Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Arq Bras Cardiol ; 121(6): e20230675, 2024.
Article in Portuguese, English | MEDLINE | ID: mdl-38958296

ABSTRACT

BACKGROUND: The anthracycline chemotherapeutic antibiotic doxorubicin (DOX) can induce cumulative cardiotoxicity and lead to cardiac dysfunction. Long non-coding RNAs (lncRNAs) can function as important regulators in DOX-induced myocardial injury. OBJECTIVE: This study aims to investigate the functional role and molecular mechanism of lncRNA OXCT1 antisense RNA 1 (OXCT1-AS1) in DOX-induced myocardial cell injury in vitro. METHODS: Human cardiomyocytes (AC16) were stimulated with DOX to induce a myocardial cell injury model. OXCT1-AS1, miR-874-3p, and BDH1 expression in AC16 cells were determined by RT-qPCR. AC16 cell viability was measured by XTT assay. Flow cytometry was employed to assess the apoptosis of AC16 cells. Western blotting was used to evaluate protein levels of apoptosis-related markers. Dual-luciferase reporter assay was conducted to verify the binding ability between miR-874-3p and OXCT1-AS1 and between miR-874-3p and BDH1. The value of p<0.05 indicated statistical significance. RESULTS: OXCT1-AS1 expression was decreased in DOX-treated AC16 cells. Overexpression of OXCT1-AS1 reversed the reduction of cell viability and promotion of cell apoptosis caused by DOX. OXCT1-AS1 is competitively bound to miR-874-3p to upregulate BDH1. BDH1 overexpression restored AC16 cell viability and suppressed cell apoptosis under DOX stimulation. Knocking down BDH1 reversed OXCT1-AS1-mediated attenuation of AC16 cell apoptosis under DOX treatment. CONCLUSION: LncRNA OXCT1-AS1 protects human myocardial cells AC16 from DOX-induced apoptosis via the miR-874-3p/BDH1 axis.


FUNDAMENTO: O antibiótico quimioterápico antraciclina doxorrubicina (DOX) pode induzir cardiotoxicidade cumulativa e levar à disfunção cardíaca. RNAs não codificantes longos (lncRNAs) podem funcionar como importantes reguladores na lesão miocárdica induzida por DOX. OBJETIVO: Este estudo tem como objetivo investigar o papel funcional e o mecanismo molecular do RNA antisense lncRNA OXCT1 1 (OXCT1-AS1) na lesão celular miocárdica induzida por DOX in vitro. MÉTODOS: Cardiomiócitos humanos (AC16) foram estimulados com DOX para induzir um modelo de lesão celular miocárdica. A expressão de OXCT1-AS1, miR-874-3p e BDH1 em células AC16 foi determinada por RT-qPCR. A viabilidade das células AC16 foi medida pelo ensaio XTT. A citometria de fluxo foi empregada para avaliar a apoptose de células AC16. Western blotting foi utilizado para avaliar os níveis proteicos de marcadores relacionados à apoptose. O ensaio repórter de luciferase dupla foi conduzido para verificar a capacidade de ligação entre miR-874-3p e OXCT1-AS1 e entre miR-874-3p e BDH1. O valor de p<0,05 indicou significância estatística. RESULTADOS: A expressão de OXCT1-AS1 foi diminuída em células AC16 tratadas com DOX. A superexpressão de OXCT1-AS1 reverteu a redução da viabilidade celular e a promoção da apoptose celular causada pela DOX. OXCT1-AS1 está ligado competitivamente ao miR-874-3p para regular positivamente o BDH1. A superexpressão de BDH1 restaurou a viabilidade das células AC16 e suprimiu a apoptose celular sob estimulação com DOX. A derrubada do BDH1 reverteu a atenuação da apoptose de células AC16 mediada por OXCT1-AS1 sob tratamento com DOX. CONCLUSÃO: LncRNA OXCT1-AS1 protege células miocárdicas humanas AC16 da apoptose induzida por DOX através do eixo miR-874-3p/BDH1.


Subject(s)
Apoptosis , Doxorubicin , MicroRNAs , Myocytes, Cardiac , RNA, Long Noncoding , Humans , Doxorubicin/pharmacology , RNA, Long Noncoding/genetics , Apoptosis/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Survival/drug effects , Reproducibility of Results , Blotting, Western , Flow Cytometry , RNA, Competitive Endogenous
2.
Inhal Toxicol ; 36(4): 217-227, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38713814

ABSTRACT

OBJECTIVE: The present work concentrated on validating whether sinomenine alleviates bleomycin (BLM)-induced pulmonary fibrosis, inflammation, and oxidative stress. METHODS: A rat model of pulmonary fibrosis was constructed through intratracheal injection with 5 mg/kg BLM, and the effects of 30 mg/kg sinomenine on pulmonary inflammation, fibrosis, apoptosis, and 4-hydroxynonenal density were evaluated by hematoxylin and eosin staining, Masson's trichrome staining, TUNEL staining, and immunohistochemistry. Hydroxyproline content and concentrations of inflammatory cytokines and oxidative stress markers were detected using corresponding kits. MRC-5 cells were treated with 10 ng/ml PDGF, and the effects of 1 mM sinomenine on cell proliferation were assessed by EdU assays. The mRNA expression of inflammatory cytokines and the protein levels of collagens, fibrosis markers, and key markers involved in the TLR4/NLRP3/TGFß signaling were tested with RT-qPCR and immunoblotting analysis. RESULTS: Sinomenine attenuated pulmonary fibrosis and inflammation while reducing hydroxyproline content and the protein expression of collagens and fibrosis markers in BLM-induced pulmonary fibrosis rats. Sinomenine reduced apoptosis in lung samples of BLM-challenged rats by increasing Bcl-2 and reducing Bax and cleaved caspase-3 protein expression. In addition, sinomenine alleviated inflammatory response and oxidative stress in rats with pulmonary fibrosis induced by BLM. Moreover, sinomenine inhibited the TLR4/NLRP3/TGFß signaling pathway in lung tissues of BLM-stimulated rats. Furthermore, TLR4 inhibitor, TAK-242, attenuated PDGF-induced fibroblast proliferation and collagen synthesis in MRC-5 cells. CONCLUSION: Sinomenine attenuates BLM-caused pulmonary fibrosis, inflammation, and oxidative stress by inhibiting the TLR4/NLRP3/TGFß signaling, indicating that sinomenine might become a therapeutic candidate to treat pulmonary fibrosis.


Subject(s)
Bleomycin , Morphinans , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Pulmonary Fibrosis , Signal Transduction , Toll-Like Receptor 4 , Transforming Growth Factor beta , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Morphinans/pharmacology , Morphinans/therapeutic use , Bleomycin/toxicity , Oxidative Stress/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta/metabolism , Male , Signal Transduction/drug effects , Rats, Sprague-Dawley , Cell Line , Rats , Humans , Lung/drug effects , Lung/pathology , Lung/metabolism
3.
Mol Med ; 29(1): 144, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880599

ABSTRACT

BACKGROUND: Ferroptosis plays an essential role in lipopolysaccharide (LPS)-induced acute lung injury (ALI). Meteorin-like/Meteorin-ß (Metrnß) is a protein secreted by skeletal muscle and adipose tissue and plays a role in cardiovascular diseases. However, its role in acute lung injury has not been elucidated. METHODS: In this study, we used an adenovirus (Ad) delivery system to overexpress or knockdown Metrnß in lung tissue to examine the role of Metrnß in LPS-induced acute lung injury. RESULTS: We found that ferroptosis was increased during LPS-induced ALI. The expression of Metrnß was reduced in ALI lung tissue. Overexpression of Metrnß in lung tissue alleviated LPS-induced lung injury, inflammation, and ferroptosis. Moreover, Metrnß knockout in lung tissue accelerated LPS-induced ALI, inflammation, and ferroptosis. We also cultured MLE-12 cells and transfected the cells with Ad-Metrnß or Metrnß siRNA. Metrnß overexpression ameliorated LPS-induced MLE cell death, inflammation and ferroptosis, while Metrnß knockdown aggregated cell survival and decreased inflammation and ferroptosis. Moreover, we found that Metrnß enhanced ferroptosis-related Gpx4 expression and reduced ferroportin and ferritin levels. Furthermore, we found that Metrnß positively regulated SIRT1 transcription thus inhibited P53, increased SLC7A11 expression. When we used the ferroptosis inhibitor ferrostatin-1, the deteriorating effects of Metrnß knockout were abolished in ALI mice. Moreover, SIRT1 knockout also abolished the protective effects of Metrnß overexpression in vivo. CONCLUSIONS: Taken together, Metrnß could protect LPS-induced ALI by activating SIRT1-P53- SLC7A11 mediated ferroptosis inhibition.


Subject(s)
Acute Lung Injury , Ferroptosis , Animals , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Suppressor Protein p53
4.
Biomark Med ; 15(6): 401-412, 2021 04.
Article in English | MEDLINE | ID: mdl-33733809

ABSTRACT

Aim: We aimed to investigate the association of long noncoding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) expression with acute respiratory distress syndrome (ARDS) risk and its prognostic value for 28-day mortality in sepsis patients. Materials & methods: LncRNA PVT1 expression from 109 sepsis patients and 100 health controls was detected. General sepsis severity was assessed using acute physiology and chronic health evaluation II score and sequential organ failure assessment score. Results: LncRNA PVT1 had an acceptable predictive value for higher ARDS risk, then was identified as an independent risk factor for sepsis ARDS; LncRNA PVT1 expression positively correlated with general disease severity in sepsis patients; LncRNA PVT1 was overexpressed in 28-day deaths compared with 28-day survivors in sepsis patients. Conclusion: LncRNA PVT1 may facilitate the surveillance of ARDS, general disease severity and the prediction of mortality in sepsis patients.


Subject(s)
RNA, Long Noncoding/genetics , Respiratory Distress Syndrome/mortality , Sepsis/mortality , Case-Control Studies , Female , Humans , Male , Middle Aged , Prognosis , RNA, Long Noncoding/blood , ROC Curve , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Risk Factors , Sepsis/complications , Severity of Illness Index
5.
J Clin Lab Anal ; 34(1): e23013, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31495986

ABSTRACT

BACKGROUND: This study aimed to investigate the correlation of pro-angiogenic microRNA (miRNA) expressions with rapid angiographic stenotic progression (RASP) and restenosis risks in coronary artery disease (CAD) patients underwent percutaneous coronary intervention (PCI) with drug-eluting stents (DES). METHODS: A total of 286 CAD patients underwent PCI with DES were consecutively recruited in this study. Plasma samples were collected before PCI operation, and 14 pro-angiogenic miRNAs were measured by real-time quantitative reverse transcription-polymerase chain reaction. Rapid angiographic stenotic progression at nontarget lesions and restenosis at stented lesions were evaluated by quantitative coronary angiography at 12 months after PCI operation. RESULTS: The occurrence rates of RASP and restenosis were 39.5% and 22.4%, respectively. Let-7f, miR-19a, miR-19b-1, miR-92a, miR-126, miR-210, and miR-296 were decreased in RASP patients than non-RASP patients, among which let-7f, miR-19a, miR-126, miR-210, and miR-296 independently correlated with lower RASP occurrence by multivariate analysis, followed by receiver-operating characteristic (ROC) curve exhibited that these five miRNAs showed great value in predicting RASP risk with area under curve (AUC) 0.879 (95% CI: 0.841-0.917). Besides, let-7f, miR-19a, miR-92a, miR-126, miR-130a, and miR-210 were reduced in restenosis patients than non-restenosis patients, among them miR-19a, miR-126, miR-210, and miR-378 independently correlated with lower restenosis occurrence by multivariate analysis, followed by ROC curve disclosed that these four miRNAs had good value in predicting restenosis risk with AUC 0.776 (95% CI: 0.722-0.831). CONCLUSIONS: Circulating let-7f, miR-19a, miR-126, miR-210, and miR-296 independently correlate with reduced RASP risk, while miR-19a, miR-126, miR-210, and miR-378 independently correlate with decreased restenosis risk in CAD patients underwent PCI with DES.


Subject(s)
Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Coronary Angiography , Coronary Artery Disease/surgery , Coronary Restenosis/etiology , Coronary Stenosis/diagnostic imaging , Gene Expression Regulation , Percutaneous Coronary Intervention/adverse effects , Biomarkers/blood , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Coronary Restenosis/blood , Coronary Restenosis/genetics , Coronary Stenosis/blood , Coronary Stenosis/genetics , Female , Humans , Male , Middle Aged , Neovascularization, Physiologic/genetics , ROC Curve , Risk Factors
6.
J Clin Lab Anal ; 33(6): e22917, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115097

ABSTRACT

AIM: This study aimed to investigate the correlation of lnc-ANRIL/miR-125a axis with risk, severity, inflammation, and prognosis of sepsis. METHODS: A hundred and twenty-six sepsis patients and 125 healthy controls were recruited, and then, blood samples were collected, and plasma was separated for lnc-ANRIL, miR-125a, lnc-ANRIL/miR-125a axis, and inflammatory cytokine level detections. In addition, basic characteristics, 28-day mortality, and accumulating survival of sepsis patients were recorded. RESULTS: Plasma lnc-ANRIL expression was increased, miR-125a expression was decreased, and lnc-ANRIL/miR-125a axis level was elevated in sepsis patients compared with healthy controls, and all of them had good value for predicting sepsis risk with AUCs of 0.800, 0.817, and 0.843, respectively. Lnc-ANRIL and lnc-ANRIL/miR-125a axis were positively correlated with biochemical index levels including CRP and PCT levels, disease severity scale scores, and pro-inflammatory cytokine levels in sepsis patients, while miR-125a displayed the opposite trend. Lnc-ANRIL and lnc-ANRIL/miR-125a axis expressions were elevated, while miR-125a expression was declined in deaths compared with survivors, and all of them predicted 28-day mortality in sepsis patients with AUCs of 0.765, 0.745, and 0.785, respectively. Subsequently, the Kaplan-Meier analysis revealed that patients with high lnc-ANRIL, low miR-125a, and high lnc-ANRIL/miR-125a axis levels presented with worse accumulating survival. In addition, multivariate regression model analyses revealed that high plasma lnc-ANRIL/miR-125a axis was an independent predictive factor for both increased 28-day mortality and worse accumulating survival. CONCLUSION: Circulating lnc-ANRIL/miR-125a axis was upregulated and could serve as a biomarker for severity, inflammation, and prognosis in sepsis patients.


Subject(s)
MicroRNAs/blood , RNA, Long Noncoding/blood , Sepsis/etiology , Sepsis/mortality , Adult , Aged , Case-Control Studies , Cell-Free Nucleic Acids/blood , Cytokines/blood , Female , Humans , Inflammation/blood , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Prognosis , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...