Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Biol Chem ; 287(35): 29442-56, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22736770

RESUMEN

Ada3 protein is an essential component of histone acetyl transferase containing coactivator complexes conserved from yeast to human. We show here that germline deletion of Ada3 in mouse is embryonic lethal, and adenovirus-Cre mediated conditional deletion of Ada3 in Ada3(FL/FL) mouse embryonic fibroblasts leads to a severe proliferation defect which was rescued by ectopic expression of human Ada3. A delay in G(1) to S phase of cell cycle was also seen that was due to accumulation of Cdk inhibitor p27 which was an indirect effect of c-myc gene transcription control by Ada3. We further showed that this defect could be partially reverted by knocking down p27. Additionally, drastic changes in global histone acetylation and changes in global gene expression were observed in microarray analyses upon loss of Ada3. Lastly, formation of abnormal nuclei, mitotic defects and delay in G(2)/M to G(1) transition was seen in Ada3 deleted cells. Taken together, we provide evidence for a critical role of Ada3 in embryogenesis and cell cycle progression as an essential component of HAT complex.


Asunto(s)
Ciclo Celular/fisiología , Embrión de Mamíferos/embriología , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción/metabolismo , Acetilación , Animales , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Noqueados , Factores de Transcripción/genética
2.
Breast Cancer Res Treat ; 137(3): 721-31, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23288344

RESUMEN

Transcriptional activation by estrogen receptor (ER) is a key step to breast oncogenesis. Given previous findings that ADA3 is a critical component of HAT complexes that regulate ER function and evidence that overexpression of other ER coactivators such as SRC-3 is associated with clinical outcomes in breast cancer, the current study was designed to assess the potential significance of ADA3 expression/localization in human breast cancer patients. In this study, we analyzed ADA3 expression in breast cancer tissue specimens and assessed the correlation of ADA3 staining with cancer progression and patient outcome. Tissue microarrays prepared from large series of breast cancer patients with long-term follow-ups were stained with anti-ADA3 monoclonal antibody using immunohistochemistry. Samples were analyzed for ADA3 expression followed by correlation with various clinicopathological parameters and patients' outcomes. We report that breast cancer specimens show predominant nuclear, cytoplasmic, or mixed nuclear + cytoplasmic ADA3 staining patterns. Predominant nuclear ADA3 staining correlated with ER+ status. While predominant cytoplasmic ADA3 staining negatively correlated with ER+ status, but positively correlated with ErbB2, EGFR, and Ki67. Furthermore, a positive correlation of cytoplasmic ADA3 was observed with higher histological grade, mitotic counts, Nottingham Prognostic Index, and positive vascular invasion. Patients with nuclear ADA3 and ER positivity have better breast cancer specific survival and distant metastasis free survival. Significantly, cytoplasmic expression of ADA3 showed a strong positive association with reduced BCSS and DMFS in ErbB2+/EGFR+ patients. Although in multivariate analyses ADA3 expression was not an independent marker of survival, predominant nuclear ADA3 staining in breast cancer tissues correlates with ER+ expression and together serves as a marker of good prognosis, whereas predominant cytoplasmic ADA3 expression correlates with ErbB2+/EGFR+ expression and together is a marker of poor prognosis. Thus, ADA3 cytoplasmic localization together with ErbB2+/EGFR+ status may serve as better prognostic marker than individual proteins to predict survival of patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Citoplasma/metabolismo , Factores de Transcripción/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Núcleo Celular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Transporte de Proteínas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Carga Tumoral , Adulto Joven
3.
Breast Cancer Res Treat ; 134(1): 171-80, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22270930

RESUMEN

Uncontrolled proliferation is one of the hallmarks of breast cancer. We have previously identified the human Ecd protein (human ortholog of Drosophila Ecdysoneless, hereafter called Ecd) as a novel promoter of mammalian cell cycle progression, a function related to its ability to remove the repressive effects of Rb-family tumor suppressors on E2F transcription factors. Given the frequent dysregulation of cell cycle regulatory components in human cancer, we used immunohistochemistry of paraffin-embedded tissues to examine Ecd expression in normal breast tissue versus tissues representing increasing breast cancer progression. Initial studies of a smaller cohort without outcomes information showed that Ecd expression was barely detectable in normal breast tissue and in hyperplasia of breast, but high levels of Ecd were detected in benign breast hyperplasia, ductal carcinoma in situ (DCIS) and infiltrating ductal carcinoma (IDCs) of the breast. In this cohort of 104 IDC patients, Ecd expression levels showed a positive correlation with higher grade (P=0.04). Further analyses of Ecd expression using a larger, independent cohort (954) confirmed these results, with a strong positive correlation of elevated Ecd expression with higher histological grade (P=0.013), mitotic index (P=0.032), and Nottingham Prognostic Index score (P=0.014). Ecd expression was positively associated with HER2/neu (P=0.002) overexpression, a known marker of poor prognosis in breast cancer. Significantly, increased Ecd expression showed a strong positive association with shorter breast cancer specific survival (BCSS) (P=0.008) and disease-free survival (DFS) (P=0.003) in HER2/neu overexpressing patients. Taken together, our results reveal Ecd as a novel marker for breast cancer progression and show that levels of Ecd expression predict poorer survival in Her2/neu overexpressing breast cancer patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Proteínas Portadoras/metabolismo , Receptor ErbB-2/metabolismo , Adolescente , Adulto , Anciano , Especificidad de Anticuerpos , Biomarcadores de Tumor/genética , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/patología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Expresión Génica , Humanos , Hiperplasia/metabolismo , Estimación de Kaplan-Meier , Persona de Mediana Edad , Pronóstico , Receptor ErbB-2/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Adulto Joven
4.
Adv Exp Med Biol ; 720: 135-44, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21901624

RESUMEN

Recent molecular profiling has identified six major subtypes of breast cancers that exhibit different survival outcomes for patients. To address the origin of different subtypes of breast cancers, we have now identified, isolated, and immortalized (using hTERT) mammary stem/progenitor cells which maintain their stem/progenitor properties even after immortalization. Our decade long research has shown that these stem/progenitor cells are highly susceptible to oncogenesis. Given the emerging evidence that stem/progenitor cells are precursors of cancers and that distinct subtypes of breast cancer have different survival outcome, these cellular models provide novel tools to understand the oncogenic process leading to various subtypes of breast cancers and for future development of novel therapeutic strategies to treat different subtypes of breast cancers.


Asunto(s)
Neoplasias de la Mama/clasificación , Técnicas de Cultivo de Célula , Transformación Celular Neoplásica , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Separación Celular , Células Epiteliales/clasificación , Femenino , Humanos , Células Madre Neoplásicas/fisiología , Células Madre/fisiología , Telomerasa/genética
5.
J Biol Chem ; 284(39): 26402-10, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19640839

RESUMEN

The Ecdysoneless (Ecd) protein is required for cell-autonomous roles in development and oogenesis in Drosophila, but the function of its evolutionarily conserved mammalian orthologs is not clear. To study the cellular function of Ecd in mammalian cells, we generated Ecd(lox/lox) mouse embryonic fibroblast cells from Ecd floxed mouse embryos. Cre-mediated deletion of Ecd in Ecd(lox/lox) mouse embryonic fibroblasts led to a proliferative block due to a delay in G(1)-S cell cycle progression; this defect was reversed by the introduction of human Ecd. Loss of Ecd led to marked down-regulation of E2F target gene expression. Furthermore, Ecd directly bound to Rb at the pocket domain and competed with E2F for binding to hypophosphorylated Rb. Our results demonstrate that mammalian Ecd plays a role in cell cycle progression via the Rb-E2F pathway.


Asunto(s)
Proteínas Portadoras/fisiología , Ciclo Celular/fisiología , Proliferación Celular , Fibroblastos/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Femenino , Fibroblastos/citología , Fase G1/fisiología , Expresión Génica , Humanos , Immunoblotting , Inmunoprecipitación , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Unión Proteica , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Fase S/fisiología , Factores de Tiempo
6.
Biol Chem ; 391(1): 9-19, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19919181

RESUMEN

Ecdysoneless (Ecd) is an evolutionarily conserved protein and its function is essential for embryonic development in Drosophila and cell growth in yeast. However, its function has remained unknown until recently. Studies in yeast suggested a potential role of Ecd in transcription; however, Ecd lacks a DNA-binding domain. Using a GAL4-luciferase reporter assay and a GAL4 DNA-binding domain fusion with Ecd or its mutants, we present evidence that human Ecd has a transactivation activity in its C-terminal region. Importantly, further analyses using point mutants showed that a single amino acid change at either Asp-484 or Leu-489 essentially completely abolishes the transactivation activity of Ecd. We further demonstrate that Ecd interacts with p300, a histone acetyltransferase, and coexpression of Ecd with p300 enhances the Ecd-mediated transactivation activity. Ecd localizes to both nucleus and cytoplasm and shuttles between the nucleus and cytoplasm; however, it exhibits strong nuclear export. Based on previous yeast studies and evidence provided here, we suggest that Ecd functions as a transcriptional regulator. Our results indicate an important function of human Ecd and provide a basis to explore the transcriptional partners of Ecd.


Asunto(s)
Proteínas Portadoras/fisiología , Activación Transcripcional , Secuencia de Aminoácidos , Núcleo Celular/enzimología , Citoplasma/enzimología , Análisis Mutacional de ADN , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia
7.
Dev Biol ; 316(2): 191-9, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18334253

RESUMEN

The lin-12/Notch signaling pathway is conserved from worms to humans and is a master regulator of metazoan development. Here, we demonstrate that lin-12/Notch gain-of-function (gf) animals display precocious alae at the L4 larval stage with a significant increase in let-7 expression levels. Furthermore, lin-12(gf) animals display a precocious and higher level of let-7 gfp transgene expression in seam cells at L3 stage. Interestingly, lin-12(gf) mutant rescued the lethal phenotype of let-7 mutants similar to other known heterochronic mutants. We propose that lin-12/Notch signaling pathway functions in late developmental timing, upstream of or in parallel to the let-7 heterochronic pathway. Importantly, the human microRNA let-7a was also upregulated in various human cell lines in response to Notch 1 activation, suggesting an evolutionarily conserved cross-talk between let-7 and the canonical lin-12/Notch signaling pathway.


Asunto(s)
Caenorhabditis elegans/genética , MicroARNs/genética , Mutación , Animales , Animales Modificados Genéticamente , Northern Blotting , Caenorhabditis elegans/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Larva , Fenotipo , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Nat Neurosci ; 20(12): 1694-1707, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29184203

RESUMEN

Haploinsufficiency of the AT-rich interactive domain 1B (ARID1B) gene causes autism spectrum disorder and intellectual disability; however, the neurobiological basis for this is unknown. Here we generated Arid1b-knockout mice and examined heterozygotes to model human patients. Arid1b-heterozygous mice showed a decreased number of cortical GABAergic interneurons and reduced proliferation of interneuron progenitors in the ganglionic eminence. Arid1b haploinsufficiency also led to an imbalance between excitatory and inhibitory synapses in the cerebral cortex. Furthermore, we found that Arid1b haploinsufficiency suppressed histone H3 lysine 9 acetylation (H3K9ac) overall and particularly reduced H3K9ac of the Pvalb promoter, resulting in decreased transcription. Arid1b-heterozygous mice exhibited abnormal cognitive and social behaviors, which were rescued by treatment with a positive allosteric GABAA receptor modulator. Our results demonstrate a critical role for Arid1b in interneuron development and behavior and provide insight into the pathogenesis of autism spectrum disorder and intellectual disability.


Asunto(s)
Conducta Animal/fisiología , Corteza Cerebral/crecimiento & desarrollo , Interneuronas , Vías Nerviosas/crecimiento & desarrollo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/psicología , Reacción de Prevención , Recuento de Células , Corteza Cerebral/citología , Cognición , Epigénesis Genética/genética , Miedo/fisiología , Haploinsuficiencia , Discapacidad Intelectual/genética , Discapacidad Intelectual/psicología , Ratones , Ratones Noqueados , Vías Nerviosas/citología , Optogenética/métodos , Conducta Social , Sinapsis/ultraestructura , Ácido gamma-Aminobutírico/fisiología
9.
Cell Cycle ; 14(7): 990-1000, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25616580

RESUMEN

The mammalian ortholog of Drosophila ecdysoneless (Ecd) gene product regulates Rb-E2F interaction and is required for cell cycle progression. Ecd is overexpressed in breast cancer and its overexpression predicts shorter survival in patients with ErbB2-positive tumors. Here, we demonstrate Ecd knock down (KD) in human mammary epithelial cells (hMECs) induces growth arrest, similar to the impact of Ecd Knock out (KO) in mouse embryonic fibroblasts. Furthermore, whole-genome mRNA expression analysis of control vs. Ecd KD in hMECs demonstrated that several of the top 40 genes that were down-regulated were E2F target genes. To address the role of Ecd in mammary oncogenesis, we overexpressed Ecd and/or mutant H-Ras in hTERT-immortalized hMECs. Cell cycle analyses revealed hMECs overexpressing Ecd+Ras showed incomplete arrest in G1 phase upon growth factor deprivation, and more rapid cell cycle progression in growth factor-containing medium. Analyses of cell migration, invasion, acinar structures in 3-D Matrigel and anchorage-independent growth demonstrated that Ecd+Ras-overexpressing cells exhibit substantially more dramatic transformed phenotype as compared to cells expressing vector, Ras or Ecd. Under conditions of nutrient deprivation, Ecd+Ras-overexpressing hMECs exhibited better survival, with substantial upregulation of the autophagy marker LC3 both at the mRNA and protein levels. Significantly, while hMECs expressing Ecd or mutant Ras alone did not form tumors in NOD/SCID mice, Ecd+Ras-overexpressing hMECs formed tumors, clearly demonstrating oncogenic cooperation between Ecd and mutant Ras. Collectively, we demonstrate an important co-oncogenic role of Ecd in the progression of mammary oncogenesis through promoting cell survival.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/fisiología , Glándulas Mamarias Humanas/patología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Animales , Autofagia , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Células Cultivadas , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , Proteínas Asociadas a Microtúbulos/metabolismo , Trasplante de Neoplasias
10.
Cell Cycle ; 11(22): 4266-74, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23095635

RESUMEN

Cell cycle regulation and DNA repair following damage are essential for maintaining genome integrity. DNA damage activates checkpoints in order to repair damaged DNA prior to exit to the next phase of cell cycle. Recently, we have shown the role of Ada3, a component of various histone acetyltransferase complexes, in cell cycle regulation, and loss of Ada3 results in mouse embryonic lethality. Here, we used adenovirus-Cre-mediated Ada3 deletion in Ada3(fl/fl) mouse embryonic fibroblasts (MEFs) to assess the role of Ada3 in DNA damage response following exposure to ionizing radiation (IR). We report that Ada3 depletion was associated with increased levels of phospho-ATM (pATM), γH2AX, phospho-53BP1 (p53BP1) and phospho-RAD51 (pRAD51) in untreated cells; however, radiation response was intact in Ada3(-/-) cells. Notably, Ada3(-/-) cells exhibited a significant delay in disappearance of DNA damage foci for several critical proteins involved in the DNA repair process. Significantly, loss of Ada3 led to enhanced chromosomal aberrations, such as chromosome breaks, fragments, deletions and translocations, which further increased upon DNA damage. Notably, the total numbers of aberrations were more clearly observed in S-phase, as compared with G1 or G2 phases of cell cycle with IR. Lastly, comparison of DNA damage in Ada3(fl/fl) and Ada3(-/-) cells confirmed higher residual DNA damage in Ada3(-/-) cells, underscoring a critical role of Ada3 in the DNA repair process. Taken together, these findings provide evidence for a novel role for Ada3 in maintenance of the DNA repair process and genomic stability.


Asunto(s)
Inestabilidad Genómica , Factores de Transcripción/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Aberraciones Cromosómicas , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Cariotipificación , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Recombinasa Rad51/metabolismo , Radiación Ionizante , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
11.
Clin Cancer Res ; 18(22): 6188-98, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22977192

RESUMEN

PURPOSE: To study the expression and function of a novel cell-cycle regulatory protein, human ecdysoneless (Ecd), during pancreatic cancer pathogenesis. EXPERIMENTAL DESIGN: Immunohistochemical expression profiling of Ecd was done in nonneoplastic normal pancreatic tissues and pancreatic ductal adenocarcinoma lesions (from tissue microarray and Rapid Autopsy program) as well as precancerous PanIN lesions and metastatic organs. To analyze the biological significance of Ecd in pancreatic cancer progression, Ecd was stably knocked down in pancreatic cancer cell line followed by in vitro and in vivo functional assays. RESULTS: Normal pancreatic ducts showed very weak to no Ecd expression compared to significant positive expression in pancreatic cancer tissues (mean ± SE composite score: 0.3 ± 0.2 and 3.8 ± 0.2 respectively, P < 0.0001) as well as in PanIN precursor lesions with a progressive increase in Ecd expression with increasing dysplasia (PanIN-1-PanIN-3). Analysis of matched primary tumors and metastases from patients with pancreatic cancer revealed that Ecd is highly expressed in both primary pancreatic tumor and in distant metastatic sites. Furthermore, knockdown of Ecd suppressed cell proliferation in vitro and tumorigenicity of pancreatic cancer cells in mice orthotopic tumors. Microarray study revealed that Ecd regulates expression of glucose transporter GLUT4 in pancreatic cancer cells and was subsequently shown to modulate glucose uptake, lactate production, and ATP generation by pancreatic cancer cells. Finally, knockdown of Ecd also reduced level of pAkt, key signaling molecule known to regulate aerobic glycolysis in cancer cells. CONCLUSION: Ecd is a novel tumor-promoting factor that is differentially expressed in pancreatic cancer and potentially regulates glucose metabolism within cancer cells.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas Portadoras/genética , Transformación Celular Neoplásica/metabolismo , Glucólisis , Neoplasias Pancreáticas/metabolismo , Animales , Carcinoma Ductal Pancreático/secundario , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Proliferación Celular , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Metástasis Linfática , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Páncreas/metabolismo , Neoplasias Pancreáticas/patología , Análisis de Matrices Tisulares , Carga Tumoral , Regulación hacia Arriba
12.
J Biol Chem ; 282(12): 8812-20, 2007 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-17272277

RESUMEN

The p53 tumor suppressor protein functions as a critical component of genotoxic stress response by regulating the expression of effector gene products that control the fate of a cell following DNA damage. Unstressed cells maintain p53 at low levels through regulated degradation, and p53 levels and activity are rapidly elevated upon genotoxic stress. Biochemical mechanisms that control the levels and activity of p53 are therefore of great interest. We and others have recently identified hAda3 (human homologue of yeast alteration/deficiency in activation 3) as a p53-interacting protein and enhancer of p53 activity. Here, we show that endogenous levels of p53 and Ada3 interact with each other, and by using inducible overexpression and short hairpin RNA-mediated knockdown strategies we demonstrate that hAda3 stabilizes p53 protein by promoting its acetylation. Use of a p53 mutant with mutations of known p300/CREB-binding protein acetylation sites demonstrated that hAda3-dependent acetylation is required for increase in p53 stability and target gene induction. Importantly, we demonstrate that endogenous hAda3 is essential for DNA damage-induced acetylation and stabilization of p53 as well as p53 target gene induction. Overall, our results establish hAda3, a component of coactivator complexes that include histone acetyltransferase p300/CREB-binding protein, as a critical mediator of acetylation-dependent stabilization and activation of p53 upon genotoxic stress in mammalian cells.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Proteína de Unión a CREB/química , Línea Celular Tumoral , Daño del ADN , Genes p53 , Humanos , Mutación , Interferencia de ARN , Retroviridae/metabolismo , Factores de Tiempo , Activación Transcripcional , Transfección , Proteína p53 Supresora de Tumor/química , Ubiquitina/química , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA