Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol In Vitro ; 97: 105808, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38484921

RESUMEN

The use of millimeter waves (MMW) will exponentially grow in the coming years due to their future utilization in 5G/6G networks. The question of possible biological effects at these frequencies has been raised. In this present study, we aimed to investigate gene expression changes under exposure to MMW using the Bulk RNA Barcoding and sequencing (BRB-seq) technology. To address this issue, three exposure scenarios were performed aiming at: i) comparing the cellular response of two primary culture of keratinocytes (HEK and NHEK) and one keratinocyte derivate cell line (HaCaT) exposed to MMW; ii) exploring the incident power density dose-effect on gene expression in HaCaT cell line; and, iii) studying the exposure duration at the new ICNIRP exposure limit for the general population. With the exception of heat effect induced by high power MMW (over 10 mW/cm2), those exposure scenarios have not enabled us to demonstrate important gene expression changes in the different cell populations studied. Very few differentially genes were observed between MMW exposed samples and heat shock control, and most of them were significantly associated with heat shock response that may reflect small differences in the heat generation. Together these results show that acute exposure to MMW has no effects on the transcriptional landscape of human keratinocyte models under athermal conditions.


Asunto(s)
Queratinocitos , Humanos , Queratinocitos/metabolismo , Línea Celular
2.
Mol Oncol ; 17(7): 1302-1323, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36808875

RESUMEN

Oestrogen receptor-alpha (ERα) positivity is intimately associated with the development of hormone-dependent breast cancers. A major challenge in the treatment of these cancers is to understand and overcome the mechanisms of endocrine resistance. Recently, two distinct translation programmes using specific transfer RNA (tRNA) repertoires and codon usage frequencies were evidenced during cell proliferation and differentiation. Considering the phenotype switch of cancer cells to more proliferating and less-differentiated states, we can speculate that the changes in the tRNA pool and codon usage that likely occur make the ERα coding sequence no longer adapted, impacting translational rate, co-translational folding and the resulting functional properties of the protein. To verify this hypothesis, we generated an ERα synonymous coding sequence whose codon usage was optimized to the frequencies observed in genes expressed specifically in proliferating cells and then investigated the functional properties of the encoded receptor. We demonstrate that such a codon adaptation restores ERα activities to levels observed in differentiated cells, including: (a) an enhanced contribution exerted by transactivation function 1 (AF1) in ERα transcriptional activity; (b) enhanced interactions with nuclear receptor corepressor 1 and 2 [NCoR1 and NCoR2 (also known as SMRT) respectively], promoting repressive capability; and (c) reduced interactions with SRC proto-oncogene, non-receptor tyrosine kinase (Src) and phosphoinositide 3-kinase (PI3K) p85 kinases, inhibiting MAPK and AKT signalling pathway.


Asunto(s)
Neoplasias , Receptores de Estrógenos , Receptores de Estrógenos/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Mutación Silenciosa , Línea Celular Tumoral , Codón/genética , Neoplasias/genética
3.
Cancers (Basel) ; 14(19)2022 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-36230857

RESUMEN

Estrogen receptor-alpha (ERα) is the driving transcription factor in 70% of breast cancers and its activity is associated with hormone dependent tumor cell proliferation and survival. Given the recurrence of hormone resistant relapses, understanding the etiological factors fueling resistance is of major clinical interest. Hypoxia, a frequent feature of the solid tumor microenvironment, has been described to promote endocrine resistance by triggering ERα down-regulation in both in vitro and in vivo models. Yet, the consequences of hypoxia on ERα genomic activity remain largely elusive. In the present study, transcriptomic analysis shows that hypoxia regulates a fraction of ERα target genes, underlying an important regulatory overlap between hypoxic and estrogenic signaling. This gene expression reprogramming is associated with a massive reorganization of ERα cistrome, highlighted by a massive loss of ERα binding sites. Profiling of enhancer acetylation revealed a hormone independent enhancer activation at the vicinity of genes harboring hypoxia inducible factor (HIFα) binding sites, the major transcription factors governing hypoxic adaptation. This activation counterbalances the loss of ERα and sustains hormone-independent gene expression. We describe hypoxia in luminal ERα (+) breast cancer as a key factor interfering with endocrine therapies, associated with poor clinical prognosis in breast cancer patients.

4.
Mol Cell Endocrinol ; 530: 111282, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33894309

RESUMEN

The Myocardin-related transcription factor A [MRTFA, also known as Megakaryoblastic Leukemia 1 (MKL1))] is a major actor in the epithelial to mesenchymal transition (EMT). We have previously shown that activation and nuclear accumulation of MRTFA mediate endocrine resistance of estrogen receptor alpha (ERα) positive breast cancers by initiating a partial transition from luminal to basal-like phenotype and impairing ERα cistrome and transcriptome. In the present study, we deepen our understanding of the mechanism by monitoring functional changes in the receptor's activity. We demonstrate that MRTFA nuclear accumulation down-regulates the expression of the unliganded (Apo-)ERα and causes a redistribution of the protein localization from its normal nuclear place to the entire cell volume. This phenomenon is accompanied by a shift in Apo-ERα monomer/dimer ratio towards the monomeric state, leading to significant functional consequences on ERα activities. In particular, the association of Apo-ERα with chromatin is drastically decreased, and the remaining ERα binding sites are substantially less enriched in ERE motifs than in control conditions. Monitored by proximity Ligation Assay, ERα interactions with P160 family coactivators are partly impacted when MRTFA accumulates in the nucleus, and those with SMRT and NCOR1 corepressors are abolished. Finally, ERα interactions with kinases such as c-src and PI3K are increased, thereby enhancing MAP Kinase and AKT activities. In conclusion, the activation and nuclear accumulation of MRTFA in ERα positive breast cancer cells remodels both ERα location and functions by shifting its activity from nuclear genome regulation to extra-nuclear non-genomic signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Sitios de Unión , Neoplasias de la Mama/genética , Cromatina/metabolismo , Transición Epitelial-Mesenquimal , Receptor alfa de Estrógeno/química , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Transporte de Proteínas
5.
Int J Mol Sci ; 22(1)2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33466512

RESUMEN

Approximately 80% of breast cancer (BC) cases express the estrogen receptor (ER), and 30-40% of these cases acquire resistance to endocrine therapies over time. Hyperactivation of Akt is one of the mechanisms by which endocrine resistance is acquired. Apigenin (Api), a flavone found in several plant foods, has shown beneficial effects in cancer and chronic diseases. Here, we studied the therapeutic potential of Api in the treatment of ER-positive, endocrine therapy-resistant BC. To achieve this objective, we stably overexpressed the constitutively active form of the Akt protein in MCF-7 cells (named the MCF-7/Akt clone). The proliferation of MCF-7/Akt cells is partially independent of estradiol (E2) and exhibits an incomplete response to the anti-estrogen agent 4-hydroxytamoxifen, demonstrating the resistance of these cells to hormone therapy. Api exerts an antiproliferative effect on the MCF-7/Akt clone. Api inhibits the proliferative effect of E2 by inducing G2/M phase cell cycle arrest and apoptosis. Importantly, Api inhibits the Akt/FOXM1 signaling pathway by decreasing the expression of FOXM1, a key transcription factor involved in the cell cycle. Api also alters the expression of genes regulated by FOXM1, including cell cycle-related genes, particularly in the MCF-7/Akt clone. Together, our results strengthen the therapeutic potential of Api for the treatment of endocrine-resistant BC.


Asunto(s)
Apigenina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Forkhead Box M1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/antagonistas & inhibidores , Línea Celular Tumoral , Células Endocrinas/efectos de los fármacos , Células Endocrinas/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Células MCF-7 , Transducción de Señal/efectos de los fármacos
7.
Biochim Biophys Acta Gene Regul Mech ; 1863(5): 194507, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32113984

RESUMEN

Estrogen receptor (ERα) is central in driving the development of hormone-dependent breast cancers. A major challenge in treating these cancers is to understand and overcome endocrine resistance. The Megakaryoblastic Leukemia 1 (MKL1, MRTFA) protein is a master regulator of actin dynamic and cellular motile functions, whose nuclear translocation favors epithelial-mesenchymal transition. We previously demonstrated that nuclear accumulation of MKL1 in estrogen-responsive breast cancer cell lines promotes hormonal escape. In the present study, we confirm through tissue microarray analysis that nuclear immunostaining of MKL1 is associated with endocrine resistance in a cohort of breast cancers and we decipher the underlining mechanisms using cell line models. We show through gene expression microarray analysis that the nuclear accumulation of MKL1 induces dedifferentiation leading to a mixed luminal/basal phenotype and suppresses estrogen-mediated control of gene expression. Chromatin immunoprecipitation of DNA coupled to high-throughput sequencing (ChIP-Seq) shows a profound reprogramming in ERα cistrome associated with a massive loss of ERα binding sites (ERBSs) generally associated with lower ERα-binding levels. Novel ERBSs appear to be associated with EGF and RAS signaling pathways. Collectively, these results highlight a major role of MKL1 in the loss of ERα transcriptional activity observed in certain cases of endocrine resistances, thereby contributing to breast tumor cells malignancy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular , Neoplasias de la Mama/genética , Estrógenos/metabolismo , Femenino , Humanos , Células MCF-7 , Unión Proteica
8.
Cell Commun Signal ; 15(1): 26, 2017 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-28666461

RESUMEN

BACKGROUND: Estrogen receptors (ER) α and ß are found in both women and men in many tissues, where they have different functions, including having roles in cell proliferation and differentiation of the reproductive tract. In addition to estradiol (E2), a natural hormone, numerous compounds are able to bind ERs and modulate their activities. Among these compounds, phytoestrogens such as isoflavones, which are found in plants, are promising therapeutics for several pathologies. Glyceollins are second metabolites of isoflavones that are mainly produced in soybean in response to an elicitor. They have potentially therapeutic actions in breast cancer by reducing the proliferation of cancer cells. However, the molecular mechanisms driving these effects remain elusive. METHODS: First, to determine the proliferative or anti-proliferative effects of glyceollins, in vivo and in vitro approaches were used. The length of epithelial duct in mammary gland as well as uterotrophy after treatment by E2 and glyceollins and their effect on proliferation of different breast cell line were assessed. Secondly, the ability of glyceollin to activate ER was assessed by luciferase assay. Finally, to unravel molecular mechanisms involved by glyceollins, transcriptomic analysis was performed on MCF-7 breast cancer cells. RESULTS: In this study, we show that synthetic versions of glyceollin I and II exert anti-proliferative effects in vivo in mouse mammary glands and in vitro in different ER-positive and ER-negative breast cell lines. Using transcriptomic analysis, we produce for the first time an integrated view of gene regulation in response to glyceollins and reveal that these phytochemicals act through at least two major pathways. One pathway involving FOXM1 and ERα is directly linked to proliferation. The other involves the HIF family and reveals that stress is a potential factor in the anti-proliferative effects of glyceollins due to its role in increasing the expression of REDD1, an mTORC1 inhibitor. CONCLUSION: Overall, our study clearly shows that glyceollins exert anti-proliferative effects by reducing the expression of genes encoding cell cycle and mitosis-associated factors and biomarkers overexpressed in cancers and by increasing the expression of growth arrest-related genes. These results reinforce the therapeutic potential of glyceollins for breast cancer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Fitoestrógenos/farmacología , Pterocarpanos/farmacología , Animales , Estradiol/metabolismo , Femenino , Humanos , Células MCF-7 , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Receptores de Estrógenos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Mol Cell Biol ; 34(13): 2418-36, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24752895

RESUMEN

Estradiol signaling is ideally suited for analyzing the molecular and functional linkages between the different layers of information directing transcriptional regulations: the DNA sequence, chromatin modifications, and the spatial organization of the genome. Hence, the estrogen receptor (ER) can bind at a distance from its target genes and engages timely and spatially coordinated processes to regulate their expression. In the context of the coordinated regulation of colinear genes, identifying which ER binding sites (ERBSs) regulate a given gene still remains a challenge. Here, we investigated the coordination of such regulatory events at a 2-Mb genomic locus containing the estrogen-sensitive trefoil factor (TFF) cluster of genes in breast cancer cells. We demonstrate that this locus exhibits a hormone- and cohesin-dependent reduction in the plasticity of its three-dimensional organization that allows multiple ERBSs to be dynamically brought to the vicinity of estrogen-sensitive genes. Additionally, by using triplex-forming oligonucleotides, we could precisely document the functional links between ER engagement at given ERBSs and the regulation of particular genes. Hence, our data provide evidence of a formerly suggested cooperation of enhancers toward gene regulation and also show that redundancy between ERBSs can occur.


Asunto(s)
Estrógenos/farmacología , Regulación de la Expresión Génica , Péptidos/genética , Receptores de Estrógenos/genética , Activación Transcripcional/efectos de los fármacos , Sitios de Unión/genética , Neoplasias de la Mama/genética , Factor de Unión a CCCTC , Proteínas de Ciclo Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN , Femenino , Humanos , Hibridación Fluorescente in Situ , Células MCF-7 , Reacción en Cadena de la Polimerasa Multiplex , Proteínas Nucleares/genética , Oligonucleótidos/genética , Fosfoproteínas/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Interferencia de ARN , ARN Interferente Pequeño , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Transcripción Genética/efectos de los fármacos , Factor Trefoil-2 , Cohesinas
10.
Mol Cell Endocrinol ; 390(1-2): 34-44, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24721635

RESUMEN

Estrogen receptor alpha (ERα) is generally considered to be a good prognostic marker because almost 70% of ERα-positive tumors respond to anti-hormone therapies. Unfortunately, during cancer progression, mammary tumors can escape from estrogen control, resulting in resistance to treatment. In this study, we demonstrate that activation of the actin/megakaryoblastic leukemia 1 (MKL1) signaling pathway promotes the hormonal escape of estrogen-sensitive breast cancer cell lines. The actin/MKL1 signaling pathway is silenced in differentiated ERα-positive breast cancer MCF-7 and T47D cell lines and active in ERα-negative HMT-3522 T4-2 and MDA-MB-231 breast cancer cells, which have undergone epithelial-mesenchymal transition. We showed that MKL1 activation in MCF-7 cells, either by modulating actin dynamics or using MKL1 mutants, down-regulates ERα expression and abolishes E2-dependent cell growth. Interestingly, the constitutively active form of MKL1 represses PR and HER2 expression in these cells and increases the expression of HB-EGF, TGFß, and amphiregulin growth factors in an E2-independent manner. The resulting expression profile (ER-, PR-, HER2-) typically corresponds to the triple-negative breast cancer expression profile.


Asunto(s)
Actinas/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Estradiol/fisiología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Células MCF-7 , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Tamoxifeno/farmacología , Transactivadores , Transcripción Genética
11.
Nucleic Acids Res ; 40(17): 8255-65, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22730288

RESUMEN

Enhancers are developmentally controlled transcriptional regulatory regions whose activities are modulated through histone modifications or histone variant deposition. In this study, we show by genome-wide mapping that the newly discovered deoxyribonucleic acid (DNA) modification 5-hydroxymethylcytosine (5hmC) is dynamically associated with transcription factor binding to distal regulatory sites during neural differentiation of mouse P19 cells and during adipocyte differentiation of mouse 3T3-L1 cells. Functional annotation reveals that regions gaining 5hmC are associated with genes expressed either in neural tissues when P19 cells undergo neural differentiation or in adipose tissue when 3T3-L1 cells undergo adipocyte differentiation. Furthermore, distal regions gaining 5hmC together with H3K4me2 and H3K27ac in P19 cells behave as differentiation-dependent transcriptional enhancers. Identified regions are enriched in motifs for transcription factors regulating specific cell fates such as Meis1 in P19 cells and PPARγ in 3T3-L1 cells. Accordingly, a fraction of hydroxymethylated Meis1 sites were associated with a dynamic engagement of the 5-methylcytosine hydroxylase Tet1. In addition, kinetic studies of cytosine hydroxymethylation of selected enhancers indicated that DNA hydroxymethylation is an early event of enhancer activation. Hence, acquisition of 5hmC in cell-specific distal regulatory regions may represent a major event of enhancer progression toward an active state and participate in selective activation of tissue-specific genes.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Elementos de Facilitación Genéticos , Células 3T3-L1 , 5-Metilcitosina/análogos & derivados , Animales , Sitios de Unión , Línea Celular Tumoral , Cromatina/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Neurogénesis/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo
12.
Genome Res ; 21(4): 555-65, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21233399

RESUMEN

Transcription factors (TFs) bind specifically to discrete regions of mammalian genomes called cis-regulatory elements. Among those are enhancers, which play key roles in regulation of gene expression during development and differentiation. Despite the recognized central regulatory role exerted by chromatin in control of TF functions, much remains to be learned regarding the chromatin structure of enhancers and how it is established. Here, we have analyzed on a genomic-scale enhancers that recruit FOXA1, a pioneer transcription factor that triggers transcriptional competency of these cis-regulatory sites. Importantly, we found that FOXA1 binds to genomic regions showing local DNA hypomethylation and that its cell-type-specific recruitment to chromatin is linked to differential DNA methylation levels of its binding sites. Using neural differentiation as a model, we showed that induction of FOXA1 expression and its subsequent recruitment to enhancers is associated with DNA demethylation. Concomitantly, histone H3 lysine 4 methylation is induced at these enhancers. These epigenetic changes may both stabilize FOXA1 binding and allow for subsequent recruitment of transcriptional regulatory effectors. Interestingly, when cloned into reporter constructs, FOXA1-dependent enhancers were able to recapitulate their cell type specificity. However, their activities were inhibited by DNA methylation. Hence, these enhancers are intrinsic cell-type-specific regulatory regions of which activities have to be potentiated by FOXA1 through induction of an epigenetic switch that includes notably DNA demethylation.


Asunto(s)
Elementos de Facilitación Genéticos , Epigenómica , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Animales , Sitios de Unión/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Humanos , Ratones , Modelos Genéticos , Neuronas/citología , Neuronas/metabolismo
13.
J Biol Chem ; 284(49): 33729-39, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19826002

RESUMEN

Although involved in processes leading to the emergence and development of hormone-dependent breast cancers, the estrogen receptor alpha (ERalpha) also prevents transformed cells from progressing toward a more aggressive phenotype. The transcriptional activity of ERalpha is mediated through two transactivation functions, called activation function 1 and 2, whose respective involvement varies in a cell-specific manner. Here, we identify the Rho/megakaryoblastic leukemia 1 (MKL1) signaling pathway as a main actor in controlling the cell-specific activity of both transactivation functions of ERalpha. Notably, we show that, when the coregulator MKL1 is sequestered in an inactive form by unpolymerized actin, the transcriptional activity of ERalpha mainly relies on the activation function 1. The activation of MKL1, which results from its dissociation from unpolymerized actin, promoted by the ability of Rho to support polymeric actin accumulation, silences the activation function 1 of ERalpha and allows the receptor to mainly act through its activation function 2. Importantly, this switch in the respective contribution exerted by both transactivation functions is correlated with an impaired ability of ERalpha to efficiently transactivate estrogen-regulated reporter genes. MKL1 is further shown to be present on estrogen-responsive genes in vivo. Interestingly, the Rho/MKL1 signaling pathway is activated during the epithelial-mesenchymal transition. A reduced transactivation efficiency of ERalpha, resulting from the activation of this pathway, may therefore suppress the protective role exerted by ERalpha toward tumor progression and invasiveness.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , Leucemia Megacarioblástica Aguda/metabolismo , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Unión al GTP rho/biosíntesis , Actinas/química , Línea Celular Tumoral , ADN/química , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transducción de Señal , Transactivadores , Transcripción Genética , Activación Transcripcional , Proteínas de Unión al GTP rho/metabolismo
14.
Biol Reprod ; 80(5): 889-96, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19129512

RESUMEN

The brain of teleosts is known for its strong aromatase expression, exhibiting unique features compared with other vertebrates. Among these features is the high sensitivity of aromatase B (the product of cyp19a1b) to estrogens. This effect involves the binding of estrogen receptors on an estrogen-responsive element (ERE) of the cyp19a1b promoter. Given the presence of potential androgen-responsive elements (AREs) on this promoter, in vivo and in vitro effects of androgens were studied. Using immunohistochemistry and quantitative PCR on zebrafish embryos, we found that cyp19a1b is upregulated by testosterone, an aromatizable androgen, and by 5alpha-dihydrotestosterone (DHT), a nonaromatizable androgen, suggesting a potential androgenic regulation of cyp19a1b through androgen receptors (ARs). To assess a putative direct regulation of the cyp19a1b gene by ARs, we transfected U251MG cells with zebrafish AR together with a luciferase reporter gene driven by 3000 bp of the proximal cyp19a1b promoter containing the ERE and potential AREs. Interestingly, although zebrafish AR activated luciferase reporter genes controlled by AREs, they failed to induce the cyp19a1b-luciferase construct. These data suggest that the androgenic regulation of cyp19a1b does not involve AR. We further showed that regulation of the cyp19a1b gene by testosterone is, in fact, due to aromatization, whereas the effect of DHT involves conversion into 5alpha-androstane-3beta,17beta-diol (betadiol), a metabolite of DHT with known estrogenic activity. The blockage of the androgen regulation of cyp19a1b expression using antiestrogens further confirmed the involvement of estrogen receptors in mediating these effects.


Asunto(s)
Andrógenos/farmacología , Aromatasa/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Receptores de Estrógenos/metabolismo , Proteínas de Pez Cebra/genética , Andrógenos/metabolismo , Animales , Aromatasa/metabolismo , Secuencia de Bases , Línea Celular , Dihidrotestosterona/metabolismo , Dihidrotestosterona/farmacología , Genes Reporteros , Humanos , Larva/efectos de los fármacos , Larva/metabolismo , Luciferasas/genética , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Testosterona/metabolismo , Testosterona/farmacología , Transfección , Regulación hacia Arriba/efectos de los fármacos , Proteínas de Pez Cebra/metabolismo
15.
Endocrinology ; 150(1): 200-11, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18772239

RESUMEN

A precise description of the mechanisms by which estrogen receptor-alpha (ERalpha) exerts its influences on cellular growth and differentiation is still pending. Here, we report that the differentiation of PC12 cells is profoundly affected by ERalpha. Importantly, depending upon its binding to 17beta-estradiol (17betaE2), ERalpha is found to exert different effects on pathways involved in nerve growth factor (NGF) signaling. Indeed, upon its stable expression in PC12 cells, unliganded ERalpha is able to partially inhibit the neurite outgrowth induced by NGF. This process involves a repression of MAPK and phosphatidylinositol 3-kinase/Akt signaling pathways, which leads to a negative regulation of markers of neuronal differentiation such as VGF and NFLc. This repressive action of unliganded ERalpha is mediated by its D domain and does not involve its transactivation and DNA-binding domains, thereby suggesting that direct transcriptional activity of ERalpha is not required. In contrast with this repressive action occurring in the absence of 17betaE2, the expression of ERalpha in PC12 cells allows 17betaE2 to potentiate the NGF-induced neurite outgrowth. Importantly, 17betaE2 has no impact on NGF-induced activity of MAPK and Akt signaling pathways. The mechanisms engaged by liganded ERalpha are thus unlikely to rely on an antagonism of the inhibition mediated by the unliganded ERalpha. Furthermore, 17betaE2 enhances NGF-induced response of VGF and NFLc neuronal markers in PC12 clones expressing ERalpha. This stimulatory effect of 17betaE2 requires the transactivation functions of ERalpha and its D domain, suggesting that an estrogen-responsive element-independent transcriptional mechanism is potentially relevant for the neuritogenic properties of 17betaE2 in ERalpha-expressing PC12 cells.


Asunto(s)
División Celular/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Células PC12/citología , Animales , Células Clonales/citología , Células Clonales/efectos de los fármacos , Dietilestilbestrol/farmacología , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Amplificación de Genes , Variación Genética , Ligandos , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Células PC12/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Transcripción Genética/efectos de los fármacos
16.
Diabetes ; 55(6): 1605-13, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731822

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) alpha is a transcription factor controlling lipid and glucose homeostasis. PPARalpha-deficient (-/-) mice are protected from high-fat diet-induced insulin resistance. However, the impact of PPARalpha in the pathophysiological setting of obesity-related insulin resistance is unknown. Therefore, PPARalpha(-/-) mice in an obese (ob/ob) background were generated. PPARalpha deficiency did not influence the growth curves of the obese mice but surprisingly resulted in a severe, age-dependent hyperglycemia. PPARalpha deficiency did not aggravate peripheral insulin resistance. By contrast, PPARalpha(-/-) ob/ob mice developed pancreatic beta-cell dysfunction characterized by reduced mean islet area and decreased insulin secretion in response to glucose in vitro and in vivo. In primary human pancreatic islets, PPARalpha agonist treatment prevented fatty acid-induced impairment of glucose-stimulated insulin secretion, apoptosis, and triglyceride accumulation. These results indicate that PPARalpha improves the adaptative response of the pancreatic beta-cell to pathological conditions. PPARalpha could thus represent a promising target in the prevention of type 2 diabetes.


Asunto(s)
Resistencia a la Insulina/fisiología , Islotes Pancreáticos/metabolismo , PPAR alfa/fisiología , Páncreas/metabolismo , Adulto , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Peso Corporal , Relación Dosis-Respuesta a Droga , Expresión Génica , Glucosa/farmacología , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Técnicas In Vitro , Insulina/metabolismo , Resistencia a la Insulina/genética , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Persona de Mediana Edad , PPAR alfa/genética , Palmitatos/farmacología , Páncreas/citología , Páncreas/efectos de los fármacos , Triglicéridos/metabolismo
17.
Circ Res ; 98(3): 361-9, 2006 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-16397146

RESUMEN

Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase used in the prevention of cardiovascular disease (CVD). In addition to their cholesterol-lowering activities, statins exert pleiotropic antiinflammatory effects, which might contribute to their beneficial effects not only on CVD but also on lipid-unrelated immune and inflammatory diseases, such as rheumatoid arthritis, asthma, stroke, and transplant rejection. However, the molecular mechanisms involved in these antiinflammatory properties of statins are unresolved. Here we show that the peroxisome proliferator-activated receptor (PPAR) alpha mediates antiinflammatory effects of simvastatin in vivo in models of acute inflammation. The inhibitory effects of statins on lipopolysaccharide-induced inflammatory response genes were abolished in PPARalpha-deficient macrophages and neutrophils. Moreover, simvastatin inhibited PPARalpha phosphorylation by lipopolysaccharide-activated protein kinase C (PKC) alpha. A constitutive active form of PKCalpha inhibited nuclear factor kappaB transrepression by PPARalpha whereas simvastatin enhanced transrepression activity of wild-type PPARalpha, but not of PPARalpha mutated in its PKC phosphorylation sites. These data indicate that the acute antiinflammatory effect of simvastatin occurs via PPARalpha by a mechanism involving inhibition of PKCalpha inactivation of PPARalpha transrepression activity.


Asunto(s)
Edema/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , PPAR alfa/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Transducción de Señal/fisiología , Animales , Extremidades/irrigación sanguínea , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/deficiencia , PPAR alfa/genética , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología
18.
J Clin Invest ; 115(11): 3228-38, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16239970

RESUMEN

Vascular SMC proliferation is a crucial event in occlusive cardiovascular diseases. PPARalpha is a nuclear receptor controlling lipid metabolism and inflammation, but its role in the regulation of SMC growth remains to be established. Here, we show that PPARalpha controls SMC cell-cycle progression at the G1/S transition by targeting the cyclin-dependent kinase inhibitor and tumor suppressor p16(INK4a) (p16), resulting in an inhibition of retinoblastoma protein phosphorylation. PPARalpha activates p16 gene transcription by both binding to a canonical PPAR-response element and interacting with the transcription factor Sp1 at specific proximal Sp1-binding sites of the p16 promoter. In a carotid arterial-injury mouse model, p16 deficiency results in an enhanced SMC proliferation underlying intimal hyperplasia. Moreover, PPARalpha activation inhibits SMC growth in vivo, and this effect requires p16 expression. These results identify an unexpected role for p16 in SMC cell-cycle control and demonstrate that PPARalpha inhibits SMC proliferation through p16. Thus, the PPARalpha/p16 pathway may be a potential pharmacological target for the prevention of cardiovascular occlusive complications of atherosclerosis.


Asunto(s)
Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidores de Crecimiento/fisiología , Músculo Liso Vascular/patología , PPAR alfa/fisiología , Túnica Íntima/patología , Regulación hacia Arriba , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Regulación hacia Abajo/genética , Fase G1/genética , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/genética , Células HeLa , Humanos , Hiperplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , PPAR alfa/deficiencia , PPAR alfa/genética , Fosforilación , Proteína de Retinoblastoma/metabolismo , Fase S/genética , Transcripción Genética/fisiología , Túnica Íntima/citología , Regulación hacia Arriba/genética
19.
J Biol Chem ; 280(33): 29971-9, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15899888

RESUMEN

The liver plays a central role in the control of blood glucose homeostasis by maintaining a balance between glucose production and utilization. The farnesoid X receptor (FXR) is a bile acid-activated nuclear receptor. Hepatic FXR expression is regulated by glucose and insulin. Here we identify a role for FXR in the control of hepatic carbohydrate metabolism. When submitted to a controlled fasting-refeeding schedule, FXR(-/-) mice displayed an accelerated response to high carbohydrate refeeding with an accelerated induction of glycolytic and lipogenic genes and a more pronounced repression of gluconeogenic genes. Plasma insulin and glucose levels were lower in FXR(-/-) mice upon refeeding the high-carbohydrate diet. These alterations were paralleled by decreased hepatic glycogen content. Hepatic insulin sensitivity was unchanged in FXR(-/-) mice. Treatment of isolated primary hepatocytes with a synthetic FXR agonist attenuated glucose-induced mRNA expression as well as promoter activity of L-type pyruvate kinase, acetyl-CoA carboxylase 1, and Spot14. Moreover, activated FXR interfered negatively with the carbohydrate response elements regions. These results identify a novel role for FXR as a modulator of hepatic carbohydrate metabolism.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Proteínas de Unión al ADN/fisiología , Ayuno/fisiología , Hígado/metabolismo , Factores de Transcripción/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Gluconeogénesis , Glucólisis , Hepatocitos/metabolismo , Lípidos/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Factores de Transcripción/genética
20.
Arterioscler Thromb Vasc Biol ; 25(3): 585-91, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15618549

RESUMEN

OBJECTIVE: The objective of this trial was to study the effects of fenofibrate (FF) and gemfibrozil (GF), the most commonly used fibrates, on high-density lipoprotein (HDL) and apolipoprotein (apo) A-I. METHODS AND RESULTS: In a head-to-head double-blind clinical trial, both FF and GF decreased triglycerides and increased HDL cholesterol levels to a similar extent, whereas plasma apoA-I only increased after FF but not GF. Results in human (h) apoA-Itransgenic (hA-ITg) peroxisome proliferator-activated receptor (PPAR) alpha-/- mice demonstrated that PPARalpha mediates the effects of FF and GF on HDL in vivo. Although plasma and hepatic mRNA levels of hapoA-I increased more pronouncedly after FF than GF in hA-ITgPPARalpha+/+ mice, both fibrates induced acylCoAoxidase mRNA similarly. FF and GF transactivated PPARalpha with similar activity and affinity on a DR-1 PPAR response element, but maximal activation on the hapoA-I DR-2 PPAR response element was significantly lower for GF than for FF. Moreover, GF induced recruitment of the coactivator DRIP205 on the DR-2 site less efficiently than did FF. CONCLUSIONS: Both GF and FF exert their effects on HDL through PPARalpha. Whereas FF behaves as a full agonist, GF appears to act as a partial agonist due to a differential recruitment of coactivators to the promoter. These observations provide an explanation for the differences in the activity of these fibrates on apoA-I.


Asunto(s)
Apolipoproteína A-I/sangre , Fenofibrato/administración & dosificación , Gemfibrozilo/administración & dosificación , Hiperlipidemias/tratamiento farmacológico , Hipolipemiantes/administración & dosificación , PPAR alfa/metabolismo , Adolescente , Adulto , Anciano , Animales , Apolipoproteína A-I/genética , HDL-Colesterol/sangre , Femenino , Humanos , Hiperlipidemias/metabolismo , Hígado/fisiología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , PPAR alfa/genética , ARN Mensajero/metabolismo , Especificidad de la Especie , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...