Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 150(2): 132-150, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38557054

RESUMEN

BACKGROUND: An imbalance of antiproliferative BMP (bone morphogenetic protein) signaling and proliferative TGF-ß (transforming growth factor-ß) signaling is implicated in the development of pulmonary arterial hypertension (PAH). The posttranslational modification (eg, phosphorylation and ubiquitination) of TGF-ß family receptors, including BMPR2 (bone morphogenetic protein type 2 receptor)/ALK2 (activin receptor-like kinase-2) and TGF-ßR2/R1, and receptor-regulated Smads significantly affects their activity and thus regulates the target cell fate. BRCC3 modifies the activity and stability of its substrate proteins through K63-dependent deubiquitination. By modulating the posttranslational modifications of the BMP/TGF-ß-PPARγ pathway, BRCC3 may play a role in pulmonary vascular remodeling, hence the pathogenesis of PAH. METHODS: Bioinformatic analyses were used to explore the mechanism by which BRCC3 deubiquitinates ALK2. Cultured pulmonary artery smooth muscle cells (PASMCs), mouse models, and specimens from patients with idiopathic PAH were used to investigate the rebalance between BMP and TGF-ß signaling in regulating ALK2 phosphorylation and ubiquitination in the context of pulmonary hypertension. RESULTS: BRCC3 was significantly downregulated in PASMCs from patients with PAH and animals with experimental pulmonary hypertension. BRCC3, by de-ubiquitinating ALK2 at Lys-472 and Lys-475, activated receptor-regulated Smad1/5/9, which resulted in transcriptional activation of BMP-regulated PPARγ, p53, and Id1. Overexpression of BRCC3 also attenuated TGF-ß signaling by downregulating TGF-ß expression and inhibiting phosphorylation of Smad3. Experiments in vitro indicated that overexpression of BRCC3 or the de-ubiquitin-mimetic ALK2-K472/475R attenuated PASMC proliferation and migration and enhanced PASMC apoptosis. In SM22α-BRCC3-Tg mice, pulmonary hypertension was ameliorated because of activation of the ALK2-Smad1/5-PPARγ axis in PASMCs. In contrast, Brcc3-/- mice showed increased susceptibility of experimental pulmonary hypertension because of inhibition of the ALK2-Smad1/5 signaling. CONCLUSIONS: These results suggest a pivotal role of BRCC3 in sustaining pulmonary vascular homeostasis by maintaining the integrity of the BMP signaling (ie, the ALK2-Smad1/5-PPARγ axis) while suppressing TGF-ß signaling in PASMCs. Such rebalance of BMP/TGF-ß pathways is translationally important for PAH alleviation.


Asunto(s)
Hipertensión Pulmonar , Músculo Liso Vascular , Miocitos del Músculo Liso , Animales , Humanos , Masculino , Ratones , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , PPAR gamma/metabolismo , PPAR gamma/genética , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/patología , Hipertensión Arterial Pulmonar/genética , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Transducción de Señal , Ubiquitinación , Remodelación Vascular
2.
BMC Cardiovasc Disord ; 24(1): 349, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987688

RESUMEN

PURPOSE: Glycolysis and immune metabolism play important roles in acute myocardial infarction (AMI). Therefore, this study aimed to identify and experimentally validate the glycolysis-related hub genes in AMI as diagnostic biomarkers, and further explore the association between hub genes and immune infiltration. METHODS: Differentially expressed genes (DEGs) from AMI peripheral blood mononuclear cells (PBMCs) were analyzed using R software. Glycolysis-related DEGs (GRDEGs) were identified and analyzed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) for functional enrichment. A protein-protein interaction network was constructed using the STRING database and visualized using Cytoscape software. Immune infiltration analysis between patients with AMI and stable coronary artery disease (SCAD) controls was performed using CIBERSORT, and correlation analysis between GRDEGs and immune cell infiltration was performed. We also plotted nomograms and receiver operating characteristic (ROC) curves to assess the predictive accuracy of GRDEGs for AMI occurrence. Finally, key genes were experimentally validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting using PBMCs. RESULTS: A total of 132 GRDEGs and 56 GRDEGs were identified on the first day and 4-6 days after AMI, respectively. Enrichment analysis indicated that these GRDEGs were mainly clustered in the glycolysis/gluconeogenesis and metabolic pathways. Five hub genes (HK2, PFKL, PKM, G6PD, and ALDOA) were selected using the cytoHubba plugin. The link between immune cells and hub genes indicated that HK2, PFKL, PKM, and ALDOA were significantly positively correlated with monocytes and neutrophils, whereas G6PD was significantly positively correlated with neutrophils. The calibration curve, decision curve analysis, and ROC curves indicated that the five hub GRDEGs exhibited high predictive value for AMI. Furthermore, the five hub GRDEGs were validated by RT-qPCR and western blotting. CONCLUSION: We concluded that HK2, PFKL, PKM, G6PD, and ALDOA are hub GRDEGs in AMI and play important roles in AMI progression. This study provides a novel potential immunotherapeutic method for the treatment of AMI.


Asunto(s)
Biología Computacional , Redes Reguladoras de Genes , Glucólisis , Infarto del Miocardio , Mapas de Interacción de Proteínas , Humanos , Glucólisis/genética , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/diagnóstico , Perfilación de la Expresión Génica , Bases de Datos Genéticas , Transcriptoma , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Valor Predictivo de las Pruebas , Masculino , Persona de Mediana Edad , Hexoquinasa/genética , Femenino , Estudios de Casos y Controles , Nomogramas , Reproducibilidad de los Resultados
3.
J Adv Nurs ; 79(8): 3082-3091, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36978259

RESUMEN

AIMS: This study aimed to measure sleep quality and its possible association with fear of progression (FOP) in patients with chronic heart failure (HF). DESIGN: A cross-sectional study. METHODS: A total of 254 patients with chronic HF were recruited from two tertiary hospitals in China. The sociodemographic and clinical data of participants were collected using a general information questionnaire. The Pittsburgh Sleep Quality Index and Fear of Progression Questionnaire-Short Form were used to evaluate sleep quality over 1 month and FOP. Pearson correlation and hierarchical regression were conducted to analyse the relationship between sleep quality and FOP. RESULTS: Of all participants, 85.8% had poor sleep quality, with a mean score of 12.3 ± 4.2 (possible score could be 0 to 21). The severity of FOP was positively correlated with poor sleep quality. HF hospitalization in the past year, numbers of HF drugs, monthly income and total score of FOP were strong predictors of decreased sleep quality, accounting for 24.2% of the variance in the sleep quality of these patients. CONCLUSIONS: Patients with chronic HF generally reported poor sleep quality, which should be highly concerned for medical workers. Alleviating FOP as a therapeutic strategy may improve sleep quality. IMPACT: It is urgent to raise clinical attention that Chinese patients with chronic HF have poor sleep quality, which is not just due to the symptoms of HF itself. FOP is an important psychological factor influencing sleep quality in patients with chronic HF, which has not been explored in China. This study provides a new perspective on targeted interventions for poor sleep quality in chronic HF. PATIENT AND PUBLIC INVOLVEMENT: No patient or public involvement.


Asunto(s)
Insuficiencia Cardíaca , Calidad del Sueño , Humanos , Estudios Transversales , Miedo/psicología , Enfermedad Crónica , Encuestas y Cuestionarios , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/psicología
4.
J Biol Chem ; 295(8): 2324-2335, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31953322

RESUMEN

Defects in energy metabolism in either the retina or the immediately adjacent retinal pigment epithelium (RPE) underlie retinal degeneration, but the metabolic dependence between retina and RPE remains unclear. Nitrogen-containing metabolites such as amino acids are essential for energy metabolism. Here, we found that 15N-labeled ammonium is predominantly assimilated into glutamine in both the retina and RPE/choroid ex vivo [15N]Ammonium tracing in vivo show that, like the brain, the retina can synthesize asparagine from ammonium, but RPE/choroid and the liver cannot. However, unless present at toxic concentrations, ammonium cannot be recycled into glutamate in the retina and RPE/choroid. Tracing with 15N-labeled amino acids show that the retina predominantly uses aspartate transaminase for de novo synthesis of glutamate, glutamine, and aspartate, whereas RPE uses multiple transaminases to utilize and synthesize amino acids. Retina consumes more leucine than RPE, but little leucine is catabolized. The synthesis of serine and glycine is active in RPE but limited in the retina. RPE, but not the retina, uses alanine as mitochondrial substrates through mitochondrial pyruvate carrier. However, when the mitochondrial pyruvate carrier is inhibited, alanine may directly enter the retinal mitochondria but not those of RPE. In conclusion, our results demonstrate that the retina and RPE differ in nitrogen metabolism and highlight that the RPE supports retinal metabolism through active amino acid metabolism.


Asunto(s)
Nitrógeno/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Aminoácidos/metabolismo , Compuestos de Amonio/farmacología , Animales , Coroides/metabolismo , Masculino , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Isótopos de Nitrógeno/metabolismo , Especificidad de Órganos/efectos de los fármacos , Piruvatos/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Retinaldehído/metabolismo
5.
Circulation ; 142(12): 1190-1204, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32755395

RESUMEN

BACKGROUND: Angiotensin-converting enzyme 2 (ACE2) converts angiotensin II, a potent vasoconstrictor, to angiotensin-(1-7) and is also a membrane protein that enables coronavirus disease 2019 (COVID-19) infectivity. AMP-activated protein kinase (AMPK) phosphorylation of ACE2 enhances ACE2 stability. This mode of posttranslational modification of ACE2 in vascular endothelial cells is causative of a pulmonary hypertension (PH)-protective phenotype. The oncoprotein MDM2 (murine double minute 2) is an E3 ligase that ubiquitinates its substrates to cause their degradation. In this study, we investigated whether MDM2 is involved in the posttranslational modification of ACE2 through its ubiquitination of ACE2, and whether an AMPK and MDM2 crosstalk regulates the pathogenesis of PH. METHODS: Bioinformatic analyses were used to explore E3 ligase that ubiquitinates ACE2. Cultured endothelial cells, mouse models, and specimens from patients with idiopathic pulmonary arterial hypertension were used to investigate the crosstalk between AMPK and MDM2 in regulating ACE2 phosphorylation and ubiquitination in the context of PH. RESULTS: Levels of MDM2 were increased and those of ACE2 decreased in lung tissues or pulmonary arterial endothelial cells from patients with idiopathic pulmonary arterial hypertension and rodent models of experimental PH. MDM2 inhibition by JNJ-165 reversed the SU5416/hypoxia-induced PH in C57BL/6 mice. ACE2-S680L mice (dephosphorylation at S680) showed PH susceptibility, and ectopic expression of ACE2-S680L/K788R (deubiquitination at K788) reduced experimental PH. Moreover, ACE2-K788R overexpression in mice with endothelial cell-specific AMPKα2 knockout mitigated PH. CONCLUSIONS: Maladapted posttranslational modification (phosphorylation and ubiquitination) of ACE2 at Ser-680 and Lys-788 is involved in the pathogenesis of pulmonary arterial hypertension and experimental PH. Thus, a combined intervention of AMPK and MDM2 in the pulmonary endothelium might be therapeutically effective in PH treatment.


Asunto(s)
Peptidil-Dipeptidasa A/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Hipertensión Arterial Pulmonar/patología , Ubiquitinación , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Enzima Convertidora de Angiotensina 2 , Animales , Susceptibilidad a Enfermedades , Células Endoteliales/citología , Células Endoteliales/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidil-Dipeptidasa A/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas
6.
Int Heart J ; 62(1): 23-32, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33518662

RESUMEN

The aim of this study was to explore potential predictive biomarkers and therapeutic targets of post-infarct heart failure (HF) using bioinformatics analyses.CEL raw data of GSE59867 and GSE62646 were downloaded from the GEO database. Differentially expressed genes (DEGs) between patients with ST-segment elevation myocardial infarction (STEMI) and those with stable coronary artery disease (CAD) at admission and DEGs between admission and 6 months after myocardial infarction (MI) in patients with STEMI were analyzed. A gene ontology (GO) analysis and a gene set enrichment analysis (GSEA) were performed, and a protein-protein interaction network was constructed. Critical genes were further analyzed.In total, 147 DEGs were screened between STEMI and CAD at admission, and 62 DEGs were identified in patients with STEMI between admission and 6 months after MI. The results of GO and GSEA indicate that neutrophils, neutrophil-related immunity responses, and monocytes/macrophages play important roles in MI pathogenesis. SLED1 expression was higher in patients with HF than in those without HF at admission and 1 month after MI. GSEA indicates that mTORC1 activation, E2F targets, G2M checkpoint, and MYC targets v1 inhibition may play key roles in the development of post-infarct HF. Furthermore, SLED1 may be involved in the development of post-infarct HF by activating mTORC1 and inhibiting E2F targets, G2M checkpoint, and MYC targets v1.SLED1 may be a novel biomarker of post-infarct HF and may serve as a potential therapeutic target in this disease.


Asunto(s)
Proteína Mayor Básica del Eosinófilo/metabolismo , Insuficiencia Cardíaca/metabolismo , Infarto del Miocardio con Elevación del ST/complicaciones , Biomarcadores/metabolismo , Proteína Mayor Básica del Eosinófilo/genética , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/genética , Humanos , Mapas de Interacción de Proteínas , Infarto del Miocardio con Elevación del ST/genética , Infarto del Miocardio con Elevación del ST/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 38(6): 1309-1320, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29622561

RESUMEN

OBJECTIVE: Recently, we have demonstrated that acute glucosamine-induced augmentation of protein O-linked ß-N-acetylglucosamine (O-GlcNAc) levels inhibits inflammation in isolated vascular smooth muscle cells and neointimal formation in a rat model of carotid injury by interfering with NF-κB (nuclear factor-κB) signaling. However, the specific molecular target for O-GlcNAcylation that is responsible for glucosamine-induced vascular protection remains unclear. In this study, we test the hypothesis that increased A20 (also known as TNFAIP3 [tumor necrosis factor α-induced protein 3]) O-GlcNAcylation is required for glucosamine-mediated inhibition of inflammation and vascular protection. APPROACH AND RESULTS: In cultured rat vascular smooth muscle cells, both glucosamine and the selective O-linked N-acetylglucosaminidase inhibitor thiamet G significantly increased A20 O-GlcNAcylation. Thiamet G treatment did not increase A20 protein expression but did significantly enhance binding to TAX1BP1 (Tax1-binding protein 1), a key regulatory protein for A20 activity. Adenovirus-mediated A20 overexpression further enhanced the effects of thiamet G on prevention of TNF-α (tumor necrosis factor-α)-induced IκB (inhibitor of κB) degradation, p65 phosphorylation, and increases in DNA-binding activity. A20 overexpression enhanced the inhibitory effects of thiamet G on TNF-α-induced proinflammatory cytokine expression and vascular smooth muscle cell migration and proliferation, whereas silencing endogenous A20 by transfection of specific A20 shRNA significantly attenuated these inhibitory effects. In balloon-injured rat carotid arteries, glucosamine treatment markedly inhibited neointimal formation and p65 activation compared with vehicle treatment. Adenoviral delivery of A20 shRNA to the injured arteries dramatically reduced balloon injury-induced A20 expression and inflammatory response compared with scramble shRNA and completely abolished the vascular protection of glucosamine. CONCLUSIONS: These results suggest that O-GlcNAcylation of A20 plays a key role in the negative regulation of NF-κB signaling cascades in TNF-α-treated vascular smooth muscle cells in culture and in acutely injured arteries, thus protecting against inflammation-induced vascular injury.


Asunto(s)
Antiinflamatorios/farmacología , Traumatismos de las Arterias Carótidas/prevención & control , Proteínas de Unión al ADN/metabolismo , Glucosamina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Acetilglucosamina/metabolismo , Animales , Antiinflamatorios/metabolismo , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Glucosamina/metabolismo , Glicosilación , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
8.
Biochem Biophys Res Commun ; 485(1): 69-75, 2017 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-28202417

RESUMEN

Triptolide is the predominant active component of the Chinese herb Tripterygium wilfordii Hook F (TwHF) that has been widely used to treat several chronic inflammatory diseases due to its immunosuppressive, anti-inflammatory, and anti-proliferative properties. In the present study, we elucidated the cardioprotective effects of triptolide against cardiac dysfunction and myocardial remodeling in chronic pressure-overloaded hearts. Furthermore, the potential mechanisms of triptolide were investigated. For this purpose, C57/BL6 mice were anesthetized and subjected to transverse aortic constriction (TAC) or sham operation. Six weeks after the operation, all mice were randomly divided into 4 groups: sham-operated with vehicle group, TAC with vehicle group, and TAC with triptolide (20 or 100 µg/kg/day intraperitoneal injection) groups. Our data showed that the levels of NLRP3 inflammasome were significantly increased in the TAC group and were associated with increased inflammatory mediators and profibrotic factor production, resulting in myocardial fibrosis, cardiomyocyte hypertrophy, and impaired cardiac function. Triptolide treatment attenuated TAC-induced myocardial remodeling, improved cardiac diastolic and systolic function, inhibited the NLRP3 inflammasome and downstream inflammatory mediators (IL-1ß, IL-18, MCP-1, VCAM-1), activated the profibrotic TGF-ß1 pathway, and suppressed macrophage infiltration in a dose-dependent manner. Our study demonstrated that the protective effect of triptolide against pressure overload in the heart may act by inhibiting the NLRP3 inflammasome-induced inflammatory response and activating the profibrotic pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Diterpenos/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Inflamasomas/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fenantrenos/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Antiinflamatorios/química , Diterpenos/química , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Compuestos Epoxi/química , Compuestos Epoxi/uso terapéutico , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/inmunología , Hipertrofia Ventricular Izquierda/patología , Inmunidad Innata/efectos de los fármacos , Inflamasomas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/inmunología , Miocardio/metabolismo , Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Fenantrenos/química , Tripterygium/química
9.
Cell Physiol Biochem ; 40(5): 908-920, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27941310

RESUMEN

BACKGROUND: We have recently demonstrated that activated transforming growth factor-ß (TGF-ß) signaling suppresses myocardial peroxisome proliferator-activated receptor γ (PPARγ) expression in the pressure overloaded heart. In this study, we aim to further define the molecular mechanisms that underlie TGF-ß-induced PPARγ transcriptional inhibition. METHODS: Adult mouse cardiac fibroblasts were isolated and cultured. PPARγ promoter activity was measured by the dual-Luciferase reporter assay. Interactions between transcription factors and the target gene were identified. RESULTS: In cultured cardiac fibroblasts transfected with a plasmid containing a human PPARγ promoter, co-transfection of Smad3 and Smad4, but not Smad2, plasmids significantly enhanced TGF-ß1-induced inhibition of PPARγ promoter activity. Promoter deletion analysis and site-directed mutagenesis assays defined two Smad binding elements on the promoter of the PPARγ gene. Utilizing chromatin immunoprecipitation analysis and DNA-affinity precipitation methods, we demonstrated that the transcriptional regulatory complex consisting of Smad3, mSin3A and HDAC1 bound to the promoter of the PPARγ gene in cardiac fibroblasts in response to TGF-ß1 stimulation. Either silencing endogenous mSin3A expression by Lentivirus-mediated transduction of mSin3A shRNA or pretreatment with the specific HDAC1 inhibitor MS-275 effectively attenuated TGF-ß-induced transcriptional suppression of PPARγ. CONCLUSION: These results suggest that TGF-ß1-induced inhibition of PPARγ transcription depends on formation of a functional transcriptional regulatory complex that includes Smad3, mSin3A and HDAC1 at the PPARγ promoter.


Asunto(s)
Fibroblastos/metabolismo , Histona Desacetilasa 1/metabolismo , Miocardio/citología , PPAR gamma/genética , Proteínas Represoras/metabolismo , Proteína smad3/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Sitios de Unión , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Complejos Multiproteicos/metabolismo , PPAR gamma/metabolismo , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Complejo Correpresor Histona Desacetilasa y Sin3 , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo
10.
Am J Pathol ; 182(5): 1932-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23499549

RESUMEN

The ApoA-I mimetic peptide D-4F has demonstrated potent atheroprotective actions in vivo and in vitro. We investigated the effect of R-D4F (ie, the D-4F peptide with reverse order of amino acids) on intimal hyperplasia after vascular injury in a mouse model of carotid artery ligation. Adult male C57BL/6J mice were pretreated intraperitoneally with vehicle, D-4F (1 mg/kg), or R-D4F (1 mg/kg or 5 mg/kg) daily for 3 days; the mice were then subjected to left carotid artery ligation. All treatments were continued for 28 days after surgery. Neither D-4F nor R-D4F treatment affected serum lipid levels. Morphometric analysis showed that the occluded vessels had significant neointimal formation, compared with the uninjured arteries in vehicle-treated mice. Like the D-4F treatment, R-D4F treatment significantly (P < 0.05) inhibited intimal hyperplasia (-42%), local neutrophil and macrophage infiltration, and mRNA expression of the proinflammatory mediator monocyte chemotactic protein 1 (-55%) and vascular cell adhesion protein 1 (-53%), compared with vehicle. Furthermore, the vasoprotective effect of high-dose R-D4F was significantly enhanced, compared with the low dose. In cultured mouse RAW 264.7 macrophages, pretreatment with R-D4F also effectively inhibited lipopolysaccharide-induced leukocyte integrin CD11b expression, a key molecule for leukocyte infiltration. Taken together, these results suggest that R-D4F has significant anti-inflammatory features and facilitates prevention of neointimal formation after vascular injury in mice.


Asunto(s)
Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Neointima/tratamiento farmacológico , Neointima/prevención & control , Péptidos/farmacología , Péptidos/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Antígeno CD11b/metabolismo , Estenosis Carotídea/sangre , Estenosis Carotídea/tratamiento farmacológico , Estenosis Carotídea/patología , Línea Celular , Inflamación/sangre , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ligadura , Lípidos/sangre , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neointima/sangre , Neointima/patología , Péptidos/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Free Radic Biol Med ; 221: 1-12, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38734270

RESUMEN

Ischemia-reperfusion (I/R) injury, a multifaceted pathological process, occurs when the prolongation of reperfusion duration triggers ferroptosis-mediated myocardial damage. Isoliquiritigenin (ISL), a single flavonoid from licorice, exhibits a wide range of pharmacological impacts, but its function in ferroptosis caused by myocardial I/R injury remains unclear. This study delved into the protective effect of ISL on myocardial I/R injury-induced ferroptosis and its mechanism. Neonatal mouse cardiomyocytes (NMCM) underwent hypoxia/reoxygenation (H/R) to simulate the pathological process of myocardial I/R. ISL significantly attenuated H/R-triggered production of reactive oxygen species in NMCM, reduced the expression of malondialdehyde and the activity of lactate dehydrogenase, enhanced superoxide dismutase and catalase activity, and increased the expression of nuclear factor E2-related factor 2 (Nrf2) and its downstream heme oxygenase 1 (HO-1), thereby mitigating oxidative stress damage. CCK8 experiment revealed that the ferroptosis inhibitor Ferrostatin-1 significantly improved myocardial cell viability after 24 h of reoxygenation, and ISL treatment showed a similar effect. ISL reduced intracellular free iron accumulation, up-regulated glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11) expression, and inhibited lipid peroxidation accumulation, thereby alleviating ferroptosis. The Nrf2-specific inhibitor ML385 counteracted ISL's defensive role against H/R-triggered oxidative stress damage and ferroptosis. In vivo experiments further confirmed that by regulating the translocation of Nrf2 into the nucleus, ISL treatment increased the levels of HO-1, GPX4, and SLC7A11, inhibited the expression of ACSL4, Drp1 to exert the antioxidant role, alleviated mitochondrial damage, and ferroptosis, ultimately reducing myocardial infarction area and injury induced by I/R. ML385 nearly abolished ISL's protective impact on the I/R model by inhibiting Nrf2 function. In summary, ISL is capable of mitigating oxidative stress, mitochondrial damage, and cardiomyocyte ferroptosis caused by I/R, thereby reducing myocardial injury. A key mechanism includes triggering the Nrf2/HO-1/SLC7A11/GPX4 pathway to prevent oxidative stress damage and cardiomyocyte ferroptosis caused by I/R.


Asunto(s)
Chalconas , Ferroptosis , Hemo-Oxigenasa 1 , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Chalconas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Ratones , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ferroptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Masculino , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
12.
J Ethnopharmacol ; 335: 118612, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39047883

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Eclipta prostrata (Linn.) is a traditional medicinal Chinese herb that displays multiple biological activities, such as encompassing immunomodulatory, anti-inflammatory, anti-tumor, liver-protective, antioxidant, and lipid-lowering effects. Ecliptasaponin A (ESA), a pentacyclic triterpenoid saponin isolated from Eclipta prostrata (Linn.), has been demonstrated to exert superior anti-inflammatory activity against many inflammatory disorders. AIM OF THE STUDY: Inflammation plays a critical role in acute myocardial infarction (AMI). This study aims to explore the treatment effects of ESA in AMI, as well as the underlying mechanism. METHODS: An AMI mouse model was established in mice via left anterior descending coronary artery (LAD) ligation. After surgery, ESA was injected at doses of 0.5, 1.25, and 2.5 mg/kg, respectively. Myocardial infarction size, cardiomyocyte apoptosis and cardiac echocardiography were studied. The potential mechanism of action of ESA was investigated by RNA-seq, Western blot, surface plasmon resonance (SPR), molecular docking, and immunofluorescence staining. RESULTS: ESA treatment not only significantly reduced myocardial infarct size, decreased myocardial cell apoptosis, and inhibited inflammatory cell infiltration, but also facilitated to improve cardiac function. RNA-seq and Western blot analysis proved that ESA treatment-induced differential expression genes mainly enriched in HMGB1/TLR4/NF-κB pathway. Consistently, ESA treatment resulted into the down-regulation of IL-1ß, IL-6, and TNF-α levels after AMI. Furthermore, SPR and molecular docking results showed that ESA could bind directly to HMGB1, thereby impeding the activation of the downstream TLR4/NF-κB pathway. The immunofluorescence staining and Western blot results at the cellular level also demonstrated that ESA inhibited the activation of the HMGB1/TLR4/NF-κB pathway in H9C2 cells. CONCLUSION: Our study was the first to demonstrate a cardiac protective role of ESA in AMI. Mechanism study indicated that the treatment effects of ESA are mainly attributed to its anti-inflammatory activity that was mediated by the HMGB1/TLR4/NF-κB pathway.

13.
Circulation ; 125(12): 1533-41, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22361324

RESUMEN

BACKGROUND: Interleukin-8 (IL8) receptors IL8RA and IL8RB on neutrophil membranes bind to IL8 and direct neutrophil recruitment to sites of inflammation, including acutely injured arteries. This study tested whether administration of IL8RA- and/or IL8RB-transduced rat aortic endothelial cells (ECs) accelerates adhesion of ECs to the injured surface, thus suppressing inflammation and neointima formation in balloon-injured rat carotid arteries. We tested the hypothesis that targeted delivery of ECs by overexpressing IL8RA and IL8RB receptors prevents inflammatory responses and promotes structural recovery of arteries after endoluminal injury. METHODS AND RESULTS: Young adult male rats received balloon injury of the right carotid artery and were transfused intravenously with ECs (total, 1.5×10(6) cells at 1, 3, and 5 hours after injury) transduced with adenoviral vectors carrying IL8RA, IL8RB, and IL8RA/RB (dual transduction) genes, AdNull (empty vector), or vehicle (no EC transfusion). ECs overexpressing IL8Rs inhibited proinflammatory mediators expression significantly (by 60% to 85%) and reduced infiltration of neutrophils and monocytes/macrophages into injured arteries at 1 day after injury, as well as stimulating a 2-fold increase in reendothelialization at 14 days after injury. IL8RA-EC, IL8RB-EC, and IL8RA/RB-EC treatment reduced neointima formation dramatically (by 80%, 74%, and 95%) at 28 days after injury. CONCLUSIONS: ECs with overexpression of IL8RA and/or IL8RB mimic the behavior of neutrophils that target and adhere to injured tissues, preventing inflammation and neointima formation. Targeted delivery of ECs to arteries with endoluminal injury provides a novel strategy for the prevention and treatment of cardiovascular disease.


Asunto(s)
Traumatismos de las Arterias Carótidas/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Neointima/metabolismo , Receptores de Interleucina-8A/biosíntesis , Receptores de Interleucina-8B/biosíntesis , Animales , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/prevención & control , Células Endoteliales/patología , Células Endoteliales/trasplante , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Inflamación/metabolismo , Inflamación/patología , Masculino , Neointima/patología , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/fisiología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/fisiología
14.
Am J Physiol Heart Circ Physiol ; 305(4): H590-8, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23771691

RESUMEN

The endothelium is a dynamic component of the cardiovascular system that plays an important role in health and disease. This study tested the hypothesis that targeted delivery of endothelial cells (ECs) overexpressing neutrophil membrane IL-8 receptors IL8RA and IL8RB reduces acute myocardial infarction (MI)-induced left ventricular (LV) remodeling and dysfunction and increases neovascularization in the area at risk surrounding the infarcted tissue. MI was created by ligating the left anterior descending coronary artery in 12-wk-old male Sprague-Dawley rats. Four groups of rats were studied: group 1: sham-operated rats without MI or EC transfusion; group 2: MI rats with intravenous vehicle; group 3: MI rats with transfused ECs transduced with empty adenoviral vector (Null-EC); and group 4: MI rats with transfused ECs overexpressing IL8RA/RB (1.5 × 106 cells post-MI). Two weeks after MI, LV function was assessed by echocardiography; infarct size was assessed by triphenyltetrazolium chloride (live tissue) and picrosirus red (collagen) staining, and capillary density and neutrophil infiltration in the area at risk were measured by CD31 and MPO immunohistochemical staining, respectively. When compared with the MI + vehicle and MI-Null-EC groups, transfusion of IL8RA/RB-ECs decreased neutrophil infiltration and pro-inflammatory cytokine expression and increased capillary density in the area at risk, decreased infarct size, and reduced MI-induced LV dysfunction. These findings provide proof of principle that targeted delivery of ECs is effective in repairing injured cardiac tissue. Targeted delivery of ECs to infarcted hearts provides a potential novel strategy for the treatment of acute MI in humans.


Asunto(s)
Células Endoteliales/trasplante , Terapia Genética/métodos , Infarto del Miocardio/terapia , Miocardio/metabolismo , Receptores de Interleucina-8/biosíntesis , Remodelación Ventricular , Adenoviridae/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Vectores Genéticos , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/inmunología , Miocardio/patología , Neovascularización Fisiológica , Infiltración Neutrófila , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-8/genética , Proteínas Recombinantes de Fusión/biosíntesis , Factores de Tiempo , Transducción Genética , Transfección , Regulación hacia Arriba , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/inmunología , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda
15.
Eur J Cardiovasc Nurs ; 22(6): 638-646, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36748202

RESUMEN

AIMS: Studies have shown that symptom perception is associated with fear of progression (FOP) in many diseases and regulated by psychological factors. Whether the association also occurs in patients with chronic heart failure (HF) remains unclear, as do the specific mechanisms involved. This study aimed to explore the multiple mediation effects of self-care confidence and mental resilience on the relationship between symptom perception and FOP in Chinese patients with chronic HF. METHODS AND RESULTS: A cross-sectional study was conducted on 247 patients with chronic HF recruited from two hospitals in Yangzhou, China. The sociodemographic and clinical data and self-reported questionnaires including heart failure somatic perception, fear of progression, self-care confidence, and mental resilience were collected. Data analysis relating to correlations and mediating effects was carried out by SPSS 26.0 and PROCESS v3.3 macro. Fear of progression was positively correlated with symptom perception (r = 0.599, P < 0.01), but negatively correlated with self-care confidence (r = -0.663, P < 0.01), mental resilience-strength (r = -0.521, P < 0.01), and mental resilience-toughness (r = -0.596, P < 0.01). The relationship between symptom perception and FOP was mediated by self-care confidence [effect = 0.095, 95% confidence interval (CI) (0.054-0.142)] and mental resilience-toughness [effect = 0.033, 95% CI (0.006-0.074)], respectively, and together in serial [effect = 0.028, 95% CI (0.011-0.050)]. The proportion of the mediating effect accounting for the total effect was 31.0%. CONCLUSION: Self-care confidence and mental resilience-toughness were multiple mediators of the association between symptom perception and FOP in patients with chronic HF. Interventions targeted at strengthening self-care confidence and mental resilience may be beneficial for the reduction of FOP, especially with regard to toughness.


Asunto(s)
Insuficiencia Cardíaca , Análisis de Mediación , Humanos , Estudios Transversales , Miedo/psicología , Enfermedad Crónica , Insuficiencia Cardíaca/psicología , Percepción
16.
Clin Cardiol ; 46(7): 729-736, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37114367

RESUMEN

BACKGROUND: Our recent study has demonstrated that kinesiophobia is common in Chinese inpatients with chronic heart failure (CHF). Symptoms of heart failure (HF), coping mode, self-efficacy for exercise (SEE), and social support have been reported to be associated with kinesiophobia. However, little is known about the relationships between these four variables and kinesiophobia in older patients with CHF. OBJECTIVE: To test a model of influencing factors of kinesiophobia in older CHF patients. METHODS: A cross-sectional design was conducted from January 2021 to October 2021. The general information questionnaire, the Chinese version of the Tampa Scale for Kinesiophobia Heart (TSK-SV Heart-C), Symptom Status Questionnaire-Heart Failure, SEE, the Medical Coping Modes Questionnaire, and Social Support Rating Scale were used. Spearman correlation analysis and structural equation model (SEM) were performed for data analysis. RESULTS: A total of 270 older patients with CHF were recruited. Symptom status of HF (r = 0.455, p < .01), avoidance coping mode (r = 0.393, p <.01), and yielding coping mode (r = 0.439, p < .01) were positively correlated with kinesiophobia, while SEE (r = -0.530, p < .01), facing coping mode (r = -0.479, p < .01), and social support (r = -0.464, p < .01) were negatively correlated with kinesiophobia. SEM analysis showed that social support could affect kinesiophobia through the mediating variables of symptom status of HF, avoidance coping mode, and exercise self-efficacy. CONCLUSIONS: Symptoms of HF, coping mode, SEE, and social support may play role in kinesiophobia in older CHF patients. We should pay more attention to the synergies among these four variables in the improvement of kinesiophobia.


Asunto(s)
Insuficiencia Cardíaca , Trastornos Fóbicos , Humanos , Anciano , Trastornos Fóbicos/diagnóstico , Miedo , Kinesiofobia , Estudios Transversales , Enfermedad Crónica , Encuestas y Cuestionarios , Insuficiencia Cardíaca/diagnóstico
17.
Cell Discov ; 9(1): 118, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38012147

RESUMEN

Hydroxycarboxylic acid receptor 2 (HCAR2) belongs to the family of class A G protein-coupled receptors with key roles in regulating lipolysis and free fatty acid formation in humans. It is deeply involved in many pathophysiological processes and serves as an attractive target for the treatment of cardiovascular, neoplastic, autoimmune, neurodegenerative, inflammatory, and metabolic diseases. Here, we report four cryo-EM structures of human HCAR2-Gi1 complexes with or without agonists, including the drugs niacin (2.69 Å) and acipimox (3.23 Å), the highly subtype-specific agonist MK-6892 (3.25 Å), and apo form (3.28 Å). Combined with molecular dynamics simulation and functional analysis, we have revealed the recognition mechanism of HCAR2 for different agonists and summarized the general pharmacophore features of HCAR2 agonists, which are based on three key residues R1113.36, S17945.52, and Y2847.43. Notably, the MK-6892-HCAR2 structure shows an extended binding pocket relative to other agonist-bound HCAR2 complexes. In addition, the key residues that determine the ligand selectivity between the HCAR2 and HCAR3 are also illuminated. Our findings provide structural insights into the ligand recognition, selectivity, activation, and G protein coupling mechanism of HCAR2, which shed light on the design of new HCAR2-targeting drugs for greater efficacy, higher selectivity, and fewer or no side effects.

18.
Am J Physiol Heart Circ Physiol ; 303(5): H513-22, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22777418

RESUMEN

Acute increases in cellular protein O-linked N-acetyl-glucosamine (O-GlcNAc) modification (O-GlcNAcylation) have been shown to have protective effects in the heart and vasculature. We hypothesized that d-glucosamine (d-GlcN) and Thiamet-G, two agents that increase protein O-GlcNAcylation via different mechanisms, inhibit TNF-α-induced oxidative stress and vascular dysfunction by suppressing inducible nitric oxide (NO) synthase (iNOS) expression. Rat aortic rings were incubated for 3h at 37°C with d-GlcN or its osmotic control l-glucose (l-Glc) or with Thiamet-G or its vehicle control (H(2)O) followed by the addition of TNF-α or vehicle (H(2)O) for 21 h. After incubation, rings were mounted in a myograph to assess arterial reactivity. Twenty-four hours of incubation of aortic rings with TNF-α resulted in 1) a hypocontractility to 60 mM K(+) solution and phenylephrine, 2) blunted endothelium-dependent relaxation responses to ACh and substance P, and 3) unaltered relaxing response to the Ca(2+) ionophore A-23187 and the NO donor sodium nitroprusside compared with aortic rings cultured in the absence of TNF-α. d-GlcN and Thiamet-G pretreatment suppressed the TNF-α-induced hypocontractility and endothelial dysfunction. Total protein O-GlcNAc levels were significantly higher in aortic segments treated with d-GlcN or Thiamet-G compared with controls. Expression of iNOS protein was increased in TNF-α-treated rings, and this was attenuated by pretreatment with either d-GlcN or Thiamet-G. Dense immunostaining for nitrotyrosylated proteins was detected in the endothelium and media of the aortic wall, suggesting enhanced peroxynitrite production by iNOS. These findings demonstrate that acute increases in protein O-GlcNAcylation prevent TNF-α-induced vascular dysfunction, at least in part, via suppression of iNOS expression.


Asunto(s)
Acetilglucosamina/metabolismo , Aorta Torácica/metabolismo , Enfermedades de la Aorta/prevención & control , Inflamación/metabolismo , Procesamiento Proteico-Postraduccional , Vasoconstricción , Vasodilatación , Acilación , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/inmunología , Aorta Torácica/fisiopatología , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/fisiopatología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Inflamación/inmunología , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Masculino , Miografía , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , beta-N-Acetilhexosaminidasas/metabolismo
19.
Mol Biol Rep ; 39(4): 4445-54, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21947883

RESUMEN

Circulating endothelial progenitor cells (EPCs) have a critical role in endothelial maintenance and repair. Apolipoprotein A-I mimetic peptide D-4F has been shown to posses anti-atherogenic properties via sequestration of oxidized phospholipids, induction of remodeling of high density lipoprotein and promotion of cholesterol efflux from macrophage-derived foam cells. In this study, we test the effects of D-4F on EPC biology. EPCs were isolated from the peripheral venous blood of healthy male volunteers and characterized by 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine-labeled acetylated LDL uptake and ulex europaeus agglutinin binding and flow cytometry. Cell proliferation, migration, adhesion, nitric oxide production and endothelial nitric oxide synthase (eNOS) expression in the absence and presence of D-4F or simvastatin (as a positive control), were assayed. We demonstrated that D-4F significantly enhanced EPC proliferation, migration and adhesion in a dose-dependent manner compared with vehicle. However, all of the favorable effects of D-4F on EPCs were dramatically attenuated by preincubation with NOS inhibitor L-NAME. Further, D-4F also increased nitric oxide production in culture supernatant and the levels of eNOS expression and phosphorylation. The stimulatory effects of D-4F (10 µg/ml) on EPC biology were comparable to 0.5 µM simvastatin. These results suggest that eNOS/NO pathway mediates the functional modulation of EPC biology in response to D-4F treatment and support the notion that the beneficial role of D-4F on EPCs may be one of the important components of its anti-atherogenic potential.


Asunto(s)
Apolipoproteína A-I/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Células Madre/citología , Adulto , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Óxido Nítrico/biosíntesis , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/enzimología
20.
PeerJ ; 10: e13436, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35586138

RESUMEN

Background: Intimal hyperplasia is a common pathological process of restenosis following angioplasty, atherosclerosis, pulmonary hypertension, vein graft stenosis, and other proliferative diseases. This study aims to screen for potential novel gene targets and mechanisms related to vascular intimal hyperplasia through an integrated microarray analysis of the Gene Expression Omnibus Database (GEO) database. Material and Methods: The gene expression profile of the GSE56143 dataset was downloaded from the Gene Expression Omnibus database. Functional enrichment analysis, protein-protein interaction (PPI) network analysis, and the transcription factor (TF)-target gene regulatory network were used to reveal the biological functions of differential genes (DEGs). Furthermore, the expression levels of the top 10 key DEGs were verified at the mRNA and protein level in the carotid artery 7 days after ligation. Results: A total of 373 DEGs (199 upregulated DEGs and 174 downregulated DEGs) were screened. These DEGs were significantly enriched in biological processes, including immune system process, cell adhesion, and several pathways, which were mainly associated with cell adhesion molecules and the regulation of the actin cytoskeleton. The top 10 key DEGs (Ptprc, Fn1, Tyrobp, Emr1, Itgb2, Itgax, CD44, Ctss, Ly86, and Aif1) acted as key genes in the PPI network. The verification of these key DEGs at the mRNA and protein levels was consistent with the results of the above-mentioned bioinformatics analysis. Conclusion: The present study identified key genes and pathways involved in intimal hyperplasia induced by carotid artery ligation. These results improved our understanding of the mechanisms underlying the development of intimal hyperplasia and provided candidate targets.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Humanos , Perfilación de la Expresión Génica/métodos , Hiperplasia/genética , Mapas de Interacción de Proteínas/genética , Arterias Carótidas/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA