Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36834872

RESUMEN

The retinoid-related orphan receptor α (RORα) is one subfamily of nuclear hormone receptors (NRs). This review summarizes the understanding and potential effects of RORα in the cardiovascular system and then analyzes current advances, limitations and challenges, and further strategy for RORα-related drugs in cardiovascular diseases. Besides regulating circadian rhythm, RORα also influences a wide range of physiological and pathological processes in the cardiovascular system, including atherosclerosis, hypoxia or ischemia, myocardial ischemia/reperfusion injury, diabetic cardiomyopathy, hypertension, and myocardial hypertrophy. In terms of mechanism, RORα was involved in the regulation of inflammation, apoptosis, autophagy, oxidative stress, endoplasmic reticulum (ER) stress, and mitochondrial function. Besides natural ligands for RORα, several synthetic RORα agonists or antagonists have been developed. This review mainly summarizes protective roles and possible mechanisms of RORα against cardiovascular diseases. However, there are also several limitations and challenges of current research on RORα, especially the difficulties on the transformability from the bench to the bedside. By the aid of multidisciplinary research, breakthrough progress on RORα-related drugs to combat cardiovascular disorder may appear.


Asunto(s)
Enfermedades Cardiovasculares , Cardiomiopatías Diabéticas , Humanos , Cardiomegalia , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Receptores Citoplasmáticos y Nucleares , Retinoides
2.
BMC Pulm Med ; 22(1): 95, 2022 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-35305612

RESUMEN

BACKGROUND: Lung ultrasound (LUS) is a useful tool for assessing the severity of lung disease, without radiation exposure. However, there is little data on the practicality of LUS in assessing the severity of bronchopulmonary dysplasia (BPD) and evaluating short-term clinical outcomes. We adapted a LUS score to evaluate BPD severity and assess the reliability of mLUS score correlated with short-term clinical outcomes. METHODS: Prospective diagnostic accuracy study was designed to enroll preterm infants with gestational age < 34 weeks. Lung ultrasonography was performed at 36 weeks postmenstrual age. The diagnostic and predictive values of new modified lung ultrasound (mLUS) scores based on eight standard sections were compared with classic lung ultrasound (cLUS) scores. RESULTS: A total of 128 infants were enrolled in this cohort, including 30 without BPD; 31 with mild BPD; 23 with moderate BPD and 44 with severe BPD. The mLUS score was significantly correlated with the short-term clinical outcomes, superior to cLUS score. The mLUS score well correlated with moderate and severe BPD (AUC = 0.813, 95% CI 0.739-0.888) and severe BPD (AUC = 0.801, 95% CI 0.728-0.875), which were superior to cLUS score. The ROC analysis of mLUS score to evaluate the other short-term outcomes also showed significant superiority to cLUS score. The optimal cutoff points for mLUS score were 14 for moderate and severe BPD and 16 for severe BPD. CONCLUSIONS: The mLUS score correlates significantly with short-term clinical outcomes and well evaluates these outcomes in preterm infants.


Asunto(s)
Displasia Broncopulmonar , Displasia Broncopulmonar/diagnóstico por imagen , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Pulmón/diagnóstico por imagen , Estudios Prospectivos , Reproducibilidad de los Resultados , Ultrasonografía
3.
J Paediatr Child Health ; 58(8): 1396-1406, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35524688

RESUMEN

OBJECTIVES: To investigate the risk profile of preterm birth (PTB) in 2018 in China. METHOD: A prospective multicentre case-control study was conducted in 15 hospitals located in seven provinces throughout three geographical areas (the Eastern, South-Central and North-Western regions) in China. A total of 3147 preterm (<37+0 weeks) and 3147 term (37+0 to 41+6 weeks) live-birth mothers were included. Designed questionnaires were used to investigate maternal and fetal information. We calculated multivariable logistic regression and population attributable risk (PAR). RESULTS: Iatrogenic PTB accounted for 48.1% of preterm mothers. Multivariable analysis showed PTB was significantly associated with six categories of maternal and fetal factors, adverse life-style and psychological conditions (adjusted odds ratio (aOR) 2.063, 95% confidence interval (CI) 1.601-2.657) had the highest PAR% (60.1%). High school and below education level (PAR% = 25.8%), living in town or village (PAR% = 24.4%), low pregnant weight gain (PAR% = 16.8%), hypertensive disorders in pregnancy (aOR: 5.010, 95% CI: 4.039-6.216, PAR% = 15.3%), placental abnormality (aOR: 4.242, 95% CI: 3.454-5.211, PAR% = 14.1%) and multiple pregnancy (aOR: 10.990, 95% CI: 7.743-15.599, PAR% = 11.8%) were significantly associated with PTB with high PAR% value. CONCLUSION: The main risk factors for PTB in China were placental abnormality, hypertensive disorders in pregnancy and multiple pregnancy. Adverse life-style and psychological conditions and socio-economic disadvantage had high public health significance.


Asunto(s)
Hipertensión Inducida en el Embarazo , Nacimiento Prematuro , Estudios de Casos y Controles , China/epidemiología , Femenino , Humanos , Recién Nacido , Placenta , Embarazo , Nacimiento Prematuro/epidemiología , Nacimiento Prematuro/etiología , Estudios Prospectivos , Factores de Riesgo , Encuestas y Cuestionarios
4.
Acta Pharmacol Sin ; 42(2): 230-241, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32770173

RESUMEN

Sirtuin 3 (SIRT3) is a potential therapeutic target for cardiovascular, metabolic, and other aging-related diseases. In this study, we investigated the role of SIRT3 in diabetic cardiomyopathy (DCM). Mice were injected with streptozotocin (STZ, 60 mg/kg, ip) to induce diabetes mellitus. Our proteomics analysis revealed that SIRT3 expression in the myocardium of diabetic mice was lower than that of control mice, as subsequently confirmed by real-time PCR and Western blotting. To explore the role of SIRT3 in DCM, SIRT3-knockout mice and 129S1/SvImJ wild-type mice were injected with STZ. We found that diabetic mice with SIRT3 deficiency exhibited aggravated cardiac dysfunction, increased lactate dehydrogenase (LDH) level in the serum, decreased adenosine triphosphate (ATP) level in the myocardium, exacerbated myocardial injury, and promoted myocardial reactive oxygen species (ROS) accumulation. Neonatal rat cardiomyocytes were transfected with SIRT3 siRNA, then exposed to high glucose (HG, 25.5 mM). We found that downregulation of SIRT3 further increased LDH release, decreased ATP level, suppressed the mitochondrial membrane potential, and elevated oxidative stress in HG-treated cardiomyocytes. SIRT3 deficiency further raised expression of necroptosis-related proteins including receptor-interacting protein kinase 1 (RIPK1), RIPK3, and cleaved caspase 3, and upregulated the expression of inflammation-related proteins including NLR family pyrin domain-containing protein 3 (NLRP3), caspase 1 p20, and interleukin-1ß both in vitro and in vivo. Collectively, SIRT3 deficiency aggravated hyperglycemia-induced mitochondrial damage, increased ROS accumulation, promoted necroptosis, possibly activated the NLRP3 inflammasome, and ultimately exacerbated DCM in the mice. These results suggest that SIRT3 can be a molecular intervention target for the prevention and treatment of DCM.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Cardiomiopatías Diabéticas/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sirtuina 3/genética , Animales , Diabetes Mellitus Experimental/genética , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Miocitos Cardíacos/patología , Necroptosis/genética , Estrés Oxidativo/genética , Ratas , Ratas Sprague-Dawley , Estreptozocina
5.
Int J Mol Sci ; 22(21)2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34769322

RESUMEN

Myocardial ischemia or hypoxia can induce myocardial fibroblast proliferation and myocardial fibrosis. Hydrogen sulfide (H2S) is a gasotransmitter with multiple physiological functions. In our present study, primary cardiac fibroblasts were incubated with H2S donor sodium hydrosulfide (NaHS, 50 µM) for 4 h followed by hypoxia stimulation (containing 5% CO2 and 1% O2) for 4 h. Then, the preventive effects on cardiac fibroblast proliferation and the possible mechanisms were investigated. Our results showed that NaHS reduced the cardiac fibroblast number, decreased the hydroxyproline content; inhibited the EdU positive ratio; and down-regulated the expressions of α-smooth muscle actin (α-SMA), the antigen identified by monoclonal antibody Ki67 (Ki67), proliferating cell nuclear antigen (PCNA), collagen I, and collagen III, suggesting that hypoxia-induced cardiac fibroblasts proliferation was suppressed by NaHS. NaHS improved the mitochondrial membrane potential and attenuated oxidative stress, and inhibited dynamin-related protein 1 (DRP1), but enhanced optic atrophy protein 1 (OPA1) expression. NaHS down-regulated receptor interacting protein kinase 1 (RIPK1) and RIPK3 expression, suggesting that necroptosis was alleviated. NaHS increased the sirtuin 3 (SIRT3) expressions in hypoxia-induced cardiac fibroblasts. Moreover, after SIRT3 siRNA transfection, the inhibitory effects on cardiac fibroblast proliferation, oxidative stress, and necroptosis were weakened. In summary, necroptosis inhibition by exogenous H2S alleviated hypoxia-induced cardiac fibroblast proliferation via SIRT3.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Corazón/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Hipoxia/fisiopatología , Necroptosis , Sirtuinas/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Gasotransmisores/farmacología , Corazón/fisiopatología , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Sirtuinas/genética
6.
Int J Mol Sci ; 21(18)2020 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-32933152

RESUMEN

Dihydromyricetin (DHY), a flavonoid component isolated from Ampelopsis grossedentata, exerts versatile pharmacological activities. However, the possible effects of DHY on diabetic vascular endothelial dysfunction have not yet been fully elucidated. In the present study, male C57BL/6 mice, wild type (WT) 129S1/SvImJ mice and sirtuin 3 (SIRT3) knockout (SIRT3-/-) mice were injected with streptozotocin (STZ, 60 mg/kg/day) for 5 consecutive days. Two weeks later, DHY were given at the doses of 250 mg/kg by gavage once daily for 12 weeks. Fasting blood glucose (FBG) and glycosylated hemoglobin (HbA1c) level, endothelium-dependent relaxation of thoracic aorta, reactive oxygen species (ROS) production, SIRT3, and superoxide dismutase 2 (SOD2) protein expressions, as well as mitochondrial Deoxyribonucleic Acid (mtDNA) copy number, in thoracic aorta were detected. Our study found that DHY treatment decreased FBG and HbA1c level, improved endothelium-dependent relaxation of thoracic aorta, inhibited oxidative stress and ROS production, and enhanced SIRT3 and SOD2 protein expression, as well as mtDNA copy number, in thoracic aorta of diabetic mice. However, above protective effects of DHY were unavailable in SIRT3-/- mice. The study suggested DHY improved endothelial dysfunction in diabetic mice via oxidative stress inhibition in a SIRT3-dependent manner.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Flavonoles/farmacología , Estrés Oxidativo/efectos de los fármacos , Sirtuina 3/metabolismo , Enfermedades Vasculares/tratamiento farmacológico , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Glucemia/efectos de los fármacos , ADN/metabolismo , Variaciones en el Número de Copia de ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Enfermedades Vasculares/metabolismo
7.
Cell Biochem Biophys ; 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39259410

RESUMEN

To investigate the mechanism of Wen Fei Fu Yang Qu Tan Fang (WFFYQTF) in the treatment of chronic obstructive pulmonary disease (COPD) using network pharmacology and pharmacodynamics. The TCMSP database was utilized to identify the chemical components and molecular targets of WFFYQTF. Cytoscape software was employed to construct a "drug component-target" network. COPD risk genes and intersecting molecular targets of WFFYQTF were identified using GeneCards, OMIM, and DisGeNET databases. The STRING website was the place where protein-protein interaction (PPI) analysis was performed. Cytoscape topological analysis was applied for screening out key targets of WFFYQTF. GO and KEGG enrichment analyses were conducted using the DAVID database to elucidate the treatment targets of COPD with WFFYQTF. A total of 136 active components of WFFYQTF were identified, including key components such as quercetin, kaempferol, and luteolin, which were found to be particularly significant. Additionally, 412 drug targets and 7121 COPD risk genes were screened out, and 323 treatment targets of COPD with WFFYQTF were determined by Wayne analysis. Core targets identified via PPI analysis included SRC, STAT3, AKT1, HSP90AA1, and JUN. Pathways such as the hypoxia responce, inflammatory response, PI3K/AKT pathway, TH17 pathway and MAPK pathway were obtained with GO and KEGG enrichment analyses. Molecular docking results suggested that quercetin could be soundly bound to STAT3 and AKT1, and kaempferol to SRC. WFFYQTF can effectively impede COPD progression through the coordinated action of multiple components, targets, and pathways during treatment.

8.
Pediatr Pulmonol ; 58(9): 2551-2558, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37294069

RESUMEN

OBJECTIVE: Lung ultrasound (LUS) is a useful and radiation-free diagnostic tool for predicting bronchopulmonary dysplasia, which is a risk factor for late respiratory disease. However, data on the relationship of LUS with late respiratory disease was scarce. This study aims to determine whether LUS is associated with late respiratory disease during early childhood. METHODS: This prospective cohort study enrolled preterm infants born before 32 weeks of gestation. LUS was performed at 36 weeks' postmenstrual age. The predictive values of a modified lung ultrasound (mLUS) score based on eight standard sections were assessed to predict late respiratory disease, defined as a physician diagnosis of bronchopulmonary dysplasia deterioration, asthma, reactive airway disease, bronchiolitis, pneumonia, or respiratory-related hospitalization during the first 2 years of life. RESULTS: A total of 94 infants completed follow-up, of whom 74.5% met the late respiratory disease criteria. The mLUS scores were significantly associated with late respiratory disease (adjusted odds ratio: 1.23, CI: 1.10-1.38, p < 0.001). The mLUS scores also well predicted late respiratory disease (AUC = 0.820, 95% CI: 0.733-0.907). These scores were superior to the classic lung ultrasound score (p = 0.02) and as accurate as the modified NICHD-defined bronchopulmonary dysplasia classification (p = 0.91). A mLUS score ≥14 was the optimal cutoff point for predicting late respiratory disease. CONCLUSION: The modified lung ultrasound score correlates significantly with late respiratory disease and well predicts it in preterm infants during the first 2 years of life.


Asunto(s)
Displasia Broncopulmonar , Enfermedades Respiratorias , Lactante , Recién Nacido , Humanos , Preescolar , Recien Nacido Prematuro , Displasia Broncopulmonar/complicaciones , Estudios Prospectivos , Pulmón/diagnóstico por imagen , Enfermedades Respiratorias/complicaciones
9.
Biochem Pharmacol ; 215: 115748, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37591449

RESUMEN

Diabetic cardiomyopathy (DCM) is one serious and common complication in diabetes without effective treatments. Hydrogen sulfide (H2S) fights against a variety of cardiovascular diseases including DCM. Retinoic acid-related orphan receptor α (RORα) has protective effects on cardiovascular system. However, whether RORα mediates the protective effect of H2S against DCM remains unknown. The present research was to explore the roles and mechanisms of RORα in H2S against DCM. The study demonstrated that H2S donor sodium hydrosulfide (NaHS) alleviated cell injury but enhanced RORα expression in high glucose (HG)-stimulated cardiomyocytes. However, NaHS no longer had the protective effect on attenuating cell damage and oxidative stress, improving mitochondrial membrane potential, inhibiting necroptosis and enhanced signal transducer and activator of transcription 3 (STAT3) Ser727 phosphorylation in HG-stimulated cardiomyocytes after RORα siRNA transfection. Moreover, NaHS improved cardiac function, attenuated myocardial hypertrophy and fibrosis, alleviated oxidative stress, inhibited necroptosis, but increased STAT3 phosphorylation in wild type (WT) mice but not in RORα knockout mice (a spontaneous staggerer mice, sg/sg mice) with diabetes. Additionally, NaHS increased RORα promoter activity in cardiomyocytes with HG stimulation, which was related to the binding sites of E2F transcription factor 1 (E2F1) in the upstream region of RORα promoter. NaHS enhanced E2F1 expression and increased the binding of E2F1 to RORα promoter in cardiomyocytes with HG stimulation. In sum, H2S promoted RORα transcription via E2F1 to alleviate necroptosis and protect against DCM. It is helpful to propose a novel therapeutic implication for DCM.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Sulfuro de Hidrógeno , Animales , Ratones , Sulfuro de Hidrógeno/farmacología , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/prevención & control , Receptor alfa de Ácido Retinoico , Sitios de Unión , Ratones Noqueados , Tretinoina
10.
Biomed Pharmacother ; 168: 115802, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918258

RESUMEN

Diabetes mellitus is a metabolic disease caused by disorders of insulin secretion and utilization. Long-term hyperglycemia, insulin resistance, and disorders of glucose and lipid metabolism cause vascular endothelial cell damage. Endothelial dysfunction is a key feature of diabetic vascular complications such as diabetic nephropathy, retinopathy, neuropathy, and atherosclerosis. Importantly, cell death is thought to be a key factor contributing to vascular endothelial injury. Morphologically, cell death can be divided into three forms: type I apoptosis, type II autophagy, and type III necrosis. According to the difference in function, cell death can be divided into accidental cell death (ACD) and regulated cell death (RCD). RCD is a controlled process involving numerous proteins and precise signaling cascades. Multiple subroutines covered by RCD may be involved in diabetic endothelial dysfunction, including apoptosis, autophagy, necroptosis, pyroptosis, entosis, ferroptosis, ferroautophagy, parthanatos, netotic cell death, lysosome-dependent cell death, alkaliptosis, oxeiptosis, cuproptosis, and PANoptosis. This article briefly reviews the mechanism and significance of cell death associated with diabetic endothelial dysfunction, which will help deepen the understanding of diabetic endothelial cell death and provide new therapeutic ideas.


Asunto(s)
Diabetes Mellitus , Muerte Celular Regulada , Humanos , Muerte Celular , Apoptosis , Necrosis/metabolismo
11.
Antioxidants (Basel) ; 12(1)2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36671063

RESUMEN

Dihydromyricetin (DHY), the main flavonoid component in Ampelopsis grossedentata, has important benefits for health. The present study aimed to investigate the exact effects and possible mechanisms of DHY on diabetic cardiomyopathy (DCM). Male C57BL/6 mice and sirtuin 3 (SIRT3) knockout (SIRT3-KO) mice were injected with streptozotocin (STZ) to induce a diabetic model. Two weeks later, DHY (250 mg/kg) or carboxymethylcellulose (CMC) were administrated once daily by gavage for twelve weeks. We found that DHY alleviated fasting blood glucose (FBG) and triglyceride (TG) as well as glycosylated hemoglobin (HbA1c) levels; increased fasting insulin (FINS); improved cardiac dysfunction; ameliorated myocardial hypertrophy, fibrosis and injury; suppressed oxidative stress, inflammasome and necroptosis; but improved SIRT3 expression in STZ-induced mice. Neonatal rat cardiomyocytes were pre-treated with DHY (80 µM) with or without high glucose (HG) stimulation. The results showed that DHY attenuated cell damage but improved SIRT3 expression and inhibited oxidative stress, inflammasome and necroptosis in cardiomyocytes with high glucose stimulation. Moreover, the above protective effects of DHY on DCM were unavailable in SIRT3-KO mice, implying a promising medical potential of DHY for DCM treatment. In sum, DHY improved cardiac dysfunction; ameliorated myocardial hypertrophy, fibrosis and injury; and suppressed oxidative stress, inflammation and necroptosis via SIRT3 activation in STZ-induced diabetic mice, suggesting DHY may serve as a candidate for an agent to attenuate diabetic cardiomyopathy.

12.
Thromb Res ; 216: 14-21, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35679633

RESUMEN

BACKGROUND: Pulmonary embolism (PE) is a life-threatening condition associated with ~10% of deaths of hospitalized patients. Machine learning algorithms (MLAs) which predict the onset of pulmonary embolism (PE) could enable earlier treatment and improve patient outcomes. However, the extent to which they generalize to broader patient populations impacts their clinical utility. OBJECTIVE: To conduct the first large-scale external validation of a machine learning-based PE prediction model which uses EHR data from the first three hours of a patient's hospital stay to predict the occurrence of PE within the next 10 days of the inpatient stay. METHODS: This retrospective study included approximately two million adult hospital admissions across 44 medical institutions in the US from 2011 to 2017. Demographics, vital signs, and lab tests from adult inpatients at 12 institutions (n = 331,268; 3.3% PE positive) were used for training an XGBoost model. External validation of the model was conducted on patient populations from each of 32 medical institutions (total n = 1,660,715; 3.7% PE positive) without retraining. Model performance was assessed using the area under the receiver operating characteristic curve (AUROC). Backward elimination regression was used to identify correlations between characteristics of the external validation sets and AUROC. RESULTS: The model performed well (AUROC = 0.87) on the 20% hold-out subset of the training set. Despite demographic differences between the 32 external validation populations (percent PE positive: min = 1.54%, max = 6.47%), without retraining, the model had excellent discrimination, with a mean AUROC of 0.88 (min = 0.79, max = 0.93). Fixing sensitivity at 0.80, the model had a mean specificity of 0.85 (min = 0.64, max = 0.93). Backward elimination regression identified a negative association (ß = -0.015, p < 0.001) between the percentage of PE positive encounters and AUROC. CONCLUSIONS: A PE prediction model performed remarkably well across 32 different external patient populations without retraining and despite significant differences in demographic characteristics, demonstrating its generalizability and potential as a clinical decision support tool to aid PE detection and improve patient outcomes in a clinical setting.


Asunto(s)
Aprendizaje Automático , Embolia Pulmonar , Adulto , Algoritmos , Humanos , Embolia Pulmonar/diagnóstico , Curva ROC , Estudios Retrospectivos
13.
Free Radic Biol Med ; 181: 29-42, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35101564

RESUMEN

Diabetic cardiomyopathy lacks effective and novel methods. Hydrogen sulfide (H2S) as the third gasotransmitter plays an important role in the cardiovascular system. Our study was to elucidate the protective effect and possible mechanism of H2S on diabetic cardiomyopathy from the perspective of necroptosis. Leptin receptor deficiency (db/db) mice and streptozotocin (STZ)-induced diabetic cystathionine-γ-lyase (CSE) knockout (KO) mice were investigated. In addition, cardiomyocytes were stimulated with high glucose. We found that plasma H2S level, myocardial H2S production and CSE mRNA expression was impaired in the diabetic mice. CSE deficiency exacerbated diabetic cardiomyopathy, and promoted myocardial oxidative stress, necroptosis and inflammasome in STZ-induced mice. CSE inhibitor dl-propargylglycine (PAG) aggravated cell damage and oxidative stress, deteriorated necroptosis and inflammasome in cardiomyocytes with high glucose stimulation. H2S donor sodium hydrosulfide (NaHS) improved diabetic cardiomyopathy, attenuated myocardial oxidative stress, necroptosis and the NLR family pyrin domain-containing protein 3 (NLRP3) in db/db mice. NaHS also alleviated cell damage, oxidative stress, necroptosis and inflammasome in cardiomyocytes with high glucose stimulation. In Conclusion, H2S deficiency aggravated mitochondrial damage, increased reactive oxygen species accumulation, promoted necroptosis, activated NLRP3 inflammasome, and finally exacerbated diabetic cardiomyopathy. Exogenous H2S supplementation alleviated necroptosis to suppress NLRP3 inflammasome activation and attenuate diabetic cardiomyopathy via mitochondrial dysfunction improvement and oxidative stress inhibition. Our study provides the first evidence and a new mechanism that necroptosis inhibition by a pharmacological manner of H2S administration protected against diabetic cardiomyopathy. It is beneficial to provide a novel strategy for the prevention and treatment of diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Sulfuro de Hidrógeno , Animales , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/prevención & control , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Ratones , Necroptosis
14.
Stem Cells Dev ; 31(3-4): 53-66, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34913742

RESUMEN

Bronchopulmonary dysplasia (BPD) remains the most important respiratory morbidity of preterm infants with few effective preventive strategies. Administration of mesenchymal stem cells (MSC) was considered effective to prevent BPD via paracrine extracellular vesicles (EVs), while appropriate regimens of MSC-EVs and the mechanism remain unclear. Therefore, we established a hyperoxia-induced rat BPD model, and examined the effect of early intraperitoneal MSC-EVs with different doses on BPD. We found that MSC-EVs ameliorated hyperoxia-induced lung injury in a dose-dependent manner, and high-dose MSC-EVs ameliorated alveolar simplification and fibrosis. Also, MSC-EVs showed its beneficial effects on vascular growth and pulmonary hypertension. Primary AT2 cells were observed to transdifferentiate into AT1 cells when exposure to hyperoxia in vitro. Administration of MSC-EVs at the first-day culture significantly delayed the transdifferentiation of AT2 cells induced by hyperoxia. We further found that exposure to hyperoxia led to elevated expression of WNT5a mRNA and protein, a key agent in AT2 transdifferentiation, while MSC-EVs administration decreased it. Further study is warranted that MSC-EVs may delay the transdifferentiation of AT2 cells via WNT5a. These studies provide key preclinical evidence of MSC-EVs therapeutics on BPD and highlight the effect of MSC-EVs on suppressing the transdifferentiation of AT2 cells and its possible mechanism through downregulation of WNT5a.


Asunto(s)
Displasia Broncopulmonar , Vesículas Extracelulares , Hiperoxia , Células Madre Mesenquimatosas , Células Epiteliales Alveolares , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/metabolismo , Transdiferenciación Celular , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro , Ratas
15.
Oxid Med Cell Longev ; 2021: 9925771, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603602

RESUMEN

Sirtuin 3 (SIRT3) is critical in mitochondrial function and oxidative stress. Our present study investigates whether hydrogen sulfide (H2S) attenuated myocardial fibrosis and explores the possible role of SIRT3 on the protective effects. Neonatal rat cardiac fibroblasts were pretreated with NaHS followed by angiotensin II (Ang II) stimulation. SIRT3 was knocked down with siRNA technology. SIRT3 promoter activity and expression, as well as mitochondrial function, were measured. Male wild-type (WT) and SIRT3 knockout (KO) mice were intraperitoneally injected with NaHS followed by transverse aortic constriction (TAC). Myocardium sections were stained with Sirius red. Hydroxyproline content, collagen I and collagen III, α-smooth muscle actin (α-SMA), and dynamin-related protein 1 (DRP1) expression were measured both in vitro and in vivo. We found that NaHS enhanced SIRT3 promoter activity and increased SIRT3 mRNA expression. NaHS inhibited cell proliferation and hydroxyproline secretion, decreased collagen I, collagen III, α-SMA, and DRP1 expression, alleviated oxidative stress, and improved mitochondrial respiration function and membrane potential in Ang II-stimulated cardiac fibroblasts, which were unavailable after SIRT3 was silenced. In vivo, NaHS reduced hydroxyproline content, ameliorated perivascular and interstitial collagen deposition, and inhibited collagen I, collagen III, and DRP1 expression in the myocardium of WT mice but not SIRT3 KO mice with TAC. Altogether, NaHS attenuated myocardial fibrosis through oxidative stress inhibition via a SIRT3-dependent manner.


Asunto(s)
Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Sirtuina 3/metabolismo , Actinas/metabolismo , Animales , Colágeno/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Sulfuro de Hidrógeno/uso terapéutico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Noqueados , Miocardio/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Sirtuina 3/antagonistas & inhibidores , Sirtuina 3/deficiencia , Transcripción Genética/efectos de los fármacos
16.
Biochem Pharmacol ; 192: 114665, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34181898

RESUMEN

Sirtuin3 (SIRT3) is involved in reactive oxygen species (ROS), cell metabolism, apoptosis and inflammation. However, the exact role of SIRT3 in macrophages during pathophysiological process of atherosclerosis remains unclear. The present study was to investigate the possible effects and mechanisms of SIRT3 on lipid uptake and foam cells transforming in oxidized low-density lipoprotein (ox-LDL)-stimulated macrophages. Compared with wild-type (WT) mice, SIRT3 deficiency further increased foam cell formation and cellular cholesterol accumulation, exacerbated oxidative stress, impaired mitochondrial permeability potential, decreased optic atrophy 1 (OPA1) but enhanced dynamin-related protein 1 (DRP1) expression, and promoted NLR family pyrin domain-containing protein 3 (NLRP3) activation in ox-LDL-stimulated macrophages from SIRT3 knockout (KO) mice. Dihydromyricetin (DMY), a potential compound to enhance SIRT3 expression, significantly inhibited cellular cholesterol accumulation, suppressed foam cell formation, improved mitochondrial function, attenuated oxidative stress, and alleviated NLRP3 activation in ox-LDL-stimulated macrophages. Moreover, above protective effects of DMY was unavailable in macrophages from SIRT3 KO mice. Collectively, the study demonstrated the protective role of SIRT3 against oxidative stress and NLRP3 inflammasome in cholesterol accumulation and foam cell formation of macrophages with ox-LDL-stimulation, which is beneficial to provide novel strategy for atherosclerosis prevention and treatment.


Asunto(s)
Colesterol/metabolismo , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Oxidativo/fisiología , Sirtuina 3/deficiencia , Animales , Células Espumosas/patología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sirtuina 3/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda