Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Genes (Basel) ; 15(5)2024 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-38790192

RESUMEN

TR2 and TR4 (NR2C1 and NR2C2, respectively) are evolutionarily conserved nuclear orphan receptors capable of binding direct repeat sequences in a stage-specific manner. Like other nuclear receptors, TR2 and TR4 possess important roles in transcriptional activation or repression with developmental stage and tissue specificity. TR2 and TR4 bind DNA and possess the ability to complex with available cofactors mediating developmental stage-specific actions in primitive and definitive erythrocytes. In erythropoiesis, TR2 and TR4 are required for erythroid development, maturation, and key erythroid transcription factor regulation. TR2 and TR4 recruit and interact with transcriptional corepressors or coactivators to elicit developmental stage-specific gene regulation during hematopoiesis.


Asunto(s)
Hematopoyesis , Humanos , Animales , Hematopoyesis/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica
2.
J Cell Mol Med ; 25(5): 2404-2417, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33491272

RESUMEN

Drug options for the life-threatening Cushing's disease are limited, and surgical resection or radiation therapy is not invariably effective. Testicular receptor 4 (TR4) has been identified as a novel drug target to treat Cushing's disease. We built the structure model of TR4 and searched the TR4 antagonist candidate via in silico virtual screening. Bexarotene was identified as an antagonist of TR4 that can directly interact with TR4 ligand binding domain (TR4-LBD) and induces a conformational change in the secondary structure of TR4-LBD. Bexarotene suppressed AtT-20 cell growth, proopiomelanocortin (POMC) expression and adrenocorticotropin (ACTH) secretion. Mechanism dissection revealed that bexarotene could suppress TR4-increased POMC expression via promoting the TR4 translocation from the nucleus to the cytoplasm. This TR4 translocation might then result in reducing the TR4 binding to the TR4 response element (TR4RE) on the 5' promoter region of POMC. Results from in vivo mouse model also revealed that oral bexarotene administration markedly suppressed ACTH-secreting tumour growth, adrenal enlargement and the secretion of ACTH and corticosterone in mice with already established tumours. Together, these results suggest that bexarotene may be developed as a potential novel therapeutic drug to better suppress Cushing's disease.


Asunto(s)
Bexaroteno/farmacología , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/antagonistas & inhibidores , Proopiomelanocortina/metabolismo , Transducción de Señal/efectos de los fármacos , Adenoma Hipofisario Secretor de ACTH , Hormona Adrenocorticotrópica/biosíntesis , Animales , Bexaroteno/química , Sitios de Unión , Línea Celular Tumoral , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Expresión Génica , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/química , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Proopiomelanocortina/genética , Unión Proteica , Transporte de Proteínas , Relación Estructura-Actividad , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Sci Adv ; 5(10): eaax6366, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31633027

RESUMEN

Alternative lengthening of telomeres (ALT) is known to use homologous recombination (HR) to replicate telomeric DNA in a telomerase-independent manner. However, the detailed process remains largely undefined. It was reported that nuclear receptors COUP-TFII and TR4 are recruited to the enriched GGGTCA variant repeats embedded within ALT telomeres, implicating nuclear receptors in regulating ALT activity. Here, we identified a function of nuclear receptors in ALT telomere maintenance that involves a direct interaction between COUP-TFII/TR4 and FANCD2, the key protein in the Fanconi anemia (FA) DNA repair pathway. The COUP-TFII/TR4-FANCD2 complex actively induces the DNA damage response by recruiting endonuclease MUS81 and promoting the loading of the PCNA-POLD3 replication complex in ALT telomeres. Furthermore, the COUP-TFII/TR4-mediated ALT telomere pathway does not require the FA core complex or the monoubiquitylation of FANCD2, key steps in the canonical FA pathway. Thus, our findings reveal that COUP-TFII/TR4 regulates ALT telomere maintenance through a novel noncanonical FANCD2 pathway.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Telómero/metabolismo , Secuencias de Aminoácidos , Factor de Transcripción COUP II/antagonistas & inhibidores , Factor de Transcripción COUP II/genética , Línea Celular Tumoral , ADN Polimerasa III/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Fase G2 , Humanos , Mutagénesis Sitio-Dirigida , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/antagonistas & inhibidores , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Homeostasis del Telómero
4.
Biochem Biophys Res Commun ; 508(1): 210-216, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30473219

RESUMEN

The aim of this study was to investigate the effect of miR-4778-3p on the radiosensitivity of cervical cancer cells and to elucidate the underlying mechanism. Tissue samples were collected from eight patients with cervical cancer prior to chemoradiotherapy. MicroRNA chip analyses, RT-PCR, gene transfection, CCK8, wound healing and Transwell assays, colony-forming assay, western blot, and the Dual-Luciferase Reporter Assay System were used to evaluate the role of miR-4778-3p in cervical cancer radiosensitivity and its relationships with target molecules NR2C2 and Med19. Thirty-two differentially expressed miRNA molecules (fold-change > 2; p < 0.05) associated with cervical cancer radioresistance were identified. The expression of miR-4778-3p was significantly lower in recurrent or metastatic patients than in control subjects. In vitro studies using radioresistant HeLa and SiHa cervical cancer cell lines showed that miR-4778-3p upregulation significantly inhibited cell proliferation, invasiveness, and migration after irradiation. There was also a significant increase in apoptosis and a significant decrease in the proportion of cells at the G2/M phase. Further, miR-4778-3p upregulation led to increased expression of apoptosis-related molecules, such as Bax, Caspase-3, Caspase-8, and Caspase-9. Reporter gene assays showed that miR-4778-3p bound specifically to NR2C2 and Med19 and negatively regulated their expression. Thus, miR-4778-3p reduces the vitality, proliferation, and migration of radioresistant cervical cancer cells and may regulate the radiosensitivity of cervical cancer by targeting and regulating NR2C2 and Med19 expression.


Asunto(s)
Complejo Mediador/metabolismo , MicroARNs/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Neoplasias del Cuello Uterino/genética , Apoptosis , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Complejo Mediador/genética , MicroARNs/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/radioterapia
5.
Dev Comp Immunol ; 90: 152-156, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30248360

RESUMEN

Transforming growth factor-ß activated kinase 1 (TAK1) is a crucial signal transducer in multiple signaling pathways. TAK1 binds TAB1, TAB2, and TAB3, which act as activators and adaptors that specifically regulate the activation of TAK1. To date, the role of TABs is largely unknown in fish. In the present study, a TAB1 cDNA sequence was identified in grouper (Epinephelus coioides), and designated EcTAB1. The full-length open reading frame of EcTAB1 is 1, 521 bp; it encodes 506 amino acids that contains an N-terminal PP2C domain. Many important functional sites in mammalian TAB1 were conserved in TAB1 from grouper and from other fish. Multiple sequence alignment showed that EcTAB1 protein shared high sequence identity with TAB1 of other fish, especially with Stegastes partitus (95% identity). TAB1 was clustered into the same subgroup with other fish TAB1 in the phylogenetic tree. Tissue expression analysis indicated that TAB1 was widely distributed in different tissues. After infection with Cryptocaryon irritans, EcTAB1 expression was up-regulated in the infection site (gills). Besides, EcTAB1 was expressed throughout the grouper spleen (GS) cells and significantly enhanced the activation of NF-κB.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Infecciones por Cilióforos/inmunología , Cilióforos/fisiología , Enfermedades de los Peces/inmunología , Proteínas de Peces/genética , Branquias/inmunología , Perciformes/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Clonación Molecular , Proteínas de Peces/metabolismo , Branquias/parasitología , Humanos , FN-kappa B/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Filogenia , Alineación de Secuencia , Transducción de Señal , Regulación hacia Arriba
6.
Dev Comp Immunol ; 93: 18-27, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30571995

RESUMEN

Candida albicans is one of the most common fungal pathogens of humans. Currently, there are limitations in the evaluation of C. albicans infection in existing animal models, especially in terms of understanding the influence of specific infectious stages of the fungal pathogen on the host. We show that C. albicans infects, grows and invades tissues in the planarian flatworm Schmidtea mediterranea, and that the planarian responds to infection by activating components of the host innate immune system to clear and repair host tissues. We study different stages of C. albicans infection and demonstrate that planarian stem cells increase division in response to fungal infection, a process that is likely evolutionarily conserved in metazoans. Our results implicate MORN2 and TAK1/p38 signaling pathways as possible mediators of the host innate immune response to fungal infection. We propose the use of planarians as a model system to investigate host-pathogen interactions during fungal infections.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Planarias/inmunología , Animales , Candida albicans/crecimiento & desarrollo , Candidiasis/microbiología , Modelos Animales de Enfermedad , Proteínas Nucleares/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Planarias/microbiología , Células Madre/citología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Oncogene ; 37(44): 5901-5912, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29973687

RESUMEN

While TR4 nuclear receptor plays key roles to promote prostate cancer progression, its roles to alter the progression of clear cell renal cell carcinoma (ccRCC), remains unclear. Here, we demonstrate that TR4 can promote the ccRCC cell vasculogenic mimicry (VM) formation and its associated metastasis via modulating the miR490-3p/vimentin (VIM) signals. Mechanism dissection revealed that TR4 might increase the oncogene VIM expression via decreasing the miR-490-3p expression through direct binding to the TR4-response-elements (TR4REs) on the promoter region of miR-490-3p, which might then directly target the 3' UTR of VIM-mRNA to increase its protein expression. Preclinical studies using the in vivo mouse model with xenografted RCC Caki-1 cells into the sub-renal capsule of nude mice also found that TR4 could promote the ccRCC VM and its associated metastasis via modulating the miR490-3p/VIM signals. Together, results from preclinical studies using multiple RCC cell lines and the in vivo mouse model all conclude that TR4 may play a key role to promote ccRCC VM formation and metastasis and targeting the newly identified TR4/miR-490-3p/VIM signals with small molecules may help us to develop a new therapeutic approach to better suppress the ccRCC metastasis.


Asunto(s)
Carcinoma de Células Renales/genética , Neoplasias Renales/genética , MicroARNs/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Vimentina/genética , Animales , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/secundario , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/metabolismo , Neoplasias Renales/secundario , Ratones , Ratones Endogámicos BALB C , Vimentina/metabolismo
8.
Blood ; 130(23): 2537-2547, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29018082

RESUMEN

The orphan nuclear receptors TR4 (NR2C2) and TR2 (NR2C1) are the DNA-binding subunits of the macromolecular complex, direct repeat erythroid-definitive, which has been shown to repress ε- and γ-globin transcription during adult definitive erythropoiesis. Previous studies implied that TR2 and TR4 act largely in a redundant manner during erythroid differentiation; however, during the course of routine genetic studies, we observed multiple variably penetrant phenotypes in the Tr4 mutants, suggesting that indirect effects of the mutation might be masked by multiple modifying genes. To test this hypothesis, Tr4+/- mutant mice were bred into a congenic C57BL/6 background and their phenotypes were reexamined. Surprisingly, we found that homozygous Tr4 null mutant mice expired early during embryogenesis, around embryonic day 7.0, and well before erythropoiesis commences. We further found that Tr4+/- erythroid cells failed to fully differentiate and exhibited diminished proliferative capacity. Analysis of Tr4+/- mutant erythroid cells revealed that reduced TR4 abundance resulted in decreased expression of genes required for heme biosynthesis and erythroid differentiation (Alad and Alas2), but led to significantly increased expression of the proliferation inhibitory factor, cyclin dependent kinase inhibitor (Cdkn1c) These studies support a vital role for TR4 in promoting erythroid maturation and proliferation, and demonstrate that TR4 and TR2 execute distinct, individual functions during embryogenesis and erythroid differentiation.


Asunto(s)
Diferenciación Celular/genética , Células Eritroides/citología , Células Eritroides/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Anemia/sangre , Anemia/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular/genética , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Heterocigoto , Homocigoto , Linfopoyesis/genética , Ratones , Ratones Noqueados , Mutación , Mielopoyesis/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo
9.
Biochem Biophys Res Commun ; 490(2): 423-428, 2017 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-28623141

RESUMEN

Sirtuin 7 (SIRT7) is an NAD+-dependent deacetylase/deacylase, but only a limited number of SIRT7 substrates have been identified. Recently, we found that Sirt7 knockout mice are resistant to high-fat diet-induced fatty liver, and that SIRT7 positively regulates the protein level of TR4, a nuclear receptor involved in lipid metabolism, by inhibiting the CUL4B/DDB1/DCAF1 E3 ubiquitin ligase complex. However, the mechanism by which SIRT7 inhibits the E3 ubiquitin ligase complex was not identified. Here, we demonstrate that SIRT7 binds directly to DDB1 and deacetylates DDB1 at Lys1121. K1121R-DDB1 (a deacetylation-mimicking mutant) displayed reduced binding with DCAF1. The expression of TR4 protein and TR4 target genes, including Cd36, Cidea, Cidec and Pparg1, was increased in K1121R-DDB1-overexpressing Hepa1-6 cells compared to WT-DDB1-overexpressing cells. Our results indicate that the SIRT7-mediated deacetylation of K1121 attenuates the activity of the CUL4B/DDB1/DCAF1 E3 ubiquitin ligase complex by reducing binding between DDB1 and DCAF1, leading to the increased expression of TR4.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Unión Proteica , Mapas de Interacción de Proteínas , Proteolisis , Sirtuinas/genética
10.
Mol Cell Biochem ; 430(1-2): 139-147, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28210900

RESUMEN

Renilla luciferase reporter is a widely used internal control in dual luciferase reporter assay system, where its transcription is driven by a constitutively active promoter. However, the authenticity of the Renilla luciferase response in some experimental settings has recently been questioned. Testicular receptor 4 (TR4, also known as NR2C2) belongs to the subfamily 2 of nuclear receptors. TR4 binds to a direct repeat regulatory element in the promoter of a variety of target genes and plays a key role in tumorigenesis, lipoprotein regulation, and central nervous system development. In our experimental system using murine pituitary corticotroph tumor AtT20 cells to investigate TR4 actions on POMC transcription, we found that overexpression of TR4 resulted in reduced Renilla luciferase expression whereas knockdown TR4 increased Renilla luciferase expression. The TR4 inhibitory effect was mediated by the TR4 DNA-binding domain and behaved similarly to the GR and its agonist, Dexamethasone. We further demonstrated that the chimeric intron, commonly present in various Renilla plasmid backbones such as pRL-Null, pRL-SV40, and pRL-TK, was responsible for TR4's inhibitory effect. The results suggest that an intron-free Renilla luciferase reporter may provide a satisfactory internal control for TR4 at certain dose range. Our findings advocate caution on the use of Renilla luciferase as an internal control in TR4-directed studies to avoid misleading data interpretation.


Asunto(s)
Dexametasona/farmacología , Genes Reporteros , Luciferasas de Renilla/biosíntesis , Proteínas de Neoplasias/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Neoplasias Hipofisarias/metabolismo , Animales , Línea Celular Tumoral , Reacciones Falso Positivas , Luciferasas de Renilla/genética , Ratones , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/agonistas , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/genética
11.
Oncotarget ; 7(43): 69149-69158, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27708250

RESUMEN

We recently demonstrated that the orphan nuclear receptor testicular receptor 4 (TR4) is a potent regulator of corticotroph tumor growth and hormone secretion. The Ras/Raf/MEK/ERK pathway is commonly overactivated in human tumors and we have demonstrated that corticotroph tumor TR4 is activated by ERK1/2-mediated phosphorylation. We evaluated effects of MEK-162, a selective, non-ATP-competitive allosteric inhibitor of MEK1/2, on murine and human in vitro and in vivo corticotroph tumor proliferation and adrenocorticotrophic hormone (ACTH) secretion. MEK-162 treatment dose-dependently inhibited corticotroph tumor proliferation, induced apoptosis, reduced pro-opiomelanocortin (POMC) mRNA levels and inhibited ACTH secretion in vitro. Similar findings were obtained in human corticotroph tumor primary cultures (n = 5). These actions of MEK-162 were augmented in the presence of TR4 overexpression, suggesting that TR4 levels may serve as a predictive biomarker of MEK-162 corticotroph tumor responsiveness. Additionally, MEK-162 treatment reduced TR4 protein expression and blocked recruitment of TR4 to bind its consensus site on the POMC promoter (-854bp to -637bp), elucidating multiple mechanisms to control TR4 corticotroph tumor actions. In a murine corticotroph tumor in vivo model of Cushing's disease, MEK-162 treatment inhibited tumor growth and reduced tumor-derived circulating plasma ACTH, and corticosterone levels. These results demonstrate the potent actions of MEK-162 to inhibit corticotroph tumor growth and hormone secretion in vitro and in vivo via TR4-dependent and independent mechanisms, and raise the possibility of MEK-162 as a novel therapy for Cushing's disease.


Asunto(s)
Bencimidazoles/farmacología , Proliferación Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/tratamiento farmacológico , Adenoma Hipofisario Secretor de ACTH/tratamiento farmacológico , Adenoma Hipofisario Secretor de ACTH/genética , Adenoma Hipofisario Secretor de ACTH/metabolismo , Adenoma/tratamiento farmacológico , Adenoma/genética , Adenoma/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Proopiomelanocortina/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Células Tumorales Cultivadas
12.
FEBS Lett ; 590(18): 3264-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27497262

RESUMEN

Transforming growth factor-ß-activated kinase (TAK)1-binding proteins (TAB) activate nuclear factor-κB by linking TAK1 to signaling molecules. We investigated the mechanisms underlying B-cell receptor (BCR) signaling in TAB2- and TAB3-deficient and TAB3 domain deletion mutant DT40 B cell lines. Loss of TAB2 and TAB3 abolished BCR-induced inhibitor of κB kinase (IKK) activation and TAK1 binding to caspase recruitment domain membrane-associated guanylate kinase protein (CARMA)1. Deletion of TAB3, coupling of ubiquitin conjugation to ER degradation, coiled-coil, and zinc finger domains blocked IKK activation and association with CARMA1. Thus, TAB2 and TAB3 connect signaling molecules that activate IKK in BCR signaling.


Asunto(s)
Quinasa I-kappa B/metabolismo , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular , Pollos , Guanilato Ciclasa/metabolismo , Unión Proteica , Ubiquitina/metabolismo
13.
J Clin Endocrinol Metab ; 101(8): 3123-33, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27253665

RESUMEN

CONTEXT: Glucocorticoids are powerful steroid hormones that regulate development, metabolism, and immune response. However, glucocorticoid unresponsiveness or resistance is observed in the treatment of inflammatory, autoimmune, and lymphoproliferative diseases and significantly limits their efficacy. OBJECTIVE: In Cushing's disease, although some glucocorticoid-mediated suppression of pituitary-derived ACTH is seen, corticotroph tumors exhibit relative resistance to glucocorticoid action. We previously demonstrated that testicular orphan receptor 4 (TR4) binds to the pro-opiomelanocortin (POMC) promoter to induce corticotroph tumor POMC expression and ACTH secretion, and we hypothesized that TR4 may interact with glucocorticoid signaling to modulate POMC expression and action. RESULTS: Here we demonstrate that TR4 abrogates glucocorticoid receptor (GR)- or dexamethasone-mediated POMC and activator protein-1 transrepression in both murine and human pituitary corticotroph tumor cells. Co-immunoprecipitation studies indicate that TR4 and GR interact directly with each other, resulting in TR4-mediated disruption of GR binding to the POMC promoter. CONCLUSION: These results demonstrate that TR4 binds GR to play an important role in glucocorticoid-directed corticotroph tumor POMC regulation in addition to modulating glucocorticoid actions on other GR targets. Characterization of this pathway may offer important insights into glucocorticoid resistance and may identify a novel approach for the treatment of Cushing's disease and the glucocorticoid-resistant states.


Asunto(s)
Errores Innatos del Metabolismo/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/fisiología , Receptores de Glucocorticoides/deficiencia , Animales , Dexametasona/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción AP-1/metabolismo , Células Tumorales Cultivadas
14.
Biochimie ; 127: 79-85, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27109382

RESUMEN

lipid metabolism is the major causes of atherosclerosis. There is increasing evidence that miR-133a plays an important role in atherosclerosis. However, the regulatory mechanism of miR-133a in macrophages is still unclear. Several lines of evidence indicate that loss of TR4 leads to reduce lipid accumulation in liver and adipose tissues, etc, and lesional macrophages-derived TR4 can greatly increase the foam cell formation through increasing the CD36-mediated the uptake of ox-LDL. Interestingly, computational analysis suggests that TR4 may be a target gene of miR-133a. Here, we examined whether miR-133a regulates TR4 expression in ox-LDL-induced mouse RAW 264.7 macrophages, thereby affecting lipid accumulation. Using ox-LDL-treatment RAW 264.7 macrophages transfected with miR-133a mimics or inhibitors, we have showed that miR-133a can directly regulate the expression of TR4 in RAW 264.7 cells, thereby attenuates CD36-medide lipid accumulation. Furthermore, our studies suggest an additional explanation for the regulatory mechanism of miR-133a regulation to its functional target, TR4 in RAW 264.7 macrophages. Thus, our findings suggest that miR-133a may regulate lipid accumulation in ox-LDL-stimulated RAW 264.7 macrophages via TR4-CD36 pathway.


Asunto(s)
Antígenos CD36/metabolismo , Metabolismo de los Lípidos/genética , Macrófagos/citología , Macrófagos/metabolismo , MicroARNs/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Transducción de Señal/genética , Animales , Secuencia de Bases , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Ratones , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
15.
Environ Toxicol Pharmacol ; 44: 18-24, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27084994

RESUMEN

Di-(2-ethylhexyl) phthalate (DEHP) is a widely used environmental endocrine disruptor. Many studies have reported that DEHP exposure causes reproductive toxicity and cells apoptosis. However, the mechanism by which DEHP exposure causes male reproductive toxicity remains unknown. This study investigated the role of the testicular orphan nuclear receptor4 (TR4)/Bcl-2 pathway in apoptosis induced by DEHP, which resulted in reproductive damage. To elucidate the mechanism underpinning the male reproductive toxicity of DEHP, we sought to investigate apoptotic effects, expression levels of TR4/Bcl-2 pathway in GC-2spd cells, including TR4, Bcl-2 and caspase-3. GC-2spd cells were exposed to various concentrations of DEHP (0, 50, 100, or 200µM). The results indicated that, with the increase of the concentrations of DEHP, the survival rate of cell decreased gradually. DEHP exposure at over 100µM significantly induced apoptotic cell death. DEHP decreased SOD and GSH-Px activity in 200µM group. Compared to the control group, the mRNA levels of caspase-3 increased significantly, however, Bcl-2 mRNA decreased (P<0.05). In addition, there was a significant reduction in TR4, Bcl-2 and procaspase-3 protein levels. Taken together, these results lead us to speculate that in vitro exposure to DEHP might induce apoptosis in GC-2spd cells through the TR4/Bcl-2 pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Dietilhexil Ftalato/toxicidad , Disruptores Endocrinos/toxicidad , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Plastificantes/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis/fisiología , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Malondialdehído/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Superóxido Dismutasa/metabolismo
16.
Cancer Res ; 76(1): 117-26, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26677977

RESUMEN

The use of multikinase inhibitors (MKI) in oncology, such as sorafenib, is associated with a cutaneous adverse event called hand-foot skin reaction (HFSR), in which sites of pressure or friction become inflamed and painful, thus significantly impacting quality of life. The pathogenesis of MKI-induced HFSR is unknown, and the only available treatment options involve dose reduction or discontinuation of therapy, which have negative effects on primary disease management. To investigate the underlying mechanisms by which sorafenib promotes keratinocyte cytotoxicity and subsequent HFSR induction, we performed a transporter-directed RNAi screen in human epidermal keratinocytes and identified SLC22A20 (OAT6) as an uptake carrier of sorafenib. Further investigations into the intracellular mechanism of sorafenib activity through in situ kinome profiling identified the mitogen-activated protein kinase MAP3K7 (TAK1) as a target of sorafenib that induces cell death. Finally, we demonstrate that sorafenib induced keratinocyte injury in vivo and that this effect could be reversed by cotreatment with the OAT6 inhibitor probenecid. Collectively, our findings reveal a novel pathway that regulates the entry of some MKIs into keratinocytes and explains the basis underlying sorafenib-induced skin toxicity, with important implications for the therapeutic management of HFSR.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Niacinamida/análogos & derivados , Transportadores de Anión Orgánico/metabolismo , Compuestos de Fenilurea/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Enfermedades de la Piel/inducido químicamente , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Femenino , Células Hep G2 , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos C57BL , Niacinamida/farmacocinética , Niacinamida/toxicidad , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Transportadores de Anión Orgánico/genética , Compuestos de Fenilurea/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Distribución Aleatoria , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología , Sorafenib , Transfección
17.
J Steroid Biochem Mol Biol ; 157: 27-40, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26159912

RESUMEN

Nuclear receptors are defined as a family of ligand regulated transcription factors [1-6]. While this definition reflects that ligand binding is a key property of nuclear receptors, it is still a heated subject of debate if all the nuclear receptors (48 human members) can bind ligands (ligands referred here to both physiological and synthetic ligands). Recent studies in nuclear receptor structure biology and pharmacology have undoubtedly increased our knowledge of nuclear receptor functions and their regulation. As a result, they point to new avenues for the discovery and development of nuclear receptor regulators, including nuclear receptor ligands. Here we review the recent literature on orphan nuclear receptor structural analysis and ligand identification, particularly on the orphan nuclear receptors that do not heterodimerize with retinoid X receptors, which we term as non-X orphan receptors. We also propose a speculative "retinoid hypothesis" for a subset of non-X orphan nuclear receptors, which we hope to help shed light on orphan nuclear receptor biology and drug discovery. This article is part of a Special Issue entitled 'Orphan Nuclear Receptors'.


Asunto(s)
Receptores Nucleares Huérfanos/química , Receptores Nucleares Huérfanos/metabolismo , Retinoides/metabolismo , Animales , Sitios de Unión , Factor de Transcripción COUP II/química , Factor de Transcripción COUP II/metabolismo , Receptor Nuclear Huérfano DAX-1/química , Receptor Nuclear Huérfano DAX-1/metabolismo , Humanos , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/química , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Conformación Proteica , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Retinoides/química , Factor Esteroidogénico 1/química , Factor Esteroidogénico 1/metabolismo
18.
Bioinformatics ; 31(15): 2537-44, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840044

RESUMEN

MOTIVATION: Modeling regulatory networks using expression data observed in a differentiation process may help identify context-specific interactions. The outcome of the current algorithms highly depends on the quality and quantity of a single time-course dataset, and the performance may be compromised for datasets with a limited number of samples. RESULTS: In this work, we report a multi-layer graphical model that is capable of leveraging many publicly available time-course datasets, as well as a cell lineage-specific data with small sample size, to model regulatory networks specific to a differentiation process. First, a collection of network inference methods are used to predict the regulatory relationships in individual public datasets. Then, the inferred directional relationships are weighted and integrated together by evaluating against the cell lineage-specific dataset. To test the accuracy of this algorithm, we collected a time-course RNA-Seq dataset during human erythropoiesis to infer regulatory relationships specific to this differentiation process. The resulting erythroid-specific regulatory network reveals novel regulatory relationships activated in erythropoiesis, which were further validated by genome-wide TR4 binding studies using ChIP-seq. These erythropoiesis-specific regulatory relationships were not identifiable by single dataset-based methods or context-independent integrations. Analysis of the predicted targets reveals that they are all closely associated with hematopoietic lineage differentiation.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Eritroides/metabolismo , Eritropoyesis/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Células Madre/metabolismo , Algoritmos , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Eritroides/citología , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Priones/genética , Priones/metabolismo , Curva ROC , Tamaño de la Muestra , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Cancer Gene Ther ; 21(10): 411-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25104727

RESUMEN

Docetaxel-based therapy is one of the first-line options for castration-resistant prostate cancer (CRPC). However, a large proportion of CRPC patients show different extents of docetaxel resistance. The current study aims to investigate the role of testicular nuclear receptor 4 (TR4) in docetaxel resistance in CRPC. TR4 expression level in prostate biopsy samples from CRPC patients treated with docetaxel was measured by immunohistochemistry (IHC). Alternation of TR4 expression in prostate cancer (PCa) cell line PC3 was applied to find out the influence of TR4 on half-maximal inhibitory concentration (IC50), cell viability and cell apoptosis. Patients who failed to achieve prostate-specific antigen (PSA) response (<50% PSA reduction from baseline) after docetaxel-based chemotherapy had a comparatively higher TR4 expression than those who achieved PSA response (⩾50% PSA reduction from baseline). Knocking down TR4 in PC3 cells led to a lower IC50 dose, poorer cell viability and more cell apoptosis when treated with docetaxel, whereas overexpression of TR4 in PC3 led to a higher IC50 dose, better cell viability and less cell apoptosis. TR4 enhances the chemo-resistance of docetaxel in CRPC. It may serve as a biomarker to determine the prognosis of docetaxel-based therapy and as a potential therapy target to combine with docetaxel to better suppress CRPC.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Taxoides/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Docetaxel , Relación Dosis-Respuesta a Droga , Expresión Génica , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Concentración 50 Inhibidora , Lentivirus/genética , Masculino , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción Genética
20.
Endocr Relat Cancer ; 21(3): R279-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24623743

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ, NR1C3) and testicular receptor 4 nuclear receptor (TR4, NR2C2) are two members of the nuclear receptor (NR) superfamily that can be activated by several similar ligands/activators including polyunsaturated fatty acid metabolites, such as 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, as well as some anti-diabetic drugs such as thiazolidinediones (TZDs). However, the consequences of the transactivation of these ligands/activators via these two NRs are different, with at least three distinct phenotypes. First, activation of PPARγ increases insulin sensitivity yet activation of TR4 decreases insulin sensitivity. Second, PPARγ attenuates atherosclerosis but TR4 might increase the risk of atherosclerosis. Third, PPARγ suppresses prostate cancer (PCa) development and TR4 suppresses prostate carcinogenesis yet promotes PCa metastasis. Importantly, the deregulation of either PPARγ or TR4 in PCa alone might then alter the other receptor's influences on PCa progression. Knocking out PPARγ altered the ability of TR4 to promote prostate carcinogenesis and knocking down TR4 also resulted in TZD treatment promoting PCa development, indicating that both PPARγ and TR4 might coordinate with each other to regulate PCa initiation, and the loss of either one of them might switch the other one from a tumor suppressor to a tumor promoter. These results indicate that further and detailed studies of both receptors at the same time in the same cells/organs may help us to better dissect their distinct physiological roles and develop better drug(s) with fewer side effects to battle PPARγ- and TR4-related diseases including tumor and cardiovascular diseases as well as metabolic disorders.


Asunto(s)
Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/prevención & control , Miembro 2 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , PPAR gamma/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/prevención & control , Animales , Regulación de la Expresión Génica , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...