Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther Nucleic Acids ; 34: 102071, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38046397

RESUMEN

Heart failure is a leading cause of death and is often accompanied by activation of quiescent cardiac myofibroblasts, which results in cardiac fibrosis. In this study, we aimed to identify novel circular RNAs that regulate cardiac fibrosis. We applied transverse aortic constriction (TAC) for 1, 4, and 8 weeks in mice. RNA sequencing datasets were obtained from cardiac fibroblasts isolated by use of a Langendorff apparatus and then further processed by use of selection criteria such as differential expression and conservation in species. CircSMAD4 was upregulated by TAC in mice or by transforming growth factor (TGF)-ß1 in primarily cultured human cardiac fibroblasts. Delivery of si-circSMAD4 attenuated myofibroblast activation and cardiac fibrosis in mice treated with isoproterenol (ISP). si-circSmad4 significantly reduced cardiac fibrosis and remodeling at 8 weeks. Mechanistically, circSMAD4 acted as a sponge against the microRNA miR-671-5p in a sequence-specific manner. miR-671-5p was downregulated during myofibroblast activation and its mimic form attenuated cardiac fibrosis. miR-671-5p mimic destabilized fibroblast growth factor receptor 2 (FGFR2) mRNA in a sequence-specific manner and interfered with the fibrotic action of FGFR2. The circSMAD4-miR-671-5p-FGFR2 pathway is involved in the differentiation of cardiac myofibroblasts and thereby the development of cardiac fibrosis.

2.
Mol Ther Nucleic Acids ; 27: 645-655, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35036071

RESUMEN

Vascular calcification (VC), or calcium deposition inside the blood vessels, is common in patients with atherosclerosis, cardiovascular disease, and chronic kidney disease. Although several treatments are available to reduce calcification, the incidence of VC continues to rise. Recently, there have been several reports describing the regulation of circular RNAs (circRNAs) in various diseases. However, the role of circRNAs in VC has not yet been fully explored. Here, we investigated the function of circSmoc1-2, one of the circRNAs generated from the Smoc1 gene, which is downregulated in response to VC. CircSmoc1-2 is localized primarily to the cytoplasm and is resistant to exonuclease digestion. Inhibition of circSmoc1-2 worsens VC, while overexpression of circSmoc1-2 reduces VC, suggesting that circSmoc1-2 can prevent calcification. We went on to investigate the mechanism of circSmoc1-2 as a microRNA sponge and noted that miR-874-3p, the predicted target of circSmoc1-2, promotes VC, while overexpression of circSmoc1-2 reduces VC by suppressing miR-874-3p. Additionally, we identified the potential mRNA target of miR-874-3p as Adam19. In conclusion, we revealed that the circSmoc1-2/miR-874-3p/Adam19 axis regulates VC, suggesting that circSmoc1-2 may be a novel therapeutic target in the treatment of VC.

3.
Exp Mol Med ; 53(11): 1781-1791, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34845330

RESUMEN

Vascular calcification increases morbidity and mortality in patients with cardiovascular and renal diseases. Previously, we reported that histone deacetylase 1 prevents vascular calcification, whereas its E3 ligase, mouse double minute 2 homolog (MDM2), induces vascular calcification. In the present study, we identified the upstream regulator of MDM2. By utilizing cellular models and transgenic mice, we confirmed that E3 ligase activity is required for vascular calcification. By promoter analysis, we found that both msh homeobox 1 (Msx1) and msh homeobox 2 (Msx2) bound to the MDM2 promoter region, which resulted in transcriptional activation of MDM2. The expression levels of both Msx1 and Msx2 were increased in mouse models of vascular calcification and in calcified human coronary arteries. Msx1 and Msx2 potentiated vascular calcification in cellular and mouse models in an MDM2-dependent manner. Our results establish a novel role for MSX1/MSX2 in the transcriptional activation of MDM2 and the resultant increase in MDM2 E3 ligase activity during vascular calcification.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Transcripción MSX1/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Ubiquitina-Proteína Ligasas/genética , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Animales , Biomarcadores , Calcio/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Elementos de Respuesta , Ubiquitina-Proteína Ligasas/metabolismo , Calcificación Vascular/patología
4.
Mol Ther Nucleic Acids ; 22: 627-639, 2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33230462

RESUMEN

Vascular calcification, the ectopic deposition of calcium in blood vessels, develops in association with various metabolic diseases and atherosclerosis and is an independent predictor of morbidity and mortality associated with these diseases. Herein, we report that reduction of microRNA-27a-3p (miR-27a-3p) causes an increase in activating transcription factor 3 (ATF3), a novel osteogenic transcription factor, in vascular smooth muscle cells. Both microRNA (miRNA) and mRNA microarrays were performed with rat vascular smooth muscle cells, and reciprocally regulated pairs of miRNA and mRNA were selected after bioinformatics analysis. Inorganic phosphate significantly reduced the expression of miR-27a-3p in A10 cells. The transcript level was also reduced in vitamin D3-administered mouse aortas. miR-27a-3p mimic reduced calcium deposition, whereas miR-27a-3p inhibitor increased it. The Atf3 mRNA level was upregulated in a cellular vascular calcification model, and miR-27a-3p reduced the Atf3 mRNA and protein levels. Transfection with Atf3 could recover the miR-27a-3p-induced reduction of calcium deposition. Our results suggest that reduction of miR-27a-3p may contribute to the development of vascular calcification by de-repression of ATF3.

5.
J Cell Mol Med ; 24(18): 10542-10550, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32783377

RESUMEN

Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.


Asunto(s)
Calcio/metabolismo , Histona Desacetilasas/efectos de los fármacos , MicroARNs/fisiología , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/etiología , Regiones no Traducidas 3' , Animales , Aorta Torácica/citología , Línea Celular , Simulación por Computador , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/inmunología , Regulación hacia Abajo , Regulación de la Expresión Génica , Genes Reporteros , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , MicroARNs/genética , Análisis por Micromatrices , Músculo Liso Vascular/citología , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , Fosfatos/toxicidad , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/prevención & control
6.
Mol Ther Nucleic Acids ; 19: 31-41, 2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-31790973

RESUMEN

Circular RNAs (circRNAs) are generally formed by back splicing and are expressed in various cells. Vascular calcification (VC), a common complication of chronic kidney disease (CKD), is often associated with cardiovascular disease. The relationship between circRNAs and VC has not yet been studied. Inorganic phosphate (Pi) was used to treat rat vascular smooth muscle cells to induce VC. circRNAs were identified by analyzing RNA sequencing (RNA-seq) data, and their expression change during VC was validated. The selected circRNAs, including circSamd4a, circSmoc1-1, circMettl9, and circUxs1, were resistant to RNase R digestion and mostly localized in the cytoplasm. While silencing circSamd4a promoted VC, overexpressing it reduced VC in calcium assay and Alizarin red S (ARS) staining. In addition, microRNA (miRNA) microarray, luciferase reporter assay, and calcium assay suggested that circSamd4a could act as a miRNA suppressor. Our data show that circSamd4a has an anti-calcification role by functioning as a miRNA sponge. Moreover, mRNAs that can interact with miRNAs were predicted from RNA-seq and bioinformatics analysis, and the circSamd4a-miRNA-mRNA axis involved in VC was verified by luciferase reporter assay and calcium assay. Since circSamd4a is conserved in humans, it can serve as a novel therapeutic target in resolving VC.

7.
Sci Rep ; 9(1): 5848, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971745

RESUMEN

Vascular calcification is characterized by the accumulation of hydroxyapatite crystals, which is a result of aberrant mineral metabolism. Although many clinical studies have reported its adverse effects on cardiovascular morbidity, the molecular mechanism of vascular calcification, especially the involvement of long noncoding RNAs (lncRNAs), is not yet reported. From the transcriptomic analysis, we discovered hundreds of lncRNAs differentially expressed in rat vascular smooth muscle cells (VSMCs) treated with inorganic phosphate, which mimics vascular calcification. We focused on Lrrc75a-as1 and elucidated its transcript structure and confirmed its cytoplasmic localization. Our results showed that calcium deposition was elevated after knockdown of Lrrc75a-as1, while its overexpression inhibited calcium accumulation in A10 cells. In addition, Lrrc75a-as1 attenuated VSMCs calcification by decreasing the expression of osteoblast-related factors. These findings suggest that Lrrc75a-as1 acts as a negative regulator of vascular calcification, and may serve as a possible therapeutic target in vascular calcification.


Asunto(s)
ARN Largo no Codificante/metabolismo , Calcificación Vascular/patología , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Calcio/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Redes Reguladoras de Genes , Humanos , MicroARNs/química , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Ratas , Alineación de Secuencia , Calcificación Vascular/genética
8.
Exp Mol Med ; 50(1): e427, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29328071

RESUMEN

Sumoylation, the conjugation of a small ubiquitin-like modifier (SUMO) protein to a target, has diverse cellular effects. However, the functional roles of the SUMO modification during myogenesis have not been fully elucidated. Here, we report that basal sumoylation of histone deacetylase 1 (HDAC1) enhances the deacetylation of MyoD in undifferentiated myoblasts, whereas further sumoylation of HDAC1 contributes to switching its binding partners from MyoD to Rb to induce myocyte differentiation. Differentiation in C2C12 skeletal myoblasts induced new immunoblot bands above HDAC1 that were gradually enhanced during differentiation. Using SUMO inhibitors and sumoylation assays, we showed that the upper band was caused by sumoylation of HDAC1 during differentiation. Basal deacetylase activity was not altered in the SUMO modification-resistant mutant HDAC1 K444/476R (HDAC1 2R). Either differentiation or transfection of SUMO1 increased HDAC1 activity that was attenuated in HDAC1 2R. Furthermore, HDAC1 2R failed to deacetylate MyoD. Binding of HDAC1 to MyoD was attenuated by K444/476R. Binding of HDAC1 to MyoD was gradually reduced after 2 days of differentiation. Transfection of SUMO1 induced dissociation of HDAC1 from MyoD but potentiated its binding to Rb. SUMO1 transfection further attenuated HDAC1-induced inhibition of muscle creatine kinase luciferase activity that was reversed in HDAC1 2R. HDAC1 2R failed to inhibit myogenesis and muscle gene expression. In conclusion, HDAC1 sumoylation plays a dual role in MyoD signaling: enhancement of HDAC1 deacetylation of MyoD in the basally sumoylated state of undifferentiated myoblasts and dissociation of HDAC1 from MyoD during myogenesis.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Proteína MioD/metabolismo , Mioblastos/metabolismo , Acetilación , Animales , Diferenciación Celular/fisiología , Línea Celular , Histona Desacetilasa 1/genética , Ratones , Desarrollo de Músculos , Músculo Esquelético/citología , Miogenina/genética , Regiones Promotoras Genéticas , Transducción de Señal , Sumoilación
9.
FEBS Lett ; 591(7): 1041-1052, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28235243

RESUMEN

S100 calcium-binding protein A4 (S100A4) induces proliferation and migration of vascular smooth muscle cells (VSMCs). We aimed to find the microRNA regulating S100A4 expression. S100A4 transcripts are abruptly increased in the acute phase of carotid arterial injury 1 day later (at day 1) but gradually decreases at days 7 and 14. Bioinformatics analysis reveals that miR-124 targets S100A4. VSMC survival is attenuated by miR-124 mimic but increased by miR-124 inhibitor. miR-124 decreases immediately after carotid arterial injury but dramatically increases at days 7 and 14. miR-124 inhibitor-induced cell proliferation is blocked by S100A4 siRNA, whereas miR-124-induced cell death is recovered by S100A4. Our findings suggest that miR-124 is a novel regulator of VSMC proliferation and may play a role in the development of neointimal proliferation.


Asunto(s)
Proliferación Celular/genética , Regulación de la Expresión Génica , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Proteína de Unión al Calcio S100A4/genética , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Western Blotting , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Línea Celular , Inmunohistoquímica , Masculino , Músculo Liso Vascular/citología , Neointima/genética , Neointima/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión al Calcio S100A4/metabolismo , Homología de Secuencia de Aminoácido , Factores de Tiempo
10.
Nat Commun ; 7: 10492, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26832969

RESUMEN

Vascular calcification (VC) is often associated with cardiovascular and metabolic diseases. However, the molecular mechanisms linking VC to these diseases have yet to be elucidated. Here we report that MDM2-induced ubiquitination of histone deacetylase 1 (HDAC1) mediates VC. Loss of HDAC1 activity via either chemical inhibitor or genetic ablation enhances VC. HDAC1 protein, but not mRNA, is reduced in cell and animal calcification models and in human calcified coronary artery. Under calcification-inducing conditions, proteasomal degradation of HDAC1 precedes VC and it is mediated by MDM2 E3 ubiquitin ligase that initiates HDAC1 K74 ubiquitination. Overexpression of MDM2 enhances VC, whereas loss of MDM2 blunts it. Decoy peptide spanning HDAC1 K74 and RG 7112, an MDM2 inhibitor, prevent VC in vivo and in vitro. These results uncover a previously unappreciated ubiquitination pathway and suggest MDM2-mediated HDAC1 ubiquitination as a new therapeutic target in VC.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Calcificación Vascular/metabolismo , Animales , Calcio , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Humanos , Masculino , Ratones , Músculo Liso Vascular/citología , Proteínas Proto-Oncogénicas c-mdm2/genética , Ratas , Ubiquitinación
11.
Cell Signal ; 27(11): 2241-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26232617

RESUMEN

High glucose-insulted bone marrow-derived mesenchymal stem cells (BMCs) showed impaired angiogenesis along with downregulation of stem cell factor (SCF). This study was designed to determine the involvement of microRNAs (miR), which are actively involved in the physiological function of stem cells. We observed that miR-34c was significantly induced by high glucose treatment and blunted tube formation of BMCs. Stem cell factor (SCF) was confirmed as a target of miR-34c by 3'-UTR promoter analysis and Western blot. SCF knockdown by siRNA induced Krüppel-like factor 4 (KLF4) and resulted in the blockade of angiogenesis of BMCs. Sequentially, KLF4 overexpression completely blocked tube formation through inducing PAI-1 (plasminogen activator inhibitor-1). To study the action of miR-34c in terms of the therapeutic potential of BMCs, myocardial infarction (MI) was induced by ligation of the coronary artery in nude mice, BMCs transfected with miR-control or miR-34c were injected into the infarcted myocardium 7 days later, and histological studies were performed 2 weeks later. Cardiac fibrosis was 18.24±4.7% in the miR-34c-BMC group and 10.01±0.2% in the miR-control-BMC group (p<0.05). Cardiac function and vessel density were decreased in the miR-34c-BMC group compared with the miR-con-BMC group. Particularly, miR-34c-BMCs failed to incorporate into vessels. Our results show that the angiogenic activity of BMCs is finely regulated by the miR-34c-SCF-KLF4 axis, which is a potent translational target for optimizing the therapeutic activity of autologous BMCs for cardiac repair.


Asunto(s)
Glucosa/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/genética , Infarto del Miocardio/terapia , Factor de Células Madre/genética , Animales , Células de la Médula Ósea/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Vasos Coronarios/cirugía , Regulación hacia Abajo , Fibrosis/patología , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/biosíntesis , Infarto del Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factor de Células Madre/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Cell Signal ; 27(6): 1056-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683915

RESUMEN

The fine balance between proliferation and differentiation of vascular smooth muscle cells (VSMCs) is indispensable for the maintenance of healthy blood vessels, whereas an increase in proliferation participates in pathologic cardiovascular events such as atherosclerosis and restenosis. Here we report that microRNA-34c (miR-34c) targets stem cell factor (SCF) to inhibit VSMC proliferation and neointimal hyperplasia. In an animal model, miR-34c was significantly increased in the rat carotid artery after catheter injury. Transient transfection of miR-34c to either VSMCs or A10 cells inhibited cell survival by inducing apoptosis, which was accompanied by an increase in expression of p21, p27, and Bax. Transfection of miR-34c also attenuated VSMC migration. Bioinformatics showed that SCF is a target candidate of miR-34c. miR-34c down-regulated luciferase activity driven by a vector containing the 3'-untranslated region of SCF in a sequence-specific manner. Forced expression of SCF in A10 cells induced proliferation and migration, whereas knocking-down of SCF reduced cell survival and migration. miR-34c antagomir-induced VSMC proliferation was blocked by SCF siRNA. Delivery of miR-34c to rat carotid artery attenuated the expression of SCF and blocked neointimal hyperplasia. These results suggest that miR-34c is a new modulator of VSMC proliferation and that it inhibits neointima formation by regulating SCF.


Asunto(s)
Arterias Carótidas/patología , MicroARNs/metabolismo , Factor de Células Madre/metabolismo , Regiones no Traducidas 3' , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Secuencia de Bases , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Hiperplasia , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima , Oligonucleótidos Antisentido/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Alineación de Secuencia , Factor de Células Madre/antagonistas & inhibidores , Factor de Células Madre/genética
13.
Cell Signal ; 26(10): 2240-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25025573

RESUMEN

Skeletal muscle atrophy results from the net loss of muscular proteins and organelles and is caused by pathologic conditions such as nerve injury, immobilization, cancer, and other metabolic diseases. Recently, ubiquitination-mediated degradation of skeletal-muscle-specific transcription factors was shown to be involved in muscle atrophy, although the mechanisms have yet to be defined. Here we report that ret finger protein (RFP), also known as TRIM27, works as an E3 ligase in Pax7-induced degradation of MyoD. Muscle injury induced by sciatic nerve transection up-regulated RFP and RFP physically interacted with both Pax7 and MyoD. RFP and Pax7 synergistically reduced the protein amounts of MyoD but not the mRNA. RFP-induced reduction of MyoD protein was blocked by proteasome inhibitors. The Pax7-induced reduction MyoD was attenuated by RFP siRNA and by MG132, a proteasome inhibitor. RFPΔR, an RFP construct that lacks the RING domain, failed to reduce MyoD amounts. RFP ubiquitinated MyoD, but RFPΔR failed to do so. Forced expression of RFP, but not RFPΔR, enhanced Pax7-induced ubiquitination of MyoD, whereas RFP siRNA blocked the ubiquitination. Sciatic nerve injury-induced muscle atrophy as well the reduction in MyoD was attenuated in RFP knockout mice. Taken together, our results show that RFP works as a novel E3 ligase in the Pax7-mediated degradation of MyoD in response to skeletal muscle atrophy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Atrofia Muscular/patología , Proteína MioD/metabolismo , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX7/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Leupeptinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Atrofia Muscular/metabolismo , Proteína MioD/química , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Factor de Transcripción PAX7/química , Inhibidores de Proteasas/farmacología , Unión Proteica , Proteolisis/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Regeneración , Ubiquitina-Proteína Ligasas , Ubiquitinación/efectos de los fármacos
14.
Circ Res ; 115(5): 493-503, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015078

RESUMEN

RATIONALE: Small heterodimer partner (SHP; NR0B2) is an atypical orphan nuclear receptor that lacks a conventional DNA-binding domain. Through interactions with other transcription factors, SHP regulates diverse biological events, including glucose metabolism in liver. However, the role of SHP in adult heart diseases has not yet been demonstrated. OBJECTIVE: We aimed to investigate the role of SHP in adult heart in association with cardiac hypertrophy. METHODS AND RESULTS: The roles of SHP in cardiac hypertrophy were tested in primary cultured cardiomyocytes and in animal models. SHP-null mice showed a hypertrophic phenotype. Hypertrophic stresses repressed the expression of SHP, whereas forced expression of SHP blocked the development of hypertrophy in cardiomyocytes. SHP reduced the protein amount of Gata6 and, by direct physical interaction with Gata6, interfered with the binding of Gata6 to GATA-binding elements in the promoter regions of natriuretic peptide precursor type A. Metformin, an antidiabetic agent, induced SHP and suppressed cardiac hypertrophy. The metformin-induced antihypertrophic effect was attenuated either by SHP small interfering RNA in cardiomyocytes or in SHP-null mice. CONCLUSIONS: These results establish SHP as a novel antihypertrophic regulator that acts by interfering with GATA6 signaling. SHP may participate in the metformin-induced antihypertrophic response.


Asunto(s)
Cardiomegalia/prevención & control , Factor de Transcripción GATA6/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Sitios de Unión , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Modelos Animales de Enfermedad , Factor de Transcripción GATA6/genética , Regulación de la Expresión Génica , Genotipo , Células HEK293 , Humanos , Masculino , Metformina/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenotipo , Regiones Promotoras Genéticas , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de los fármacos , Transfección
15.
Korean Circ J ; 44(3): 177-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24876859

RESUMEN

BACKGROUND AND OBJECTIVES: Diabetes is reported to reduce the function or number of progenitor cells. We compared the gene expression patterns of bone marrow-derived mesenchymal stem cells from diabetic (DM-BMCs) and healthy (non-DM-BMCs) rats and suggested Angiopoietin-like 4 (Angptl4) could be a responsible factor for impaired angiogenesis of DM-BMCs. SUBJECTS AND METHODS: BMCs were isolated from DM or non-DM rat, and in vitro angiogenesis activity was compared by tube formation assay on Matrigel and complementary deoxyribonucleic acid expression was analyzed by microarray with or without oxytocin treatment. Human BMCs (hBMCs) were treated with high glucose, and were performed polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay. Angptl4 plasmid DNA and micro ribonucleic acid-132 (miR-132) were transfected to immortalized hBMCs. RESULTS: In vitro angiogenesis assay showed the impaired tube formation in DM-BMCs, and slightly recovery by oxytocin treatment. Angptl4, an adipokine, was upregulated in DM-BMCs compared to non-DM-BMCs. Oxytocin treatment reduced Angptl4 in DM-BMCs. In hBMCs, overexpression of Angptl4 attenuated the tube formation. In addition to Angptl4, miR-132 was increased by high glucose treatment. Collectively, high glucose resulted in impaired tube formation through miR-132 induction and Angptl4 upregulation in BMCs. CONCLUSION: Our results show that the angiogenic activity of BMCs is impaired by high glucose stress, which would be mediated by Angptl4 and miR-132.

16.
Circ Res ; 114(7): 1133-43, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24526703

RESUMEN

RATIONALE: Histone deacetylases (HDACs) are closely involved in cardiac reprogramming. Although the functional roles of class I and class IIa HDACs are well established, the significance of interclass crosstalk in the development of cardiac hypertrophy remains unclear. OBJECTIVE: Recently, we suggested that casein kinase 2α1-dependent phosphorylation of HDAC2 leads to enzymatic activation, which in turn induces cardiac hypertrophy. Here we report an alternative post-translational activation mechanism of HDAC2 that involves acetylation of HDAC2 mediated by p300/CBP-associated factor/HDAC5. METHODS AND RESULTS: Hdac2 was acetylated in response to hypertrophic stresses in both cardiomyocytes and a mouse model. Acetylation was reduced by a histone acetyltransferase inhibitor but was increased by a nonspecific HDAC inhibitor. The enzymatic activity of Hdac2 was positively correlated with its acetylation status. p300/CBP-associated factor bound to Hdac2 and induced acetylation. The HDAC2 K75 residue was responsible for hypertrophic stress-induced acetylation. The acetylation-resistant Hdac2 K75R showed a significant decrease in phosphorylation on S394, which led to the loss of intrinsic activity. Hdac5, one of class IIa HDACs, directly deacetylated Hdac2. Acetylation of Hdac2 was increased in Hdac5-null mice. When an acetylation-mimicking mutant of Hdac2 was infected into cardiomyocytes, the antihypertrophic effect of either nuclear tethering of Hdac5 with leptomycin B or Hdac5 overexpression was reduced. CONCLUSIONS: Taken together, our results suggest a novel mechanism by which the balance of HDAC2 acetylation is regulated by p300/CBP-associated factor and HDAC5 in the development of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Histona Desacetilasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Ratones , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Factores de Transcripción p300-CBP/genética
17.
Atherosclerosis ; 229(2): 348-55, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23880186

RESUMEN

OBJECTIVE: The proliferation and remodeling of vascular smooth muscle cells (VSMCs) is an important pathological event in atherosclerosis and restenosis. Here we report that microRNA-132 (miR-132) blocks vascular smooth muscle cells (VSMC) proliferation by inhibiting the expression of LRRFIP1 [leucine-rich repeat (in Flightless 1) interacting protein-1]. METHODS AND RESULTS: MicroRNA microarray revealed that miR-132 was upregulated in the rat carotid artery after catheter injury, which was further confirmed by quantitative real-time RT-PCR. Transfection of a miR-132 mimic significantly inhibited the proliferation of VSMCs, whereas transfection of a miR-132 antagomir increased it. miR-132 mimic inhibited VSMC migration and induced apoptosis. miR-132 mimic increased the protein amounts of both p27 and smooth muscle (SM) α-actin, whereas it decreased SM α-actin and Bcl2. Bioinformatics showed that LRRFIP1 is a target candidate of miR-132. miR-132 down-regulated luciferase activity driven by a vector containing the 3'-untranslated region of Lrrfip1 in a sequence-specific manner. LRRFIP1 induced VSMC proliferation and increased phosphorylation of ERK. Immunohistochemical analysis revealed that Lrrfip1 was clearly expressed along with the basal laminar area of smooth muscle, and its expression pattern was disrupted 7 days after arterial injury. LRRFIP1 mRNA was decreased 14 days after injury. Delivery of miR-132 to rat carotid artery reduced LRRFIP1 expression and attenuated neointimal proliferation in carotid artery injury models. CONCLUSIONS: Our results suggest that miR-132 is a novel regulator of VSMC proliferation that represses neointimal formation by inhibiting LRRFIP1 expression.


Asunto(s)
Aterosclerosis/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Proteínas de Unión al ARN/metabolismo , Actinas/metabolismo , Animales , Apoptosis/fisiología , Aterosclerosis/patología , Secuencia de Bases , Cateterismo/efectos adversos , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Hiperplasia/metabolismo , Hiperplasia/patología , Masculino , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Neointima/patología , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley
18.
FEBS Lett ; 587(15): 2385-92, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23770100

RESUMEN

Vascular smooth muscle cell (VSMC) proliferation plays a key role in neointimal hyperplasia and restenosis. Here we report the role of the microRNA miR-142-5p and its downstream target genes on the proliferation of cultured VSMCs. miR-142-5p promoted VSMC proliferation by down-regulating B cell translocation gene 3 (BTG3). We found that BTG3 inhibited the expression of cell cycle regulatory genes and cell growth. As shown by luciferase reporter assay, miR-142-5p bound directly to the 3'-untranslated region of BTG3. Overexpression of miR-142-5p induced expression of cell cycle regulatory genes. Thus, BTG3, a novel, direct target of miR-142-5p, negatively regulates VSMC proliferation.


Asunto(s)
Ciclo Celular/fisiología , Proliferación Celular , Regulación hacia Abajo , MicroARNs/genética , Músculo Liso Vascular/citología , Proteínas/fisiología , Animales , Células Cultivadas , Masculino , Proteínas/genética , Ratas , Ratas Sprague-Dawley
19.
Mol Cell Biol ; 32(14): 2917-33, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22615488

RESUMEN

Histone lysine methylation and demethylation are considered critical steps in transcriptional regulation. In this report, we performed chromatin immunoprecipitation with microarray technology (ChIP-chip) analysis to examine the genome-wide occupancy of H3K9-me2 during all-trans-retinoic acid (ATRA)-induced differentiation of HL-60 promyelocytic leukemia cells. Using this approach, we found that KDM3B, which contains a JmjC domain, was downregulated during differentiation through the recruitment of a corepressor complex. Furthermore, KDM3B displayed histone H3K9-me1/2 demethylase activity and induced leukemogenic oncogene lmo2 expression via a synergistic interaction with CBP. Here, we found that KDM3B repressed leukemia cell differentiation and was upregulated in blood cells from acute lymphoblastic leukemia (ALL)-type leukemia patients. The combined results of this study provide evidence that the H3K9-me1/2 demethylase KDM3B might play a role in leukemogenesis via activation of lmo2 through interdependent actions with the histone acetyltransferase (HAT) complex containing CBP.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas con Dominio LIM/genética , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Proteínas Proto-Oncogénicas/genética , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , ADN de Neoplasias/genética , Regulación Leucémica de la Expresión Génica , Células HEK293 , Células HL-60 , Histona Acetiltransferasas/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Células K562 , Leucemia Promielocítica Aguda/patología , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncogenes , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Activación Transcripcional , Tretinoina/farmacología
20.
J Biol Chem ; 286(40): 34733-42, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21832073

RESUMEN

Histone lysine methylation, as one of the most important factors in transcriptional regulation, is associated with a various physiological conditions. Using a bioinformatics search, we identified and subsequently cloned mouse SET domain containing 3 (SETD3) with SET (Su(var)3-9, Enhancer-of-zeste and Trithorax) and Rubis-subs-bind domains. SETD3 is a novel histone H3K4 and H3K36 methyltransferase with transcriptional activation activity. SETD3 is expressed abundantly in muscular tissues and, when overexpressed, activates transcription of muscle-related genes, myogenin, muscle creatine kinase (MCK), and myogenic factor 6 (Myf6), thereby inducing muscle cell differentiation. Conversely, knockdown of SETD3 by shRNA significantly retards muscle cell differentiation. In this study, SETD3 was recruited to the myogenin gene promoter along with MyoD where it activated transcription. Together, these data indicate that SETD3 is a H3K4/K36 methyltransferase and plays an important role in the transcriptional regulation of muscle cell differentiation.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/fisiología , Músculos/metabolismo , Animales , Diferenciación Celular , Cromatina/química , Biología Computacional/métodos , Regulación de la Expresión Génica , Histona Metiltransferasas , Histonas/química , Ratones , Miogenina/química , Plásmidos/metabolismo , Conformación Proteica , Ratas , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...