Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 320(2): H867-H880, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33356961

RESUMEN

Dysfunction of late endothelial progenitor cells (EPCs) has been suggested to be associated with hypertension. ß2-Adrenergic receptor (ß2AR) is a novel and key target for EPC homing. Here, we proposed that attenuated ß2AR signaling contributes to EPCs dysfunction, whereas enhanced ß2AR signaling restores EPCs' functions in hypertension. EPCs derived from hypertensive patients exhibited reduced cell number, impaired in vitro migratory and adhesion abilities, and impaired re-endothelialization after transplantation in nude mice with carotid artery injury. ß2AR expression of EPCs from hypertensive patients was markedly downregulated, whereas the phosphorylation of the p38 mitogen-activated protein kinase (p38-MAPK) was elevated. The cleaved caspase-3 levels were elevated in EPCs. The overexpression of ß2AR in EPCs from hypertensive patients inhibited p38-MAPK signaling, whereas it enhanced in vitro EPC proliferation, migration, and adhesion and in vivo re-endothelialization. The ß2AR-mediated effects were attenuated by treating the EPCs with a neutralizing monoclonal antibody against ß2AR, which could be partially antagonized by the p38-MAPK inhibitor SB203580. Moreover, shear stress stimulation, a classic nonpharmacological intervention, increased the phosphorylation levels of ß2AR and enhanced the in vitro and in vivo functions of EPCs from hypertensive patients. Collectively, the current investigation demonstrated that impaired ß2AR/p38-MAPK/caspase-3 signaling at least partially reduced the re-endothelialization capacity of EPCs from hypertensive patients. Restoration of ß2AR expression and shear stress treatment could improve their endothelial repair capacity by regulating the p38-MAPK/caspase-3 signaling pathway. The clinical significance of ß2AR in endothelium repair still requires further investigation.NEW & NOTEWORTHY Impaired ß2-adrenergic receptor (ß2AR) expression with an elevation of p38-MAPK/caspase-3 signaling at least partially contributes to the decline of re-endothelialization capacity of late endothelial progenitor cells (EPCs) from hypertensive patients. ß2AR gene transfer and shear stress treatment improve the late EPC-mediated enhancement of the re-endothelialization capacity in hypertensive patients through activating ß2AR/p38-MAPK/caspase-3 signaling. The present study is the first to reveal the potential molecular mechanism of the impaired endothelium-reparative capacity of late EPCs in hypertension after vascular injury and strongly suggests that ß2AR is a novel and crucial therapeutic target for increasing EPC-mediated re-endothelialization capacity in hypertension.


Asunto(s)
Traumatismos de las Arterias Carótidas/prevención & control , Proliferación Celular , Células Progenitoras Endoteliales/metabolismo , Hipertensión/metabolismo , Repitelización , Receptores Adrenérgicos beta 2/metabolismo , Animales , Apoptosis , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Estudios de Casos y Controles , Caspasa 3/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipertensión/patología , Masculino , Ratones Desnudos , Persona de Mediana Edad , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
J Recept Signal Transduct Res ; 41(1): 6-14, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32605511

RESUMEN

Acute myocardial infarction (AMI) represents a severe coronary heart disease with relatively high rate of mortality and usually can lead to the damage of the myocardial tissues. Reperfusion of the ischemic myocardial tissues can minimize AMI-induced damage. As far as we know, the molecular mechanisms underlying ischemia/reperfusion (I/R)-induced injury remains elusive. This study was undertaken to explore the role of miR-1247-3p in regulating myocardial I/R injury. The hypoxia/reoxygenation (H/R)-treated H9c2 cells showed a decreased cell viability and mitochondrial membrane potential with an increase in the apoptosis; furthermore, miR-1247-3p was down-regulated in these cells. MiR-1247-3p overexpression attenuated H/R-induced H9c2 cell injury; while miR-1247-3p knockdown in H9c2 cells exhibited similar effects being observed in H/R-treated cells. The bioinformatics prediction revealed Bcl-2-like protein 11 (BCL2L11) and caspase-2 were two potential targets for miR-1247-3p, and functional assays confirmed that miR-1247-3p targeted both BCL2L11 and caspase-2 3' untranslated regions, which lead to the repressed expression of these genes. Silencing of BCL2L11 and caspase-2 both, respectively, counteracted the H9c2 cell injury caused by H/R treatment. Moreover, BCL2L11 and caspase-2 overexpression, respectively, impaired the protective effects of miR-1247-3p overexpression on H/R-treated H9c2 cells. The data in the present investigation revealed that miR-1247-3p restoration exhibited protective effects on H/R-induced cardiomyocyte injury through targeting BCL2L11 and caspase-2, implying that miR-1247-3p along with caspase-2/BCL2L11 signaling may provide novel sight for a better understating of I/R-induced myocardial damage. The role of miR-1247-3p might be further confirmed in animal models and clinical studies.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Caspasa 2/genética , MicroARNs/genética , Miocardio/metabolismo , Daño por Reperfusión/genética , Animales , Apoptosis/genética , Hipoxia de la Célula/genética , Supervivencia Celular/genética , Regulación de la Expresión Génica/genética , Humanos , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Sustancias Protectoras/farmacología , Ratas , Daño por Reperfusión/patología , Transducción de Señal/genética
3.
J Bioenerg Biomembr ; 52(3): 155-164, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32291604

RESUMEN

Myocardial infarction represents one of the severe cardiovascular diseases and is one of the high-risk factors for mortality, and ischemia/reperfusion (I/R) injury is one of the risk factors that contribute to the high mortality of myocardial infarction. MicroRNAs have been proven as key regulators in various diseases including myocardial infarction. The present study was sought to determine the role of miR-703 in the myocardial I/R injury and to explore detailed mechanisms. Hypoxia/reoxygenation (H/R) treatment repressed cell viability, increased cytotoxicity and pyroptosis in mouse cardiomyocytes. More importantly, we found that miR-703 was suppressed in mouse cardiomyocytes upon H/R stimulation. Restoration of miR-703 expression in mouse cardiomyocytes counteracted the H/R-induced cytotoxicity and pyroptosis in mouse cardiomyocytes; and the effects of miR-703 inhibition on cell viability and pyroptosis were similar to that of H/R treatment in mouse cardiomyocytes. In a further investigation, we found that NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) was targeted and repressed by miR-703 in mouse cardiomyocytes. NLRP3 knockdown also attenuated H/R-induced cytotoxicity and pyroptosis in mouse cardiomyocytes. In the mechanistic perspective, NLRP3 enforced expression disrupted the protective effects of miR-703 restoration on the H/R-induced cytotoxicity and pyroptosis in mouse cardiomyocytes. Our results for the first time demonstrate the protective actions of miR-703 in the H/R-induced mouse cardiomyocyte injury. More importantly, miR-703 protects against H/R-induced cardiomyocyte injury via inhibiting the NLRP3/caspase-1-mediated pyroptosis.


Asunto(s)
Caspasa 1/metabolismo , MicroARNs/uso terapéutico , Miocitos Cardíacos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/fisiología , Animales , Hipoxia de la Célula , Ratones , Transfección
4.
Environ Sci Pollut Res Int ; 29(3): 3498-3509, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34389950

RESUMEN

This study examined the effects of Stmn2 on phenotype transformation of vascular smooth muscle in vascular injury via RNA sequencing and experimental validation. Total RNA was extracted for RNA sequencing after 1, 3 and 5 days of injury to screen the differentially expressed genes (DEGs). Western blot was used to detect the protein expression of Stmn2 and its associated targets. The morphological changes of carotid arteries in rats were examined by hematoxylin and eosin (H&E) staining. The expression of vascular smooth muscle cell (VSMC) phenotype markers smooth muscle alpha-actin (α-SMA), vimentin and OPN were detected by immunohistochemistry. DEGs were related to the extracellular matrix and other cell components outside the plasma membrane. They were associated with protein binding, cytoskeleton protein binding, signal receptor binding and other molecular functions, actin cytoskeleton regulation and other Kyoto Encyclopedia of Genes and Genomes pathways. Stmn2 was identified as the hub gene of actin cytoskeleton pathway and vascular disease, and its expression followed the trend of decreasing initially and increasing afterwards during the progress of vascular injury. Western blot assay showed that the expression of Stmn2 and Tubulin decreased immediately after vascular injury; Stmn2 overexpression significantly up-regulated the expression of osteopontin and α-SMA and vimentin in VSMCs. The results of morphology analysis and immunostaining also showed that Stmn2 overexpression promoted the intima thickening and enhanced the proliferating cell nuclear antigen expression in the injured vascular tissues. In conclusion, our results implied that Stmn2 may play a potential role in vascular injury, which may be associated with VSMC phenotype transformation. Further studies are warranted to determine detailed molecular mechanisms of Stmn2 in vascular injury.


Asunto(s)
Músculo Liso Vascular , Lesiones del Sistema Vascular , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Fenotipo , Ratas , Análisis de Secuencia de ARN , Lesiones del Sistema Vascular/genética
5.
Front Cell Dev Biol ; 9: 647763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34055778

RESUMEN

Myocardial infarction (MI) remains a leading cause of morbidity and mortality worldwide. Endothelial progenitor cell (EPC)-derived exosomes have been found to be effective in alleviating MI, while the detailed mechanisms remain unclear. The present study aimed to determine the protective effects of EPC-derived exosomal miR-1246 and miR-1290 on MI-induced injury and to explore the underlying molecular mechanisms. The exosomes were extracted from EPCs; gene expression levels were determined by quantitative real-time PCR, and protein expression levels were determined by western blot and immunofluorescence staining, respectively. The angiogenesis and proliferation of human cardiac fibroblasts (HCFs) were determined by tube formation assay and immunofluorescence staining of PKH67, respectively. Luciferase reporter, CHIP, and EMSA assays determined the interaction between miR-1246/1290 and the targeted genes (EFL5 and SP1). The protective effects of miR-1246/1290 on MI were evaluated in a rat model of MI. EPC-derived exosomes significantly upregulated miR-1246 and miR-1290 expression and promoted phenotypic changes of fibroblasts to endothelial cells, angiogenesis, and proliferation in HCFs. Exosomes from EPCs with miR-1246 or miR-1290 mimics transfection promoted phenotypic changes of fibroblasts to endothelial cells and angiogenesis in HCFs, while exosomes from EPCs with miR-1246 or miR-1290 knockdown showed opposite effects in HCFs. Mechanistically, miR-1246 and miR-1290 from EPC-derived exosomes induced upregulation of ELF5 and SP1, respectively, by targeting the promoter regions of corresponding genes. Overexpression of both ELF5 and SP1 enhanced phenotypic changes of fibroblasts to endothelial cells and angiogenesis in HCFs pretreated with exosomes from EPCs with miR-1246 or miR-1290 mimics transfection, while knockdown of both EFL5 and SP1 exerted the opposite effects in HCFs. Both ELF5 and SP1 can bind to the promoter of CD31, leading to the upregulation of CD31 in HCFs. Furthermore, in vivo animal studies showed that exosomes from EPCs with miR-1246 or miR-1290 overexpression attenuated the MI-induced cardiac injury in the rats and caused an increase in ELF5, SP1, and CD31 expression, respectively, but suppressed α-SMA expression in the cardiac tissues. In conclusion, our study revealed that miR-1246 and miR-1290 in EPC-derived exosomes enhanced in vitro and in vivo angiogenesis in MI, and these improvements may be associated with amelioration of cardiac injury and cardiac fibrosis after MI.

6.
Aging (Albany NY) ; 12(13): 12930-12942, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32611835

RESUMEN

This study evaluated the benefit of dual therapy in reducing ischemic events in atrial fibrillation (AF) patients presenting with acute coronary syndrome (ACS) or undergoing percutaneous coronary intervention (PCI). We searched PubMed, Cochrane Library, and ClinicalTrials.gov for randomized controlled trials (RCTs) comparing dual and triple therapies (oral anticoagulation plus aspirin and P2Y12 inhibitor) for AF patients with ACS or those undergoing PCI. The composite primary outcome included all-cause death, myocardial infarction (MI), stent thrombosis (ST), or stroke. Relative risk (RR) and the corresponding 95% confidence interval (CI) was used as the measure of effect size. Four RCTs with 10,969 patients were included. Dual therapy had a higher event rate of primary outcome than triple therapy (RR, 1.15; 95%CI, 1.03-1.28; P<0.0001). Dual therapy was associated with significantly higher MI risk, insignificantly higher ST risk, and significantly lower major bleeding risk than triple therapy (RR1.23, 95%CI 1.01-1.49, P = 0.036; RR 1.43, 95 %CI 0.98-2.09, P = 0.064; and RR0.58, 95%CI 0.45-0.76, P<0.0001, respectively). Dual antithrombotic therapy was associated with higher ischemic risk but lower major bleeding risk than triple therapy. The data suggest that antithrombotic regimens should be based on tradeoffs between ischemia and bleeding risk.


Asunto(s)
Síndrome Coronario Agudo , Fibrilación Atrial , Fibrinolíticos , Intervención Coronaria Percutánea , Anciano , Femenino , Fibrinolíticos/efectos adversos , Fibrinolíticos/uso terapéutico , Humanos , Masculino
7.
Biomed Pharmacother ; 113: 108773, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30889484

RESUMEN

OBJECTIVES: This study was to evaluate platelet reactivity over time among patients with chronic kidney disease (CKD) receiving standard dose of clopidogrel after percutaneous coronary intervention (PCI). The effect of CYP2C19 loss-of-function genotypes on platelet reactivity was also determined. METHODS: Patients with CKD (n = 138) on maintenance dose of clopidogrel after PCI were enrolled. Platelet reactivity was assessed by measuring P2Y12 reaction units (PRU) with VerifyNow P2Y12 assay, and platelet reactivity index (PRI) with flow cytometric using vasodilator-stimulated phosphoprotein (VASP) at baseline and 2 weeks later, respectively. The genotypes of CYP2C19 were also measured concurrently. RESULTS: The proportion of patients with high platelet reactivity (HPR) ranged from 23.2% to 59.4%, and almost 1 in 5 patients had a dual conversion between HPR and non-HPR status. Patients carrying CYP2C19 loss-of-function genotypes showed a higher platelet reactivity than non-carriers, but with an undetermined HPR status between the first and second visits. The individual switch of HPR to non-HPR status existed in both loss-of-function genotype carriers and non-carriers. CONCLUSIONS: HPR conversions occur in a significant proportion of CKD patients with maintenance doses of clopidogrel treatment post-PCI, and this conversion was not confined to CYP2C19 loss-of-function genotype carriers. Risk stratification for treatment adjustment in personalized antiplatelet therapy should be investigated in future research.


Asunto(s)
Plaquetas/efectos de los fármacos , Enfermedad de la Arteria Coronaria/terapia , Inhibidores de Agregación Plaquetaria/farmacología , Insuficiencia Renal Crónica/fisiopatología , Anciano , Plaquetas/metabolismo , Clopidogrel/farmacología , Citocromo P-450 CYP2C19/genética , Femenino , Citometría de Flujo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Intervención Coronaria Percutánea/métodos , Stents , Factores de Tiempo
8.
Endocrinology ; 143(3): 829-36, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11861504

RESUMEN

We and others demonstrated that the mRNAs encoding GATA-binding proteins, GATA-1 and GATA-4, were detected in mouse and rat testis, and in isolated rat Sertoli cells and testicular tumor cell lines derived from Leydig and Sertoli cells. In this study, we investigated the possible effects of gonadotropins and cAMP on the expression of GATA-binding protein genes in testicular cells. Unexpectedly, FSH negatively regulated GATA-1 (but not GATA-4) mRNA in a dose-dependent manner in primary cultures of rat Sertoli cells isolated from 21-d-old animals. GATA-1 mRNA was also negatively regulated by cAMP in a dose- and time-dependent manner in MA-10, a mouse Leydig tumor cell line. When 0.3 mM cAMP was administered to MA-10 cell cultures for 4 h, more than 95% of the GATA-1 mRNA and protein was abolished. The reduction of GATA-1 mRNA by cAMP can be mimicked by treatment with forskolin, which elevates intracellular cAMP levels. The inhibitory effect of cAMP was specific to the GATA-1 gene, given that GATA-4 and alpha-tubulin mRNA levels were not changed by any of the cAMP treatments. Inhibin alpha-subunit mRNA, on the other hand, was evidently increased by cAMP treatment in both MA-10 and Sertoli cells. However, inhibin alpha-subunit mRNA levels were elevated at 60-90 min before the suppression of GATA-1 mRNA detected. The inhibitory effect of cAMP on GATA-1 mRNA and protein was shown to be specific to testicular cells. The GATA-1 mRNA expressed in MEL, a mouse erythroid leukemia cell line, was not affected by cAMP. The reduction of GATA-1 mRNA by cAMP can be prevented when a translational inhibitor, cycloheximide, is added. In summary, we demonstrated that gonadotropins via cAMP negatively regulate the mRNA and protein levels of GATA-1, but not GATA-4, in testicular cells. The inhibitory effect on GATA-1 gene expression was specific to testicular cells and was not observed in erythroid cells.


Asunto(s)
AMP Cíclico/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Gonadotropinas/fisiología , Testículo/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Animales , Northern Blotting , Western Blotting , Núcleo Celular/metabolismo , Regulación hacia Abajo/fisiología , Células Precursoras Eritroides/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Tumor de Células de Leydig/metabolismo , Masculino , Ratones , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Neoplasias Testiculares/metabolismo , Testículo/citología
9.
J Androl ; 25(4): 479-93, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15223837

RESUMEN

In an attempt to identify new sperm-specific genes that are involved in sperm maturation, fertilization, and embryo development, such as the mammalian ortholog of the sperm-supplied protein gene, spe-11, in Caenorhabditis elegans, we cloned and characterized a new spermatid-specific protein gene, ssp411, from adult rat testes. The ssp411 cDNA shared >85% sequence identity with an unnamed human protein, FLJ21347, and an uncharacterized mouse testicular protein called transcript increased in spermiogenesis 78 (TISP78). A 2.8-kb ssp411 mRNA was expressed in a testis-specific and age-dependent manner; the mRNA was evident at 28 days and remained at high levels throughout adulthood. An SSP411 protein of molecular weight 88 000 was detected in testicular extracts by Western blot analysis. Ssp411 mRNA and SSP411 protein, as analyzed by in situ hybridization and immunohistochemistry, were both expressed in a stage-dependent fashion during the cycle of the seminiferous epithelium. The ssp411 mRNA was predominantly localized to round and elongated spermatids, with maximal expression at stages VII-XII. The SSP411 protein was mainly observed in elongated spermatids and reached its highest levels during stages V-VI. A conserved thioredoxin-like domain was detected in the N-terminal region of SSP411 and its orthologs. An analysis of the predicted 3-dimensional structural modeling and folding pattern further suggested that SSP411 is identifiable as a member of thioredoxin family. In summary, we have identified a new rat spermatid protein gene, ssp411, and its orthologs in human and mouse and demonstrated that SSP411 might belong to a testis-specific thioredoxin family. This suggests that SSP411 may play a role in sperm maturation, fertilization, and/or embryo development, as has been shown in thioredoxin family.


Asunto(s)
Espermátides/fisiología , Tiorredoxinas/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Secuencia de Consenso , Humanos , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Tiorredoxinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA