Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 13(7): 595, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35817766

RESUMEN

Age-related hearing loss (ARHL) is the most common sensory impairment mainly caused by degeneration of sensory hair cells in the cochlea with no causal medical treatment available. Auditory function and sensory hair cell survival critically depend on the Kv7.4 (KCNQ4) channel, a voltage-gated potassium channel expressed in outer hair cells (OHCs), with its impaired function or reduced activity previously associated with ARHL. Here, we investigated the effect of a potent small-molecule Kv7.4 agonist on ARHL in the senescence-accelerated mouse prone 8 (SAMP8) model. For the first time in vivo, we show that Kv7.4 activation can significantly reduce age-related threshold shifts of auditory brainstem responses as well as OHC loss in the SAMP8 model. Pharmacological activation of Kv7.4 thus holds great potential as a therapeutic approach for ARHL as well as other hearing impairments related to Kv7.4 function.


Asunto(s)
Células Ciliadas Auditivas Externas , Pérdida Auditiva , Animales , Cóclea , Modelos Animales de Enfermedad , Audición , Pérdida Auditiva/tratamiento farmacológico , Ratones , Canales de Potasio
2.
Cell Death Dis ; 10(10): 747, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31582725

RESUMEN

The serine-threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) modulates important cellular functions during development, acting as a signal integrator of a wide variety of stress signals, and as a regulator of transcription factors and cofactors. We have previously demonstrated that HIPK2 binds and phosphorylates High-Mobility Group A1 (HMGA1), an architectural chromatinic protein ubiquitously expressed in embryonic tissues, decreasing its binding affinity to DNA. To better define the functional role of HIPK2 and HMGA1 interaction in vivo, we generated mice in which both genes are disrupted. About 50% of these Hmga1/Hipk2 double knock-out (DKO) mice die within 12 h of life (P1) for respiratory failure. The DKO mice present an altered lung morphology, likely owing to a drastic reduction in the expression of surfactant proteins, that are required for lung development. Consistently, we report that both HMGA1 and HIPK2 proteins positively regulate the transcriptional activity of the genes encoding the surfactant proteins. Moreover, these mice display an altered expression of thyroid differentiation markers, reasonably because of a drastic reduction in the expression of the thyroid-specific transcription factors PAX8 and FOXE1, which we demonstrate here to be positively regulated by HMGA1 and HIPK2. Therefore, these data indicate a critical role of HIPK2/HMGA1 cooperation in lung and thyroid development and function, suggesting the potential involvement of their impairment in the pathogenesis of human lung and thyroid diseases.


Asunto(s)
Proteína HMGA1a/genética , Proteínas Serina-Treonina Quinasas/genética , Enfermedades Respiratorias/genética , Glándula Tiroides/anomalías , Animales , Animales Recién Nacidos , Desarrollo Embrionario , Eliminación de Gen , Regulación de la Expresión Génica , Proteína HMGA1a/metabolismo , Células HeLa , Humanos , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Asociadas a Surfactante Pulmonar , Enfermedades Respiratorias/patología , Glándula Tiroides/embriología , Glándula Tiroides/patología
4.
Cell Death Differ ; 24(11): 1948-1962, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28777374

RESUMEN

High Mobility Group A1 (HMGA1) is an architectural chromatin protein whose overexpression is a feature of malignant neoplasias with a causal role in cancer initiation and progression. HMGA1 promotes tumor growth by several mechanisms, including increase of cell proliferation and survival, impairment of DNA repair and induction of chromosome instability. Autophagy is a self-degradative process that, by providing energy sources and removing damaged organelles and misfolded proteins, allows cell survival under stress conditions. On the other hand, hyper-activated autophagy can lead to non-apoptotic programmed cell death. Autophagy deregulation is a common feature of cancer cells in which has a complex role, showing either an oncogenic or tumor suppressor activity, depending on cellular context and tumor stage. Here, we report that depletion of HMGA1 perturbs autophagy by different mechanisms. HMGA1-knockdown increases autophagosome formation by constraining the activity of the mTOR pathway, a major regulator of autophagy, and transcriptionally upregulating the autophagy-initiating kinase Unc-51-like kinase 1 (ULK1). Consistently, functional experiments demonstrate that HMGA1 binds ULK1 promoter region and negatively regulates its transcription. On the other hand, the increase in autophagosomes is not associated to a proportionate increase in their maturation. Overall, the effects of HMGA1 depletion on autophagy are associated to a decrease in cell proliferation and ultimately impact on cancer cells viability. Importantly, silencing of ULK1 prevents the effects of HMGA1-knockdown on cellular proliferation, viability and autophagic activity, highlighting how these effects are, at least in part, mediated by ULK1. Interestingly, this phenomenon is not restricted to skin cancer cells, as similar results have been observed also in HeLa cells silenced for HMGA1. Taken together, these results clearly indicate HMGA1 as a key regulator of the autophagic pathway in cancer cells, thus suggesting a novel mechanism through which HMGA1 can contribute to cancer progression.


Asunto(s)
Autofagia , Proteína HMGA1a/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Proteína 5 Relacionada con la Autofagia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Proliferación Celular , Supervivencia Celular , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Transcripción Genética
5.
Cell Cycle ; 15(6): 812-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26889953

RESUMEN

The High Mobility Group A1 proteins (HMGA1) are nonhistone chromatinic proteins with a critical role in development and cancer. We have recently reported that HMGA1 proteins are able to increase the expression of spindle assembly checkpoint (SAC) genes, thus impairing SAC function and causing chromosomal instability in cancer cells. Moreover, we found a significant correlation between HMGA1 and SAC genes expression in human colon carcinomas. Here, we report that mouse embryonic fibroblasts null for the Hmga1 gene show downregulation of Bub1, Bub1b, Mad2l1 and Ttk SAC genes, and present several features of chromosomal instability, such as nuclear abnormalities, binucleation, micronuclei and karyotypic alterations. Interestingky, also MEFs carrying only one impaired Hmga1 allele present karyotypic alterations. These results indicate that HMGA1 proteins regulate SAC genes expression and, thereby, genomic stability also in embryonic cells.


Asunto(s)
Inestabilidad Cromosómica , Fibroblastos/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Proteína HMGA1a/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/patología , Embrión de Mamíferos , Fibroblastos/patología , Regulación de la Expresión Génica , Prueba de Complementación Genética , Proteína HMGA1a/deficiencia , Cariotipo , Ratones , Ratones Noqueados , Micronúcleos con Defecto Cromosómico , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
6.
Oncotarget ; 6(19): 17342-53, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26009897

RESUMEN

The mitotic spindle assembly checkpoint (SAC) is an essential control system of the cell cycle that contributes to mantain the genomic stability of eukaryotic cells. SAC genes expression is often deregulated in cancer cells, leading to checkpoint impairment and chromosome instability. The mechanisms responsible for the transcriptional regulation and deregulation of these genes are still largely unknown. Herein we identify the nonhistone architectural nuclear proteins High Mobility Group A1 (HMGA1), whose overexpression is a feature of several human malignancies and has a key role in cancer progression, as transcriptional regulators of SAC genes expression. In particular, we show that HMGA1 proteins are able to increase the expression of the SAC genes Ttk, Mad2l1, Bub1 and Bub1b, binding to their promoter regions. Consistently, HMGA1-depletion induces SAC genes downregulation associated to several mitotic defects. In particular, we observed a high number of unaligned chromosomes in metaphase, a reduction of prometaphase time, a delay of anaphase, a higher cytokinesis time and a higher percentage of cytokinesis failure by using live-cell microscopy. Finally, a significant direct correlation between HMGA1 and SAC genes expression was detected in human colon carcinomas indicating a novel mechanism by which HMGA1 contributes to cancer progression.


Asunto(s)
Inestabilidad Cromosómica/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas HMGB/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunohistoquímica , Ratones , Células 3T3 NIH , Reacción en Cadena de la Polimerasa , Transfección
7.
Oncotarget ; 6(12): 10320-34, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25868975

RESUMEN

HIPK2, a cell fate decision kinase inactivated in several human cancers, is thought to exert its oncosuppressing activity through its p53-dependent and -independent apoptotic function. However, a HIPK2 role in cell proliferation has also been described. In particular, HIPK2 is required to complete cytokinesis and impaired HIPK2 expression results in cytokinesis failure and tetraploidization. Since tetraploidy may yield to aneuploidy and chromosomal instability (CIN), we asked whether unscheduled tetraploidy caused by loss of HIPK2 might contribute to tumorigenicity. Here, we show that, compared to Hipk2+/+ mouse embryo fibroblasts (MEFs), hipk2-null MEFs accumulate subtetraploid karyotypes and develop CIN. Accumulation of these defects inhibits proliferation and spontaneous immortalization of primary MEFs whereas increases tumorigenicity when MEFs are transformed by E1A and Harvey-Ras oncogenes. Upon mouse injection, E1A/Ras-transformed hipk2-null MEFs generate tumors with genetic alterations resembling those of human cancers derived by initial tetraploidization events, such as pancreatic adenocarcinoma. Thus, we evaluated HIPK2 expression in different stages of pancreatic transformation. Importantly, we found a significant correlation among reduced HIPK2 expression, high grade of malignancy, and high nuclear size, a marker of increased ploidy. Overall, these results indicate that HIPK2 acts as a caretaker gene, whose inactivation increases tumorigenicity and causes CIN by cytokinesis failure.


Asunto(s)
Carcinogénesis/patología , Inestabilidad Cromosómica , Citocinesis/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Femenino , Células HeLa , Humanos , Ratones , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transfección
8.
Oncotarget ; 5(18): 8341-54, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25268743

RESUMEN

The High Mobility Group A (HMGA) are nuclear proteins that participate in the organization of nucleoprotein complexes involved in chromatin structure, replication and gene transcription. HMGA overexpression is a feature of human cancer and plays a causal role in cell transformation. Since non-coding RNAs and pseudogenes are now recognized to be important in physiology and disease, we investigated HMGA1 pseudogenes in cancer settings using bioinformatics analysis. Here we report the identification and characterization of two HMGA1 non-coding pseudogenes, HMGA1P6 and HMGA1P7. We show that their overexpression increases the levels of HMGA1 and other cancer-related proteins by inhibiting the suppression of their synthesis mediated by microRNAs. Consistently, embryonic fibroblasts from HMGA1P7-overexpressing transgenic mice displayed a higher growth rate and reduced susceptibility to senescence. Moreover, HMGA1P6 and HMGA1P7 were overexpressed in human anaplastic thyroid carcinomas, which are highly aggressive, but not in differentiated papillary carcinomas, which are less aggressive. Lastly, the expression of the HMGA1 pseudogenes was significantly correlated with HMGA1 protein levels thereby implicating HMGA1P overexpression in cancer progression. In conclusion, HMGA1P6 and HMGA1P7 are potential proto-oncogenic competitive endogenous RNAs.


Asunto(s)
Proteínas HMGA/genética , MicroARNs/genética , Neoplasias Ováricas/genética , Seudogenes/genética , Carcinoma Anaplásico de Tiroides/genética , Neoplasias de la Tiroides/genética , Animales , Apoptosis , Proliferación Celular , Senescencia Celular , Biología Computacional , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Proteínas HMGA/metabolismo , Humanos , Células MCF-7 , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Interferencia de ARN , Carcinoma Anaplásico de Tiroides/metabolismo , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Factores de Tiempo , Transfección
10.
J Clin Endocrinol Metab ; 97(5): E710-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22399519

RESUMEN

CONTEXT: We have previously demonstrated that a set of micro-RNA (miRNA) is significantly down-regulated in anaplastic thyroid carcinomas with respect to normal thyroid tissues and to differentiated thyroid carcinomas. OBJECTIVE: The objective was to evaluate the role of two of these down-regulated miRNA, miR-25 and miR-30d, in thyroid carcinogenesis. DESIGN: miR-25 and miR-30d expression was restored in the ACT-1, 8505c, and FRO anaplastic thyroid cell lines, and their effects on cell proliferation, migration, and target expression were evaluated. RESULTS: We report that miR-25 and miR-30d target the polycomb protein enhancer of zeste 2 (EZH2) that has oncogenic activity and is drastically up-regulated in anaplastic thyroid carcinomas but not in the differentiated ones. Ectopic expression of miR-25 and miR-30d inhibited proliferation and colony formation of anaplastic thyroid carcinoma cells by inducing G2/M-phase cell-cycle arrest. Finally, we found an inverse correlation between the expression of these miRNA and the EZH2 protein levels in anaplastic thyroid carcinomas, suggesting a critical role of these miRNA in regulating EZH2 expression also in vivo. CONCLUSION: The down-regulation of miR-25 and miR-30d could contribute to the process of thyroid cancer progression, leading to the development of anaplastic carcinomas targeting EZH2 mRNA.


Asunto(s)
Carcinoma/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , MicroARNs/genética , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/genética , Factores de Transcripción/genética , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Expresión Génica/genética , Humanos , MicroARNs/metabolismo , Complejo Represivo Polycomb 2 , Glándula Tiroides/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Factores de Transcripción/metabolismo
11.
J Cell Physiol ; 227(12): 3749-55, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22392906

RESUMEN

It is well established that estrogens participate in the control of normal spermatogenesis and endogenous or environmental estrogens are involved in pathological germ cell proliferation including testicular germ cell tumors. The effects of estrogen are now known to be mediated by estrogen receptor-α (ERα) and ERß subtypes, but only ERß has been found in human germ cells of normal testis. However, its expression was markedly diminished in human testicular seminomas. The expression and the possible interaction of ERß and HMGA1 were studied in normal germ cells and in human testicular seminomas. GC1 and TCam-2 germ cell lines, were used; in addition, a tissue micro-array (TMA) was built using the most representative areas from 35 cases of human testicular seminomas. The expression and the interaction of ERß and HMGA1 were observed by using immunoprecipitation and Western blot analyses in combination with immunocytochemistry and immunofluorescence analyses. Here, we show that ERß interacts with HMGA1 in normal germ cells, while down regulation of ERß associates with transcriptional co-regulator HMGA1 over-expression and cytoplasmic localization both in human testicular seminomas and in TCam-2 cell line. In addition, we show that 17ß-oestradiol induces an HMGA1 increased cytoplasmic expression associated to an ERß down-regulation in TCam-2 cell line. Taken together, our results suggest that exposure to estrogens or estrogen-mimics, in some as of yet undefined manner, diminishes the ERß-mediated growth restraint in human testicular seminoma, probably due to the HMGA1 cytoplasmic delocalization associated with ERß down-regulation.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína HMGA1a/metabolismo , Seminoma/metabolismo , Animales , Línea Celular Tumoral , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Proteína HMGA1a/genética , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Masculino , Ratones , Análisis por Matrices de Proteínas , Unión Proteica , Neoplasias Testiculares/metabolismo
12.
J Biol Chem ; 286(33): 29005-29013, 2011 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-21715331

RESUMEN

HIPK2 is a serine/threonine kinase that acts as a coregulator of an increasing number of factors involved in cell survival and proliferation during development and in response to different types of stress. Here we report on a novel target of HIPK2, the cyclin-dependent kinase inhibitor p27(kip1). HIPK2 phosphorylates p27(kip1) in vitro and in vivo at serine 10, an event that accounts for 80% of the total p27(kip1) phosphorylation and plays a crucial role in the stability of the protein. Indeed, HIPK2 depletion by transient or stable RNA interference in tumor cells of different origin was consistently associated with strong reduction of p27(kip1) phosphorylation at serine 10 and of p27(kip1) stability. An initial evaluation of the functional relevance of this HIPK2-mediated regulation of p27(kip1) revealed a contribution to cell motility, rather than to cell proliferation, but only in cells that do not express wild-type p53.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Portadoras/genética , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Humanos , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Serina/genética , Serina/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
J Pathol ; 224(1): 110-20, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21381029

RESUMEN

Oestrogen exposure has been linked to a risk for the development of testicular germ cell cancers. The effects of oestrogen are now known to be mediated by oestrogen receptor-α (ERα) and ERß subtypes, but only ERß has been found in human germ cells of normal testis. However, its expression was markedly diminished in seminomas, embryonal cell carcinomas and mixed germ cell tumours, but remains high in teratomas. PATZ1 is a recently discovered zinc finger protein that, due to the presence of the POZ domain, acts as a transcriptional repressor affecting the basal activity of different promoters. We have previously described that PATZ1 plays a crucial role in normal male gametogenesis and that its up-regulation and mislocalization could be associated with the development of testicular germ cell tumours. Here we show that ERß interacts with PATZ1 in normal germ cells, while down-regulation of ERß associates with transcriptional co-regulator PATZ1 delocalization in human testicular seminomas. In addition, we show that the translocation of PATZ1 from the cytoplasm into the nucleus is regulated by cAMP, which also induces increased expression and nuclear localization of ERß, while this effect is counteracted by using the anti-oestrogen ICI 182-780.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Seminoma/metabolismo , Neoplasias Testiculares/metabolismo , Antineoplásicos Hormonales/farmacología , Núcleo Celular/metabolismo , AMP Cíclico/farmacología , Citoplasma/metabolismo , Regulación hacia Abajo , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Fulvestrant , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/genética , Seminoma/patología , Translocación Genética/efectos de los fármacos , Células Tumorales Cultivadas
14.
Cancer Res ; 70(13): 5379-88, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20530667

RESUMEN

We have previously described a mechanism through which the high-mobility group A1 (HMGA1) proteins inhibit p53-mediated apoptosis by delocalizing the p53 proapoptotic activator homeodomain-interacting protein kinase 2 from the nucleus to the cytoplasm. By this mechanism, HMGA1 modulates the transcription of p53 target genes such as Mdm2, p21(waf1), and Bax, inhibiting apoptosis. Here, we report that HMGA1 antagonizes the p53-mediated transcriptional repression of another apoptosis-related gene, Bcl-2, suggesting a novel mechanism by which HMGA1 counteracts apoptosis. Moreover, HMGA1 overexpression promotes the reduction of Brn-3a binding to the Bcl-2 promoter, thereby blocking the Brn-3a corepressor function on Bcl-2 expression following p53 activation. Consistently, a significant direct correlation between HMGA1 and Bcl-2 overexpression has been observed in human breast carcinomas harboring wild-type p53. Therefore, this study suggests a novel mechanism, based on Bcl-2 induction, by which HMGA1 overexpression contributes to the escape from apoptosis leading to neoplastic transformation.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Genes bcl-2 , Proteína HMGA1a/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Regulación hacia Abajo , Femenino , Proteína HMGA1a/biosíntesis , Humanos , Ratones , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...