Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 3225, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270532

RESUMEN

Certain cellular processes are dose-dependent, requiring specific quantities or stoichiometries of gene products, as exemplified by haploinsufficiency and sex-chromosome dosage compensation. Understanding dosage-sensitive processes requires tools to quantitatively modulate protein abundance. Here we present CasTuner, a CRISPR-based toolkit for analog tuning of endogenous gene expression. The system exploits Cas-derived repressors that are quantitatively tuned by ligand titration through a FKBP12F36V degron domain. CasTuner can be applied at the transcriptional or post-transcriptional level using a histone deacetylase (hHDAC4) fused to dCas9, or the RNA-targeting CasRx, respectively. We demonstrate analog tuning of gene expression homogeneously across cells in mouse and human cells, as opposed to KRAB-dependent CRISPR-interference systems, which exhibit digital repression. Finally, we quantify the system's dynamics and use it to measure dose-response relationships of NANOG and OCT4 with their target genes and with the cellular phenotype. CasTuner thus provides an easy-to-implement tool to study dose-responsive processes in their physiological context.


Asunto(s)
Sistemas CRISPR-Cas , Factores de Transcripción , Ratones , Humanos , Animales , Sistemas CRISPR-Cas/genética , Factores de Transcripción/metabolismo , Genes Homeobox , Expresión Génica
2.
Mol Cell ; 82(1): 190-208.e17, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34932975

RESUMEN

Developmental genes such as Xist, which initiates X chromosome inactivation, are controlled by complex cis-regulatory landscapes, which decode multiple signals to establish specific spatiotemporal expression patterns. Xist integrates information on X chromosome dosage and developmental stage to trigger X inactivation in the epiblast specifically in female embryos. Through a pooled CRISPR screen in differentiating mouse embryonic stem cells, we identify functional enhancer elements of Xist at the onset of random X inactivation. Chromatin profiling reveals that X-dosage controls the promoter-proximal region, while differentiation cues activate several distal enhancers. The strongest distal element lies in an enhancer cluster associated with a previously unannotated Xist-enhancing regulatory transcript, which we named Xert. Developmental cues and X-dosage are thus decoded by distinct regulatory regions, which cooperate to ensure female-specific Xist upregulation at the correct developmental time. With this study, we start to disentangle how multiple, functionally distinct regulatory elements interact to generate complex expression patterns in mammals.


Asunto(s)
Elementos de Facilitación Genéticos , Sitios Genéticos , Células Madre Embrionarias de Ratones/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Inactivación del Cromosoma X , Cromosoma X , Animales , Diferenciación Celular , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regulación hacia Arriba
3.
Sci Rep ; 11(1): 17764, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34493753

RESUMEN

Endothelial-mesenchymal transition (EndMT) is a form of endothelial dysfunction wherein endothelial cells acquire a mesenchymal phenotype and lose endothelial functions, which contributes to the pathogenesis of intimal hyperplasia and atherosclerosis. The mitogen activated protein kinase 7 (MAPK7) inhibits EndMT and decreases the expression of the histone methyltransferase Enhancer-of-Zeste homologue 2 (EZH2), thereby maintaining endothelial quiescence. EZH2 is the catalytic subunit of the Polycomb Repressive Complex 2 that methylates lysine 27 on histone 3 (H3K27me3). It is elusive how the crosstalk between MAPK7 and EZH2 is regulated in the endothelium and if the balance between MAPK7 and EZH2 is disturbed in vascular disease. In human coronary artery disease, we assessed the expression levels of MAPK7 and EZH2 and found that with increasing intima/media thickness ratio, MAPK7 expression decreased, whereas EZH2 expression increased. In vitro, MAPK7 activation decreased EZH2 expression, whereas endothelial cells deficient of EZH2 had increased MAPK7 activity. MAPK7 activation results in increased expression of microRNA (miR)-101, a repressor of EZH2. This loss of EZH2 in turn results in the increased expression of the miR-200 family, culminating in decreased expression of the dual-specificity phosphatases 1 and 6 who may repress MAPK7 activity. Transfection of endothelial cells with miR-200 family members decreased the endothelial sensitivity to TGFß1-induced EndMT. In endothelial cells there is reciprocity between MAPK7 signaling and EZH2 expression and disturbances in this reciprocal signaling associate with the induction of EndMT and severity of human coronary artery disease.


Asunto(s)
Transdiferenciación Celular/fisiología , Enfermedad de la Arteria Coronaria/patología , Endotelio Vascular/patología , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Mesodermo/patología , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Transducción de Señal/fisiología , Túnica Íntima/patología , Regiones no Traducidas 3'/genética , Enfermedad de la Arteria Coronaria/enzimología , Estenosis Coronaria/enzimología , Estenosis Coronaria/patología , Fosfatasa 1 de Especificidad Dual/biosíntesis , Fosfatasa 1 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/biosíntesis , Fosfatasa 6 de Especificidad Dual/genética , Endotelio Vascular/enzimología , Activación Enzimática , Regulación de la Expresión Génica , Genes Reporteros , Código de Histonas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hiperplasia , Mesodermo/enzimología , MicroARNs/biosíntesis , MicroARNs/genética , Túnica Media/patología
4.
Curr Opin Chem Biol ; 57: 75-81, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32619853

RESUMEN

Epigenetic editing refers to the locus-specific targeting of epigenetic enzymes to rewrite the local epigenetic landscape of an endogenous genomic site, often with the aim of transcriptional reprogramming. Implementing clustered regularly interspaced short palindromic repeat-dCas9 greatly accelerated the advancement of epigenetic editing, yielding preclinical therapeutic successes using a variety of epigenetic enzymes. Here, we review the current applications of these epigenetic editing tools in mammals and shed light on biochemical improvements that facilitate versatile applications.


Asunto(s)
Sistemas CRISPR-Cas , Epigénesis Genética , Edición Génica/métodos , Animales , Metilación de ADN , Epigenómica/métodos , Genoma , Código de Histonas , Humanos
5.
Int J Mol Sci ; 21(10)2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32455614

RESUMEN

Epigenetic editing, an emerging technique used for the modulation of gene expression in mammalian cells, is a promising strategy to correct disease-related gene expression. Although epigenetic reprogramming results in sustained transcriptional modulation in several in vivo models, further studies are needed to develop this approach into a straightforward technology for effective and specific interventions. Important goals of current research efforts are understanding the context-dependency of successful epigenetic editing and finding the most effective epigenetic effector(s) for specific tasks. Here we tested whether the fibrosis- and cancer-associated PLOD2 gene can be repressed by the DNA methyltransferase M.SssI, or by the non-catalytic Krüppel associated box (KRAB) repressor directed to the PLOD2 promoter via zinc finger- or CRISPR-dCas9-mediated targeting. M.SssI fusions induced de novo DNA methylation, changed histone modifications in a context-dependent manner, and led to 50%-70% reduction in PLOD2 expression in fibrotic fibroblasts and in MDA-MB-231 cancer cells. Targeting KRAB to PLOD2 resulted in the deposition of repressive histone modifications without DNA methylation and in almost complete PLOD2 silencing. Interestingly, both long-term TGFß1-induced, as well as unstimulated PLOD2 expression, was completely repressed by KRAB, while M.SssI only prevented the TGFß1-induced PLOD2 expression. Targeting transiently expressed dCas9-KRAB resulted in sustained PLOD2 repression in HEK293T and MCF-7 cells. Together, these findings point to KRAB outperforming DNA methylation as a small potent targeting epigenetic effector for silencing TGFß1-induced and uninduced PLOD2 expression.


Asunto(s)
Silenciador del Gen , Heterocromatina/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Adulto , Células Cultivadas , ADN-Citosina Metilasas/genética , ADN-Citosina Metilasas/metabolismo , Epigénesis Genética , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Células MCF-7 , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Factor de Crecimiento Transformador beta/metabolismo
6.
Methods Mol Biol ; 1767: 167-185, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29524134

RESUMEN

In a swift revolution, CRISPR/Cas9 has reshaped the means and ease of interrogating biological questions. Particularly, mutants that result in a nuclease-deactivated Cas9 (dCas9) provide scientists with tools to modulate transcription of genomic loci at will by targeting transcriptional effector domains. To interrogate the temporal order of events during transcriptional regulation, rapidly inducible CRISPR/dCas9 systems provide previously unmet molecular tools. In only a few years of time, numerous light and chemical-inducible switches have been applied to CRISPR/dCas9 to generate dCas9 switches. As these inducible switch systems are able to modulate dCas9 directly at the protein level, they rapidly affect dCas9 stability, activity, or target binding and subsequently rapidly influence downstream transcriptional events. Here we review the current state of such biotechnological CRISPR/dCas9 enhancements. Specifically we provide details on their flaws and strengths and on the differences in molecular design between the switch systems. With this we aim to provide a selection guide for researchers with keen interest in rapid temporal control over transcriptional modulation through the CRISPR/dCas9 system.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Activación Transcripcional , Animales , Humanos , Inteínas , Modelos Moleculares , ARN Guía de Kinetoplastida/genética , Transcripción Genética
7.
Physiol Rep ; 5(17)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28904079

RESUMEN

l-Ascorbic acid (AA), generally known as vitamin C, is a crucial cofactor for a variety of enzymes, including prolyl-3-hydroxylase (P3H), prolyl-4-hydroxylase (P4H), and lysyl hydroxylase (LH)-mediated collagen maturation. Here, we investigated whether AA has additional functions in the regulation of the myofibroblast phenotype, besides its function in collagen biosynthesis. We found that AA positively influences TGFß1-induced expression of COL1A1, ACTA2, and COL4A1 Moreover, we demonstrated that AA promotes αSMA stress fiber formation as well as the synthesis and deposition of collagens type I and IV Additionally, AA amplified the contractile phenotype of the myofibroblasts, as seen by increased contraction of a 3D collagen lattice. Moreover, AA increased the expression of several TGFß1-induced genes, including DDR1 and CCN2 Finally, we demonstrated that the mechanism of AA action seems independent of Smad2/3 signaling.


Asunto(s)
Ácido Ascórbico/farmacología , Miofibroblastos/efectos de los fármacos , Fenotipo , Vitaminas/farmacología , Actinas/genética , Actinas/metabolismo , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 1/metabolismo , Humanos , Miofibroblastos/citología , Miofibroblastos/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/farmacología
8.
Sci Rep ; 7: 45047, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28344354

RESUMEN

Chronic obstructive pulmonary disease (COPD) constitutes a major health burden. Studying underlying molecular mechanisms could lead to new therapeutic targets. Macrophages are orchestrators of COPD, by releasing pro-inflammatory cytokines. This process relies on transcription factors such as NF-κB, among others. NF-κB is regulated by lysine acetylation; a post-translational modification installed by histone acetyltransferases and removed by histone deacetylases (HDACs). We hypothesized that small molecule HDAC inhibitors (HDACi) targeting class I HDACs members that can regulate NF-κB could attenuate inflammatory responses in COPD via modulation of the NF-κB signaling output. MS-275 is an isoform-selective inhibitor of HDAC1-3. In precision-cut lung slices and RAW264.7 macrophages, MS-275 upregulated the expression of both pro- and anti-inflammatory genes, implying mixed effects. Interestingly, anti-inflammatory IL10 expression was upregulated in these model systems. In the macrophages, this was associated with increased NF-κB activity, acetylation, nuclear translocation, and binding to the IL10 promoter. Importantly, in an in vivo model of cigarette smoke-exposed C57Bl/6 mice, MS-275 robustly attenuated inflammatory expression of KC and neutrophil influx in the lungs. This study highlights for the first time the potential of isoform-selective HDACi for the treatment of inflammatory lung diseases like COPD.


Asunto(s)
Antiinflamatorios/farmacología , Benzamidas/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Interleucina-10/metabolismo , Macrófagos/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Piridinas/farmacología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Antiinflamatorios/uso terapéutico , Benzamidas/uso terapéutico , Línea Celular , Inhibidores de Histona Desacetilasas/uso terapéutico , Interleucina-10/genética , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/etiología , Piridinas/uso terapéutico
9.
Am J Physiol Lung Cell Mol Physiol ; 312(3): L334-L347, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011616

RESUMEN

Airway mucus hypersecretion contributes to the morbidity and mortality in patients with chronic inflammatory lung diseases. Reducing mucus production is crucial for improving patients' quality of life. The transcription factor SAM-pointed domain-containing Ets-like factor (SPDEF) plays a critical role in the regulation of mucus production and, therefore, represents a potential therapeutic target. This study aims to reduce lung epithelial mucus production by targeted silencing SPDEF using the novel strategy, epigenetic editing. Zinc fingers and CRISPR/dCas platforms were engineered to target repressors (KRAB, DNA methyltransferases, histone methyltransferases) to the SPDEF promoter. All constructs were able to effectively suppress both SPDEF mRNA and protein expression, which was accompanied by inhibition of downstream mucus-related genes [anterior gradient 2 (AGR2), mucin 5AC (MUC5AC)]. For the histone methyltransferase G9A, and not its mutant or other effectors, the obtained silencing was mitotically stable. These results indicate efficient SPDEF silencing and downregulation of mucus-related gene expression by epigenetic editing, in human lung epithelial cells. This opens avenues for epigenetic editing as a novel therapeutic strategy to induce long-lasting mucus inhibition.


Asunto(s)
Epigénesis Genética , Células Epiteliales/metabolismo , Edición Génica , Pulmón/citología , Moco/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Secuencia de Bases , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Regulación hacia Abajo/genética , Silenciador del Gen , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Modelos Biológicos , Mucina 5AC/metabolismo , Regiones Promotoras Genéticas/genética , Dominios Proteicos , Proteínas Proto-Oncogénicas c-ets/metabolismo , Dedos de Zinc
10.
Crit Rev Biochem Mol Biol ; 52(1): 74-95, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28006962

RESUMEN

Collagen is a macromolecule that has versatile roles in physiology, ranging from structural support to mediating cell signaling. Formation of mature collagen fibrils out of procollagen α-chains requires a variety of enzymes and chaperones in a complex process spanning both intracellular and extracellular post-translational modifications. These processes include modifications of amino acids, folding of procollagen α-chains into a triple-helical configuration and subsequent stabilization, facilitation of transportation out of the cell, cleavage of propeptides, aggregation, cross-link formation, and finally the formation of mature fibrils. Disruption of any of the proteins involved in these biosynthesis steps potentially result in a variety of connective tissue diseases because of a destabilized extracellular matrix. In this review, we give a revised overview of the enzymes and chaperones currently known to be relevant to the conversion of lysine and proline into hydroxyproline and hydroxylysine, respectively, and the O-glycosylation of hydroxylysine and give insights into the consequences when these steps are disrupted.


Asunto(s)
Colágenos Fibrilares/metabolismo , Animales , Artrogriposis/metabolismo , Artrogriposis/patología , Enfermedades del Tejido Conjuntivo/metabolismo , Enfermedades del Tejido Conjuntivo/patología , Síndrome de Ehlers-Danlos/metabolismo , Síndrome de Ehlers-Danlos/patología , Colágenos Fibrilares/análisis , Glicosilación , Humanos , Hidroxilación , Hidroxilisina/análisis , Hidroxilisina/metabolismo , Hidroxiprolina/análisis , Hidroxiprolina/metabolismo , Lisina/análisis , Lisina/metabolismo , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Prolina/análisis , Prolina/metabolismo , Pliegue de Proteína
11.
Nat Commun ; 7: 12284, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27506838

RESUMEN

Histone modifications reflect gene activity, but the relationship between cause and consequence of transcriptional control is heavily debated. Recent developments in rewriting local histone codes of endogenous genes elucidated instructiveness of certain marks in regulating gene expression. Maintenance of such repressive epigenome editing is controversial, while stable reactivation is still largely unexplored. Here we demonstrate sustained gene re-expression using two types of engineered DNA-binding domains fused to a H3K4 methyltransferase. Local induction of H3K4me3 is sufficient to allow re-expression of silenced target genes in various cell types. Maintenance of the re-expression is achieved, but strongly depends on the chromatin microenvironment (that is, DNA methylation status). We further identify H3K79me to be essential in allowing stable gene re-expression, confirming its role in epigenetic crosstalk for stable reactivation. Our approach uncovers potent epigenetic modifications to be directly written onto genomic loci to stably activate any given gene.


Asunto(s)
Metilación de ADN/genética , Silenciador del Gen , Histonas/genética , Activación Transcripcional , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Ingeniería Genética/métodos , Histonas/metabolismo , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Dominios Proteicos/genética
12.
Proc Natl Acad Sci U S A ; 113(26): 7142-7, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27298363

RESUMEN

Collagens are subjected to extensive posttranslational modifications, such as lysine hydroxylation. Bruck syndrome (BS) is a connective tissue disorder characterized at the molecular level by a loss of telopeptide lysine hydroxylation, resulting in reduced collagen pyridinoline cross-linking. BS results from mutations in the genes coding for lysyl hydroxylase (LH) 2 or peptidyl-prolyl cis-trans isomerase (PPIase) FKBP65. Given that the immunophilin FKBP65 does not exhibit LH activity, it is likely that LH2 activity is somehow dependent on FKPB65. In this report, we provide insights regarding the interplay between LH2 and FKBP65. We found that FKBP65 forms complexes with LH2 splice variants LH2A and LH2B but not with LH1 and LH3. Ablating the catalytic activity of FKBP65 or LH2 did not affect complex formation. Both depletion of FKBP65 and inhibition of FKBP65 PPIase activity reduced the dimeric (active) form of LH2 but did not affect the binding of monomeric (inactive) LH2 to procollagen Iα1. Furthermore, we show that LH2A and LH2B cannot form heterodimers with each other but are able to form heterodimers with LH1 and LH3. Collectively, our results indicate that FKBP65 is linked to pyridinoline cross-linking by specifically mediating the dimerization of LH2. Moreover, FKBP65 does not interact with LH1 and LH3, explaining why in BS triple-helical hydroxylysines are not affected. Our results provide a mechanistic link between FKBP65 and the loss of pyridinolines and may hold the key to future treatments for diseases related to collagen cross-linking anomalies, such as fibrosis and cancer.


Asunto(s)
Aminoácidos/química , Artrogriposis/metabolismo , Colágeno Tipo I/química , Colágeno/química , Reactivos de Enlaces Cruzados/química , Osteogénesis Imperfecta/metabolismo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Aminoácidos/metabolismo , Artrogriposis/enzimología , Artrogriposis/genética , Colágeno/genética , Colágeno/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Dimerización , Humanos , Osteogénesis Imperfecta/enzimología , Osteogénesis Imperfecta/genética , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas de Unión a Tacrolimus/genética
13.
J Biol Chem ; 290(47): 28465-28476, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26432637

RESUMEN

PLOD2 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2) hydroxylates lysine residues in collagen telopeptides and is essential for collagen pyridinoline cross-link formation. PLOD2 expression and subsequent pyridinoline cross-links are increased in fibrotic pathologies by transforming growth factor ß-1 (TGFß1). In this report we examined the molecular processes underlying TGFß1-induced PLOD2 expression. We found that binding of the TGFß1 pathway related transcription factors SMAD3 and SP1-mediated TGFß1 enhanced PLOD2 expression and could be correlated to an increase of acetylated histone H3 and H4 at the PLOD2 promoter. Interestingly, the classical co-activators of SMAD3 complexes, p300 and CBP, were not responsible for the enhanced H3 and H4 acetylation. Depletion of SMAD3 reduced PLOD2 acetylated H3 and H4, indicating that another as of yet unidentified histone acetyltransferase binds to SMAD3 at PLOD2. Assessing histone methylation marks at the PLOD2 promoter depicted an increase of the active histone mark H3K79me2, a decrease of the repressive H4K20me3 mark, but no role for the generally strong transcription-related modifications: H3K4me3, H3K9me3 and H3K27me3. Collectively, our findings reveal that TGFß1 induces a SP1- and SMAD3-dependent recruitment of histone modifying enzymes to the PLOD2 promoter other than the currently known TGFß1 downstream co-activators and epigenetic modifications. This also suggests that additional activation strategies are used downstream of the TGFß1 pathway, and hence their unraveling could be of great importance to fully understand TGFß1 activation of genes.


Asunto(s)
Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Acetilación , Células Cultivadas , Histonas/metabolismo , Humanos , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad3/metabolismo , Factor de Transcripción Sp1/metabolismo
14.
Cell Signal ; 27(8): 1589-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25917318

RESUMEN

Smooth muscle-22α (SM22α), encoded by transgelin (TAGLN), is expressed in mesenchymal lineage cells, including myofibroblasts and smooth muscle cells. It is an F-actin binding protein that regulates the organization of actin cytoskeleton, cellular contractility and motility. SM22α is crucial for the maintenance of smooth muscle cell phenotype and its function. SM22α is also expressed in the processes of mesenchymal transition of epithelial (EMT) or endothelial cells (EndMT). The expression of TAGLN/SM22α is induced by transforming growth factor-ß (TGFß) signaling and enhanced by concomitant interleukin-1ß (IL-1ß) signaling. We investigated the epigenetic regulation of TAGLN expression by enhancer of zeste homolog-2 (EZH2), the methyltransferase of Polycomb, in the context of TGFß and IL-1ß signaling in endothelial cells. We demonstrate that the expression of EZH2 in endothelial cells was regulated by the inflammatory cytokine IL-1ß. A decrease in both expression and activity of EZH2 led to an increase in TAGLN expression. Inhibition of EZH2 augmented TGFß2-induced SM22α expression. The decrease of EZH2 levels in endothelial cells co-stimulated with IL-1ß and TGFß2 correlated with decreased H3K27me3 levels at the TAGLN proximal promoter. Moreover, the SM22α expression increased. Taken together, this suggests that EZH2 regulates the chromatin structure at the TAGLN promoter through tri-methylation of H3K27. EZH2 therefore acts as an epigenetic integrator of IL-1ß and TGFß2 signaling, providing an example of how cellular signaling can be resolved at the level of epigenetic regulation. Since IL-1ß and TGFß2 represent the pro-inflammatory and pro-fibrotic conditions during vascular fibroproliferative disease, we surmise that EZH2, as the molecule that integrates their signaling, could also be a promising target for development of future therapy.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Interleucina-1beta/farmacología , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Factor de Crecimiento Transformador beta2/farmacología , Línea Celular Transformada , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación de la Expresión Génica , Histonas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , Regiones Promotoras Genéticas , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , Remodelación Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...