Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Biol Chem ; 288(16): 11047-65, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23426362

RESUMEN

The androgen receptor (AR) is essential for diverse aspects of prostate development and function. Molecular mechanisms by which prostate cancer (PC) cells redirect AR signaling to genes that primarily support growth are unclear. A systematic search for critical AR-tethering proteins led to ELK1, an ETS transcription factor of the ternary complex factor subfamily. Although genetically redundant, ELK1 was obligatory for AR-dependent growth and clonogenic survival in both hormone-dependent PC and castration-recurrent PC cells but not for AR-negative cell growth. AR required ELK1 to up-regulate a major subset of its target genes that was strongly and primarily enriched for cell growth functions. AR functioned as a coactivator of ELK1 by association through its A/B domain, bypassing the classical mechanism of ELK1 activation by phosphorylation and without inducing ternary complex target genes. The ELK1-AR synergy per se was ligand-independent, although it required ligand for nuclear localization of AR as targeting the AR A/B domain to the nucleus recapitulated the action of hormone; accordingly, Casodex was a poor antagonist of the synergy. ELK3, the closest substitute for ELK1 in structure/function and genome recognition, did not interact with AR. ELK1 thus directs selective and sustained gene induction that is a substantial and critical component of growth signaling by AR in PC cells. The ELK1-AR interaction offers a functionally tumor-selective drug target.


Asunto(s)
Núcleo Celular/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Adolescente , Adulto , Antagonistas de Andrógenos/farmacología , Anilidas/farmacología , Núcleo Celular/genética , Núcleo Celular/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Humanos , Masculino , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Estructura Terciaria de Proteína , Receptores Androgénicos/genética , Compuestos de Tosilo/farmacología , Proteína Elk-1 con Dominio ets/genética
2.
Biochemistry ; 48(34): 8249-60, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19639961

RESUMEN

The glycosyl-phosphatidylinositol anchored folate receptor (FR) mediates selective delivery of a broad range of experimental drugs to the receptor-rich tumors, but molecular mechanisms controlling FR internalization have not been adequately studied. FR quantitatively recycles between the cell surface and endocytic compartments via a Cdc42-dependent pinocytic pathway. Protein kinase C (PKC) activators including diacylglycerol and phorbol ester have previously been reported to increase the proportion of FR on the cell surface. Here we identify the alpha-subtype of PKC as the mediator of phorbol ester action on FR recycling and provide evidence that activated PKCalpha is recruited to FR-rich membrane microdomains where, in association with its receptor RACK1, it inhibits FR internalization; the activation state of Cdc42 remains unaltered. We also show that the PKC substrate, annexin II, is required for FR internalization. The studies clarify a molecular mechanism for the regulation of FR recycling through PKC which could potentially be exploited for effective drug delivery.


Asunto(s)
Proteínas Portadoras/metabolismo , Endocitosis , Proteína Quinasa C-alfa/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Anexina A2/metabolismo , Secuencia de Bases , Línea Celular , Detergentes/farmacología , Endocitosis/efectos de los fármacos , Receptores de Folato Anclados a GPI , Humanos , Ligandos , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C-alfa/genética , Transporte de Proteínas/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Proteína de Unión al GTP cdc42/metabolismo
3.
Oncogene ; 37(26): 3528-3548, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29563610

RESUMEN

Oncogene-induced senescence (OIS) is an intrinsic tumor suppression mechanism that requires the p53 and RB pathways and post-translational activation of C/EBPß through the RAS-ERK cascade. We previously reported that in transformed/proliferating cells, C/EBPß activation is inhibited by G/U-rich elements (GREs) in its 3'UTR. This mechanism, termed "3'UTR regulation of protein activity" (UPA), maintains C/EBPß in a low-activity state in tumor cells and thus facilitates senescence bypass. Here we show that C/EBPß UPA is overridden by AMPK signaling. AMPK activators decrease cytoplasmic levels of the GRE binding protein HuR, which is a key UPA component. Reduced cytoplasmic HuR disrupts 3'UTR-mediated trafficking of Cebpb transcripts to the peripheral cytoplasm-a fundamental feature of UPA-thereby stimulating C/EBPß activation and growth arrest. In primary cells, oncogenic RAS triggers a Ca++-CaMKKß-AMPKα2-HuR pathway, independent of AMPKα1, that is essential for C/EBPß activation and OIS. This axis is disrupted in cancer cells through down-regulation of AMPKα2 and CaMKKß. Thus, CaMKKß-AMPKα2 signaling constitutes a key tumor suppressor pathway that activates a novel UPA-cancelling mechanism to unmask the cytostatic and pro-senescence functions of C/EBPß.


Asunto(s)
Regiones no Traducidas 3'/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Senescencia Celular/fisiología , Neoplasias/patología , Proteínas ras/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Proteína 1 Similar a ELAV/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Noqueados , Células 3T3 NIH
4.
Mol Cell Biol ; 36(5): 693-713, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26667036

RESUMEN

The integrated stress response (ISR) controls cellular adaptations to nutrient deprivation, redox imbalances, and endoplasmic reticulum (ER) stress. ISR genes are upregulated in stressed cells, primarily by the bZIP transcription factor ATF4 through its recruitment to cis-regulatory C/EBP:ATF response elements (CAREs) together with a dimeric partner of uncertain identity. Here, we show that C/EBPγ:ATF4 heterodimers, but not C/EBPß:ATF4 dimers, are the predominant CARE-binding species in stressed cells. C/EBPγ and ATF4 associate with genomic CAREs in a mutually dependent manner and coregulate many ISR genes. In contrast, the C/EBP family members C/EBPß and C/EBP homologous protein (CHOP) were largely dispensable for induction of stress genes. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferate poorly and exhibit oxidative stress due to reduced glutathione levels and impaired expression of several glutathione biosynthesis pathway genes. Cebpg(-/-) mice (C57BL/6 background) display reduced body size and microphthalmia, similar to ATF4-null animals. In addition, C/EBPγ-deficient newborns die from atelectasis and respiratory failure, which can be mitigated by in utero exposure to the antioxidant, N-acetyl-cysteine. Cebpg(-/-) mice on a mixed strain background showed improved viability but, upon aging, developed significantly fewer malignant solid tumors than WT animals. Our findings identify C/EBPγ as a novel antioxidant regulator and an obligatory ATF4 partner that controls redox homeostasis in normal and cancerous cells.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Estrés Oxidativo , Factor de Transcripción Activador 4/análisis , Factor de Transcripción Activador 4/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/análisis , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular , Femenino , Feto/anomalías , Feto/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Glutatión/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Neoplasias/genética , Neoplasias/metabolismo , Multimerización de Proteína , Elementos de Respuesta , Factor de Transcripción CHOP/metabolismo
5.
Mol Endocrinol ; 25(4): 621-34, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21330406

RESUMEN

A need for androgen response elements (AREs) for androgen receptor (AR)-dependent growth of hormone depletion-insensitive prostate cancer is generally presumed. In such cells, androgen-independent activation by AR of certain genes has been attributed to selective increases in basal associations of AR with putative enhancers. We examined the importance of AR binding to DNA in prostate cancer cells in which proliferation in the absence of hormone was profoundly (∼ 90%) dependent on endogenous AR and where the receptor was not up-regulated or mutated but was predominantly nuclear. Here, ARE-mediated promoter activation and the binding of AR to a known ARE in the chromatin remained entirely androgen dependent, and the cells showed an androgen-responsive gene expression profile with an unaltered sensitivity to androgen dose. In the same cells, a different set of genes primarily enriched for cell division functions was activated by AR independently of hormone and significantly overlapped the signature gene overexpression profile of hormone ablation-insensitive clinical tumors. After knockdown of endogenous AR, hormone depletion-insensitive cell proliferation and AR apoprotein-dependent gene expression were rescued by an AR mutant that was unable to bind to ARE but that could transactivate through a well-established AR tethering protein. Hormone depletion-insensitive AR binding sites in the chromatin were functional, binding, and responding to both the wild-type and the mutant AR and lacked enrichment for canonical or noncanonical ARE half-sites. Therefore, a potentially diverse set of ARE-independent mechanisms of AR interactions with target genes must underlie truly hormone depletion-insensitive gene regulation and proliferation in prostate cancer.


Asunto(s)
Andrógenos/deficiencia , Neoplasias de la Próstata/metabolismo , Elementos de Respuesta , Andrógenos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , Receptores Androgénicos/metabolismo , Transducción de Señal
6.
J Steroid Biochem Mol Biol ; 122(5): 333-40, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20817090

RESUMEN

The folate receptor α (FRα) is critical for normal embryonic and fetal development. The receptor has a relatively narrow tissue specificity which includes the visceral endoderm and the placenta and mediates delivery of folate, inadequacy of which results in termination of pregnancy or developmental defects. We have previously reported that the FRα gene is negatively and directly regulated by estrogen and positively but indirectly by progesterone and glucocorticoid. To further investigate hormonal control of this gene and in view of the growing evidence for the importance of the androgen receptor (AR) in endometrial and placental functions, we examined the response of the FRα gene to androgen. Here we demonstrate that the FRα gene is directly activated by androgen. The P4 promoter of the FRα gene is the target of hormone-dependent activation by the androgen receptor (AR) in a manner that is co-activator-dependent. The site of functional association of AR in the FRα gene maps to a 35bp region occurring ∼1500bp upstream of the target promoter. The functional elements within this region are an androgen response element (ARE) half-site and a non-canonical C/EBP element that cooperate to recruit AR in a manner that is dependent on the DNA-bound C/EBPα. Since the placenta is rich in C/EBPα, the findings underscore the multiplicity of mechanisms by which the FRα gene is under the exquisite control of steroid hormones.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Receptores de Folato Anclados a GPI/metabolismo , Receptores Androgénicos/fisiología , Femenino , Células HeLa , Humanos , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas/fisiología , Receptores Androgénicos/genética , Activación Transcripcional , Transfección
7.
Prostate ; 68(11): 1206-14, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18481268

RESUMEN

BACKGROUND: C/EBPalpha is a critical mediator of terminal differentiation and a tumor suppressor through its strong antiproliferative actions on cell cycle regulatory proteins. C/EBPalpha also appears to regulate androgen receptor (AR) AR signaling. There, is a paucity of information on the expression and sub-cellular localization of C/EBPalpha in normal mouse and human prostate and in prostate cancer. METHODS: Immunohistochemistry of tissues including tissue arrays, quantitative polymerase chain reaction and mRNA expression database mining. RESULTS: In the mouse prostate epithelium, C/EBPalpha was present at 1 week postnatal localized in the cytosol, began to show nuclear localization at 8 weeks and continued to show prominent nuclear expression at 10 weeks and beyond; C/EBPalpha mRNA was expressed at all ages. In humans, C/EBPalpha showed prominent nuclear localization from peripubescence up to middle age but was sequestered in the cytosol in older individuals; the mRNA level for C/EBPalpha remained essentially unchanged. Most prostate adenocarcinomas expressed a range of levels of C/EBPalpha mRNA and protein that were relatively high in metastatic tumors in a manner that correlated with AR expression; however, most cells showed C/EBPalpha sequestered in the cytosol. CONCLUSIONS: Temporal changes in sub-cellular localization of C/EBPalpha are consistent with a role in prostate differentiation and as a prostate tumor suppressor; the cytoplasmic sequestration of C/EBPalpha, unique to older human prostates, is arguably a permissive condition for the greater frequency of proliferative disorders of the prostate. In malignant prostate C/EBPalpha may be available to regulate AR signaling through transient changes in its sub-cellular localization.


Asunto(s)
Adenocarcinoma/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Próstata/crecimiento & desarrollo , Próstata/fisiología , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Animales , Núcleo Celular/metabolismo , Citosol/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Próstata/patología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA