Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 13(6): e1002165, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26057340

RESUMEN

Low reproducibility rates within life science research undermine cumulative knowledge production and contribute to both delays and costs of therapeutic drug development. An analysis of past studies indicates that the cumulative (total) prevalence of irreproducible preclinical research exceeds 50%, resulting in approximately US$28,000,000,000 (US$28B)/year spent on preclinical research that is not reproducible-in the United States alone. We outline a framework for solutions and a plan for long-term improvements in reproducibility rates that will help to accelerate the discovery of life-saving therapies and cures.


Asunto(s)
Investigación Biomédica/economía , Reproducibilidad de los Resultados , Investigación Biomédica/normas
4.
Elife ; 132024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39140332

RESUMEN

Antibodies are used in many areas of biomedical and clinical research, but many of these antibodies have not been adequately characterized, which casts doubt on the results reported in many scientific papers. This problem is compounded by a lack of suitable control experiments in many studies. In this article we review the history of the 'antibody characterization crisis', and we document efforts and initiatives to address the problem, notably for antibodies that target human proteins. We also present recommendations for a range of stakeholders - researchers, universities, journals, antibody vendors and repositories, scientific societies and funders - to increase the reproducibility of studies that rely on antibodies.


Asunto(s)
Anticuerpos , Investigación Biomédica , Reproducibilidad de los Resultados , Humanos , Animales
6.
J Biol Chem ; 286(16): 14554-63, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21349840

RESUMEN

Liver X receptors (LXRs) play a critical role in regulating lipid synthesis and transport in numerous tissues. In the skin, activation of LXR induces keratinocyte differentiation and improves epidermal permeability barrier homeostasis. To elucidate the mechanism of LXR action in skin, we mapped its cistrome by identifying LXRß-RXRα binding sites using ChIP-on-chip in normal human epidermal keratinocytes (NHEKs). The cistrome was integrated with transcription data to obtain a global view of LXR action in keratinocyte biology. Here, we identify 2035 LXRß-RXRα binding sites containing 4794 LXR response elements in NHEKs and show the presence of consensus heterodimer active regions in genes involved in keratinocyte lipid transport/synthesis and terminal differentiation. Bioinformatics analysis of the cistrome revealed an enrichment of AP1 cis-regulatory motifs in the vicinity of the LXRß-RXRα binding sites. Importantly, we have demonstrated a direct interaction between LXR and Jun/Fos, indicating that the cooperation between LXR and AP1 may orchestrate keratinocyte differentiation. Finally, we corroborated these results by genome-wide mapping of the c-Fos and c-Jun cistromes in NHEKs, demonstrating that 77% of all the LXRß-RXRα binding regions show the presence of AP1 motifs at adjacent locations. Our findings provide new insight into the mechanism of LXR action in keratinocyte differentiation, lipid production and barrier formation, further strengthening the validation of LXR as a potential therapeutic target for skin disorders including skin aging, psoriasis, and atopic dermatitis.


Asunto(s)
Receptores Nucleares Huérfanos/química , Receptores X Retinoide/química , Factor de Transcripción AP-1/química , Animales , Sitios de Unión , Diferenciación Celular , Dimerización , Regulación de la Expresión Génica , Genoma , Humanos , Queratinocitos/citología , Receptores X del Hígado , Ratones , Ratones Noqueados , Transducción de Señal , Piel/metabolismo
7.
EMBO J ; 27(3): 535-45, 2008 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18219273

RESUMEN

Estrogen deficiency in menopause is a major cause of osteoporosis in women. Estrogen acts to maintain the appropriate ratio between bone-forming osteoblasts and bone-resorbing osteoclasts in part through the induction of osteoclast apoptosis. Recent studies have suggested a role for Fas ligand (FasL) in estrogen-induced osteoclast apoptosis by an autocrine mechanism involving osteoclasts alone. In contrast, we describe a paracrine mechanism in which estrogen affects osteoclast survival through the upregulation of FasL in osteoblasts (and not osteoclasts) leading to the apoptosis of pre-osteoclasts. We have characterized a cell-type-specific hormone-inducible enhancer located 86 kb downstream of the FasL gene as the target of estrogen receptor-alpha induction of FasL expression in osteoblasts. In addition, tamoxifen and raloxifene, two selective estrogen receptor modulators that have protective effects in bone, induce apoptosis in pre-osteoclasts by the same osteoblast-dependent mechanism. These results demonstrate that estrogen protects bone by inducing a paracrine signal originating in osteoblasts leading to the death of pre-osteoclasts and offer an important new target for the prevention and treatment of osteoporosis.


Asunto(s)
Huesos/metabolismo , Estrógenos/fisiología , Proteína Ligando Fas/biosíntesis , Proteína Ligando Fas/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Células 3T3 , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Técnicas de Cocultivo , Proteína Ligando Fas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Receptor fas/fisiología
8.
J Biol Chem ; 285(22): 17054-64, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20356837

RESUMEN

Selective androgen receptor modulators (SARMs) are androgen receptor (AR) ligands that induce anabolism while having reduced effects in reproductive tissues. In various experimental contexts SARMs fully activate, partially activate, or even antagonize the AR, but how these complex activities translate into tissue selectivity is not known. Here, we probed receptor function using >1000 synthetic AR ligands. These compounds produced a spectrum of activities in each assay ranging from 0 to 100% of maximal response. By testing different classes of compounds in ovariectomized rats, we established that ligands that transactivated a model promoter 40-80% of an agonist, recruited the coactivator GRIP-1 <15%, and stabilized the N-/C-terminal interdomain interaction <7% induced bone formation with reduced effects in the uterus and in sebaceous glands. Using these criteria, multiple SARMs were synthesized including MK-0773, a 4-aza-steroid that exhibited tissue selectivity in humans. Thus, AR activated to moderate levels due to reduced cofactor recruitment, and N-/C-terminal interactions produce a fully anabolic response, whereas more complete receptor activation is required for reproductive effects. This bimodal activation provides a molecular basis for the development of SARMs.


Asunto(s)
Andrógenos/metabolismo , Azaesteroides/farmacología , Antagonistas de Hormonas/farmacología , Receptores Androgénicos/química , Transcripción Genética , Animales , Azaesteroides/química , Células COS , Línea Celular Tumoral , Química Farmacéutica/métodos , Chlorocebus aethiops , Diseño de Fármacos , Femenino , Humanos , Ligandos , Masculino , Modelos Biológicos , Estructura Terciaria de Proteína , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroides/metabolismo , Activación Transcripcional
9.
Mol Pharmacol ; 78(6): 1046-58, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20837678

RESUMEN

The liver X receptors (LXRα and LXRß) are members of the nuclear receptor superfamily that function as key transcriptional regulators of a number of biological processes, including cholesterol homeostasis, lipid metabolism, and keratinocyte differentiation. Natural ligands that activate LXRs include oxysterol derivatives such as 25-hydroxycholesterol, 27-hydroxycholesterol, 22(R)-hydroxycholesterol, 20(S)-hydroxycholesterol, and 24(S),25-epoxycholesterol. Related oxysterols, such as 5α,6α-epoxycholesterol (5,6-EC) are present in a number of foods and have been shown to induce atherosclerosis in animal models. Intriguingly, these oxysterols have also been detected in atherosclerotic plaques. Using a variety of biochemical and cellular assays, we demonstrate that 5,6-EC is the first dietary modulator and an endogenous LXR ligand with cell and gene context-dependent antagonist, agonist, and inverse agonist activities. In a multiplexed LXR-cofactor peptide interaction assay, 5,6-EC induced the recruitment of a number of cofactor peptides onto both LXRα and LXRß and showed an EC(50) of approximately 2 µM in peptide recruitment. Furthermore, 5,6-EC bound to LXRα in a radiolabeled ligand displacement assay (EC(50) = 76 nM), thus demonstrating it to be one of the most potent natural LXRα ligands known to date. Analysis of endogenous gene expression in various cell-based systems indicated the potential of 5,6-EC to antagonize LXR-mediated gene expression. Furthermore, it also induced the expression of some LXR-responsive genes in keratinocytes. These results clearly demonstrate that 5,6-EC is an LXR modulator that may play a role in the development of lipid disorders, such as atherosclerosis, by antagonizing the agonistic action of endogenous LXR ligands.


Asunto(s)
Colesterol/análogos & derivados , Receptores Nucleares Huérfanos/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Colesterol/química , Colesterol/metabolismo , Colesterol/fisiología , Humanos , Receptores X del Hígado , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/deficiencia , Unión Proteica/fisiología , Transporte de Proteínas/fisiología
10.
Mol Pharmacol ; 77(5): 744-50, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20110405

RESUMEN

One of the many harmful factors faced by the skin is solar UV radiation, which damages skin by inducing chronic low-grade inflammation through increased expression of proinflammatory cytokines, metalloproteinases (MMPs) and cyclooxygenase-2 (COX-2). Estrogen receptors (ERs) alpha and beta are ligand-dependent transcription factors that are expressed in skin, and an ERbeta agonist has previously shown efficacy in vivo in models of pain and inflammation. Because ERbeta does not carry the breast and uterine proliferation liabilities of ERalpha, we decided to explore the possibility of using ERbeta as a target for photoaging. We show that ERbeta-selective compounds suppressed the expression of cytokines and MMPs in activated keratinocytes and fibroblast-based in vitro models of photoaging. Furthermore, in activated dermal fibroblasts, ERbeta-selective compounds also inhibited COX-2. These activities of ERbeta ligands in skin cells correlated with the expression levels of ERbeta and showed reversal by treatment with a potent synthetic ER antagonist. Furthermore, the pharmacology of ERbeta-selective compound was observed in wild-type but not in skin cells obtained from ERbeta knockout mice. Finally, we demonstrate that a synthetic ERbeta agonist inhibited UV-induced photodamage and skin wrinkle formation in a murine model of photoaging. Therefore, the potential of an ERbeta ligand to regulate multiple pathways underlying the cause of photoaging suggests ERbeta to be a novel therapeutic target for the prevention and treatment of photoaging.


Asunto(s)
Receptor beta de Estrógeno/fisiología , Envejecimiento/efectos de la radiación , Animales , Citocinas/genética , Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/genética , Femenino , Fibroblastos/fisiología , Humanos , Ligandos , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Pelados , Ratones Noqueados , Envejecimiento de la Piel/genética , Envejecimiento de la Piel/fisiología , Luz Solar/efectos adversos , Rayos Ultravioleta/efectos adversos
11.
Mol Cell Biol ; 27(5): 1904-13, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17194752

RESUMEN

Estrogen actions are mediated by a complex interface of direct control of gene expression (the so-called "genomic action") and by regulation of cell signaling/phosphorylation cascades, referred to as the "nongenomic," or extranuclear, action. We have previously described the identification of MNAR (modulator of nongenomic action of estrogen receptor) as a novel scaffold protein that regulates estrogen receptor alpha (ERalpha) activation of cSrc. In this study, we have investigated the role of MNAR in 17beta-estradiol (E2)-induced activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Consistent with our previous results, a direct correlation was established between MNAR expression levels and E2-induced activation of PI3 and Akt kinases. Endogenous MNAR, ERalpha, cSrc, and p85, the regulatory subunit of PI3 kinase, interacted in MCF7 cells treated with E2. The interaction between p85 and MNAR required activation of cSrc and MNAR phosphorylation on Tyr 920. Consequently, the mutation of this tyrosine to alanine (Y920A) abrogated the interaction between MNAR and p85 and the E2-induced activation of the PI3K/Akt pathway, which was required for the E2-induced protection of MCF7 cells from apoptosis. Nonetheless, the Y920A mutant potentiated the E2-induced activation of the Src/MAPK pathway and MCF7 cell proliferation, as observed with the wild-type MNAR. These results provide new and important insights into the molecular mechanisms of E2-induced regulation of cell proliferation and apoptosis.


Asunto(s)
Estrógenos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Estrógenos/metabolismo , Alanina/metabolismo , Sustitución de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoprecipitación , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transfección , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
12.
Mol Cell Biol ; 27(21): 7486-96, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17709391

RESUMEN

The orphan nuclear receptor Nurr1 is essential for the development and maintenance of midbrain dopaminergic neurons, the cells that degenerate during Parkinson's disease, by promoting the transcription of genes involved in dopaminergic neurotransmission. Since Nurr1 lacks a classical ligand-binding pocket, it is not clear which factors regulate its activity and how these factors are affected during disease pathogenesis. Since Wnt signaling via beta-catenin promotes the differentiation of Nurr1(+) dopaminergic precursors in vitro, we tested for functional interactions between these systems. We found that beta-catenin and Nurr1 functionally interact at multiple levels. In the absence of beta-catenin, Nurr1 is associated with Lef-1 in corepressor complexes. Beta-catenin binds Nurr1 and disrupts these corepressor complexes, leading to coactivator recruitment and induction of Wnt- and Nurr1-responsive genes. We then identified KCNIP4/calsenilin-like protein as being responsive to concurrent activation by Nurr1 and beta-catenin. Since KCNIP4 interacts with presenilins, the Alzheimer's disease-associated proteins that promote beta-catenin degradation, we tested the possibility that KCNIP4 induction regulates beta-catenin signaling. KCNIP4 induction limited beta-catenin activity in a presenilin-dependent manner, thereby serving as a negative feedback loop; furthermore, Nurr1 inhibition of beta-catenin activity was absent in PS1(-/-) cells or in the presence of small interfering RNAs specific to KCNIP4. These data describe regulatory convergence between Nurr1 and beta-catenin, providing a mechanism by which Nurr1 could be regulated by Wnt signaling.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Presenilina-1/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Ratas , Proteínas Represoras/metabolismo , beta Catenina/metabolismo
13.
Mol Endocrinol ; 23(1): 74-85, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19036900

RESUMEN

The use of estrogen-based therapies and the selective estrogen receptor (ER) modulator (SERM), raloxifene, which are approved for postmenopausal osteoporosis, is associated with side effects such as uterine/breast hyperproliferation, thromboembolism, and hot flashes. A combination of a new SERM, bazedoxifene (BZA), and Premarin (conjugated estrogens; CE) is under investigation to mitigate the estrogen/SERM side effects with promising results in Phase III clinical trials. To explore the mechanism of BZA/CE action, we investigated the recruitment of cofactor peptides to ERalpha by components of CE and a mixture containing the 10 major components of CE with or without three different SERMs. Here, we demonstrate differential recruitment of cofactor peptides to ERalpha by the individual CE components using a multiplex nuclear receptor-cofactor peptide interaction assay. We show that estrone and equilin are partial agonists in comparison with 17beta-estradiol in recruiting cofactor peptides to ERalpha. Further, CE was more potent than 17beta-estradiol in mediating ERalpha interaction with cofactor peptides. Interestingly, BZA was less potent than other SERMs in antagonizing the CE-mediated cofactor peptide recruitment to ERalpha. Finally, in accordance with these biochemical findings, 17beta-estradiol and CE, as well as SERM/CE combinations, showed differential gene regulation patterns in MCF-7 cells. In addition, BZA showed antagonism of a unique set of CE-regulated genes and did not down-regulate the expression of a number of CE-regulated genes, the expression of which was effectively antagonized by the other two SERMs. These results indicate that SERMs in combination with CE exhibit differential pharmacology, and therefore, combinations of other SERMs and estrogen preparations may not yield the same beneficial effects that are observed in clinic by pairing BZA with CE.


Asunto(s)
Estrógenos Conjugados (USP)/farmacología , Indoles/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/farmacología , Línea Celular , Interacciones Farmacológicas , Quimioterapia Combinada , Estradiol/administración & dosificación , Estradiol/efectos adversos , Estradiol/farmacología , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Estrógenos Conjugados (USP)/administración & dosificación , Estrógenos Conjugados (USP)/efectos adversos , Femenino , Perfilación de la Expresión Génica , Humanos , Indoles/administración & dosificación , Indoles/efectos adversos , Ligandos , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/metabolismo , Estructura Terciaria de Proteína , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Activación Transcripcional/efectos de los fármacos
14.
Mol Endocrinol ; 22(11): 2407-19, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18787039

RESUMEN

Liver X receptors (LXRalpha and -beta) are liposensors that exert their metabolic effects by orchestrating the expression of macrophage genes involved in lipid metabolism and inflammation. LXRs are also expressed in other tissues, including skin, where their natural oxysterol ligands induce keratinocyte differentiation and improve epidermal barrier function. To extend the potential use of LXR ligands to dermatological indications, we explored the possibility of using LXR as a target for skin aging. We demonstrate that LXR signaling is down-regulated in cell-based models of photoaging, i.e. UV-activated keratinocytes and TNFalpha-activated dermal fibroblasts. We show that a synthetic LXR ligand inhibits the expression of cytokines and metalloproteinases in these in vitro models, thus indicating its potential in decreasing cutaneous inflammation associated with the etiology of photoaging. Furthermore, a synthetic LXR ligand induces the expression of differentiation markers, ceramide biosynthesis enzymes, and lipid synthesis and transport genes in keratinocytes. Remarkably, LXRbeta-null mouse skin showed some of the molecular defects that are observed in chronologically aged human skin. Finally, we demonstrate that a synthetic LXR agonist inhibits UV-induced photodamage and skin wrinkle formation in a murine model of photoaging. Therefore, the ability of an LXR ligand to modulate multiple pathways underlying the etiology of skin aging suggests that LXR is a novel target for developing potential therapeutics for photoaging and chronological skin aging indications.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Envejecimiento de la Piel/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Humanos , Técnicas In Vitro , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Ligandos , Metabolismo de los Lípidos/genética , Receptores X del Hígado , Ratones , Ratones Pelados , Ratones Noqueados , Modelos Biológicos , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Envejecimiento de la Piel/patología
15.
Mol Cell Biol ; 25(2): 797-807, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632079

RESUMEN

To uncover factors required for transcription by RNA polymerase II on chromatin, we fractionated a mammalian cell nuclear extract. We identified the histone chaperone TAF-I (also known as INHAT [inhibitor of histone acetyltransferase]), which was previously proposed to repress transcription, as a potent activator of chromatin transcription responsive to the vitamin D3 receptor or to Gal4-VP16. TAF-I associates with chromatin in vitro and can substitute for the related protein NAP-1 in assembling chromatin onto cloned DNA templates in cooperation with the remodeling enzyme ATP-dependent chromatin assembly factor (ACF). The chromatin assembly and transcriptional activation functions are distinct, however, and can be dissociated temporally. Efficient transcription of chromatin assembled with TAF-I still requires the presence of TAF-I during the polymerization reaction. Conversely, TAF-I cannot stimulate transcript elongation when added after the other factors necessary for assembly of a preinitiation complex on naked DNA. Thus, TAF-I is required to facilitate transcription at a step after chromatin assembly but before transcript elongation.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación de la Expresión Génica , Chaperonas Moleculares/metabolismo , Moldes Genéticos , Factores de Transcripción/metabolismo , Transcripción Genética , Fraccionamiento Celular , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN , Genes Reporteros , Células HeLa , Chaperonas de Histonas , Humanos , Chaperonas Moleculares/genética , ARN Polimerasa II/metabolismo , Receptores de Calcitriol/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/genética
16.
Steroids ; 73(9-10): 901-5, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18261753

RESUMEN

Estrogens play a critical role in the regulation of cellular proliferation, differentiation, and apoptosis. Evidence indicates that this regulation is mediated by a complex interface of direct control of gene expression (so-called "genomic action") and by regulation of cell-signaling/phosphorylation cascades (referred to as the "non-genomic", or "extranuclear" action). However, the mechanisms of the non-genomic action of estrogens are not well defined. We have recently described the identification of a novel scaffold protein termed MNAR (modulator of non-genomic action of estrogen receptor), that couples conventional steroid receptors with extranuclear signal transduction pathways, thus potentially providing additional and tissue- or cell-specific level of steroid hormone regulation of cell functions. We have demonstrated that the MNAR is required for ER alpha (ERa) interaction with p60(src) (Src), which leads to activation of Src/MAPK pathway. Our new data also suggest that activation of cSrc in response to E2 leads to MNAR phosphorylation, interaction with p85, and activation of the PI3 and Akt kinases. These data therefore suggest that MNAR acts as an important scaffold that integrates ERa action in regulation of important signaling pathways. ERa non-genomic action has been suggested to play a key role in estrogen-induced cardio-, neuro-, and osteo-protection. Therefore, evaluation of the molecular crosstalk between MNAR and ERa may lead to development of functionally selective ER modulators that can separate between beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS and the "detrimental", proliferative effects in reproductive tissues and organs.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Transactivadores/fisiología , Familia-src Quinasas/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Proteínas Co-Represoras , Activación Enzimática , Humanos , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Transactivadores/metabolismo , Factores de Transcripción
17.
J Cell Physiol ; 213(3): 610-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17886255

RESUMEN

By regulating activities and expression levels of key signaling molecules, estrogens control mechanisms that are responsible for crucial cellular functions. Ligand binding to estrogen receptor (ER) leads to conformational changes that regulate the receptor activity, its interaction with other proteins and DNA. In the cytoplasm, receptor interactions with kinases and scaffolding molecules regulate cell signaling cascades (extranuclear/nongenomic action). In the nucleus, estrogens control a repertoire of coregulators and other auxiliary proteins that are associated with ER, which in turn determines the nature of regulated genes and level of their expression (genomic action). The combination of genomic and nongenomic actions of estrogens ultimately confers the cell-type and tissue-type selectivity. Recent studies have revealed some important new insights into the molecular mechanisms underlying ER action, which may help to explain the functional basis of existing selective ER modulators (SERMs) and provide evidence into how ER might be selectively targeted to achieve specific therapeutic goals. In this review, we will summarize some new molecular details that relate to estrogen signaling. We will also discuss some new strategies that may potentially lead to the development of functionally selective ER modulators that can separate between the beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS as well as the "detrimental," proliferative effects in reproductive tissues and organs.


Asunto(s)
Estrógenos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antagonistas de Estrógenos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Modelos Biológicos , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/fisiología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología
18.
Curr Opin Investig Drugs ; 8(10): 821-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17907058

RESUMEN

Androgens play an important role not only in male sexual differentiation, puberty, sexual behavior and spermatogenesis, but also in the maintenance of bone architecture and muscle mass and strength. For decades, steroidal androgens have been used by hypogonadal and aging men as hormone replacement therapy, and abused by prominent athletes as anabolic agents for enhancing physical performance. The use of steroidal androgens is associated with hepatotoxicity, potential for prostate stimulation, virilizing actions and other side effects resulting from their cross-reactivity to related steroid receptors. Therefore, to utilize the therapeutic potential of the androgen receptor for the treatment of indications such as osteoporosis and frailty, several pharmaceutical and biotechnology companies are developing non-steroidal tissue-selective androgen receptor modulators (SARMs) that retain the beneficial properties of natural androgens and exhibit better therapeutic indices. This article reviews the mechanism of androgen action, novel non-steroidal ligands under development and future directions of SARM research for the discovery of novel modulators for frailty and osteoporosis.


Asunto(s)
Antagonistas de Receptores Androgénicos , Andrógenos , Anciano Frágil , Osteoporosis/tratamiento farmacológico , Anciano de 80 o más Años , Animales , Femenino , Terapia de Reemplazo de Hormonas , Humanos , Masculino , Osteoporosis Posmenopáusica/tratamiento farmacológico , Congéneres de la Testosterona/farmacología , Congéneres de la Testosterona/uso terapéutico
19.
Mol Cell Biol ; 23(2): 620-8, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12509459

RESUMEN

The interferon (IFN)-induced signal transduction and transcription activation complex, ISGF3, is assembled from three proteins, STAT1, STAT2, and IRF9. Of these components, STAT2 provides a fundamental and essential transcriptional activation function for ISGF3. In the present study, we show that ISGF3-mediated transcription is dependent on STAT2 interactions with DRIP150, a subunit of the multimeric Mediator coactivator complex. Other Mediator subunits, DRIP77 and DRIP130, were found either to bind STAT2 without augmenting ISGF3 transcriptional activity or to enhance ISGF3 transcription without binding STAT2, but only DRIP150 both enhanced IFN-dependent transcription and coimmunoprecipitated with STAT2. Endogenous DRIP150 and STAT2 were able to interact in solution, and DNA affinity chromatography and chromatin immunoprecipitation assays demonstrated that DRIP150 binds to the mature, activated ISGF3-DNA complex and is recruited to target gene promoters in an IFN-dependent fashion. IFN-dependent recruitment of DRIP130 to an ISGF3 target promoter and SRB10-STAT2 coprecipitation suggest indirect association with a multisubunit Mediator complex. The site of STAT2 interaction was mapped to DRIP150 residues 188 to 566, which are necessary and sufficient for interaction with STAT2. Expression of this DRIP150 fragment, but not DRIP150 fragments outside the STAT2 interaction region, suppressed ISGF3-mediated transcriptional activity in a dominant-negative fashion, suggesting a direct functional role of this domain in mediating STAT2-DRIP150 interactions. These findings indicate that the IFN-activated ISGF3 transcription factor regulates transcription through contact with DRIP150 and implicate the Mediator coactivator complex in IFN-activated gene regulation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interferones/farmacología , Proteínas Nucleares/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética , Línea Celular , Cromatina/metabolismo , ADN/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Genes Reporteros , Células HeLa , Humanos , Immunoblotting , Factor 3 de Genes Estimulados por el Interferón , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Interferones/metabolismo , Complejo Mediador , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Subunidades de Proteína/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transactivadores/metabolismo , Activación Transcripcional , Transfección
20.
Mol Cell Biol ; 24(18): 8288-300, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15340088

RESUMEN

Requisite levels of intracellular cholesterol and fatty acids are maintained in part by the sterol regulatory element binding proteins (SREBPs). Three major SREBP isoforms exist; SREBP-1a and SREBP-1c are expressed from overlapping mRNAs, whereas SREBP-2 is encoded by a separate gene. The active forms of SREBP-1a and SREBP-1c differ only at their extreme N termini; SREBP-1c lacks 28 aa present in SREBP-1a and instead contains 4 unique aa of its own. While the SREBP-1a and -1c isoforms differentially activate transcription, the molecular basis of this difference is unknown. Here we define the differences between these proteins that confer the enhanced activity of SREBP-1a and demonstrate that this enhancement is a direct result of its avid binding to the coactivator CREB binding protein (CBP) and the mammalian mediator complex. While previous work determined that the C/H1 zinc finger and KIX domains of CBP bind to SREBP-1a, we provide evidence that the interaction with C/H1 is important for gene activation. We further show that the association between the activation domain of SREBP-1 and mediator is through aa 500 to 824 of DRIP150. Finally, we demonstrate the recruitment of mediator to an SREBP-responsive promoter in a sterol-dependent manner.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Corticosterona , Humanos , Datos de Secuencia Molecular , Mutagénesis , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Esteroles/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA