Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(11): e0215117, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31714915

RESUMEN

PURPOSE: Cervical cancer remains the second most common cancer and cancer-related death among women in Ethiopia. This is the first study, to our knowledge, describing the demographic, and clinicopathologic characteristics of cervical cancer cases in a mainly rural, Southwestern Ethiopian population with a low literacy rate to provide data on the cervical cancer burden and help guide future prevention and intervention efforts. METHODS: A descriptive analysis of 154 cervical cancer cases at the Jimma University Teaching Hospital in Southwestern Ethiopia from January 2008 -December 2010 was performed. Demographic and clinical characteristics were obtained from patient questionnaires and cervical punch biopsies were histologically examined. RESULTS: Of the 154 participants with a histopathologic diagnosis of cervical cancer, 95.36% had not heard of cervical cancer and 89.6% were locally advanced at the time of diagnosis. Moreover, 86.4% of participants were illiterate, and 62% lived in a rural area. CONCLUSION: A majority of the 154 women with cervical cancer studied at the Jimma University Teaching Hospital in Southwestern Ethiopia were illiterate, had not heard of cervical cancer and had advanced disease at the time of diagnosis. Given the low rates of literacy and knowledge regarding cervical cancer in this population which has been shown to correlate with a decreased odds of undergoing screening, future interventions to address the cervical cancer burden here must include an effective educational component.


Asunto(s)
Neoplasias del Cuello Uterino/epidemiología , Adulto , Etiopía/epidemiología , Femenino , Conocimientos, Actitudes y Práctica en Salud , Humanos , Alfabetización , Tamizaje Masivo , Persona de Mediana Edad , Población Rural , Encuestas y Cuestionarios , Neoplasias del Cuello Uterino/diagnóstico
2.
Methods Mol Biol ; 1249: 121-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25348302

RESUMEN

Retroviral gene transfer is a highly efficient and effective method of stably introducing genetic material into the genome of specific cell types. The process involves the transfection of retroviral expression vectors into a packaging cell line, the isolation of viral particles, and the infection of target cell lines. Compared to traditional gene transfer methods such as liposome-mediated transfection, retroviral gene transfer allows for stable gene expression in cell populations without the need for lengthy selection and cloning procedures. This is particularly helpful when studying gene products that have negative effect on cell growth and viability. Here, we describe the retroviral transfer of cystatin cDNAs using HEK293-derived Phoenix packaging cells and human HeLa cervical carcinoma cells as target cells.


Asunto(s)
Cistatinas/genética , Expresión Génica , Técnicas de Transferencia de Gen , Retroviridae/metabolismo , Células HEK293 , Células HeLa , Humanos , Estándares de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética , Transfección , Ensamble de Virus
3.
J Gerontol A Biol Sci Med Sci ; 66(7): 723-31, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21636832

RESUMEN

Senescent cells exhibit altered expression of numerous genes. Identifying the significance of the changes in gene expression may help advance our understanding of the senescence biology. Here, we report on the consistent and strong upregulation of CST1 expression during cellular senescence, independent of the initial trigger. CST1 expression at both the messenger RNA and protein levels was barely detected in control cells, which included early passage proliferating, quiescent, or immortal human fibroblasts and various human tumor cell lines. Immunoblotting and immunofluorescence cytochemical studies further suggest that CST1 accumulates intracellularly, within vesicular structures. We discuss these results in light of the known function of CST1 as a potent inhibitor of lysosomal cysteine proteases.


Asunto(s)
Envejecimiento/genética , Senescencia Celular/genética , Expresión Génica , ARN/genética , Cistatinas Salivales/genética , Envejecimiento/patología , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Inmunohistoquímica , Cistatinas Salivales/biosíntesis
4.
J Reprod Immunol ; 81(1): 25-33, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19493573

RESUMEN

Trophoblast cells and many cancer cells that harbor foreign antigens may evade immunity by epigenetic silencing of key immune response genes, including MHC class I and II and CD40. Chromatin active agents, such as histone deacetylase inhibitors (HDACi), induce immune response gene expression but often the expression levels are low and the cells lack a robust antigen presentation response. We show here that pre-treatment of trophoblast cells and certain cancer cells with agents that activate stress pathways (Ras oncogene, PMA or H2O2) and induce senescence can substantially enhance the induction of immune response genes (MHC class II, CD40, MICA, MICB) by HDACi and restore a vigorous IFN-gamma response in trophoblast cells and tumor cells. These results could potentially impact the development of novel anti-cancer therapeutic strategies.


Asunto(s)
Senescencia Celular/inmunología , Coriocarcinoma/inmunología , Trofoblastos/inmunología , Neoplasias Uterinas/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Coriocarcinoma/tratamiento farmacológico , Coriocarcinoma/genética , Coriocarcinoma/patología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/inmunología , Femenino , Silenciador del Gen/efectos de los fármacos , Silenciador del Gen/inmunología , Genes MHC Clase I/genética , Genes MHC Clase I/inmunología , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Células HeLa , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/farmacología , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Estrés Oxidativo/inmunología , Embarazo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/inmunología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Trofoblastos/patología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
5.
Cancer Res ; 67(14): 6657-64, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638875

RESUMEN

The evolutionarily conserved TREX (Transcription/Export) complex physically couples transcription, messenger ribonucleoprotein particle biogenesis, RNA processing, and RNA export for a subset of genes. HPR1 encodes an essential component of the S. cerevisiae TREX complex. HPR1 loss compromises transcriptional elongation, nuclear RNA export, and genome stability. Yet, HPR1 is not required for yeast viability. Thoc1 is the recently discovered human functional orthologue of HPR1. Thoc1 is expressed at higher levels in breast cancer than in normal epithelia, and expression levels correlate with tumor size and metastatic potential. Depletion of Thoc1 protein (pThoc1) in human cancer cell lines compromises cell proliferation. It is currently unclear whether Thoc1 is essential for all mammalian cells or whether cancer cells may differ from normal cells in their dependence on Thoc1. To address this issue, we have compared the requirements for Thoc1 in the proliferation and survival of isogenic normal and oncogene-transformed cells. Neoplastic cells rapidly lose viability via apoptotic cell death on depletion of pThoc1. Induction of apoptotic cell death is coincident with increased DNA damage as indicated by the appearance of phosphorylated histone H2AX. In contrast, the viability of normal cells is largely unaffected by pThoc1 loss. Normal cells lacking Thoc1 cannot be transformed by forced expression of E1A and Ha-ras, suggesting that Thoc1 may be important for neoplastic transformation. In sum, our data are consistent with the hypothesis that cancer cells require higher levels of pThoc1 for survival than normal cells. If true, pThoc1 may provide a novel molecular target for cancer therapy.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/fisiología , Apoptosis , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica , Daño del ADN , Proteínas de Unión al ADN , Evolución Molecular , Fibroblastos/metabolismo , Genoma , Células HeLa , Humanos , Modelos Biológicos , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas de Unión al ARN
6.
J Biol Chem ; 282(4): 2666-75, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17135242

RESUMEN

Activated Ras signaling can induce a permanent growth arrest in osteosarcoma cells. Here, we report that a senescence-like growth inhibition is also achieved in human carcinoma cells upon the transduction of H-Ras(V12). Ras-induced tumor senescence can be recapitulated by the transduction of activated, but not wild-type, MEK. The ability for H-Ras(V12) to suppress tumor cell growth is drastically compromised in cells that harbor endogenous activating ras mutations. Notably, growth inhibition of tumor cells containing ras mutations can be achieved through the introduction of activated MEK. Tumor senescence induced by Ras signaling can occur in the absence of p16 or Rb and is not interrupted by the inactivation of Rb, p107, or p130 via short hairpin RNA or the transduction with HPV16 E7. In contrast, inactivation of p21 via short hairpin RNA disrupts Ras-induced tumor senescence. In summary, this study uncovers a senescence-like program activated by Ras signaling to inhibit cancer cell growth. This program appears to be intact in cancer cells that do not harbor ras mutations. Moreover, cancer cells that carry ras mutations remain susceptible to tumor senescence induced by activated MEK. These novel findings can potentially lead to the development of innovative cancer intervention.


Asunto(s)
Senescencia Celular , Neoplasias , Transducción de Señal , Proteínas ras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas , Mutación , Neoplasias/metabolismo , Neoplasias/patología , Proteínas ras/genética
7.
Carcinogenesis ; 27(2): 350-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16280331

RESUMEN

In contrast to rodent cells, normal human fibroblasts are generally resistant to neoplastic transformation in vitro. Here, we report the derivation and characterization of a spontaneously transformed cell line from normal human IMR90 fibroblasts transduced with E1A and Ras oncogenes. Unlike the parental, non-tumorigenic E1A/Ras-expressing IMR90 cells, these spontaneously transformed cells displayed aberrant growth potential in vitro and were capable of tumorigenesis in vivo. In contrast to the parental E1A/Ras-expressing cells, both the spontaneously transformed cells and cells derived from resultant tumors displayed specific t(7q;8q) and t(5q;17) structural chromosomal changes. Chromosome 8q contains c-Myc, which is capable of activating the telomerase catalytic subunit hTERT. Notably, upregulation of c-Myc, hTERT and telomerase activity were detected only in the tumorigenic cells. Transduction of Myc siRNA into the tumorigenic cells led to a concomitant downregulation of hTERT. Furthermore, transduction of Myc or hTERT into the non-tumorigenic E1A/Ras-expressing IMR90 cells was able to confer tumorigenesis on these cells. These studies suggest that the t(7;8) translocation may result in Myc overexpression and its subsequent activation of hTERT, which may contribute to the tumorigenicity of the IMR90 cells. Furthermore, this report describes additional successful neoplastic transformation of human IMR90 fibroblasts by defined genetic elements. The spontaneously transformed cells we have derived provide a valuable model system for the study of neoplastic transformation.


Asunto(s)
Transformación Celular Neoplásica , Fibroblastos , Transducción Genética , Proteínas E1A de Adenovirus/fisiología , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Genes myc , Genes ras , Predisposición Genética a la Enfermedad , Humanos , Telomerasa/biosíntesis , Translocación Genética , Células Tumorales Cultivadas
8.
J Cell Biochem ; 94(6): 1135-47, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15668906

RESUMEN

Upregulation of the p16 tumor suppressor is a hallmark of senescence in human fibroblasts. In this study, we investigated potential protein modification of p16 in senescent human fibroblasts using 2D SDS-PAGE analysis. Three distinct p16 variants with isoelectric points of 5.2, 5.4, and 5.6, were consistently detected in normal human IMR90 fibroblasts that had undergone senescence due to forced expression of oncogenic H-ras or culture passage. Moreover, in contrast to short-term serum starvation, which induces quiescence, IMR90 fibroblasts cultured in low serum for a prolonged period exhibited senescent phenotypes and expression of the three p16 variants. All three p16 variants are unlikely phosphoproteins since they failed to react with antibodies against phospho-serine, and were resistant to the treatment with phosphatases. Functionally, co-immunoprecipitation assays using antibodies against cdk4 and/or cdk6 revealed that only the two most acidic p16 variants associated with cdk4/6. Moreover, senescence induced by the forced expression of p16 in early passage IMR90 fibroblasts or osteosarcoma U2OS cells was accompanied by expression of the two most acidic p16 variants, which also associated with cdk4/6. In summary, we report that prolonged serum starvation-induced senescence may provide an additional model for studying biochemical changes in senescence, including p16 regulation. Furthermore, induction of endogenous p16 in senescent human fibroblasts correlates with the expression of three distinct p16 variants independent of protein phosphorylation. Lastly, expression of the two cdk-bound variants is sufficient to induce senescence in human cells.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Western Blotting , Medio de Cultivo Libre de Suero , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Humanos , Inmunoprecipitación , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo
9.
Cell Cycle ; 3(9): 1201-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15492501

RESUMEN

Several studies have shown that forced expression of oncogenic H-ras can induce a senescence-like permanent growth arrest in normal cells. Here we report that expression of oncogenic H-ras in human osteosarcoma U2OS cells also resulted in a senescence-like flat and enlarged cell morphology and permanent growth arrest. In contrast to normal human fibroblasts, U2OS cells were arrested independently of the p16 and ARF tumor suppressors. Treatment with a MEK inhibitor or a p38MAPK inhibitor interrupted oncogenic H-ras-induced growth arrest in U2OS cells, suggesting that activation of MAPK pathways is important. To further determine whether this process is unique to oncogenic H-ras signaling, we examined the effect of oncogenic K-ras on normal cells and human osteosarcoma cells. Similar to oncogenic H-ras, oncogenic K-ras also induced senescence in normal fibroblasts, while transforming immortalized mouse fibroblasts. However, in contrast to oncogenic H-ras, oncogenic K-ras failed to induce a permanent growth arrest in osteosarcoma U2OS cells. Additionally, cells transduced with oncogenic K-ras exhibited distinguishable cellular changes compared to those transduced with oncogenic H-ras. In summary, we report for the first time that oncogenic H-ras signaling can trigger a senescence-like growth arrest in tumor cells, independent of the p16 and ARF tumor suppressors. This result suggests that tumor cells may harbor a senescence-like program that can be activated by ras signaling. Moreover, our study uncovered a cell type-dependent differential response to oncogenic K-ras, as compared to oncogenic H-ras.


Asunto(s)
Senescencia Celular/genética , Genes cdc/fisiología , Genes ras/genética , Neoplasias/genética , Neoplasias/metabolismo , Proteínas ras/genética , Animales , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Forma de la Célula/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Senescencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Genes cdc/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción Genética , Proteínas ras/metabolismo
10.
Oncogene ; 23(57): 9238-46, 2004 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-15489886

RESUMEN

Senescence irreversibly arrests the proliferation of cells that have sustained significant cellular stress. Replicative senescence, due to the shortening and dysfunction of telomeres, appears to provide a barrier to the immortalization of cells and development of cancer. In normal human fibroblasts, senescence induced by oncogenic H-ras displays a nearly identical cellular phenotype to that of replicative senescence, suggesting the activation of a common senescence mechanism. In this study, we investigated the gene expression profile of oncogenic H-ras-induced senescent human diploid fibroblasts. We found altered gene expression of various cell cycle regulators in both oncogenic H-ras-induced senescent cells and replicative senescent cells. Similar to replicative senescent cells, H-ras-induced senescent cells exhibited specific downregulation of genes involved in G2/M checkpoint control and contained tetraploid cells that were arrested in a G1 state. This observation suggests that the inactivation of G2/M checkpoints may be involved in senescence and may play a role in the generation of senescent G1 tetraploid cells. Lastly, we have identified two genes, topoisomerase IIalpha and HDAC9, whose expression was specifically altered under several conditions associated with senescence, suggesting that these two molecules may be novel biomarkers for senescent human fibroblasts.


Asunto(s)
Senescencia Celular/fisiología , Proteína Oncogénica p21(ras)/fisiología , Antígenos de Neoplasias , Ciclo Celular , Ensayo Cometa , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
11.
Cell ; 113(6): 703-16, 2003 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12809602

RESUMEN

Cellular senescence is an extremely stable form of cell cycle arrest that limits the proliferation of damaged cells and may act as a natural barrier to cancer progression. In this study, we describe a distinct heterochromatic structure that accumulates in senescent human fibroblasts, which we designated senescence-associated heterochromatic foci (SAHF). SAHF formation coincides with the recruitment of heterochromatin proteins and the retinoblastoma (Rb) tumor suppressor to E2F-responsive promoters and is associated with the stable repression of E2F target genes. Notably, both SAHF formation and the silencing of E2F target genes depend on the integrity of the Rb pathway and do not occur in reversibly arrested cells. These results provide a molecular explanation for the stability of the senescent state, as well as new insights into the action of Rb as a tumor suppressor.


Asunto(s)
Proteínas de Ciclo Celular , Senescencia Celular/genética , Proteínas de Unión al ADN , Células Eucariotas/metabolismo , Silenciador del Gen/fisiología , Heterocromatina/genética , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Sitios de Unión/genética , Línea Celular , Tamaño de la Célula/genética , ADN/genética , ADN/metabolismo , Factores de Transcripción E2F , Células Eucariotas/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Marcación de Gen , Genes p16/fisiología , Heterocromatina/metabolismo , Heterocromatina/ultraestructura , Humanos , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
12.
Proc Natl Acad Sci U S A ; 100(9): 5431-6, 2003 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-12702766

RESUMEN

The tumor suppressor p53 is regulated in part by binding to cellular proteins. We used p53 as bait in the yeast two-hybrid system and isolated homeodomain-interacting protein kinase 1 (HIPK1) as a p53-binding protein. Deletion analysis showed that amino acids 100-370 of p53 and amino acids 885-1093 of HIPK1 were sufficient for HIPK1-p53 interaction. HIPK1 was capable of autophosphorylation and specific serine phosphorylation of p53. The HIPK1 gene was highly expressed in human breast cancer cell lines and oncogenically transformed mouse embryonic fibroblasts. HIPK1 was localized to human chromosome band 1p13, a site frequently altered in cancers. Gene-targeted HIPK1-/- mice were grossly normal but oncogenically transformed HIPK1 -/- mouse embryonic fibroblasts exhibited reduced transcription of Mdm2 and were more susceptible than transformed HIPK1+/+ cells to apoptosis induced by DNA damage. Carcinogen-treated HIPK1 -/- mice developed fewer and smaller skin tumors than HIPK1+/+ mice. HIPK1 may thus play a role in tumorigenesis, perhaps by means of the regulation of p53 and/or Mdm2.


Asunto(s)
Proteínas Portadoras/genética , Marcación de Gen , Proteínas Quinasas/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Northern Blotting , Proteínas Portadoras/metabolismo , Humanos , Ratones , Ratones Noqueados , Unión Proteica , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Técnicas del Sistema de Dos Híbridos
13.
Mol Cell Biol ; 22(10): 3497-508, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11971980

RESUMEN

Oncogenic activation of the mitogen-activated protein (MAP) kinase cascade in murine fibroblasts initiates a senescence-like cell cycle arrest that depends on the ARF/p53 tumor suppressor pathway. To investigate whether p53 is sufficient to induce senescence, we introduced a conditional murine p53 allele (p53(val135)) into p53-null mouse embryonic fibroblasts and examined cell proliferation and senescence in cells expressing p53, oncogenic Ras, or both gene products. Conditional p53 activation efficiently induced a reversible cell cycle arrest but was unable to induce features of senescence. In contrast, coexpression of oncogenic ras or activated mek1 with p53 enhanced both p53 levels and activity relative to that observed for p53 alone and produced an irreversible cell cycle arrest that displayed features of cellular senescence. p19(ARF) was required for this effect, since p53(-/-) ARF(-/-) double-null cells were unable to undergo senescence following coexpression of oncogenic Ras and p53. Although the levels of exogenous p53 achieved in ARF-null cells were relatively low, the stabilizing effects of p19(ARF) on p53 could not explain the cooperation between oncogenic Ras and p53 in promoting senescence. Hence, enforced p53 expression without oncogenic ras in p53(-/-) mdm2(-/-) double-null cells produced extremely high p53 levels but did not induce senescence. Taken together, our results indicate that oncogenic activation of the MAP kinase pathway in murine fibroblasts converts p53 into a senescence inducer through both quantitative and qualitative mechanisms.


Asunto(s)
Senescencia Celular/fisiología , Genes ras/genética , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Nucleares , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular/fisiología , Fraccionamiento Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Embrión de Mamíferos/fisiología , Activación Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Genes p53 , MAP Quinasa Quinasa 1 , Ratones , Ratones Noqueados , Microscopía Fluorescente , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Temperatura , Proteína p14ARF Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA