Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Lab Invest ; 102(5): 505-514, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35066566

RESUMO

LncRNAs and miRNAs are correlated with the pathogenesis of myocardial ischemia-reperfusion injury (MIRI). Whether lncRNA ROR or miR-185-5p plays a crucial role in MIRI is still unclear. In in-vitro, human cardiac myocytes (HCMs) were treated with hypoxia/reoxygenation (H/R). Wistar rats were used to set up an in-vitro I/R model by means of recanalization after ligation. Evaluation of the myocardial injury marker lactate dehydrogenase (LDH) in HCMs cells was performed. The expression of miR-185-5p and ROR, IL-1ß, and IL-18 were detected by qRT-PCR. ELISA was also performed to evaluate the secretion of IL-1ß and IL-18. Western blotting was carried out to determine CDK6, NLRP3, GSDMD-N, ASC, and cleaved-caspase1 protein expression. The relationship between miR-185-5p and CDK6 or ROR was confirmed by a dual-luciferase reporter assay. Our findings revealed that H/R treated HCMs showed a significantly decreased miR-185-5p expression and increased expression of CDK6 and ROR. ROR knockdown reduced H/R induced pyroptosis and inflammation, while knockdown of miR-185-5p accelerated the effect. Furthermore, miR-185-5p was negatively regulated and absorbed by ROR in HCMs. Overexpression of miR-185-5p reversed the H/R-induced cell pyroptosis and upregulation of LDH, IL-1ß, and IL-18. In HCMs, miR-185-5p was also negatively regulated and related to CDK6 expression. Moreover, overexpression of CDK6 significantly inhibited the effects of miR-185-5p mimics on the inflammatory response and pyroptosis of HCMs. Knockdown of ROR alleviated H/R-induced myocardial injury by elevating miR-185-5p and inhibiting CDK6 expression. Taken together, our results show that the ROR/miR-185-5p/CDK6 axis modulates cell pyroptosis induced by H/R and the inflammatory response of HCMs.


Assuntos
MicroRNAs , Traumatismo por Reperfusão Miocárdica , RNA Longo não Codificante , Animais , Hipóxia , Interleucina-18 , MicroRNAs/genética , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , RNA Longo não Codificante/genética , Ratos , Ratos Wistar
2.
3 Biotech ; 11(7): 335, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34221806

RESUMO

Limb ischemia reperfusion (I/R) triggers local or systemic injury, and whether the process is mediated by pyroptosis remains unclear, we aimed to explore whether pyroptosis was involved in the process of rapamycin alleviating lung injury induced by I/R and investigate the molecular mechanisms. The histopathology of lung injury induced by I/R was confirmed by hematoxylin-eosin (HE) staining, and malondialdehyde (MDA), superoxide dismutase (SOD), and the expression of pyroptosis related molecules were detected. RNA sequencing was used to mine key long non-coding RNAs (lncRNAs). The model of lipopolysaccharide (LPS)-induced L2 cell damage was also used to explore the effect and mechanism of rapamycin on lncRNA. Rapamycin treatment alleviated I/R-induced lung histopathologically injury and increased the concentration of MDA while decreased activity of SOD and expression of NLRP3, Caspase-1, interleukin-1ß (IL-1ß), and IL-18 in rat. A total of 63 differentially expressed lncRNAs (DElncRNAs) were identified from IR + Rap group compared with IR group, and these DElncRNAs were mainly involved in cell adhesion molecules (CAMs) and endocytosis pathway. The lncRNA LOC102553434 and its target gene MMP9 were most significantly up-regulated in I/R-injured rat. In vitro experiments showed that LPS induction caused a significant increase in LOC102553434, MMP9, IL-1ß, and IL-18 in L2 cells, but rapamycin treatment significantly reversed the effects. After interfering with the expression of LOC102553434 in the LPS-injured cells pretreated with rapamycin, cell proliferation significantly increased, and the expression of MMP, NLRP3 and caspase-1 were significantly decreased. Rapamycin protects the lung from limb I/R injury by regulating LOC102553434 expression and inhibiting pyroptosis pathway. LOC102553434 plays a role in promoting pyroptosis and thus provides a target for clinical treatment of I/R-induced lung injury. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s13205-021-02708-9.

3.
Kaohsiung J Med Sci ; 37(7): 594-603, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33611829

RESUMO

Chronic obstructive pulmonary disease (COPD) is a chronic respiratory disorder. Although numerous studies on COPD have been conducted, therapeutic strategies for COPD are limited, and its pathological mechanism is still unclear. The present study aimed to explore the role of DNA methyltransferase 3a (DNMT3a) in dendritic cells (DCs) and the possible role of the Th-17/Treg cell balance in COPD. Immature DCs (iDCs) were induced and cocultured with CD4+ T cells. An in vitro COPD model was established by treatment with cigarette smoke extract (CSE). DNMT3a or allograft inflammatory factor 1 (AIF1) and c-Jun N-terminal kinase (JNK) were inhibited and overexpressed, respectively, by transfection with sh-DNMT3a or sh-AIF1 and JNK overexpression plasmids. The 3- (4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was used to measure cell viability. The Th17/Treg cell ratio was determined by flow cytometry. The expression levels of DNMT3a, c-Jun and AIF1 were measured using RT-qPCR or western blotting. Chromatin immunoprecipitation (CHIP) was used to confirm the interaction between c-Jun and the AIF1 promoter region. CSE stimulation promoted the expression of DNMT3a, and AIF1, and the ratio of p-c-Jun/c-Jun in iDCs. Besides, the iDC-mediated differentiation of Th17 cells was in a dose-dependent manner. However, knockdown of DNMT3a or AIF1 reversed the above effects caused by CSE. Inhibition of c-Jun signaling by treatment with the JNK inhibitor SP600125 also suppressed the iDC-mediated differentiation of Th17 cells, which was promoted by CSE. CHIP analysis showed that c-Jun could bind to the promoter region of AIF1. DNMT3a could regulate the iDC-mediated Th17/Treg balance by regulating the c-Jun/AIF1 axis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , DNA Metiltransferase 3A/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fumaça , Fumar/efeitos adversos , Linfócitos T Reguladores/metabolismo , Aloenxertos , Animais , Antracenos/farmacologia , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Células Cultivadas , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doença Pulmonar Obstrutiva Crônica/genética , Transdução de Sinais , Sais de Tetrazólio/farmacologia , Células Th17/metabolismo , Tiazóis/farmacologia
4.
RSC Adv ; 10(67): 40690-40696, 2020 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-35519227

RESUMO

Using TiOiPr4 with a pyrazole ligand for one-pot LA polymerization improved catalytic activity compared with using TiOiPr4 only. At 60 °C, TiOiPr4 with furPz exhibited a higher catalytic activity (approximately 3-fold) than TiOiPr4. At room temperature, TiOiPr4 with BuPz exhibited a higher catalytic activity (approximately 17-fold) than TiOiPr4. High molecular mass PLA (M nGPC = 51 100, and D = 1.10) could be produced by using TiOiPr4 with furPz in melt polymerization ([TiOiPr4] : [furPz] = 1000 : 1 : 1 at 100 °C, 240 min). The crystal structure of MePz2Ti2OiPr7 revealed the cooperative activation between two Ti atoms during LA polymerization.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA