Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Genes Genomics ; 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38847971

RESUMEN

BACKGROUND: RNA-binding proteins (RBPs) perform various biological functions in humans and are associated with several diseases, including cancer. Therefore, RBPs have emerged as novel therapeutic targets. Although recent investigations have shown that RBPs have crucial functions in breast cancer (BC), detailed research is underway to determine the RBPs that are closely related to cancers. OBJECTIVE: To provide an insight into estrogen receptor (ER) regulation by cold-inducible RNA binding protein (CIRBP) as a novel therapeutic target. RESULTS: By analyzing the genomic data, we identified a potential RBP in BC. We found that CIRBP is highly correlated with ER function and influences clinical outcomes, such as patient survival and endocrine therapy responsiveness. In addition, CIRBP influences the proliferation of BC cells by directly binding to ER-RNA. CONCLUSION: Our results suggest that CIRBP is a novel upstream regulator of ER and that the interplay between CIRBP and ER may be associated with the clinical relevance of BC.

2.
J Biol Chem ; 287(36): 30769-80, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22798067

RESUMEN

Hormonal therapies, mainly combinations of anti-androgens and androgen deprivation, have been the mainstay treatment for advanced prostate cancer because the androgen-androgen receptor (AR) system plays a pivotal role in the development and progression of prostate cancers. However, the emergence of androgen resistance, largely due to inefficient anti-hormone action, limits the therapeutic usefulness of these therapies. Here, we report that 6-(3,4-dihydro-1H-isoquinolin-2-yl)-N-(6-methylpyridin-2-yl)nicotinamide (DIMN) acts as a novel anti-androgenic compound that may be effective in the treatment of both androgen-dependent and androgen-independent prostate cancers. Through AR structure-based virtual screening using the FlexX docking model, fifty-four compounds were selected and further screened for AR antagonism via cell-based tests. One compound, DIMN, showed an antagonistic effect specific to AR with comparable potency to that of the classical AR antagonists, hydroxyflutamide and bicalutamide. Consistent with their anti-androgenic activity, DIMN inhibited the growth of androgen-dependent LNCaP prostate cancer cells. Interestingly, the compound also suppressed the growth of androgen-independent C4-2 and CWR22rv prostate cancer cells, which express a functional AR, but did not suppress the growth of the AR-negative prostate cancer cells PPC-1, DU145, and R3327-AT3.1. Taken together, the results suggest that the synthetic compound DIMN is a novel anti-androgen and strong candidate for useful therapeutic agent against early stage to advanced prostate cancer.


Asunto(s)
Antagonistas de Receptores Androgénicos/química , Antagonistas de Receptores Androgénicos/farmacología , Isoquinolinas/química , Isoquinolinas/farmacología , Modelos Moleculares , Niacinamida/análogos & derivados , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/síntesis química , Animales , Células COS , Chlorocebus aethiops , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células HeLa , Humanos , Isoquinolinas/síntesis química , Masculino , Ratones , Niacinamida/síntesis química , Niacinamida/química , Niacinamida/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Relación Estructura-Actividad
3.
Biochem Biophys Res Commun ; 422(2): 327-32, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22575506

RESUMEN

Steroidogenesis in the testis is regulated by a negative feedback mechanism through the hypothalamus-pituitary-testis axis. Recent studies suggest that besides this long-loop regulation, testicular steroidogenesis is also locally regulated by androgen. However, the molecular mechanism behind this additional regulatory pathway has been poorly addressed. In the present study, we demonstrate that liganded androgen receptor (AR) suppresses the transcriptional activity of Nur77 on steroidogenic enzyme gene promoters, affecting testicular steroidogenesis. AR physically interacts and colocalizes with Nur77 in the nucleus in the presence of androgen. AR inhibits Nur77 transactivation by competing mainly with coactivators such as SRC-1 for Nur77 binding. These results suggest that androgen, through binding to AR, directly acts as a signal inhibiting the expression of steroidogenic enzyme genes in Leydig cells, eventually resulting in decreased testicular steroidogenesis. These findings strongly support the hypothesis that androgen acts locally to regulate testicular steroidogenesis, and may provide its action mechanism.


Asunto(s)
Andrógenos/metabolismo , Regulación Enzimológica de la Expresión Génica , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores Androgénicos/metabolismo , Testículo/enzimología , Testosterona/biosíntesis , Activación Transcripcional , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células Intersticiales del Testículo/enzimología , Masculino , Coactivador 1 de Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas , Testosterona/genética
4.
Biochem Biophys Res Commun ; 421(3): 532-7, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22521644

RESUMEN

Estrogen receptors (ERs), which mediate estrogen actions, regulate cell growth and differentiation of a variety of normal tissues and hormone-responsive tumors through interaction with cellular factors. In this study, we show that thyroid transcription factor-2 (TTF-2) is expressed in mammary gland and acts as ERα co-repressor. TTF-2 inhibited ERα transactivation in a dose-dependent manner in MCF-7 breast cancer cells. In addition, TTF-2 directly bound to and formed a complex with ERα, colocalizing with ERα in the nucleus. In MCF-7/TTF-2 stable cell lines, TTF-2 repressed the expression of endogenous ERα target genes such as pS2 and cyclin D1 by interrupting ERα binding to target promoters and also significantly decreased cell proliferation. Taken together, these data suggest that TTF-2 may modulate the function of ERα as a corepressor and play a role in ER-dependent proliferation of mammary cells.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Activación Transcripcional , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Femenino , Factores de Transcripción Forkhead/genética , Humanos , Inmunoprecipitación , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/metabolismo , Ratones , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley
5.
Pharmaceuticals (Basel) ; 15(11)2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36355519

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disease that results from eczema, itching, disrupted barrier function and aberrant cutaneous immune responses. The aim of the present study was to assess the efficacy of kushenol F as an effective treatment for AD via the suppression of thymic stromal lymphopoietin (TSLP) production. The results of the present study demonstrated that the clinical symptoms of AD were less severe and there was reduced ear thickening and scratching behavior in kushenol F-treated Dermatophagoides farinae extract (DFE)/1-chloro-2,4-dinitrochlorobenzene (DNCB)-induced AD mice. Histopathological analysis demonstrated that kushenol F decreased the DFE/DNCB-induced infiltration of eosinophil and mast cells and TSLP protein expression levels. Furthermore, kushenol F-treated mice exhibited significantly lower concentrations of serum histamine, IgE and IgG2a compared with the DFE/DNCB-induced control mice. Kushenol F also significantly decreased phosphorylated NF-κB and IKK levels and the mRNA expression levels of IL-1ß and IL-6 in cytokine combination-induced human keratinocytes. The results of the present study suggested that kushenol F may be a potential therapeutic candidate for the treatment of AD via reducing TSLP levels.

6.
J Biol Chem ; 285(29): 22360-9, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20472563

RESUMEN

ARR19 (androgen receptor corepressor-19 kDa), a leucine-rich protein whose expression is down-regulated by luteinizing hormone and cAMP, is differentially expressed during the development of Leydig cells and inhibits testicular steroidogenesis by reducing the expression of steroidogenic enzymes. However, the molecular events behind the suppression of testicular steroidogenesis are unknown. In the present study, we demonstrate that ARR19 inhibits the transactivation of orphan nuclear receptor Nur77, which is one of the major transcription factors that regulate the expression of steroidogenic enzyme genes in Leydig cells. ARR19 physically interacts with Nur77 and suppresses Nur77-induced promoter activity of steroidogenic enzyme genes including StAR, P450c17, and 3beta-HSD in Leydig cells. Transient transfection and chromatin immunoprecipitation assays revealed that ARR19-mediated reduced expression of steroidogenic enzyme genes was likely due to the interference of SRC-1 recruitment to Nur77 protein on the promoter of steroidogenic enzyme genes. These findings suggest that ARR19 acts as a novel coregulator of Nur77, in turn regulating Nur77-induced testicular steroidogenesis, and may play an important role in the development and function of testicular Leydig cells.


Asunto(s)
Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteínas Represoras/metabolismo , Esteroides/biosíntesis , Testículo/metabolismo , Activación Transcripcional/genética , Adenoviridae/metabolismo , Animales , Unión Competitiva , Núcleo Celular/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Proteínas con Dominio MARVEL , Masculino , Proteínas de la Membrana , Ratones , Coactivador 1 de Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Represoras/química , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Testículo/citología
7.
J Biol Chem ; 284(27): 18021-32, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19398553

RESUMEN

ARR19 (androgen receptor corepressor of 19 kDa), which encodes for a leucine-rich protein, is expressed abundantly in the testis. Further analyses revealed that ARR19 was expressed in Leydig cells, and its expression was differentially regulated during Leydig cell development. Adenovirus-mediated overexpression of ARR19 in Leydig cells inhibited testicular steroidogenesis, down-regulating the expression of steroidogenic enzymes, which suggests that ARR19 is an antisteroidogenic factor. Interestingly, cAMP/luteinizing hormone attenuated ARR19 expression in a fashion similar to that of GATA-1, which was previously reported to be down-regulated by cAMP. Sequence analysis of the Arr19 promoter revealed the presence of two putative GATA-1 binding motifs. Further analyses with 5' deletion and point mutants of putative GATA-1 binding motifs showed that these GATA-1 binding sites were critical for high promoter activity. CREB-binding protein coactivated GATA-1 and markedly increased the activity of the Arr19 promoter. Both GATA-1 and CREB-binding proteins occupied the GATA-1 motifs within the Arr19 promoter, which was repressed by cAMP treatment. Altogether, these findings demonstrate that ARR19 is the target gene of GATA-1 and suggest that ARR19 gene expression in testicular Leydig cells is regulated by luteinizing hormone/cAMP signaling via the control of GATA-1 expression, resulting in the control of testicular steroidogenesis.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Células Intersticiales del Testículo/fisiología , Proteínas Represoras/genética , Testículo/fisiología , Andrógenos/biosíntesis , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica , Hormona Luteinizante/metabolismo , Proteínas con Dominio MARVEL , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/fisiología , Proteínas Represoras/metabolismo , Testículo/citología , Testículo/embriología , Activación Transcripcional/fisiología
8.
Cancer Sci ; 101(9): 2019-25, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20608937

RESUMEN

Estrogen receptors play a key role in breast cancer development. One of the current therapeutic strategies for the treatment of estrogen receptor (ER)-α-positive breast cancers relies on the blockade of ERα transcriptional activity. In the present study, we characterized Hakai, originally characterized as an E-cadherin binding protein, as a strong blockade of ERα in breast cancer cells. We showed that Hakai inhibited the transcriptional activity of ERα by binding directly to ERα. The DNA-binding domain of ERα was found to be responsible for its interaction with Hakai. Hakai competed with ERα coactivators, such as steroid receptor coactivator-1 (SRC-1) and glucocoriticord receptor interacting protein-1 (GRIP-1), for the modulation of ERα transactivation, while its ubiquitin-ligase activity was not required. Further, overexpression of Hakai inhibited the proliferation and migration of breast cancer cells. Taken together, these results suggest that Hakai is a novel corepressor of ERα and may play a negative role in the development and progression of breast cancers.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Unión Competitiva/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células COS , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Chlorocebus aethiops , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Inmunoprecipitación , Coactivador 2 del Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/metabolismo , Coactivador 3 de Receptor Nuclear/genética , Coactivador 3 de Receptor Nuclear/metabolismo , Coactivadores de Receptor Nuclear/genética , Unión Proteica/efectos de los fármacos , Testosterona/farmacología , Activación Transcripcional/efectos de los fármacos , Transfección , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas/genética
9.
Life (Basel) ; 10(12)2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33348749

RESUMEN

High glucose-mediated tubular injury contributes to the development and progression of diabetic nephropathy through renal tubulointerstitial fibrosis. V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein, is a complement receptor. Although the role of epithelial-mesenchymal transition (EMT) has been reported in several diseases, little is known about its relationship with VSIG4 under diabetic conditions. This study aimed to investigate the role of VSIG4 in human tubule cells stimulated by high glucose (HG, 55 mM). HG upregulated both mRNA and protein levels of VSIG4 in proximal tubule cells (HK-2 cells) and Madin Darby Canine Kidney cells. These upregulations were accompanied by increased expression of mesenchymal markers such as fibronectin, N-cadherin, matrix metalloproteinase 9, and vimentin, and by decreased expression of the epithelial marker, E-cadherin. The siRNA-mediated inhibition of VSIG4 in HK-2 cells restored the dysregulation of EMT in cells. Interestingly, VSIG4 inhibition did not affect the expression of transforming growth factor (TGF)-ß, whereas inhibition of TGF-ß reduced VSIG4 expression, subsequently suppressing fibrosis markers. These findings suggest that VSIG4 plays an important role in mediating renal tubular EMT through the downstream action of HG-induced TGF-ß activation.

10.
Arch Dermatol Res ; 312(1): 59-67, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31602487

RESUMEN

Acne is a chronic skin disease of the pilosebaceous unit resulting from Propionibacterium acnes (P. acnes), a commensal microorganism. Although numerous therapies are available for acne, there is still a need for the development of effective therapies. Erythroid differentiation regulator 1 (Erdr1) has been suggested to be beneficial during inflammatory skin diseases. In the current study, we first showed that Erdr1 expression level was lower in inflammatory acne skin compared to the normal skin, suggesting that Erdr1 was negatively regulated in acne skin. To evaluate the effect of Erdr1 further, Erdr1 was injected subcutaneously in the acne mouse model. Results revealed that the necrotic lesions by inflamed acne were dramatically decreased and collagen synthesis and fibroblasts activation were induced by Erdr1. In addition, Erdr1 reduced the infiltration of inflammatory cells in vivo and accelerated collagen production in P. acnes-treated human dermal fibroblasts through TGF-ß/Smad signaling. Taken together, Erdr1 enhanced wound healing through acceleration of collagen synthesis and activation of fibroblasts in acne skin, suggesting its potential use for acne improvement.


Asunto(s)
Acné Vulgar/patología , Colágeno/biosíntesis , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/farmacología , Propionibacterium acnes , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/farmacología , Cicatrización de Heridas/fisiología , Acné Vulgar/microbiología , Animales , Células Cultivadas , Femenino , Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Pelados , Piel/metabolismo , Proteínas Supresoras de Tumor/genética
11.
Mol Cancer Res ; 6(2): 314-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18245227

RESUMEN

Sterol regulatory element-binding protein-1c (SREBP-1c) is a basic helix-loop-helix transcription factor that plays an important role in lipid homeostasis. Here, we show that SREBP-1c regulates androgen receptor (AR) transactivation through direct interaction with AR and represses androgen-dependent growth of prostatic cells. Transient transfection studies show that SREBP-1c specifically inhibits the transactivation of AR. Chromatin immunoprecipitation assays reveal that SREBP-1c is recruited with AR onto the endogenous AR target promoter. Moreover, adenovirus-mediated overexpression of SREBP-1c decreases the mRNA level of the prostate-specific antigen gene, an endogenous target gene of AR, supporting SREBP-1c modulation of AR transactivation. In vivo and in vitro protein interaction assays show that SREBP-1c directly interacts with AR through the activation function-1 domain of AR. In addition, transfection studies and glutathione S-transferase pull-down competition experiments reveal that the SREBP-1c-mediated repression of AR transactivation is accomplished through competition with certain AR coactivators for AR interaction. The SREBP-1c-mediated inhibition of AR transactivation also involves the recruitment of histone deacetylase 1. Finally, adenovirus-mediated overexpression of SREBP-1c inhibits androgen-induced proliferation of prostatic cells in vitro and in vivo, and small interfering RNA-mediated down-regulation of SREBP-1 enhances androgen-induced proliferation of prostatic cells as well as the transactivation of AR. Taken together, these results suggest that SREBP-1c acts as an AR corepressor and may play an important role in the regulation of AR-dependent prostatic cell growth.


Asunto(s)
Andrógenos/farmacología , Próstata/metabolismo , Próstata/patología , Receptores Androgénicos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Próstata/efectos de los fármacos , Próstata/enzimología , Unión Proteica/efectos de los fármacos , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Reproduction ; 137(2): 345-51, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19017724

RESUMEN

Spermatogenesis is a complex process that produces haploid motile sperms from diploid spermatogonia through dramatic morphological and biochemical changes. P-type ATPases, which support a variety of cellular processes, have been shown to play a role in the functioning of sperm. In this study, we isolated one putative androgen-regulated gene, which is the previously reported sperm-specific aminophospholipid transporter (Atp8b3, previously known as Saplt), and explored its expression pattern in murine testis and its biochemical characteristics as a P-type ATPase. Atp8b3 is exclusively expressed in the testis and its expression is developmentally regulated during testicular development. Immunohistochemistry of the testis reveals that Atp8b3 is expressed only in germ cells, especially haploid spermatids, and the protein is localized in developing acrosomes. As expected, from its primary amino acid sequence, ATP8B3 has an ATPase activity and is phosphorylated by an ATP-producing acylphosphate intermediate, which is a signature property of the P-Type ATPases. Together, ATP8B3 may play a role in acrosome development and/or in sperm function during fertilization.


Asunto(s)
Adenosina Trifosfatasas/análisis , Proteínas de Transferencia de Fosfolípidos/análisis , ATPasas de Translocación de Protón/metabolismo , Espermatogénesis/fisiología , Testículo/enzimología , Acrosoma/enzimología , Animales , Células COS , Chlorocebus aethiops , Inmunohistoquímica , Masculino , Ratones , Microscopía Fluorescente , ATPasas de Translocación de Protón/análisis , Espermátides/enzimología
13.
Mol Endocrinol ; 22(3): 623-35, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18032697

RESUMEN

The male sex steroid, testosterone (T), is synthesized from cholesterol in the testicular Leydig cell under control of the pituitary gonadotropin LH. Unlike most cells that use cholesterol primarily for membrane synthesis, steroidogenic cells have additional requirements for cholesterol, because it is the essential precursor for all steroid hormones. Little is known about how Leydig cells satisfy their specialized cholesterol requirements for steroid synthesis. We show that in mice with a unique hypomorphic androgen mutation, which disrupts the feedback loop governing T synthesis, that genes involved in cholesterol biosynthesis/uptake and steroid biosynthesis are up-regulated. We identify LH as the central regulatory molecule that controls both steroidogenesis and Leydig cell cholesterol homeostasis in vivo. In addition to the primary defect caused by high levels of LH, absence of T signaling exacerbates the lipid homeostasis defect in Leydig cells by eliminating a short feedback loop. We show that T signaling can affect the synthesis of steroids and modulates the expression of genes involved in de novo cholesterol synthesis. Surprisingly, accumulation of active sterol response element-binding protein 2 is not required for up-regulation of genes involved in cholesterol biosynthesis and uptake in Leydig cells.


Asunto(s)
Colesterol/metabolismo , Hormona Luteinizante/biosíntesis , Receptores Androgénicos/metabolismo , Testículo/metabolismo , Testosterona/biosíntesis , Animales , Northern Blotting , Células Cultivadas , Colesterol/biosíntesis , Colesterol/genética , AMP Cíclico/farmacología , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/sangre , Masculino , Ratones , Oxidorreductasas/biosíntesis , Oxidorreductasas/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Receptores de LDL/biosíntesis , Receptores de LDL/genética , Receptores Depuradores de Clase B/biosíntesis , Receptores Depuradores de Clase B/genética , Organismos Libres de Patógenos Específicos , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Testículo/citología , Testosterona/sangre , Regulación hacia Arriba
14.
J Steroid Biochem Mol Biol ; 112(1-3): 117-21, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18832037

RESUMEN

We previously demonstrated that the expression of Mullerian inhibiting substance (MIS) in Sertoli cells is downregulated by tumor necrosis factor alpha (TNF-alpha), which is secreted by meiotic germ cells, in mouse testes. Several studies have reported that MIS that is secreted by Sertoli cells inhibits steroidogenesis and, thus, the synthesis of testosterone in testicular Leydig cells. Here, we demonstrate that in TNF-alpha knockout testes, which show high levels of MIS, steroidogenesis is decreased compared to that in wild-type testes. The levels of testosterone and the mRNA levels of steroidogenesis-related genes were significantly lower after puberty in TNF-alpha knockout testes than in wild-type testes. Furthermore, the number of sperm was reduced in TNF-alpha knockout mice. Histological analysis revealed that spermatogenesis is also delayed in TNF-alpha knockout testes. In conclusion, TNF-alpha knockout mice show reduced testicular steroidogenesis, which is likely due to the high level of testicular MIS compared to that seen in wild-type mice.


Asunto(s)
Células de Sertoli/fisiología , Testículo/fisiología , Testosterona/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Animales , Hormona Antimülleriana/biosíntesis , Masculino , Ratones , Ratones Noqueados , Espermatogénesis , Testículo/citología , Factor de Necrosis Tumoral alfa/genética
15.
Mol Cell Biol ; 25(12): 4841-52, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15923603

RESUMEN

The SWI3-related gene product (SRG3), a component of the mouse SWI/SNF complex, has been suggested to have an alternative function. Here, we demonstrate that in the prostate transactivation of the androgen receptor (AR) is modulated by SRG3 in multiple ways. The expression of SRG3, which is developmentally regulated in the prostate, is induced by androgen through AR. SRG3 in turn enhances the transactivation of AR, providing a positive feedback regulatory loop. The SRG3 coactivation of AR transactivation is achieved through the recruitment of coactivator SRC-1, the protein level of which is upregulated by SRG3, providing another pathway of positive regulation. Interestingly, SRG3 coactivation of AR transactivation is fully functional in BRG1/BRM-deficient C33A cells and the AR/SRG3/SRC-1 complex formed in vivo contains neither BRG1 nor BRM protein, suggesting the possibility of an SRG3 function independent of the SWI/SNF complex. Importantly, the AR/SRG3/SRC-1 complex occupies androgen response elements on the endogenous SRG3 and PSA promoter in an androgen-dependent manner in mouse prostate and LNCaP cells, respectively, inducing gene expression. These results suggest that the multiple positive regulatory mechanisms of AR transactivation by SRG3 may be important for the rapid proliferation of prostate cells during prostate development and regeneration.


Asunto(s)
Receptores Androgénicos/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Histona Acetiltransferasas , Humanos , Masculino , Ratones , Ratones Transgénicos , Coactivador 1 de Receptor Nuclear , Coactivadores de Receptor Nuclear , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Regiones Promotoras Genéticas , Próstata/citología , Próstata/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/genética , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Factores de Transcripción p300-CBP
16.
Sci Rep ; 8(1): 17536, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30510197

RESUMEN

The DNA damage-activated protein kinase Chk1 is known to undergo auto-phosphorylation, however the sites and functional significance of this modification remain poorly understood. We have identified two novel Chk1 auto-phosphorylation sites, threonines 378 and 382 (T378/382), located in a highly conserved motif within the C-terminal Kinase Associated 1 (KA1) domain. T378/382 occur within optimal consensus Chk1 phosphorylation motifs and substitution with phospho-mimetic aspartic acid residues results in a constitutively active mutant Chk1 kinase (Chk1-DD) that arrests cell cycle progression in G2 phase of the cell cycle in the absence of DNA damage. Remarkably, the mutant Chk1-DD protein is also subject to very rapid proteasomal degradation, with a half-life approximately one tenth that of wild-type Chk1. Consistent with this, T378/T382 auto-phosphorylation also accelerates the proteasomal degradation of constitutively active Chk1 KA1 domain structural mutants. T378/382 auto-phosphorylation and accelerated degradation of wild-type Chk1 occurs at low levels during unperturbed growth, but surprisingly, is not augmented in response to genotoxic stress. Taken together, these observations demonstrate that Chk1 T378/T382 auto-phosphorylation within the KA1 domain is linked to kinase activation and rapid proteasomal degradation, and suggest a non-canonical mechanism of regulation.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Secuencias de Aminoácidos , Línea Celular Transformada , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Humanos , Mutación , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Dominios Proteicos
17.
Toxicol In Vitro ; 46: 229-236, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28947240

RESUMEN

ABT-263 (navitoclax), a Bcl-2 family protein inhibitor, was clinically tested as an anti-cancer agent. However, the clinical trials were limited given the occurrence of resistance to monotherapy in breast cancer cells. Our study investigates the mechanisms for overcoming navitoclax resistance by combining it with an mTOR inhibitor to indirectly target survivin. The apoptotic effects of navitoclax occurred in MDA-MB-231 breast cancer cells in a time- and dose-dependent fashion, but MCF-7 cells were resistant to navitoclax treatment. The expression of Bcl-2 family genes was not altered by navitoclax, but the expression of survivin, a member of the inhibitors of apoptosis proteins (IAP) family, was downregulated, which increased death signaling in MDA-MB-231 cells. In MCF-7 cells, a navitoclax-resistant cell line, combined treatment with navitoclax and everolimus synergistically reduced survivin expression and induced cell death. These data indicate that navitoclax induces cell death in MDA-MB-231 cells but not in MCF-7 cells. Decreased survivin expression in MDA-MB-231 cells may be a primary pathway for death signaling. Combined navitoclax and everolimus treatment induces cell death by reducing the stability of survivin in MCF-7 cells. Given that survivin-targeted therapy overcomes resistance to navitoclax, this strategy could be used to treat breast cancer patients.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Sulfonamidas/farmacología , Compuestos de Anilina/administración & dosificación , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Quimioterapia Combinada , Everolimus/administración & dosificación , Everolimus/farmacología , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Survivin , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
18.
Mol Endocrinol ; 20(5): 984-95, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16455820

RESUMEN

The basic leucine zipper transcription factor, CCAAT enhancer-binding protein-alpha (C/EBPalpha), negatively regulates cell proliferation and induces terminal differentiation of various cell types. C/EBPalpha is expressed in the prostate, but its potential role in the tissue is unknown. Herein, we show that C/EBPalpha is highly expressed at the stage of growth arrest during prostate development. Furthermore, overexpression of C/EBPalpha decreases the rate of DNA synthesis in LNCaP prostate cancer cells. Investigation of the potential cross-talk between C/EBPalpha and androgen receptor (AR) that is responsible for androgen-dependent prostate proliferation demonstrates that androgen-dependent transactivation of AR is strongly repressed by C/EBPalpha. C/EBPalpha directly binds AR in vitro and forms a complex with AR in vivo. C/EBPalpha neither prevents the nuclear translocation of AR nor disrupts the N/C-terminal interaction of AR, which are both necessary for its proper transactivation activity upon ligand binding. To modulate AR transactivation, however, C/EBPalpha does compete with AR coactivators for AR binding. Additionally, C/EBPalpha is recruited onto AR-target promoters with AR and is further able to inhibit the expression of endogenous prostate-specific antigen in prostate cancer cells. Our results suggest C/EBPalpha as a potent AR corepressor and provide insight into the role of C/EBPalpha in prostate development and cancer.


Asunto(s)
Antagonistas de Receptores Androgénicos , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/análisis , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Regiones Promotoras Genéticas , Próstata/química , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/química , Neoplasias de la Próstata/genética , Ratas , Receptores Androgénicos/genética , Proteínas Represoras/análisis , Activación Transcripcional
19.
Mol Endocrinol ; 19(9): 2245-57, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15919722

RESUMEN

Androgen receptor (AR) is important in male sexual differentiation and testicular function. Here, we demonstrate the regulation of AR expression and its transactivation by the basic helix-loop-helix (bHLH) transcription factor Pod-1, the expression of which in postnatal testis reciprocally coincides with the expression of AR. Pod-1 represses the promoter activity of AR, possibly through its E-box. An AR promoter region of 169 bp, which harbors one canonical E-box, is sufficient for the Pod-1-repression and bound by purified Pod-1 proteins. Pod-1 also suppresses the transactivation of AR. Transient transfection analyses of mammalian cells show that Pod-1 represses AR transactivation in a dose-dependent manner. Furthermore, yeast two-hybrid, glutathione-S-transferase-pull-down, and co-immunoprecipitation analyses reveal that Pod-1 directly associates with AR through its N-terminal region and through the DNA binding-hinge domain of AR. Interestingly, Pod-1 recruits histone deacetylase (HDAC)-1 to inhibit both promoter activity and transactivation of AR. Overexpression of HDAC1 further inhibits the Pod-1-mediated repressions and Pod-1 directly interacts with HDAC1. Furthermore, chromatin immunoprecipitation assay reveals that HDAC1 is recruited with Pod-1 to the endogenous AR promoter and the androgen-regulated Pem promoter. Taken together, these results suggest that Pod-1, which controls AR transcription and function, may play an important role in the development and function of the testis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Receptores Androgénicos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Elementos E-Box , Histona Desacetilasa 1 , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Regiones Promotoras Genéticas , Receptores Androgénicos/genética , Eliminación de Secuencia , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos
20.
Toxicol Lett ; 258: 126-133, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27339904

RESUMEN

Sulindac has anti-neoplastic properties against colorectal cancers; however, its use as a chemopreventive agent has been limited due to toxicity and efficacy concerns. Combinatorial treatment of colorectal cancers has been attempted to maximize anti-cancer efficacy with minimal side effects by administrating NSAIDs in combination with other inhibitory compounds or drugs such as l-ascorbic acid (vitamin C), which is known to exhibit cytotoxicity towards various cancer cells at high concentrations. In this study, we evaluated a combinatorial strategy utilizing sulindac and vitamin C. The death of HCT116 cells upon combination therapy occurred via a p53-mediated mechanism. The combination therapeutic resistance developed in isogenic p53 null HCT116 cells and siRNA-mediated p53 knockdown HCT116 cells, but the exogenous expression of p53 in p53 null isogenic cells resulted in the induction of cell death. In addition, we investigated an increased level of intracellular ROS (reactive oxygen species), which was preceded by p53 activation. The expression level of PUMA (p53-upregulated modulator of apoptosis), but not Bim, was significantly increased in HCT116 cells in response to the combination treatment. Taken together, our results demonstrate that combination therapy with sulindac and vitamin C could be a novel anti-cancer therapeutic strategy for p53 wild type colon cancers.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácido Ascórbico/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Especies Reactivas de Oxígeno/agonistas , Sulindac/farmacología , Proteína p53 Supresora de Tumor/agonistas , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/metabolismo , Proteínas Reguladoras de la Apoptosis/agonistas , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma/dietoterapia , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Neoplasias del Colon/dietoterapia , Neoplasias del Colon/metabolismo , Terapia Combinada , Suplementos Dietéticos , Resistencia a Antineoplásicos , Interacciones Alimento-Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Concentración Osmolar , Oxidantes/metabolismo , Proteínas Proto-Oncogénicas/agonistas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda