Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Nucleic Acids Res ; 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39106160

RESUMEN

The androgen receptor (AR) is pivotal in prostate cancer (PCa) progression and represents a critical therapeutic target. AR-mediated gene regulation involves intricate interactions with nuclear proteins, with many mediating and undergoing post-translational modifications that present alternative therapeutic avenues. Through chromatin proteomics in PCa cells, we identified SUMO ligases together with nuclear receptor coregulators and pioneer transcription factors within the AR's protein network. Intriguingly, this network displayed a significant association with SUMO2/3. To elucidate the influence of SUMOylation on AR chromatin interactions and subsequent gene regulation, we inhibited SUMOylation using ML-792 (SUMOi). While androgens generally facilitated the co-occupancy of SUMO2/3 and AR on chromatin, SUMOi induced divergent effects dependent on the type of AR-binding site (ARB). SUMOi augmented AR's pioneer-like binding on inaccessible chromatin regions abundant in androgen response elements (AREs) and diminished its interaction with accessible chromatin regions sparse in AREs yet rich in pioneer transcription factor motifs. The SUMOi-impacted ARBs divergently influenced AR-regulated genes; those associated with AR-mediated activation played roles in negative regulation of cell proliferation, while those with AR-mediated repression were involved in pattern formation. In conclusion, our findings underscore the pervasive influence of SUMOylation in shaping AR's role in PCa cells, potentially unveiling new therapeutic strategies.

2.
Nucleic Acids Res ; 52(2): 625-642, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38015476

RESUMEN

Treatment of prostate cancer relies predominantly on the inhibition of androgen receptor (AR) signaling. Despite the initial effectiveness of the antiandrogen therapies, the cancer often develops resistance to the AR blockade. One mechanism of the resistance is glucocorticoid receptor (GR)-mediated replacement of AR function. Nevertheless, the mechanistic ways and means how the GR-mediated antiandrogen resistance occurs have remained elusive. Here, we have discovered several crucial features of GR action in prostate cancer cells through genome-wide techniques. We detected that the replacement of AR by GR in enzalutamide-exposed prostate cancer cells occurs almost exclusively at pre-accessible chromatin sites displaying FOXA1 occupancy. Counterintuitively to the classical pioneer factor model, silencing of FOXA1 potentiated the chromatin binding and transcriptional activity of GR. This was attributed to FOXA1-mediated repression of the NR3C1 (gene encoding GR) expression via the corepressor TLE3. Moreover, the small-molecule inhibition of coactivator p300's enzymatic activity efficiently restricted GR-mediated gene regulation and cell proliferation. Overall, we identified chromatin pre-accessibility and FOXA1-mediated repression as important regulators of GR action in prostate cancer, pointing out new avenues to oppose steroid receptor-mediated antiandrogen resistance.


Asunto(s)
Cromatina , Neoplasias de la Próstata , Receptores de Glucocorticoides , Humanos , Masculino , Antagonistas de Andrógenos/farmacología , Línea Celular Tumoral , Cromatina/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
3.
Endocrinology ; 163(9)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35908178

RESUMEN

The androgen receptor (AR) plays a central role in the development and maintenance of the male phenotype. The binding of androgens to the receptor induces interactions between the carboxyterminal ligand-binding domain and the highly conserved 23FQNLF27 motif in the aminoterminal domain. The role of these so-called N/C interactions in AR functioning is debated. In vitro assays show that mutating the AR in the 23FQNLF27 motif (called ARNoC) attenuates the AR transactivation of reporter genes, has no effect on ligand binding, but does affect protein-protein interactions with several AR coregulators. To test the in vivo relevance of the N/C interaction, we analyzed the consequences of the genomic introduction of the ARNoC mutation in mice. Surprisingly, the ARNoC/Y mice show a normal male development, with unaffected male anogenital distance and normal accessory sex glands, male circulating androgen levels, body composition, and fertility. The responsiveness of androgen target genes in kidney, prostate, and testes was also unaffected. We thus conclude that the N/C interactions in the AR are not essential for the development of a male phenotype under normal physiological conditions.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Andrógenos/farmacología , Animales , Ligandos , Masculino , Ratones , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Activación Transcripcional
4.
EMBO Rep ; 23(1): e53083, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34699114

RESUMEN

Here, we investigate the impact of hypoxia on the hepatic response of glucocorticoid receptor (GR) to dexamethasone (DEX) in mice via RNA-sequencing. Hypoxia causes three types of reprogramming of GR: (i) much weaker induction of classical GR-responsive genes by DEX in hypoxia, (ii) a number of genes is induced by DEX specifically in hypoxia, and (iii) hypoxia induces a group of genes via activation of the hypothalamic-pituitary-adrenal (HPA) axis. Transcriptional profiles are reflected by changed GR DNA-binding as measured by ChIP sequencing. The HPA axis is induced by hypothalamic HIF1α and HIF2α activation and leads to GR-dependent lipolysis and ketogenesis. Acute inflammation, induced by lipopolysaccharide, is prevented by DEX in normoxia but not during hypoxia, and this is attributed to HPA axis activation by hypoxia. We unfold new physiological pathways that have consequences for patients suffering from GC resistance.


Asunto(s)
Glucocorticoides , Receptores de Glucocorticoides , Animales , Dexametasona/metabolismo , Dexametasona/farmacología , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
5.
EMBO Rep ; 22(12): e52764, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34661369

RESUMEN

Whereas dimerization of the DNA-binding domain of the androgen receptor (AR) plays an evident role in recognizing bipartite response elements, the contribution of the dimerization of the ligand-binding domain (LBD) to the correct functioning of the AR remains unclear. Here, we describe a mouse model with disrupted dimerization of the AR LBD (ARLmon/Y ). The disruptive effect of the mutation is demonstrated by the feminized phenotype, absence of male accessory sex glands, and strongly affected spermatogenesis, despite high circulating levels of testosterone. Testosterone replacement studies in orchidectomized mice demonstrate that androgen-regulated transcriptomes in ARLmon/Y mice are completely lost. The mutated AR still translocates to the nucleus and binds chromatin, but does not bind to specific AR binding sites. In vitro studies reveal that the mutation in the LBD dimer interface also affects other AR functions such as DNA binding, ligand binding, and co-regulator binding. In conclusion, LBD dimerization is crucial for the development of AR-dependent tissues through its role in transcriptional regulation in vivo. Our findings identify AR LBD dimerization as a possible target for AR inhibition.


Asunto(s)
Receptores Androgénicos , Animales , Sitios de Unión/genética , Dimerización , Ligandos , Masculino , Ratones , Receptores Androgénicos/química , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Activación Transcripcional
6.
Cell Metab ; 33(9): 1763-1776.e5, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34302744

RESUMEN

Sepsis is a potentially lethal syndrome resulting from a maladaptive response to infection. Upon infection, glucocorticoids are produced as a part of the compensatory response to tolerate sepsis. This tolerance is, however, mitigated in sepsis due to a quickly induced glucocorticoid resistance at the level of the glucocorticoid receptor. Here, we show that defects in the glucocorticoid receptor signaling pathway aggravate sepsis pathophysiology by lowering lactate clearance and sensitizing mice to lactate-induced toxicity. The latter is exerted via an uncontrolled production of vascular endothelial growth factor, resulting in vascular leakage and collapse with severe hypotension, organ damage, and death, all being typical features of a lethal form of sepsis. In conclusion, sepsis leads to glucocorticoid receptor failure and hyperlactatemia, which collectively leads to a lethal vascular collapse.


Asunto(s)
Hiperlactatemia , Sepsis , Animales , Glucocorticoides , Ácido Láctico , Ratones , Receptores de Glucocorticoides/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo , Factor A de Crecimiento Endotelial Vascular
7.
Genes Dev ; 35(15-16): 1142-1160, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34244292

RESUMEN

The establishment of cell fates involves alterations of transcription factor repertoires and repurposing of transcription factors by post-translational modifications. In embryonic stem cells (ESCs), the chromatin organizers SATB2 and SATB1 balance pluripotency and differentiation by activating and repressing pluripotency genes, respectively. Here, we show that conditional Satb2 gene inactivation weakens ESC pluripotency, and we identify SUMO2 modification of SATB2 by the E3 ligase ZFP451 as a potential driver of ESC differentiation. Mutations of two SUMO-acceptor lysines of Satb2 (Satb2K →R ) or knockout of Zfp451 impair the ability of ESCs to silence pluripotency genes and activate differentiation-associated genes in response to retinoic acid (RA) treatment. Notably, the forced expression of a SUMO2-SATB2 fusion protein in either Satb2K →R or Zfp451-/- ESCs rescues, in part, their impaired differentiation potential and enhances the down-regulation of Nanog The differentiation defect of Satb2K →R ESCs correlates with altered higher-order chromatin interactions relative to Satb2wt ESCs. Upon RA treatment of Satb2wt ESCs, SATB2 interacts with ZFP451 and the LSD1/CoREST complex and gains binding at differentiation genes, which is not observed in RA-treated Satb2K →R cells. Thus, SATB2 SUMOylation may contribute to the rewiring of transcriptional networks and the chromatin interactome of ESCs in the transition of pluripotency to differentiation.


Asunto(s)
Células Madre Embrionarias , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Oncogene ; 40(27): 4567-4579, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34127815

RESUMEN

Treatment of prostate cancer confronts resistance to androgen receptor (AR)-targeted therapies. AR-associated coregulators and chromatin proteins hold a great potential for novel therapy targets. Here, we employed a powerful chromatin-directed proteomics approach termed ChIP-SICAP to uncover the composition of chromatin protein network, the chromatome, around endogenous AR in castration resistant prostate cancer (CRPC) cells. In addition to several expected AR coregulators, the chromatome contained many nuclear proteins not previously associated with the AR. In the context of androgen signaling in CRPC cells, we further investigated the role of a known AR-associated protein, a chromatin remodeler SMARCA4 and that of SIM2, a transcription factor without a previous association with AR. To understand their role in chromatin accessibility and AR target gene expression, we integrated data from ChIP-seq, RNA-seq, ATAC-seq and functional experiments. Despite the wide co-occurrence of SMARCA4 and AR on chromatin, depletion of SMARCA4 influenced chromatin accessibility and expression of a restricted set of AR target genes, especially those involved in cell morphogenetic changes in epithelial-mesenchymal transition. The depletion also inhibited the CRPC cell growth, validating SMARCA4's functional role in CRPC cells. Although silencing of SIM2 reduced chromatin accessibility similarly, it affected the expression of a much larger group of androgen-regulated genes, including those involved in cellular responses to external stimuli and steroid hormone stimulus. The silencing also reduced proliferation of CRPC cells and tumor size in chick embryo chorioallantoic membrane assay, further emphasizing the importance of SIM2 in CRPC cells and pointing to the functional relevance of this potential prostate cancer biomarker in CRPC cells. Overall, the chromatome of AR identified in this work is an important resource for the field focusing on this important drug target.


Asunto(s)
Receptores Androgénicos , Animales , Embrión de Pollo , Masculino , Próstata , Proteómica
9.
Endocr Relat Cancer ; 28(9): R231-R250, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34137734

RESUMEN

Steroid receptors (SRs) constitute an important class of signal-dependent transcription factors (TFs). They regulate a variety of key biological processes and are crucial drug targets in many disease states. In particular, estrogen (ER) and androgen receptors (AR) drive the development and progression of breast and prostate cancer, respectively. Thus, they represent the main specific drug targets in these diseases. Recent evidence has suggested that the crosstalk between signal-dependent TFs is an important step in the reprogramming of chromatin sites; a signal-activated TF can expand or restrict the chromatin binding of another TF. This crosstalk can rewire gene programs and thus alter biological processes and influence the progression of disease. Lately, it has been postulated that there may be an important crosstalk between the AR and the ER with other SRs. Especially, progesterone (PR) and glucocorticoid receptor (GR) can reprogram chromatin binding of ER and gene programs in breast cancer cells. Furthermore, GR can take the place of AR in antiandrogen-resistant prostate cancer cells. Here, we review the current knowledge of the crosstalk between SRs in breast and prostate cancers. We emphasize how the activity of ER and AR on chromatin can be modulated by other SRs on a genome-wide scale. We also highlight the knowledge gaps in the interplay of SRs and their complex interactions with other signaling pathways and suggest how to experimentally fill in these gaps.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Próstata , Receptores de Esteroides , Neoplasias de la Mama/genética , Cromatina/genética , Femenino , Humanos , Masculino , Progesterona , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Receptores de Glucocorticoides/genética , Receptores de Progesterona/genética , Receptores de Esteroides/genética
10.
J Steroid Biochem Mol Biol ; 210: 105873, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33722704

RESUMEN

Glucocorticoid (GC) receptor (GR) is a key transcription factor (TF) that regulates vital metabolic and anti-inflammatory processes. We have identified BCL6 corepressor (BCOR) as a dexamethasone-stimulated interaction partner of GR. BCOR is a component of non-canonical polycomb repressor complex 1.1 (ncPCR1.1) and linked to different developmental disorders and cancers, but the role of BCOR in GC signaling is poorly characterized. Here, using ChIP-seq we show that, GC induces genome-wide redistribution of BCOR chromatin binding towards GR-occupied enhancers in HEK293 cells. As assessed by RNA-seq, depletion of BCOR altered the expression of hundreds of GC-regulated genes, especially the ones linked to TNF signaling, GR signaling and cell migration pathways. Biotinylation-based proximity mapping revealed that GR and BCOR share several interacting partners, including nuclear receptor corepressor NCOR1. ChIP-seq showed that the NCOR1 co-occurs with both BCOR and GR on a subset of enhancers upon GC treatment. Simultaneous depletion of BCOR and NCOR1 influenced GR target gene expression in a combinatorial and gene-specific manner. Finally, we show using live cell imaging that the depletion of BCOR together with NCOR1 markedly enhances cell migration. Collectively, our data suggest BCOR as an important gene and pathway selective coregulator of GR transcriptional activity.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Receptores de Glucocorticoides/genética , Proteínas Represoras/metabolismo , Sitios de Unión , Movimiento Celular/genética , Proliferación Celular/genética , Inmunoprecipitación de Cromatina , Dexametasona/farmacología , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas/genética , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/genética
11.
Nucleic Acids Res ; 49(4): 1951-1971, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33524141

RESUMEN

Glucocorticoid receptor (GR) is an essential transcription factor (TF), controlling metabolism, development and immune responses. SUMOylation regulates chromatin occupancy and target gene expression of GR in a locus-selective manner, but the mechanism of regulation has remained elusive. Here, we identify the protein network around chromatin-bound GR by using selective isolation of chromatin-associated proteins and show that the network is affected by receptor SUMOylation, with several nuclear receptor coregulators and chromatin modifiers preferring interaction with SUMOylation-deficient GR and proteins implicated in transcriptional repression preferring interaction with SUMOylation-competent GR. This difference is reflected in our chromatin binding, chromatin accessibility and gene expression data, showing that the SUMOylation-deficient GR is more potent in binding and opening chromatin at glucocorticoid-regulated enhancers and inducing expression of target loci. Blockage of SUMOylation by a SUMO-activating enzyme inhibitor (ML-792) phenocopied to a large extent the consequences of GR SUMOylation deficiency on chromatin binding and target gene expression. Our results thus show that SUMOylation modulates the specificity of GR by regulating its chromatin protein network and accessibility at GR-bound enhancers. We speculate that many other SUMOylated TFs utilize a similar regulatory mechanism.


Asunto(s)
Cromatina/metabolismo , Receptores de Glucocorticoides/metabolismo , Sumoilación , Sitios de Unión , Regulación de la Expresión Génica , Células HEK293 , Humanos , Co-Represor 1 de Receptor Nuclear/metabolismo , Coactivador 1 de Receptor Nuclear , Mapeo de Interacción de Proteínas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación/efectos de los fármacos
12.
Mol Cell ; 81(5): 1013-1026.e11, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33548202

RESUMEN

In response to stress, human cells coordinately downregulate transcription and translation of housekeeping genes. To downregulate transcription, the negative elongation factor (NELF) is recruited to gene promoters impairing RNA polymerase II elongation. Here we report that NELF rapidly forms nuclear condensates upon stress in human cells. Condensate formation requires NELF dephosphorylation and SUMOylation induced by stress. The intrinsically disordered region (IDR) in NELFA is necessary for nuclear NELF condensation and can be functionally replaced by the IDR of FUS or EWSR1 protein. We find that biomolecular condensation facilitates enhanced recruitment of NELF to promoters upon stress to drive transcriptional downregulation. Importantly, NELF condensation is required for cellular viability under stressful conditions. We propose that stress-induced NELF condensates reported here are nuclear counterparts of cytosolic stress granules. These two stress-inducible condensates may drive the coordinated downregulation of transcription and translation, likely forming a critical node of the stress survival strategy.


Asunto(s)
Respuesta al Choque Térmico/genética , Proteínas Intrínsecamente Desordenadas/genética , Procesamiento Proteico-Postraduccional , ARN Polimerasa II/genética , Transcripción Genética , Factores de Elongación Transcripcional/genética , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Cromatina/química , Cromatina/metabolismo , Células Clonales , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Genes Reporteros , Células HEK293 , Células HeLa , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Fosforilación , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Transducción de Señal , Estrés Fisiológico , Sumoilación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/metabolismo , Proteína Fluorescente Roja
13.
Maturitas ; 141: 9-19, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33036707

RESUMEN

Ovarian deficiency, including premature ovarian insufficiency (POI) and diminished ovarian reserve (DOR), represents one of the main causes of female infertility. POI is a genetically heterogeneous condition but current understanding of its genetic basis is far from complete, with the cause remaining unknown in the majority of patients. The genes that regulate DOR have been reported but the genetic basis of DOR has not been explored in depth. Both conditions are likely to lie along a continuum of degrees of decrease in ovarian reserve. We performed genomic analysis via whole exome sequencing (WES) followed by in silico analyses and functional experiments to investigate the genetic cause of ovarian deficiency in ten affected women. We achieved diagnoses for three of them, including the identification of novel variants in STAG3, GDF9, and FANCM. We identified potentially causative FSHR variants in another patient. This is the second report of biallelic GDF9 and FANCM variants, and, combined with functional support, validates these genes as bone fide autosomal recessive "POI genes". We also identified new candidate genes, NRIP1, XPO1, and MACF1. These genes have been linked to ovarian function in mouse, pig, and zebrafish respectively, but never in humans. In the case of NRIP1, we provide functional support for the deleterious nature of the variant via SUMOylation and luciferase/ß-galactosidase reporter assays. Our study provides multiple insights into the genetic basis of POI/DOR. We have further elucidated the involvement of GDF9, FANCM, STAG3 and FSHR in POI pathogenesis, and propose new candidate genes, NRIP1, XPO1, and MACF1, which should be the focus of future studies.


Asunto(s)
Carioferinas/genética , Proteínas de Microfilamentos/genética , Proteína de Interacción con Receptores Nucleares 1/genética , Reserva Ovárica/genética , Insuficiencia Ovárica Primaria/genética , Receptores Citoplasmáticos y Nucleares/genética , Adolescente , Proteínas de Ciclo Celular/genética , ADN Helicasas/genética , Femenino , Genómica , Factor 9 de Diferenciación de Crecimiento/genética , Humanos , Infertilidad Femenina , Menopausia Prematura/genética , Enfermedades del Ovario , Secuenciación del Exoma , Adulto Joven , Proteína Exportina 1
14.
Oncogene ; 39(11): 2391-2407, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31925334

RESUMEN

We have identified BCL6 corepressor (BCOR) as a hormone-dependent interaction partner of androgen receptor (AR), a key transcription factor in the development of normal and cancerous prostate. BCOR is often mutated in cancers and hematological diseases and as a component of a non-canonical polycomb repressive complex 1 (ncPRC1.1) required for arranging many facets of cellular differentiation. However, its role in androgen signaling or prostate cancer cells remains unknown. Here, our genome-wide analyses reveal that BCOR is recruited in an androgen-dependent fashion to majority of AR-binding chromatin sites in castration-resistant prostate cancer (CRPC) cells. Interestingly, depletion of BCOR has a significant effect on the expression of androgen-repressed genes linked to regulation of cell proliferation, differentiation and development. At many of these genes, such as HOX genes, the depletion leads to a decrease in H2A K119 monoubiquitination and an increase in mRNA expression. Consistently, BCOR depletion impairs the proliferation and viability of CRPC cells, inducing their apoptosis. Collectively, our data indicate a key role for the BCOR-ncPRC1.1 complex in the corepression of an important subset of AR target genes and the regulation of prostate cancer cell proliferation.


Asunto(s)
Histonas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Andrógenos/farmacología , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/fisiología , Cromatina/genética , Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Histonas/genética , Humanos , Masculino , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/genética , Receptores Androgénicos/genética , Proteínas Represoras/genética , Ubiquitinación
15.
Cell Mol Life Sci ; 77(18): 3627-3642, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31768607

RESUMEN

Hypoxia-inducible factor (HIF), an αß dimer, is the master regulator of oxygen homeostasis with hundreds of hypoxia-inducible target genes. Three HIF isoforms differing in the oxygen-sensitive α subunit exist in vertebrates. While HIF-1 and HIF-2 are known transcription activators, HIF-3 has been considered a negative regulator of the hypoxia response pathway. However, the human HIF3A mRNA is subject to complex alternative splicing. It was recently shown that the long HIF-3α variants can form αß dimers that possess transactivation capacity. Here, we show that overexpression of the long HIF-3α2 variant induces the expression of a subset of genes, including the erythropoietin (EPO) gene, while simultaneous downregulation of all HIF-3α variants by siRNA targeting a shared HIF3A region leads to downregulation of EPO and additional genes. EPO mRNA and protein levels correlated with HIF3A silencing and HIF-3α2 overexpression. Chromatin immunoprecipitation analyses showed that HIF-3α2 binding associated with canonical hypoxia response elements in the promoter regions of EPO. Luciferase reporter assays showed that the identified HIF-3α2 chromatin-binding regions were sufficient to promote transcription by all three HIF-α isoforms. Based on these data, HIF-3α2 is a transcription activator that directly regulates EPO expression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Eritropoyetina/metabolismo , Proteínas Represoras/metabolismo , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Cromatina/metabolismo , Dimerización , Eritropoyetina/análisis , Eritropoyetina/genética , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , Empalme del ARN , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo , Activación Transcripcional
16.
Proc Natl Acad Sci U S A ; 116(26): 12942-12951, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31182584

RESUMEN

Glucocorticoid resistance (GCR) is defined as an unresponsiveness to the therapeutic effects, including the antiinflammatory ones of glucocorticoids (GCs) and their receptor, the glucocorticoid receptor (GR). It is a problem in the management of inflammatory diseases and can be congenital as well as acquired. The strong proinflammatory cytokine TNF-alpha (TNF) induces an acute form of GCR, not only in mice, but also in several cell lines: e.g., in the hepatoma cell line BWTG3, as evidenced by impaired Dexamethasone (Dex)-stimulated direct GR-dependent gene up- and down-regulation. We report that TNF has a significant and broad impact on this transcriptional performance of GR, but no impact on nuclear translocation, dimerization, or DNA binding capacity of GR. Proteome-wide proximity-mapping (BioID), however, revealed that the GR interactome was strongly modulated by TNF. One GR cofactor that interacted significantly less with the receptor under GCR conditions is p300. NFκB activation and p300 knockdown both reduced direct transcriptional output of GR whereas p300 overexpression and NFκB inhibition reverted TNF-induced GCR, which is in support of a cofactor reshuffle model. This hypothesis was supported by FRET studies. This mechanism of GCR opens avenues for therapeutic interventions in GCR diseases.


Asunto(s)
Resistencia a Medicamentos/genética , Proteína p300 Asociada a E1A/metabolismo , Glucocorticoides/farmacología , Inflamación/tratamiento farmacológico , Receptores de Glucocorticoides/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células A549 , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Dexametasona/farmacología , Dexametasona/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Proteína p300 Asociada a E1A/genética , Femenino , Técnicas de Silenciamiento del Gen , Glucocorticoides/uso terapéutico , Células HEK293 , Humanos , Inflamación/inmunología , Ratones , FN-kappa B/metabolismo , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/inmunología , ARN Interferente Pequeño/metabolismo , RNA-Seq , Receptores de Glucocorticoides/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
17.
J Steroid Biochem Mol Biol ; 192: 105382, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31145973

RESUMEN

IRF2BP2 (interferon regulatory factor-2 binding protein-2) is an uncharacterized interaction partner of glucocorticoid (GC) receptor (GR), an anti-inflammatory and metabolic transcription factor. Here, we show that GC changes the chromatin binding of IRF2BP2 in natural chromatin milieu. The GC-induced IRF2BP2-binding sites co-occur with GR binding sites and are associated with GC-induced genes. Moreover, the depletion of IRF2BP2 modulates transcription of GC-regulated genes, represses cell proliferation and increases cell movement in HEK293 cells. In A549 cells, the depletion extensively alters the responses to GC and tumor necrosis factor α (TNF), including metabolic and inflammatory pathways. Taken together, our data support the role of IRF2BP2 as a coregulator of both GR and NF-κB, potentially modulating the crosstalk between GC and TNF signaling.


Asunto(s)
Antiinflamatorios/farmacología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Inflamación/prevención & control , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Movimiento Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Factores de Transcripción/genética
18.
Nat Commun ; 10(1): 777, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30770815

RESUMEN

Androgen receptor (AR) is regulated by SUMOylation at its transactivation domain. In vitro, the SUMOylation is linked to transcriptional repression and/or target gene-selective regulation. Here, we generated a mouse model (ArKI) in which the conserved SUMO acceptor lysines of AR are permanently abolished (ArK381R, K500R). ArKI males develop normally, without apparent defects in their systemic androgen action in reproductive tissues. However, the ArKI males are infertile. Their spermatogenesis appears unaffected, but their epididymal sperm maturation is defective, shown by severely compromised motility and fertilization capacity of the sperm. Fittingly, their epididymal AR chromatin-binding and gene expression associated with sperm maturation and function are misregulated. AR is SUMOylated in the wild-type epididymis but not in the testis, which could explain the tissue-specific response to the lack of AR SUMOylation. Our studies thus indicate that epididymal AR SUMOylation is essential for the post-testicular sperm maturation and normal reproductive capability of male mice.


Asunto(s)
Epidídimo/metabolismo , Epidídimo/fisiopatología , Infertilidad Masculina/metabolismo , Infertilidad Masculina/fisiopatología , Receptores Androgénicos/metabolismo , Espermatogénesis/fisiología , Animales , Epidídimo/patología , Humanos , Infertilidad Masculina/patología , Masculino , Ratones , Receptores Androgénicos/genética , Espermatogénesis/genética , Sumoilación/genética , Sumoilación/fisiología
19.
Mol Cell Endocrinol ; 479: 117-122, 2019 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-30261210

RESUMEN

The crucial effects of androgens on the male skeleton are at least partly mediated via the androgen receptor (AR). In addition to hormone binding, the AR activity is regulated by post-translational modifications, including SUMOylation. SUMOylation is a reversible modification in which Small Ubiquitin-related MOdifier proteins (SUMOs) are attached to the AR and thereby regulate the activity of the AR and change its interactions with other proteins. To elucidate the importance of SUMOylation of AR for male bone metabolism, we used a mouse model devoid of the two AR SUMOylation sites (ARSUM-; K381R and K500R are substituted). Six-month-old male ARSUM- mice displayed significantly reduced trabecular bone volume fraction in the distal metaphyseal region of femur compared with wild type (WT) mice (BV/TV, -19.1 ±â€¯4.9%, P < 0.05). The number of osteoblasts per bone perimeter was substantially reduced (-60.5 ±â€¯7.2%, P < 0.001) while no significant effect was observed on the number of osteoclasts in the trabecular bone of male ARSUM- mice. Dynamic histomorphometric analysis of trabecular bone revealed a reduced bone formation rate (-32.6 ±â€¯7.4%, P < 0.05) as a result of reduced mineralizing surface per bone surface in ARSUM- mice compared with WT mice (-24.3 ±â€¯3.6%, P < 0.001). Furthermore, cortical bone thickness in the diaphyseal region of femur was reduced in male ARSUM- mice compared with WT mice (-7.3 ±â€¯2.0%, P < 0.05). In conclusion, mice devoid of AR SUMOylation have reduced trabecular bone mass as a result of reduced bone formation. We propose that therapies enhancing AR SUMOylation might result in bone-specific anabolic effects with minimal adverse effects in other tissues.


Asunto(s)
Huesos/anatomía & histología , Huesos/metabolismo , Receptores Androgénicos/metabolismo , Sumoilación , Animales , Huesos/diagnóstico por imagen , Hueso Esponjoso/anatomía & histología , Hueso Cortical/anatomía & histología , Masculino , Ratones , Modelos Animales , Tamaño de los Órganos , Osteogénesis , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...