Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.115
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(13): 2748-2764.e22, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37267948

RESUMEN

Ferroptosis, a cell death process driven by iron-dependent phospholipid peroxidation, has been implicated in various diseases. There are two major surveillance mechanisms to suppress ferroptosis: one mediated by glutathione peroxidase 4 (GPX4) that catalyzes the reduction of phospholipid peroxides and the other mediated by enzymes, such as FSP1, that produce metabolites with free radical-trapping antioxidant activity. In this study, through a whole-genome CRISPR activation screen, followed by mechanistic investigation, we identified phospholipid-modifying enzymes MBOAT1 and MBOAT2 as ferroptosis suppressors. MBOAT1/2 inhibit ferroptosis by remodeling the cellular phospholipid profile, and strikingly, their ferroptosis surveillance function is independent of GPX4 or FSP1. MBOAT1 and MBOAT2 are transcriptionally upregulated by sex hormone receptors, i.e., estrogen receptor (ER) and androgen receptor (AR), respectively. A combination of ER or AR antagonist with ferroptosis induction significantly inhibited the growth of ER+ breast cancer and AR+ prostate cancer, even when tumors were resistant to single-agent hormonal therapies.


Asunto(s)
Ferroptosis , Masculino , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Peroxidación de Lípido , Peróxidos , Fosfolípidos
2.
Cell ; 176(1-2): 213-226.e18, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30554876

RESUMEN

Transcriptional regulation in metazoans occurs through long-range genomic contacts between enhancers and promoters, and most genes are transcribed in episodic "bursts" of RNA synthesis. To understand the relationship between these two phenomena and the dynamic regulation of genes in response to upstream signals, we describe the use of live-cell RNA imaging coupled with Hi-C measurements and dissect the endogenous regulation of the estrogen-responsive TFF1 gene. Although TFF1 is highly induced, we observe short active periods and variable inactive periods ranging from minutes to days. The heterogeneity in inactive times gives rise to the widely observed "noise" in human gene expression and explains the distribution of protein levels in human tissue. We derive a mathematical model of regulation that relates transcription, chromosome structure, and the cell's ability to sense changes in estrogen and predicts that hypervariability is largely dynamic and does not reflect a stable biological state.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Transcripción Genética/fisiología , Receptor alfa de Estrógeno/metabolismo , Estrógenos , Expresión Génica/genética , Humanos , Modelos Teóricos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Análisis de la Célula Individual/métodos , Transcripción Genética/genética , Activación Transcripcional/fisiología , Factor Trefoil-1/genética
3.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31353221

RESUMEN

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antagonistas del Receptor de Estrógeno/farmacología , Fulvestrant/farmacología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Cinamatos/farmacología , Resistencia a Antineoplásicos , Antagonistas del Receptor de Estrógeno/uso terapéutico , Femenino , Fulvestrant/uso terapéutico , Células HEK293 , Xenoinjertos , Humanos , Indazoles/farmacología , Ligandos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Polimorfismo de Nucleótido Simple , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
4.
Cell ; 179(6): 1393-1408.e16, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31735496

RESUMEN

Behaviors are inextricably linked to internal state. We have identified a neural mechanism that links female sexual behavior with the estrus, the ovulatory phase of the estrous cycle. We find that progesterone-receptor (PR)-expressing neurons in the ventromedial hypothalamus (VMH) are active and required during this behavior. Activating these neurons, however, does not elicit sexual behavior in non-estrus females. We show that projections of PR+ VMH neurons to the anteroventral periventricular (AVPV) nucleus change across the 5-day mouse estrous cycle, with ∼3-fold more termini and functional connections during estrus. This cyclic increase in connectivity is found in adult females, but not males, and regulated by estrogen signaling in PR+ VMH neurons. We further show that these connections are essential for sexual behavior in receptive females. Thus, estrogen-regulated structural plasticity of behaviorally salient connections in the adult female brain links sexual behavior to the estrus phase of the estrous cycle.


Asunto(s)
Red Nerviosa/fisiología , Conducta Sexual Animal/fisiología , Animales , Estrógenos/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Hormonas Esteroides Gonadales/farmacología , Hipotálamo Anterior/fisiología , Masculino , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovario/metabolismo , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Receptores de Progesterona/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
5.
Cell ; 179(3): 713-728.e17, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31626771

RESUMEN

The ventrolateral subdivision of the ventromedial hypothalamus (VMHvl) contains ∼4,000 neurons that project to multiple targets and control innate social behaviors including aggression and mounting. However, the number of cell types in VMHvl and their relationship to connectivity and behavioral function are unknown. We performed single-cell RNA sequencing using two independent platforms-SMART-seq (∼4,500 neurons) and 10x (∼78,000 neurons)-and investigated correspondence between transcriptomic identity and axonal projections or behavioral activation, respectively. Canonical correlation analysis (CCA) identified 17 transcriptomic types (T-types), including several sexually dimorphic clusters, the majority of which were validated by seqFISH. Immediate early gene analysis identified T-types exhibiting preferential responses to intruder males versus females but only rare examples of behavior-specific activation. Unexpectedly, many VMHvl T-types comprise a mixed population of neurons with different projection target preferences. Overall our analysis revealed that, surprisingly, few VMHvl T-types exhibit a clear correspondence with behavior-specific activation and connectivity.


Asunto(s)
Hipotálamo/citología , Neuronas/clasificación , Conducta Social , Animales , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Hipotálamo/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Conducta Sexual Animal , Análisis de la Célula Individual , Transcriptoma
6.
Cell ; 173(1): 260-274.e25, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29551266

RESUMEN

Protein degradation plays important roles in biological processes and is tightly regulated. Further, targeted proteolysis is an emerging research tool and therapeutic strategy. However, proteome-wide technologies to investigate the causes and consequences of protein degradation in biological systems are lacking. We developed "multiplexed proteome dynamics profiling" (mPDP), a mass-spectrometry-based approach combining dynamic-SILAC labeling with isobaric mass tagging for multiplexed analysis of protein degradation and synthesis. In three proof-of-concept studies, we uncover different responses induced by the bromodomain inhibitor JQ1 versus a JQ1 proteolysis targeting chimera; we elucidate distinct modes of action of estrogen receptor modulators; and we comprehensively classify HSP90 clients based on their requirement for HSP90 constitutively or during synthesis, demonstrating that constitutive HSP90 clients have lower thermal stability than non-clients, have higher affinity for the chaperone, vary between cell types, and change upon external stimuli. These findings highlight the potential of mPDP to identify dynamically controlled degradation mechanisms in cellular systems.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Proteoma/análisis , Proteómica/métodos , Azepinas/química , Azepinas/metabolismo , Azepinas/farmacología , Línea Celular , Cromatografía Líquida de Alta Presión , Análisis por Conglomerados , Estradiol/farmacología , Humanos , Marcaje Isotópico , Células Jurkat , Células MCF-7 , Proteínas de Neoplasias/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Espectrometría de Masas en Tándem , Triazoles/química , Triazoles/metabolismo , Triazoles/farmacología
7.
Genes Dev ; 37(21-24): 998-1016, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38092521

RESUMEN

Reductions in brain kynurenic acid levels, a neuroinhibitory metabolite, improve cognitive function in diverse organisms. Thus, modulation of kynurenic acid levels is thought to have therapeutic potential in a range of brain disorders. Here we report that the steroid 5-androstene 3ß, 17ß-diol (ADIOL) reduces kynurenic acid levels and promotes associative learning in Caenorhabditis elegans We identify the molecular mechanisms through which ADIOL links peripheral metabolic pathways to neural mechanisms of learning capacity. Moreover, we show that in aged animals, which normally experience rapid cognitive decline, ADIOL improves learning capacity. The molecular mechanisms that underlie the biosynthesis of ADIOL as well as those through which it promotes kynurenic acid reduction are conserved in mammals. Thus, rather than a minor intermediate in the production of sex steroids, ADIOL is an endogenous hormone that potently regulates learning capacity by causing reductions in neural kynurenic acid levels.


Asunto(s)
Ácido Quinurénico , Esteroides , Animales , Ácido Quinurénico/farmacología , Hormonas , Mamíferos
8.
Mol Cell ; 81(6): 1160-1169.e5, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33503406

RESUMEN

Voltage-gated sodium channels are targets for many analgesic and antiepileptic drugs whose therapeutic mechanisms and binding sites have been well characterized. We describe the identification of a previously unidentified receptor site within the NavMs voltage-gated sodium channel. Tamoxifen, an estrogen receptor modulator, and its primary and secondary metabolic products bind at the intracellular exit of the channel, which is a site that is distinct from other previously characterized sodium channel drug sites. These compounds inhibit NavMs and human sodium channels with similar potencies and prevent sodium conductance by delaying channel recovery from the inactivated state. This study therefore not only describes the structure and pharmacology of a site that could be leveraged for the development of new drugs for the treatment of sodium channelopathies but may also have important implications for off-target health effects of this widely used therapeutic drug.


Asunto(s)
Modelos Moleculares , Tamoxifeno/química , Canales de Sodio Activados por Voltaje/química , Células HEK293 , Humanos
9.
EMBO J ; 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39284910

RESUMEN

Transcription factors (TFs) regulate gene expression by binding with varying strengths to DNA via their DNA-binding domain. Additionally, some TFs also interact with RNA, which modulates transcription factor binding to chromatin. However, whether RNA-mediated TF binding results in differential transcriptional outcomes remains unknown. In this study, we demonstrate that estrogen receptor α (ERα), a ligand-activated TF, interacts with RNA in a ligand-dependent manner. Defects in RNA binding lead to genome-wide loss of ERα recruitment, particularly at weaker ERα-motifs. Furthermore, ERα mobility in the nucleus increases in the absence of its RNA-binding capacity. Unexpectedly, this increased mobility coincides with robust polymerase loading and transcription of ERα-regulated genes that harbor low-strength motifs. However, highly stable binding of ERα on chromatin negatively impacts ligand-dependent transcription. Collectively, our results suggest that RNA interactions spatially confine ERα on low-affinity sites to fine-tune gene transcription.

10.
Mol Cell ; 75(6): 1103-1116.e9, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31420216

RESUMEN

The mitochondrial pathway of apoptosis is controlled by the ratio of anti- and pro-apoptotic members of the Bcl-2 family of proteins. The molecular events underlying how a given physiological stimulus changes this ratio to trigger apoptosis remains unclear. We report here that human 17-ß-estradiol (E2) and its related steroid hormones induce apoptosis by binding directly to phosphodiesterase 3A, which in turn recruits and stabilizes an otherwise fast-turnover protein Schlafen 12 (SLFN12). The elevated SLFN12 binds to ribosomes to exclude the recruitment of signal recognition particles (SRPs), thereby blocking the continuous protein translation occurring on the endoplasmic reticulum of E2-treated cells. These proteins include Bcl-2 and Mcl-1, whose ensuing decrease triggers apoptosis. The SLFN12 protein and an apoptosis activation marker were co-localized in syncytiotrophoblast of human placentas, where levels of estrogen-related hormones are high, and dynamic cell turnover by apoptosis is critical for successful implantation and placenta development.


Asunto(s)
Apoptosis/efectos de los fármacos , Estradiol/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trofoblastos/metabolismo , Adulto , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Femenino , Células HeLa , Humanos , Células MCF-7 , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ribosomas/metabolismo
11.
Mol Cell ; 75(4): 791-806.e8, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31303470

RESUMEN

YAP/TEAD are nuclear effectors of the Hippo pathway, regulating organ size and tumorigenesis largely through promoter-associated function. However, their function as enhancer regulators remains poorly understood. Through an in vivo proximity-dependent labeling (BioID) technique, we identified YAP1 and TEAD4 protein as co-regulators of ERα on enhancers. The binding of YAP1/TEAD4 to ERα-bound enhancers is augmented upon E2 stimulation and is required for the induction of E2/ERα target genes and E2-induced oncogenic cell growth. Furthermore, their enhancer binding is a prerequisite for enhancer activation marked by eRNA transcription and for the recruitment of the enhancer activation machinery component MED1. The binding of TEAD4 on active ERE-containing enhancers is independent of its DNA-binding behavior, and instead, occurs through protein-tethering trans-binding. Our data reveal a non-canonical function of YAP1 and TEAD4 as ERα cofactors in regulating cancer growth, highlighting the potential of YAP/TEAD as possible actionable drug targets for ERα+ breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Estrógenos/farmacología , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Células MCF-7 , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo , Ratones , Ratones Desnudos , Proteínas Musculares/genética , Proteínas de Neoplasias/genética , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
12.
Mol Cell ; 75(1): 154-171.e5, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31056445

RESUMEN

The epigenetic information present in mammalian gametes and whether it is transmitted to the progeny are relatively unknown. We find that many promoters in mouse sperm are occupied by RNA polymerase II (Pol II) and Mediator. The same promoters are accessible in GV and MII oocytes and preimplantation embryos. Sperm distal ATAC-seq sites containing motifs for various transcription factors are conserved in monkeys and humans. ChIP-seq analyses confirm that Foxa1, ERα, and AR occupy distal enhancers in sperm. Accessible sperm enhancers containing H3.3 and H2A.Z are also accessible in oocytes and preimplantation embryos. Furthermore, their interactions with promoters in the gametes persist during early development. Sperm- or oocyte-specific interactions mediated by CTCF and cohesin are only present in the paternal or maternal chromosomes, respectively, in the zygote and 2-cell stages. These interactions converge in both chromosomes by the 8-cell stage. Thus, mammalian gametes contain complex patterns of 3D interactions that can be transmitted to the zygote after fertilization.


Asunto(s)
Factor de Unión a CCCTC/genética , Factor Nuclear 3-beta del Hepatocito/genética , Oocitos/metabolismo , Espermatozoides/metabolismo , Cigoto/metabolismo , Animales , Secuencia de Bases , Factor de Unión a CCCTC/metabolismo , Cromatina/química , Cromatina/metabolismo , Secuencia Conservada , Embrión de Mamíferos , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Macaca mulatta , Masculino , Ratones , Oocitos/citología , Oocitos/crecimiento & desarrollo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Homología de Secuencia de Ácido Nucleico , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Dedos de Zinc/genética , Cigoto/citología , Cigoto/crecimiento & desarrollo
13.
Proc Natl Acad Sci U S A ; 121(24): e2321344121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38830107

RESUMEN

The estrogen receptor-α (ER) is thought to function only as a homodimer but responds to a variety of environmental, metazoan, and therapeutic estrogens at subsaturating doses, supporting binding mixtures of ligands as well as dimers that are only partially occupied. Here, we present a series of flexible ER ligands that bind to receptor dimers with individual ligand poses favoring distinct receptor conformations-receptor conformational heterodimers-mimicking the binding of two different ligands. Molecular dynamics simulations showed that the pairs of different ligand poses changed the correlated motion across the dimer interface to generate asymmetric communication between the dimer interface, the ligands, and the surface binding sites for epigenetic regulatory proteins. By examining the binding of the same ligand in crystal structures of ER in the agonist vs. antagonist conformers, we also showed that these allosteric signals are bidirectional. The receptor conformer can drive different ligand binding modes to support agonist vs. antagonist activity profiles, a revision of ligand binding theory that has focused on unidirectional signaling from the ligand to the coregulator binding site. We also observed differences in the allosteric signals between ligand and coregulator binding sites in the monomeric vs. dimeric receptor, and when bound by two different ligands, states that are physiologically relevant. Thus, ER conformational heterodimers integrate two different ligand-regulated activity profiles, representing different modes for ligand-dependent regulation of ER activity.


Asunto(s)
Receptor alfa de Estrógeno , Estrógenos , Simulación de Dinámica Molecular , Multimerización de Proteína , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/química , Regulación Alostérica , Humanos , Ligandos , Estrógenos/metabolismo , Estrógenos/química , Sitios de Unión , Unión Proteica , Conformación Proteica
14.
Proc Natl Acad Sci U S A ; 121(40): e2406837121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39312663

RESUMEN

Cancers develop resistance to inhibitors of oncogenes mainly due to target-centric mechanisms such as mutations and splicing. While inhibitors or antagonists force targets to unnatural conformation contributing to protein instability and resistance, activating tumor suppressors may maintain the protein in an agonistic conformation to elicit sustainable growth inhibition. Due to the lack of tumor suppressor agonists, this hypothesis and the mechanisms underlying resistance are not understood. In estrogen receptor (ER)-positive breast cancer (BC), androgen receptor (AR) is a druggable tumor suppressor offering a promising avenue for this investigation. Spatial genomics suggests that the molecular portrait of AR-expressing BC cells in tumor microenvironment corresponds to better overall patient survival, clinically confirming AR's role as a tumor suppressor. Ligand activation of AR in ER-positive BC xenografts reprograms cistromes, inhibits oncogenic pathways, and promotes cellular elasticity toward a more differentiated state. Sustained AR activation results in cistrome rearrangement toward transcription factor PROP paired-like homeobox 1, transformation of AR into oncogene, and activation of the Janus kinase/signal transducer (JAK/STAT) pathway, all culminating in lineage plasticity to an aggressive resistant subtype. While the molecular profile of AR agonist-sensitive tumors corresponds to better patient survival, the profile represented in the resistant phenotype corresponds to shorter survival. Inhibition of activated oncogenes in resistant tumors reduces growth and resensitizes them to AR agonists. These findings indicate that persistent activation of a context-dependent tumor suppressor may lead to resistance through lineage plasticity-driven tumor metamorphosis. Our work provides a framework to explore the above phenomenon across multiple cancer types and underscores the importance of factoring sensitization of tumor suppressor targets while developing agonist-like drugs.


Asunto(s)
Neoplasias de la Mama , Receptores Androgénicos , Receptores de Estrógenos , Factores de Transcripción STAT , Humanos , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/genética , Animales , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Oncogenes , Quinasas Janus/metabolismo , Ratones , Transducción de Señal , Línea Celular Tumoral , Microambiente Tumoral , Regulación Neoplásica de la Expresión Génica
15.
Proc Natl Acad Sci U S A ; 121(37): e2406854121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39231208

RESUMEN

Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by cognitive decline and learning/memory impairment associated with neuronal cell loss. Estrogen-related receptor α (ERRα) and ERRγ, which are highly expressed in the brain, have emerged as potential AD regulators, with unelucidated underlying mechanisms. Here, we identified genome-wide binding sites for ERRα and ERRγ in human neuronal cells. They commonly target a subset of genes associated with neurodegenerative diseases, including AD. Notably, Dickkopf-1 (DKK1), a Wnt signaling pathway antagonist, was transcriptionally repressed by both ERRα and ERRγ in human neuronal cells and brain. ERRα and ERRγ repress RNA polymerase II (RNAP II) accessibility at the DKK1 promoter by modulating a specific active histone modification, histone H3 lysine acetylation (H3K9ac), with the potential contribution of their corepressor. This transcriptional repression maintains Wnt signaling activity, preventing tau phosphorylation and promoting a healthy neuronal state in the context of AD.


Asunto(s)
Enfermedad de Alzheimer , Receptor Relacionado con Estrógeno ERRalfa , Péptidos y Proteínas de Señalización Intercelular , Receptores de Estrógenos , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Neuronas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Vía de Señalización Wnt/genética
16.
Annu Rev Pharmacol Toxicol ; 63: 295-320, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36662583

RESUMEN

The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and ß), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein-coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.


Asunto(s)
Estrógenos , Receptores de Estrógenos , Masculino , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Estrógenos/farmacología , Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas de Unión al GTP/metabolismo
17.
Mol Cell ; 70(4): 679-694.e7, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29775582

RESUMEN

Enhancers are thought to activate transcription by physically contacting promoters via looping. However, direct assays demonstrating these contacts are required to mechanistically verify such cellular determinants of enhancer function. Here, we present versatile cell-free assays to further determine the role of enhancer-promoter contacts (EPCs). We demonstrate that EPC is linked to mutually stimulatory transcription at the enhancer and promoter in vitro. SRC-3 was identified as a critical looping determinant for the estradiol-(E2)-regulated GREB1 locus. Surprisingly, the GREB1 enhancer and promoter contact two internal gene body SRC-3 binding sites, GBS1 and GBS2, which stimulate their transcription. Utilizing time-course 3C assays, we uncovered SRC-3-dependent dynamic chromatin interactions involving the enhancer, promoter, GBS1, and GBS2. Collectively, these data suggest that the enhancer and promoter remain "poised" for transcription via their contacts with GBS1 and GBS2. Upon E2 induction, GBS1 and GBS2 disengage from the enhancer, allowing direct EPC for active transcription.


Asunto(s)
Neoplasias de la Mama/genética , Cromatina/metabolismo , Estrógenos/farmacología , Regulación Neoplásica de la Expresión Génica , Coactivador 3 de Receptor Nuclear/metabolismo , Transcripción Genética , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cromatina/genética , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Coactivador 3 de Receptor Nuclear/genética , Regiones Promotoras Genéticas , Unión Proteica , Células Tumorales Cultivadas
18.
Mol Cell ; 70(2): 340-357.e8, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29628309

RESUMEN

Whereas the actions of enhancers in gene transcriptional regulation are well established, roles of JmjC-domain-containing proteins in mediating enhancer activation remain poorly understood. Here, we report that recruitment of the JmjC-domain-containing protein 6 (JMJD6) to estrogen receptor alpha (ERα)-bound active enhancers is required for RNA polymerase II recruitment and enhancer RNA production on enhancers, resulting in transcriptional pause release of cognate estrogen target genes. JMJD6 is found to interact with MED12 in the mediator complex to regulate its recruitment. Unexpectedly, JMJD6 is necessary for MED12 to interact with CARM1, which methylates MED12 at multiple arginine sites and regulates its chromatin binding. Consistent with its role in transcriptional activation, JMJD6 is required for estrogen/ERα-induced breast cancer cell growth and tumorigenesis. Our data have uncovered a critical regulator of estrogen/ERα-induced enhancer coding gene activation and breast cancer cell potency, providing a potential therapeutic target of ER-positive breast cancers.


Asunto(s)
Neoplasias de la Mama/enzimología , Proliferación Celular , Receptor alfa de Estrógeno/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Complejo Mediador/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Células MCF-7 , Complejo Mediador/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Unión Proteica , Transporte de Proteínas , Proteína-Arginina N-Metiltransferasas/genética , Transducción de Señal , Activación Transcripcional/efectos de los fármacos
19.
Mol Cell Proteomics ; 23(1): 100702, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38122900

RESUMEN

Estrogen receptor α (ERα) drives the transcription of genes involved in breast cancer (BC) progression, relying on coregulatory protein recruitment for its transcriptional and biological activities. Mutation of ERα as well as aberrant recruitment of its regulatory proteins contribute to tumor adaptation and drug resistance. Therefore, understanding the dynamic changes in ERα protein interaction networks is crucial for elucidating drug resistance mechanisms in BC. Despite progress in studying ERα-associated proteins, capturing subcellular transient interactions remains challenging and, as a result, significant number of important interactions remain undiscovered. In this study, we employed biotinylation by antibody recognition (BAR), an innovative antibody-based proximity labeling (PL) approach, coupled with mass spectrometry to investigate the ERα proximal proteome and its changes associated with resistance to aromatase inhibition, a key therapy used in the treatment of ERα-positive BC. We show that BAR successfully detected most of the known ERα interactors and mainly identified nuclear proteins, using either an epitope tag or endogenous antibody to target ERα. We further describe the ERα proximal proteome rewiring associated with resistance applying BAR to a panel of isogenic cell lines modeling tumor adaptation in the clinic. Interestingly, we find that ERα associates with some of the canonical cofactors in resistant cells and several proximal proteome changes are due to increased expression of ERα. Resistant models also show decreased levels of estrogen-regulated genes. Sensitive and resistant cells harboring a mutation in the ERα (Y537C) revealed a similar proximal proteome. We provide an ERα proximal protein network covering several novel ERα-proximal partners. These include proteins involved in highly dynamic processes such as sumoylation and ubiquitination difficult to detect with traditional protein interaction approaches. Overall, we present BAR as an effective approach to investigate the ERα proximal proteome in a spatial context and demonstrate its application in different experimental conditions.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Femenino , Humanos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteoma/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/uso terapéutico
20.
Proc Natl Acad Sci U S A ; 120(12): e2214225120, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36917668

RESUMEN

A murine papillomavirus, MmuPV1, infects both cutaneous and mucosal epithelia of laboratory mice and can be used to model high-risk human papillomavirus (HPV) infection and HPV-associated disease. We have shown that estrogen exacerbates papillomavirus-induced cervical disease in HPV-transgenic mice. We have also previously identified stress keratin 17 (K17) as a host factor that supports MmuPV1-induced cutaneous disease. Here, we sought to test the role of estrogen and K17 in MmuPV1 infection and associated disease in the female reproductive tract. We experimentally infected wild-type and K17 knockout (K17KO) mice with MmuPV1 in the female reproductive tract in the presence or absence of exogenous estrogen for 6 mon. We observed that a significantly higher percentage of K17KO mice cleared the virus as opposed to wild-type mice. In estrogen-treated wild-type mice, the MmuPV1 viral copy number was significantly higher compared to untreated mice by as early as 2 wk postinfection, suggesting that estrogen may help facilitate MmuPV1 infection and/or establishment. Consistent with this, viral clearance was not observed in either wild-type or K17KO mice when treated with estrogen. Furthermore, neoplastic disease progression and cervical carcinogenesis were supported by the presence of K17 and exacerbated by estrogen treatment. Subsequent analyses indicated that estrogen treatment induces a systemic immunosuppressive state in MmuPV1-infected animals and that both estrogen and K17 modulate the local intratumoral immune microenvironment within MmuPV1-induced neoplastic lesions. Collectively, these findings suggest that estrogen and K17 act at multiple stages of papillomavirus-induced disease at least in part via immunomodulatory mechanisms.


Asunto(s)
Infecciones por Papillomavirus , Ratones , Femenino , Humanos , Animales , Infecciones por Papillomavirus/genética , Queratina-17 , Ratones Transgénicos , Inmunidad , Papillomaviridae/genética , Estrógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA