marker. In order to identify candidate genes for inactivation by CpG methylation in prostate cancer, the prostate cancer cell lines LNCaP, PC3, and Du-145 were treated with 5-aza-2' deoxycytidine and trichostatin A, which leads to reversion of epigenetic silencing. By microarray analysis of 18,400 individual transcripts, several hundred genes were found to be induced when compared with cells treated with trichostatin A. Fifty re-expressed genes were selected for further analysis based on their known function, which implied a possible involvement in tumor suppression. Twelve of these genes showed a significant degree of CpG methylation in their promoters. Six genes were silenced by CpG methylation in the majority of five analyzed prostate cancer cell lines, although they displayed robust mRNA expression in normal prostate epithelial cells obtained from four different donors. In primary prostate cancer samples derived from 41 patients, the frequencies of CpG methylation detected in the promoter regions of these genes were: GPX3, 93%; SFRP1, 83%; COX2, 78%; DKK3, 68%; GSTM1, 58%; and KIP2/p57, 56%. Ectopic expression of SFRP1 or DKK3 resulted in decreased proliferation. The expression of DKK3 was accompanied by attenuation of the mitogen-activated protein kinase pathway. The high frequency of CpG methylation detected in the promoters of the identified genes suggests a potential causal involvement in prostate cancer and may prove useful for diagnostic purposes.


Asunto(s)
Silenciador del Gen , Neoplasias de la Próstata/genética , Proteínas Adaptadoras Transductoras de Señales , Línea Celular Tumoral , Quimiocinas , Islas de CpG , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/metabolismo , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética
10.
Cancer Res ; 62(21): 6255-62, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12414655

RESUMEN

Evasion of cellular senescence is required for the immortal phenotype of tumor cells. The tumor suppressor genes p16(INK4A), pRb, and p53 have been implicated in the induction of cellular senescence. To identify additional genes and pathways involved in the regulation of senescence in prostate epithelial cells (PrECs), we performed serial analysis of gene expression (SAGE). The gene expression pattern of human PrECs arrested because of senescence was compared with the pattern of early passage cells arrested because of confluence. A total of 144,137 SAGE tags representing 25,645 unique mRNA species was collected and analyzed: 157 mRNAs (70 with known function) were up-regulated and 116 (65 with known function) were down-regulated significantly in senescent PrECs (P < 0.05; fold difference >2.5). The differential regulation of an exemplary set of genes during senescence was confirmed by quantitative real-time PCR in PrECs derived from three different donors. The results presented here provide the molecular basis of the characteristic changes in morphology and proliferation observed in senescent PrECs. Furthermore, the differentially expressed genes identified in this report will be instrumental in the further analysis of cellular senescence in PrECs and may lead to the identification of tumor suppressor genes and proto-oncogenes involved in the development of prostate cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Senescencia Celular/genética , Neoplasias de la Próstata/genética , Adolescente , Ciclo Celular/genética , Movimiento Celular/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Matriz Extracelular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Sustancias de Crecimiento/biosíntesis , Sustancias de Crecimiento/genética , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Próstata/citología , Próstata/metabolismo , Próstata/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transcripción Genética
11.
Methods Mol Biol ; 1012: 145-85, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24006064

RESUMEN

The c-MYC oncogene is activated in ~50 % of all tumors, and its product, the c-MYC transcription factor, regulates numerous processes, which contribute to tumor initiation and progression. Therefore, the genome-wide characterization of c-MYC targets and their role in different tumor entities is a recurrent theme in cancer research. Recently, next-generation sequencing (NGS) has become a powerful tool to analyze mRNA and miRNA expression, as well as DNA binding of proteins in a genome-wide manner with an extremely high resolution and coverage. Since the c-MYC transcription factor regulates mRNA and miRNA expression by binding to specific DNA elements in the vicinity of promoters, NGS can be used to generate integrated representations of c-MYC-mediated regulations of gene transcription and chromatin modifications. Here, we provide protocols and examples of NGS-based analyses of c-MYC-regulated mRNA and miRNA expression, as well as of DNA binding by c-MYC. Furthermore, the validation of single c-MYC targets identified by NGS is described. Taken together, these approaches allow an accelerated and comprehensive analysis of c-MYC function in numerous cellular contexts which will further illuminate the role of this important oncogene.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , Inmunoprecipitación de Cromatina/métodos , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , MicroARNs/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/aislamiento & purificación , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN/métodos
12.
J Exp Med ; 210(7): 1331-50, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23752226

RESUMEN

The basic helix-loop-helix transcription factor AP4/TFAP4/AP-4 is encoded by a c-MYC target gene and displays up-regulation concomitantly with c-MYC in colorectal cancer (CRC) and numerous other tumor types. Here a genome-wide characterization of AP4 DNA binding and mRNA expression was performed using a combination of microarray, genome-wide chromatin immunoprecipitation, next-generation sequencing, and bioinformatic analyses. Thereby, hundreds of induced and repressed AP4 target genes were identified. Besides many genes involved in the control of proliferation, the AP4 target genes included markers of stemness (LGR5 and CD44) and epithelial-mesenchymal transition (EMT) such as SNAIL, E-cadherin/CDH1, OCLN, VIM, FN1, and the Claudins 1, 4, and 7. Accordingly, activation of AP4 induced EMT and enhanced migration and invasion of CRC cells. Conversely, down-regulation of AP4 resulted in mesenchymal-epithelial transition and inhibited migration and invasion. In addition, AP4 induction was required for EMT, migration, and invasion caused by ectopic expression of c-MYC. Inhibition of AP4 in CRC cells resulted in decreased lung metastasis in mice. Elevated AP4 expression in primary CRC significantly correlated with liver metastasis and poor patient survival. These findings imply AP4 as a new regulator of EMT that contributes to metastatic processes in CRC and presumably other carcinomas.


Asunto(s)
Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/fisiología , Transición Epitelial-Mesenquimal/fisiología , Factores de Transcripción/fisiología , Animales , Células CACO-2 , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/fisiopatología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/biosíntesis , Transición Epitelial-Mesenquimal/genética , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , ARN Interferente Pequeño/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/biosíntesis , Trasplante Heterólogo , Regulación hacia Arriba/genética
13.
Oncotarget ; 3(10): 1182-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23045412

RESUMEN

Approximately 7.5% of all colorectal cancers are considered to originate from the alternative, serrated route. Here, we investigate the expression of the c-MYC oncogene and the SIRT1 protein deacetylase by immunohistochemical staining in subgroups of colorectal serrated lesions that were characterized by different molecular alterations. The expression of c-MYC and SIRT1 correlated with the presence of KRAS and BRAF mutations and high expression of c-MYC and SIRT1 was strongly associated with higher grades of malignancy. In contrast, in the majority of serrated lesions without KRAS or BRAF mutations, c-MYC and SIRT1 expression was not found increased. In this group only a subset of mostly high grade intraepithelial neoplasia and carcinoma was characterized by elevated c-MYC and SIRT1 expression. This was associated with nuclear localization of beta-catenin, indicating that Wnt pathway activation may confer transcriptional induction of c-MYC. In summary, we established a link between oncogenic K-Ras and B-Raf, suggesting post-transcriptional regulation of c-MYC through MAPK/ERK1/2 pathway activation, as well as for Wnt signalling to the activation of the c-MYC oncogene, and consequently of SIRT1 in the serrated route. The increasing expressions with higher grades of malignancy suggest crucial functions for c-MYC and SIRT1 in the progression of serrated lesions to colorectal cancer. These functions may include antagonizing of apoptosis and senescence, which are characteristic features of serrated lesions.


Asunto(s)
Adenoma/patología , Transformación Celular Neoplásica/patología , Pólipos del Colon/patología , Neoplasias Colorrectales/patología , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-myb/metabolismo , Sirtuina 1/metabolismo , Adenoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/metabolismo , Pólipos del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , ADN de Neoplasias/genética , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Mutación/genética , Clasificación del Tumor , Invasividad Neoplásica , Reacción en Cadena de la Polimerasa , Lesiones Precancerosas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras/genética
14.
Cell Cycle ; 10(24): 4256-71, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22134354

RESUMEN

Recently, the inhibition of epithelial-mesenchymal-transition (EMT) by p53 has been described as a new mode of tumor suppression which presumably prevents metastasis. Here we report that activation of p53 down-regulates the EMT-inducing transcription factor SNAIL via induction of the miR-34a/b/c genes. Suppression of miR-34a/b/c caused up-regulation of SNAIL and cells displayed EMT markers and related features, as enhanced migration and invasion.  Ectopic miR-34a induced mesenchymal-epithelial-transition (MET) and down-regulation of SNAIL, which was mediated by a conserved miR-34a/b/c seed-matching sequence in the SNAIL 3'-UTR. miR-34a also down-regulated SLUG and ZEB1, as well as the stemness factors BMI1, CD44, CD133, OLFM4 and c-MYC. Conversely, the transcription factors SNAIL and ZEB1 bound to E-boxes in the miR-34a/b/c promoters, thereby repressing miR-34a and miR-34b/c expression. Since ectopic miR-34a prevented TGF-ß-induced EMT, the repression of miR-34 genes by SNAIL and related factors is part of the EMT program. In conclusion, the frequent inactivation of p53 and/or miR-34a/b/c found in cancer may shift the equilibrium of these reciprocal regulations towards the mesenchymal state and thereby lock cells in a metastatic state.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Retroalimentación Fisiológica/fisiología , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regiones no Traducidas 3'/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Humanos , Luciferasas , Microscopía Confocal , Mitomicina , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail
15.
Mol Cell Biol ; 28(20): 6171-81, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18710951

RESUMEN

The OTT-MAL/RBM15-MKL1 fusion protein is the result of the recurrent translocation t(1;22) in acute megakaryocytic leukemia in infants. How it contributes to the malignancy is unknown. The 3' fusion partner, MAL/MKL1/MRTF-A, is a transcriptional coactivator of serum response factor (SRF). MAL plays a key role in regulated gene expression depending on Rho family GTPases and G-actin. Here we demonstrate that OTT-MAL is a constitutive activator of SRF and target gene expression. This requires the SRF-binding motif and the MAL-derived transactivation domain. OTT-MAL localizes to the nucleus and is not regulated by upstream signaling. OTT-MAL deregulation reflects its independence from control by G-actin, which fails to interact with OTT-MAL in coimmunoprecipitation experiments. Regulation cannot be restored by reintroduction of the entire MAL N terminus into the fusion protein. OTT-MAL also caused a delayed induction of the MAL-independent, ternary complex factor-dependent target genes c-fos and egr-1 and the mitogen-activated protein kinase/Erk pathway. With testing in heterologous tissue culture systems, however, we observed considerable antiproliferative effects of OTT-MAL. Our data suggest that the deregulated activation of MAL-dependent and -independent promoters results in tissue-specific functions of OTT-MAL.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Fusión Oncogénica/metabolismo , Factor de Respuesta Sérica/metabolismo , Actinas/metabolismo , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HeLa , Humanos , Espacio Intracelular/metabolismo , Ratones , Células 3T3 NIH , Proteínas de Fusión Oncogénica/química , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Factores Complejos Ternarios/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
17.
Proc Natl Acad Sci U S A ; 104(27): 11388-93, 2007 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-17586686

RESUMEN

The p53 tumor suppressor gene encodes a transcription factor, which is translationally and posttranslationally activated after DNA damage. In a proteomic screen for p53 interactors, we found that the cullin protein Cul7 efficiently associates with p53. After DNA damage, the level of Cul7 protein increased in a caffeine-sensitive, but p53-independent, manner. Down-regulation of Cul7 by conditional microRNA expression augmented p53-mediated inhibition of cell cycle progression. Ectopic expression of Cul7 inhibited activation of p53 by DNA damaging agents and sensitized cells to adriamycin. Although Cul7 recruited the F-box protein FBX29 to p53, the combined expression of Cul7/FBX29 did not promote ubiquitination and degradation of p53 in vivo. Therefore, the inhibition of p53 activity by Cul7 is presumably mediated by alternative mechanisms. The interplay between p53 and Cul7 resembles the negative feedback loop described for p53 and Mdm2. Pharmacological modulation of Cul7 function may allow the sensitization of cancer cells expressing wild-type p53 to genotoxic agents used in cancer therapy.


Asunto(s)
Proteínas Cullin/biosíntesis , Proteínas Cullin/genética , Daño del ADN/fisiología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/fisiología , Línea Celular Tumoral , Proteínas Cullin/fisiología , Inducción Enzimática/genética , Humanos
18.
Cell Cycle ; 6(3): 339-52, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17297307

RESUMEN

Here we show that the human BubR1 and MAD2 genes, which encode inhibitors of the anaphase promoting complex (APC/C), are directly activated by the oncogenic transcription factor c-MYC via E-box sequences in their first introns. In colorectal cancer biopsies elevated expression of c-MYC correlated with increased MAD2 levels. Activation of a conditional c-MYC allele delayed progression through mitosis in pro-metaphase in a MAD2- and BubR1-dependent manner. A fraction of the daughter cells derived from extended mitotic events underwent synchronous apoptosis, which was in part mediated by BubR1. Furthermore, c-MYC activation resulted in CIN (chromosomal instability) in the diploid MIN (microsatellite instability) cell line DLD-1 and further enhanced CIN in the aneuploid CIN-line MCF7. Unexpectedly, c-MYC-induced CIN was independent of c-MYC-induced BubR1/MAD2 expression and mitotic delay. Therefore, c-MYC-induced CIN may be caused be alternative pathways. We observed that activation of c-MYC induced DNA double-strand breaks, as evidenced by formation of gamma-H2AX foci, which colocalized with foci of active DNA replication. Furthermore, c-MYC activation resulted in mitotic chromosomes exhibiting DNA damage. Therefore, oncogenic deregulation of c-MYC prevents repair of replication-stress induced DNA lesions in the G(2)-phase. We suggest that the c-MYC-mediated persistence of DNA lesions throughout mitosis leads to chromosomal missegregation and underlies c-MYC-induced CIN. The effects of deregulated c-MYC on progression through mitosis described here may have important implications for the origin of chromosomal instability in many tumor types and the sensitivity towards cancer therapeutic agents targeting DNA or the mitotic spindle.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Daño del ADN , Histonas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/genética , Anafase/genética , Anafase/fisiología , Apoptosis/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Elementos E-Box , Etopósido/metabolismo , Genes myc , Histonas/genética , Humanos , Intrones/genética , Proteínas Mad2 , Mitosis/genética , Prometafase , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Activación Transcripcional
19.
Cell Cycle ; 6(2): 189-98, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17314512

RESUMEN

Malignant melanoma is still poorly understood at the genomic level. Recently, a new technique for the high-resolution analysis of copy number changes named digital karyotyping was introduced. This approach is derived from SAGE (serial analysis of gene expression) and allows the detection of genomic amplifications and deletions, which are indicative of oncogenes and tumor suppressor genes. Four human melanoma cell lines were subjected to analysis by digital karyotyping. 828,780 genomic tags were generated and analyzed quantitatively. Thereby, we identified a somatic, homozygous deletion of 570 kbp removing exons 3-29 of the dystrophin (DMD, Duchenne muscular dystrophy) gene. Analysis of 51 melanoma cell lines further revealed a homozygous and a hemizygous deletion in DMD. Furthermore, DMD mRNA expression was downregulated with respect to primary melanocytes and accompanied by loss of dystrophin protein expression in 38 of 55 (69%) and significantly reduced in 10 of 55 (18%) melanoma cell lines. Sequence analysis of DMD cDNAs in 37 melanoma cell lines revealed six new sequence variants with a significantly lower frequency than previously described DMD polymorphisms, which may affect dystrophin function. Knock-down of DMD enhanced migration and invasion, whereas re-expression of DMD attenuated migration and induced a senescent phenotype in melanoma cell lines. Therefore, loss of DMD may critically change the migratory and proliferative capacity of melanocytes. Taken together, our results suggest that inactivation of DMD is involved in the pathogenesis of malignant melanoma.


Asunto(s)
Distrofina/genética , Silenciador del Gen , Melanoma/genética , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo/genética , Distrofina/metabolismo , Humanos , Cariotipificación/métodos , Melanoma/metabolismo , Polimorfismo Genético/genética , Análisis de Secuencia de ADN
20.
Cell Cycle ; 6(13): 1586-93, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17554199

RESUMEN

In a genome-wide screen for microRNAs regulated by the transcription factor encoded by the p53 tumor suppressor gene we found that after p53-activation the abundance of thirty-four miRNAs was significantly increased, whereas sixteen miRNAs were suppressed. The induction of miR-34a was most pronounced among all differential regulations. Also expression of the primary miR-34a transcript was induced after p53 activation and by DNA damage in a p53-dependent manner. p53 occupied an evolutionarily conserved binding site proximal to the first non-coding exon of miR-34a. Ectopic miR-34a induced apoptosis and a cell cycle arrest in the G1-phase, thereby suppressing tumor cell proliferation. Other p53-induced miRNAs identified here may also have tumor suppressive potential as they are known to suppress the anti-apoptotic factor Bcl2 (miR-15a/16) and the oncogenes RAS and HMGA2 (let-7a). Our results for the first time directly integrate the regulation of miRNA expression into the transcriptional network regulated by p53. siRNAs corresponding to p53-induced miRNAs may have potential as cancer therapeutic agents as RNA interference based therapies are currently emerging.


Asunto(s)
Apoptosis/genética , Fase G1/genética , Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/fisiología , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Mapeo Cromosómico , Daño del ADN/genética , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia de ARN , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA