Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 228: 116237, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38679211

RESUMEN

Cytochromes P450 can metabolize endogenous fatty acids, such as arachidonic acid, to bioactive lipids such as epoxyeicosatrienoic acids (EETs) that have beneficial effects. EETs protect hearts against ischemic damage, heart failure or fibrosis; however, their effects are limited by hydrolysis to less active dihydroxy oxylipins by soluble epoxide hydrolase (sEH), encoded by the epoxide hydrolase 2 gene (EPHX2, EC 3.3.2.10). Pharmacological inhibition or genetic disruption of sEH/EPHX2 have been widely studied for their impact on cardiovascular diseases. Less well studied is the role of increased EPHX2 expression, which occurs in a substantial human population that carries the EPHX2 K55R polymorphism or after induction by inflammatory stimuli. Herein, we developed a mouse model with cardiomyocyte-selective expression of human EPHX2 (Myh6-EPHX2) that has significantly increased total EPHX2 expression and activity. Myh6-EPHX2 hearts exhibit strong, cardiomyocyte-selective expression of EPHX2. EPHX2 mRNA, protein, and epoxide hydrolysis measurements suggest that Myh6-EPHX2 hearts have 12-fold increase in epoxide hydrolase activity relative to wild type (WT) hearts. This increased activity significantly decreased epoxide:diol ratios in vivo. Isolated, perfused Myh6-EPHX2 hearts were not significantly different from WT hearts in basal parameters of cardiac function; however, compared to WT hearts, Myh6-EPHX2 hearts demonstrated reduced recovery of heart contractile function after ischemia and reperfusion (I/R). This impaired recovery after I/R correlated with reduced activation of PI3K/AKT and GSK3ß signaling pathways in Myh6-EPHX2 hearts compared to WT hearts. In summary, the Myh6-EPHX2 mouse line represents a novel model of cardiomyocyte-selective overexpression of EPHX2 that has detrimental effects on cardiac function.


Asunto(s)
Epóxido Hidrolasas , Contracción Miocárdica , Epóxido Hidrolasas/genética , Epóxido Hidrolasas/metabolismo , Animales , Contracción Miocárdica/fisiología , Ratones , Ratones Transgénicos , Masculino , Miocitos Cardíacos/metabolismo , Ratones Endogámicos C57BL , Humanos , Recuperación de la Función/fisiología
2.
Sci Adv ; 10(9): eadj5107, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38427725

RESUMEN

Cell fate decisions are achieved with gene expression changes driven by lineage-specific transcription factors (TFs). These TFs depend on chromatin remodelers including the Brahma-related gene 1 (BRG1)-associated factor (BAF) complex to activate target genes. BAF complex subunits are essential for development and frequently mutated in cancer. Thus, interrogating how BAF complexes contribute to cell fate decisions is critical for human health. We examined the requirement for the catalytic BAF subunit BRG1 in neural progenitor cell (NPC) specification from human embryonic stem cells. During the earliest stages of differentiation, BRG1 was required to establish chromatin accessibility at neuroectoderm-specific enhancers. Depletion of BRG1 dorsalized NPCs and promoted precocious neural crest specification and enhanced neuronal differentiation. These findings demonstrate that BRG1 mediates NPC specification by ensuring proper expression of lineage-specific TFs and appropriate activation of their transcriptional programs.


Asunto(s)
Cromatina , Placa Neural , Humanos , Cromatina/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Placa Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
3.
Genes Dev ; 32(1): 26-41, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378787

RESUMEN

Regulation by gene-distal enhancers is critical for cell type-specific and condition-specific patterns of gene expression. Thus, to understand the basis of gene activity in a given cell type or tissue, we must identify the precise locations of enhancers and functionally characterize their behaviors. Here, we demonstrate that transcription is a nearly universal feature of enhancers in Drosophila and mammalian cells and that nascent RNA sequencing strategies are optimal for identification of both enhancers and superenhancers. We dissect the mechanisms governing enhancer transcription and discover remarkable similarities to transcription at protein-coding genes. We show that RNA polymerase II (RNAPII) undergoes regulated pausing and release at enhancers. However, as compared with mRNA genes, RNAPII at enhancers is less stable and more prone to early termination. Furthermore, we found that the level of histone H3 Lys4 (H3K4) methylation at enhancers corresponds to transcriptional activity such that highly active enhancers display H3K4 trimethylation rather than the H3K4 monomethylation considered a hallmark of enhancers. Finally, our work provides insights into the unique characteristics of superenhancers, which stimulate high-level gene expression through rapid pause release; interestingly, this property renders associated genes resistant to the loss of factors that stabilize paused RNAPII.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Elongación de la Transcripción Genética , Animales , Cromatina/química , Proteínas Cromosómicas no Histona/fisiología , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/fisiología , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Ratones , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , ARN no Traducido/biosíntesis , Sitio de Iniciación de la Transcripción , Transcripción Genética , Factores de Elongación Transcripcional/fisiología
4.
Mol Cell ; 58(6): 1101-12, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26028540

RESUMEN

Anti-sense transcription originating upstream of mammalian protein-coding genes is a well-documented phenomenon, but remarkably little is known about the regulation or function of anti-sense promoters and the non-coding RNAs they generate. Here we define at nucleotide resolution the divergent transcription start sites (TSSs) near mouse mRNA genes. We find that coupled sense and anti-sense TSSs precisely define the boundaries of a nucleosome-depleted region (NDR) that is highly enriched in transcription factor (TF) motifs. Notably, as the distance between sense and anti-sense TSSs increases, so does the size of the NDR, the level of signal-dependent TF binding, and gene activation. We further discover a group of anti-sense TSSs in macrophages with an enhancer-like chromatin signature. Interestingly, this signature identifies divergent promoters that are activated during immune challenge. We propose that anti-sense promoters serve as platforms for TF binding and establishment of active chromatin to further regulate or enhance sense-strand mRNA expression.


Asunto(s)
Cromatina/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Cromatina/metabolismo , ADN sin Sentido/genética , Regulación de la Expresión Génica , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Modelos Genéticos , Nucleosomas/genética , Nucleosomas/metabolismo , Motivos de Nucleótidos/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética
5.
Mol Cell ; 58(2): 311-322, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25773599

RESUMEN

The remarkable capacity for pluripotency and self-renewal in embryonic stem cells (ESCs) requires a finely tuned transcriptional circuitry wherein the pathways and genes that initiate differentiation are suppressed, but poised to respond rapidly to developmental signals. To elucidate transcriptional control in mouse ESCs in the naive, ground state, we defined the distribution of engaged RNA polymerase II (Pol II) at high resolution. We find that promoter-proximal pausing of Pol II is most enriched at genes regulating cell cycle and signal transduction and not, as expected, at developmental or bivalent genes. Accordingly, ablation of the primary pause-inducing factor NELF does not increase expression of lineage markers, but instead causes proliferation defects, embryonic lethality, and dysregulation of ESC signaling pathways. Indeed, ESCs lacking NELF have dramatically attenuated FGF/ERK activity, rendering them resistant to differentiation. This work thus uncovers a key role for NELF-mediated pausing in establishing the responsiveness of stem cells to developmental cues.


Asunto(s)
Células Madre Embrionarias/enzimología , Mamíferos/crecimiento & desarrollo , ARN Polimerasa III/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Mamíferos/metabolismo , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Factores de Transcripción/genética
6.
Mol Cell ; 52(4): 517-28, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24184211

RESUMEN

Metazoan gene expression is often regulated after the recruitment of RNA polymerase II (Pol II) to promoters, through the controlled release of promoter-proximally paused Pol II into productive RNA synthesis. Despite the prevalence of paused Pol II, very little is known about the dynamics of these early elongation complexes or the fate of the short transcription start site-associated (tss) RNAs they produce. Here, we demonstrate that paused elongation complexes can be remarkably stable, with half-lives exceeding 15 min at genes with inefficient pause release. Promoter-proximal termination by Pol II is infrequent, and released tssRNAs are targeted for rapid degradation. Further, we provide evidence that the predominant tssRNA species observed are nascent RNAs held within early elongation complexes. We propose that stable pausing of polymerase provides a temporal window of opportunity for recruitment of factors to modulate gene expression and that the nascent tssRNA represents an appealing target for these interactions.


Asunto(s)
Proteínas de Drosophila/genética , ARN Polimerasa II/fisiología , ARN Citoplasmático Pequeño/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Estabilidad del ARN , Transducción de Señal , Elongación de la Transcripción Genética
7.
Proc Natl Acad Sci U S A ; 110(36): 14616-21, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23950223

RESUMEN

Widespread anti-inflammatory actions of glucocorticoid hormones are mediated by the glucocorticoid receptor (GR), a ligand-dependent transcription factor of the nuclear receptor superfamily. In conjunction with its corepressor GR-interacting protein-1 (GRIP1), GR tethers to the DNA-bound activator protein-1 and NF-κB and represses transcription of their target proinflammatory cytokine genes. However, these target genes fall into distinct classes depending on the step of the transcription cycle that is rate-limiting for their activation: Some are controlled through RNA polymerase II (PolII) recruitment and initiation, whereas others undergo signal-induced release of paused elongation complexes into productive RNA synthesis. Whether these genes are differentially regulated by GR is unknown. Here we report that, at the initiation-controlled inflammatory genes in primary macrophages, GR inhibited LPS-induced PolII occupancy. In contrast, at the elongation-controlled genes, GR did not affect PolII recruitment or transcription initiation but promoted, in a GRIP1-dependent manner, the accumulation of the pause-inducing negative elongation factor. Consistently, GR-dependent repression of elongation-controlled genes was abolished specifically in negative elongation factor-deficient macrophages. Thus, GR:GRIP1 use distinct mechanisms to repress inflammatory genes at different stages of the transcription cycle.


Asunto(s)
Citocinas/genética , Regulación de la Expresión Génica , Receptores de Glucocorticoides/metabolismo , Transcripción Genética/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , Dexametasona/farmacología , Immunoblotting , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , Receptores de Glucocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 106(43): 18207-12, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19820169

RESUMEN

The kinetics and magnitude of cytokine gene expression are tightly regulated to elicit a balanced response to pathogens and result from integrated changes in transcription and mRNA stability. Yet, how a single microbial stimulus induces peak transcription of some genes (TNFalpha) within minutes whereas others (IP-10) require hours remains unclear. Here, we dissect activation of several lipopolysaccharide (LPS)-inducible genes in macrophages, an essential cell type mediating inflammatory response in mammals. We show that a key difference between the genes is the step of the transcription cycle at which they are regulated. Specifically, at TNFalpha, RNA Polymerase II initiates transcription in resting macrophages, but stalls near the promoter until LPS triggers rapid and transient release of the negative elongation factor (NELF) complex and productive elongation. In contrast, no NELF or polymerase is detectible near the IP-10 promoter before induction, and LPS-dependent polymerase recruitment is rate limiting for transcription. We further demonstrate that this strategy is shared by other immune mediators and is independent of the inducer and signaling pathway responsible for gene activation. Finally, as a striking example of evolutionary conservation, the Drosophila homolog of the TNFalpha gene, eiger, displayed all of the hallmarks of NELF-dependent polymerase stalling. We propose that polymerase stalling ensures the coordinated, timely activation the inflammatory gene expression program from Drosophila to mammals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , ARN Polimerasa II/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Evolución Molecular , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/genética , Ratones , Fosforilación , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
9.
Nat Genet ; 39(12): 1507-11, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17994021

RESUMEN

Regulation of gene expression is integral to the development and survival of all organisms. Transcription begins with the assembly of a pre-initiation complex at the gene promoter, followed by initiation of RNA synthesis and the transition to productive elongation. In many cases, recruitment of RNA polymerase II (Pol II) to a promoter is necessary and sufficient for activation of genes. However, there are a few notable exceptions to this paradigm, including heat shock genes and several proto-oncogenes, whose expression is attenuated by regulated stalling of polymerase elongation within the promoter-proximal region. To determine the importance of polymerase stalling for transcription regulation, we carried out a genome-wide search for Drosophila melanogaster genes with Pol II stalled within the promoter-proximal region. Our data show that stalling is widespread, occurring at hundreds of genes that respond to stimuli and developmental signals. This finding indicates a role for regulation of polymerase elongation in the transcriptional responses to dynamic environmental and developmental cues.


Asunto(s)
Regulación de la Expresión Génica , Genoma de los Insectos , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Inmunoprecipitación de Cromatina , Drosophila melanogaster , Activación Transcripcional
10.
Mol Cell Biol ; 25(17): 7796-802, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16107724

RESUMEN

Aneuploidy is a common feature of human tumors, often correlating with poor prognosis. The mitotic spindle checkpoint is thought to play a major role in aneuploidy suppression. To investigate the role of the spindle checkpoint in tumor suppression in vivo, we developed transgenic mice in which thymocytes express a dominant interfering fragment of Bub1, a kinase regulator of the spindle checkpoint. We report that, despite high-level expression of dominant-negative Bub1 (Bub1DN), a protein known to inhibit spindle checkpoint activity in cultured cells, thymocytes show no evidence of spindle checkpoint impairment. Transgenic animals also failed to show an increased predisposition to spontaneous tumors. Moreover, the Bub1DN transgene failed to alter the timing or characteristics of thymic lymphoma development in p53 heterozygous or homozygous null backgrounds, indicating that the lack of tumorigenesis is not due to suppression by p53-dependent checkpoints. These results indicate that overexpression of a Bub1 N-terminal fragment is insufficient to impair the spindle checkpoint in vivo or to drive tumorigenesis in the highly susceptible murine thymocyte system, either alone or in combination with G(1) checkpoint disruption.


Asunto(s)
Mutación/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Animales , Ciclo Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Regulación de la Expresión Génica , Genes Dominantes/genética , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas , Tasa de Supervivencia , Neoplasias de la Tiroides/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA