1,234 results on '"HYPOXIA/REOXYGENATION"'
Search Results
2. Cav3.2 channel regulates cerebral ischemia/reperfusion injury: a promising target for intervention.
- Author
-
Dai, Feibiao, Hu, Chengyun, Li, Xue, Zhang, Zhetao, Wang, Hongtao, Zhou, Wanjun, Wang, Jiawu, Geng, Qingtian, Dong, Yongfei, and Tang, Chaoliang
- Abstract
Calcium influx into neurons triggers neuronal death during cerebral ischemia/reperfusion injury. Various calcium channels are involved in cerebral ischemia/reperfusion injury. Cav3.2 channel is a main subtype of T-type calcium channels. T-type calcium channel blockers, such as pimozide and mibefradil, have been shown to prevent cerebral ischemia/reperfusion injury-induced brain injury. However, the role of Cav3.2 channels in cerebral ischemia/reperfusion injury remains unclear. Here, in vitro and in vivo models of cerebral ischemia/reperfusion injury were established using middle cerebral artery occlusion in mice and high glucose hypoxia/reoxygenation exposure in primary hippocampal neurons. The results showed that Cav3.2 expression was significantly upregulated in injured hippocampal tissue and primary hippocampal neurons. We further established a Cav3.2 gene-knockout mouse model of cerebral ischemia/reperfusion injury. Cav3.2 knockout markedly reduced infarct volume and brain water content, and alleviated neurological dysfunction after cerebral ischemia/reperfusion injury. Additionally, Cav3.2 knockout attenuated cerebral ischemia/reperfusion injury-induced oxidative stress, inflammatory response, and neuronal apoptosis. In the hippocampus of Cav3.2-knockout mice, calcineurin overexpression offset the beneficial effect of Cav3.2 knockout after cerebral ischemia/reperfusion injury. These findings suggest that the neuroprotective function of Cav3.2 knockout is mediated by calcineurin/nuclear factor of activated T cells 3 signaling. Findings from this study suggest that Cav3.2 could be a promising target for treatment of cerebral ischemia/reperfusion injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. miR-330-5p 调控 Nox4 对缺氧复氧大鼠胚胎心肌细胞 损伤影响的实验研究.
- Author
-
黄 毅 and 王琮翰
- Abstract
Copyright of Journal of Modern Laboratory Medicine is the property of Journal of Modern Laboratory Medicine Editorial Department and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
4. Role and mechanism of pumilio homolog 1 in hypoxia/reoxygenation induced injury of human renal tubular epithelial cells
- Author
-
Sheng-guo Hu, Yi Guo, Chao Yuan, You-kong Li, Min Wang, and Min Zhu
- Subjects
pumilio homolog 1 ,human kidney-2 ,hypoxia/reoxygenation ,oxidative stress ,apoptosis ,Internal medicine ,RC31-1245 - Abstract
Objective To explore the role and mechanism of pumilio homolog 1 (PUM1) in hypoxia/reoxygenation induced cell injury in human renal tubular epithelial cells (HK-2). Methods A hypoxia/reoxygenation (H/R) model of HK-2 cells was established in vitro. PUM1 expression was knocked down through small interfering RNA (siRNA). Cells were randomized into three groups of control, hypoxia/reoxygenation (H/R) and H/R+siRNA. Western blot (WB) method was used for detecting the expression level of PUM1 protein. Cell Count Kit 8 (CCK-8) was employed for detecting cell viability. Hydrogen peroxide (H2O2), malondialdehyde (MDA) and superoxide dismutase (SOD) were used for evaluating the levels of oxidative stress. Flow cytometry was utilized for detecting the level of cell apoptosis. Results As compared with control group, protein expression level of PUM1 in H/R 3 h group (1.76 ± 0.11 vs 0.98 ± 0.05), H/R 6 h group (2.89 ± 0.14 vs 0.98 ± 0.05) and H/R 12 h group (3.78 ± 0.08 vs 0.98 ± 0.05) gradually spiked with the prolongation of hypoxic time. As compared with H/R group, knocking down the expression of PUM1 significantly improved the cell viability (73.67 ± 3.42 vs 29.60 ± 2.94), oxidative stress [H2O2:(13.53 ± 0.85)μmol/L vs (22.43 ± 1.12)μmol/L, MDA: (16.03 ± 0.70)μmol/L vs (31.20 ± 1.50)μmol/L, SOD: (34670 ± 1800)U/L vs (5730 ± 1220)U/L] and apoptotic level [(14.89 ± 1.65)% vs (39.71 ± 1.94)%] after H/R in H/R+si-PUM1 group. Conclusion PUM1 is up-regulated in H/R induced HK-2 cells and its inhibition may alleviate H/R injury through reducing oxidative stress and lowering cell apoptosis levels.
- Published
- 2024
- Full Text
- View/download PDF
5. Regulation of receptor interacting protein kinase 1-mediated apoptosis by cordycepin attenuated hypoxia/reoxygenation-induced renal tubular epithelial cell injury
- Author
-
Xiu-tao Han, Xiu-zhao Fan, Zhi-bo Zhao, Jing-yu Zhao, Jun-hu Li, Fang Zhang, Gai-ying Jiang, Li-jun Zhang, Fa-hui Chen, Meng-qi Bai, and Xiao-shuang Zhou
- Subjects
acute kidney injury ,cordycepin ,hypoxia/reoxygenation ,network pharmacology ,apoptosis ,receptor interacting protein kinase 1 ,Internal medicine ,RC31-1245 - Abstract
Objective To explore the therapeutic mechanism of cordycepin for acute kidney injury (AKI) based upon gene chips in gene expression omnibus (GEO) database, network pharmacology, molecular docking and in vitro experiments. Methods Targeted genes of AKI were obtained from two databases, GEO (GSE87025) and GeneCards. Cordycepin-related targets were retrieved from three databases of TargetNet, BATMAN and GeneCards. The common targets of AKI and cordycepin were screened by taking the intersection of two sets. Molecular docking was performed for cordycepin and key targets after gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Cell experiments were performed for control, hypoxia/reoxygenation and hypoxia/reoxygenation+cordycepin groups for experimental verifications of the relevant targets. Results There were 12 common targets of AKI and cordycepin. GO functional annotation results indicated that these targets participated predominantly in negative regulation of apoptotic signaling pathway and necroptosis process. KEGG enrichment analysis results showed that they played some roles in signaling pathways of Toll-like receptor, necroptosis, nucleotide-binding oligomerization domain (NOD)-like receptor, nuclear factor-κB (NF-kB), tumor necrosis factor (TNF) and cell apoptosis, etc. Optimal binding energy between cordycepin and receptor interacting protein kinase 1 (RIPK1) in molecular docking was -7.1, denoting potent binding activity. Western blot indicated that cordycepin down-regulated the expressions of RIPK1, bcl-2 associated x protein (Bax) and caspase 3 and up-regulated B cell lymphoma/lewkmia-2(Bcl-2). Cell immunofluorescence results revealed that cordycepin down-regulated the expression of RIPK1. Conclusion Cordycepin may alleviate hypoxia/reoxygenation-induced renal tubular epithelial cell injury through regulating RIPK1-mediated apoptosis. Thus it achieves the therapeutic efficacy for AKI.
- Published
- 2024
- Full Text
- View/download PDF
6. Comparison of Cardioprotective Potential of Cannabidiol and β-Adrenergic Stimulation Against Hypoxia/Reoxygenation Injury in Rat Atria and Ventricular Papillary Muscles.
- Author
-
Pędzińska-Betiuk, Anna, Gergs, Ulrich, Weresa, Jolanta, Remiszewski, Patryk, Harasim-Symbor, Ewa, and Malinowska, Barbara
- Subjects
- *
RIGHT heart atrium , *LEFT heart atrium , *CARDIOVASCULAR diseases , *ATRIUMS (Architecture) , *REPERFUSION injury - Abstract
Background: Hypoxia is one of the most significant pathogenic factors in cardiovascular diseases. Preclinical studies suggest that nonpsychoactive cannabidiol (CBD) and β-adrenoceptor stimulation might possess cardioprotective potential against ischemia-reperfusion injury. The current study evaluates the influence of hypoxia-reoxygenation (H/R) on the function of atria and ventricular papillary muscles in the presence of CBD and the nonselective β-adrenoceptor agonist isoprenaline (ISO). Methods: The concentration curves for ISO were constructed in the presence of CBD (1 µM) before or after H/R. In chronic experiments (CBD 10 mg/kg, 14 days), the left atria isolated from spontaneously hypertensive (SHR) and their normotensive control (WKY) rats were subjected to H/R following ISO administration. Results: Hypoxia decreased the rate and force of contractions in all compartments. The right atria were the most resistant to hypoxia regardless of prior β-adrenergic stimulation. Previous β-adrenergic stimulation improved recovery in isolated left atria and right (but not left) papillary muscles. Acute (but not chronic) CBD administration increased the effects of ISO in left atria and right (but not left) papillary muscles. Hypertension accelerates left atrial recovery during reoxygenation. Conclusions: H/R directly modifies the function of particular cardiac compartments in a manner dependent on cardiac region and β-adrenergic prestimulation. The moderate direct cardioprotective potential of CBD and β-adrenergic stimulation against H/R is dependent on the cardiac region, and it is less than in the whole heart with preserved coronary flow. In clinical terms, our research expands the existing knowledge about the impact of cannabidiol on cardiac ischemia, the world′s leading cause of death. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. 南瓜多糖调控 Nrf2/γ-GCS 通路对缺氧/复氧心肌细胞损伤的影响.
- Author
-
郑婉, 杨珊珊, 林云, 魏俊萍, and 颜亚妮
- Subjects
- *
MEMBRANE potential , *CELL morphology , *MITOCHONDRIAL membranes , *POLYSACCHARIDES , *PROTEIN expression - Abstract
Objective:To investigate the impact of pumpkin polysaccharide (PP) on hypoxia/reoxygenation (H/R) -induced myocardial cell (H9C2) injury by regulating Nrf2/γ-GCS pathway. Methods:HR cell models were established, and the concentration of pumpkin polysaccharide (PP) was determined. H9C2 cells were grouped into Control group, H/R group, pumpkin polysaccharide group (PP group, 2 mg/L PP), Nrf2 inhibitor group (ML385 group, 2.0 μmol/L ML385), and PP+ML385 group (2.0 mg/L PP+ 2.0 μmol/L ML385), the apoptosis, mitochondrial membrane potential changes, oxidative stress (LDH, MDA, SOD, ROS), the levels of inflammatory factors (TNF-α, IL-6, IL-1β) and the protein expressions of Nrf2 and γ-GCS in H9C2 cells were detected, respectively. Results:Compared with Control group, H/R group had changes in cell morphology, the cell injury and mitochondrial membrane potential decreased, the H9C2 cell viability, SOD and protein expression levels of Nrf2 and γ -GCS were significantly decreased, while the apoptosis rate, LDH, MDA, ROS, and levels of TNF-α, IL-6 and IL-1β were significantly increased (P<0.05) ; compared with H/R group, the cell injury in PP group was reduced, the mitochondrial membrane potential was restored, the H9C2 cell viability, SOD and protein expression levels of Nrf2 and γ-GCS were significantly increased, and the apoptosis rate, LDH, MDA, ROS, and levels of TNF-α, IL-6 and IL-1β were significantly decreased (P<0.05), the effect of ML385 was opposite to that of PP, and ML385 could reverse the antioxidant and anti-inflammatory effects of PP. Conclusion:PP up-regulates the Nrf2/γ-GCS pathway to exert antioxidant and anti-inflammatory effects, and reduces H/R-induced H9C2 cell injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. ADAM10 Alleviates Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Activating the Notch Signaling Pathway.
- Author
-
Xu, Tengfei, Jiang, Shan, Liu, Tongtong, Han, Shiqiang, and Wang, Yueqiang
- Abstract
The occurrence of myocardial ischemia/reperfusion injury is commonly observed during cardiac surgery; however, there remains a dearth of effective therapeutic strategies to mitigate this injury. The a disintegrin and metallopeptidase domain 10 (ADAM10) is a transmembrane protein anchored on the cell membrane surface, and its precise mechanism of action in myocardial ischemia/reperfusion injury remains incompletely understood. This study aims to investigate the impact of ADAM10 on cardiomyocyte injury induced by hypoxia/reoxygenation (H/R) and elucidate the underlying mechanisms. The ADAM10 overexpression plasmid was transfected into H9c2 cells, which were subsequently treated with the Notch signaling pathway inhibitor DAPT and cultured under H/R conditions. Cell proliferation activity was assessed using the CCK-8 assay. The levels of LDH, SOD, and MDA were quantified through colorimetric analysis. The levels of ROS and the rate of apoptosis were measured using flow cytometry. The morphological changes in the nucleus of H9c2 cells were observed by employing Hoechst 33258 staining. The mRNA expression levels of ADAM10, Notch1, NICD, and Hes1 in H9c2 cells were determined using qRT-PCR. The expressions of Notch signaling pathway and apoptosis-related proteins were analyzed by Western blot. Overexpression of ADAM10 provided protection to H9c2 cells against injury induced by H/R, leading to an increase in SOD levels and alleviation of oxidative stress caused by the accumulation of ROS and the decrease of SOD activity. Meanwhile, overexpression of ADAM10 inhibited apoptosis in H9c2 cells exposed to H/R by regulating the expression of apoptosis-related proteins, such as Bax, Bcl-2 and Cleaved-caspase-3. Additionally, overexpression of ADAM10 facilitated the activation of the Notch1 signaling pathway in H9c2 cells exposed to H/R by upregulating the protein expression of Notch1, NICD, and Hes1. However, the protective effect of ADAM10 on H/R-induced H9c2 cells was partially reversed by DAPT. Our findings demonstrate that ADAM10 exerts protective effects in H/R-induced H9c2 cells by suppressing oxidative stress and apoptosis via the activation of the Notch signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Cirsilineol Protects Renal Tubular Epithelial Cells from Hypoxia/Reoxygenation Injury by Inhibiting Inflammation and Pyroptosis.
- Author
-
Huang Fei and Zi Liu
- Subjects
- *
EPITHELIAL cells , *IN vitro studies , *FLOW cytometry , *NF-kappa B , *KIDNEY tubules , *APOPTOSIS , *ENZYME-linked immunosorbent assay , *TOLL-like receptors , *FLAVONES , *CELL lines , *MOLECULAR structure , *CELL survival , *INFLAMMATION , *HYPOXEMIA , *IMMUNOBLOTTING - Abstract
To investigate the effects of Cirsilineol on cell viability, pyroptosis, and inflammation in hypoxia/reoxygenation-induced renal tubular cells, we utilized the renal tubular epithelial cell line HK2. The in vitro renal injury model was induced by hypoxia/reoxygenation. Cell viability in hypoxia/reoxygenation-induced HK2 cells was determined using a methylthiazolyldiphenyltetrazolium bromide assay and lactate dehydrogenase releasing kit. The effects of Cirsilineol on inflammation and pyroptosis in HK2 cells were evaluated using enzyme-linked immunosorbent assay, flow cytometry, and immunoblot assays. Our results demonstrated that Cirsilineol promoted survival in hypoxia/reoxygenation-induced tubular cells, inhibited inflammation, and reduced pyroptosis in these cells. Further investigation revealed that the protective effects of Cirsilineol are mediated by the suppression of the Toll-like receptor 4/nuclear factor kappa B pathway, which mitigates the cellular response to hypoxia/ reoxygenation injury. Our study suggests that Cirsilineol protects renal tubular epithelial cells from hypoxia/reoxygenation injury by inhibiting inflammation and pyroptosis, highlighting its potential therapeutic value in renal conditions characterized by ischemic injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. RP105 Attenuates Ischemia/Reperfusion-Induced Oxidative Stress in the Myocardium via Activation of the Lyn/Syk/STAT3 Signaling Pathway.
- Author
-
Yang, Jian, Zhai, Yuhong, Huang, Cuiyuan, Xiang, Zujin, Liu, Haiyin, Wu, Jingyi, Huang, Yifan, Liu, Li, Li, Wenqiang, Wang, Wei, Yang, Jun, and Zhang, Jing
- Subjects
- *
LACTATE dehydrogenase , *LABORATORY rats , *MYOCARDIAL ischemia , *REPERFUSION injury , *OXIDATIVE stress - Abstract
Although our previous studies have established the crucial role of RP105 in myocardial ischemia/reperfusion injury (MI/RI), its involvement in regulating oxidative stress induced by MI/RI remains unclear. To investigate this, we conducted experiments using a rat model of ischemia/reperfusion (I/R) injury. Adenovirus carrying RP105 was injected apically at multiple points, and after 72 h, the left anterior descending coronary artery was ligated for 30 min followed by 2 h of reperfusion. In vitro experiments were performed on H9C2 cells, which were transfected with recombinant adenoviral vectors for 48 h, subjected to 4 h of hypoxia, and then reoxygenated for 2 h. We measured oxidative stress markers, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities, as well as malondialdehyde (MDA) concentration, using a microplate reader. The fluorescence intensity of reactive oxygen species (ROS) in myocardial tissue was measured using a DHE probe. We also investigated the upstream and downstream components of the signal transducer and activator of transcription 3 (STAT3). Upregulation of RP105 increased SOD and GSH-Px activities, reduced MDA concentration, and inhibited ROS production in response to I/R injury in vivo and hypoxia reoxygenation (H/R) stimulation in vitro. The overexpression of RP105 led to a decrease in the myocardial enzyme LDH in serum and cell culture supernatant, as well as a reduction in infarct size. Additionally, left ventricular fraction (LVEF) and fractional shortening (LVFS) were improved in the RP105 overexpression group compared to the control. Upregulation of RP105 promoted the expression of Lyn and Syk and further activated STAT phosphorylation, which was blocked by PP2 (a Lyn inhibitor). Our findings suggest that RP105 can inhibit MI/RI-induced oxidative stress by activating STAT3 via the Lyn/Syk signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Salvianolic acid B protects against hypoxia/reoxygenation?induced renal tubular epithelial cells damage via regulating AMPK mediated ferroptosis.
- Author
-
ZHAO Jingyu, CHEN Fahui, HAO Yajie, HAN Xiutao, REN Xiya, TIAN Ruixue, and ZHOU Xiaoshuang
- Subjects
ENZYME-linked immunosorbent assay ,MEMBRANE potential ,ACUTE kidney failure ,MITOCHONDRIAL membranes ,AMP-activated protein kinases ,ELLAGIC acid - Abstract
Objective: To investigate the protective effects and mechanisms of salvianolic acid B (Sal B) on hypoxia/reoxygenation H/R-induced renal tubular epithelial cell damage. Methods In vitro experimental cells were divided into the normal control Con groupthe Sal B+Con groupthe H/R groupthe Sal B+H/R group as well as the Con groupthe H/R groupSal B+H/R groupthe H/R+CC group and the H/R+Sal B+CC group. HK-2 cell viability was detected by the CCK-8 method. Cell lysates were collected to measure the levels of GSH and malondialdehyde MDA using an enzyme-linked immunosorbent assay. The expressions of p-AMPK GPX4 ACSL4 and FSP1 were detected by Western blot. The expression of ferroptosis markers GPX4 and ACSL4 proteins was detected by cell immunofluorescence while mitochondrial membrane potential and cell apoptosis were measured by flow cytometry.Results The results of the in vitro experiments showed that Sal B had no significant toxicity on normal HK-2 cell viability within the concentration range of 20-160 μmol/L P>0.05. Compared with the model group Sal B at concentrations of 20 40 and 80 μmol/L dose-dependently increased the viability of HK-2 cells P<0.01. Compared with the model group MDA content and GSH content of the H/R+Sal B group were decreased p-AMPK GPX4 and FSP1 protein expression levels were increased ACSL4 protein expression was decreased mitochondrial membrane potential was significantly increased and apoptosis rate was significantly decreased P<0.05. Compared with the H/R+Sal B group the FSP1 and GPX4 protein expression level in the H/R+AMPK group was decreased the ACSL4 protein expression level was increased P< 0.05 and the mitochondrial membrane potential was significantly decreased P<0.05 and the addition of Sal B could reverse this situation P<0.05.Conclusion Salvianolic acid B can alleviate I/R-induced AKI and its mechanism may involve the activation of the AMPK pathway to mitigate ferroptosis in renal tubular epithelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. A New Benzofuran from the Heartwood of Dalbergia odorifera T. Chen and Its Protective Effect on Hypoxia/Reoxygenation Injury in H9c2.
- Author
-
Qingyu Zhong, Xiaowei Meng, Jiarong Li, Qing Zhu, Qiwan Zheng, Ronghua Liu, and Lanying Chen
- Subjects
- *
BENZOFURAN , *ISOFLAVONOIDS , *SINGLE crystals , *HEARTWOOD , *HYPOXEMIA - Abstract
A new benzofuran, named as (2S, 3S)-5,6-dimethoxy-3-methyl-2-(3'-hydroxyphenyl)-2,3-dihydrobenzofuran (1), along with the six known isoflavonoids, 2'-hydroxy-4',7-dimethoxyisoflavone (2), 2' ²-methoxybiochanin A (3), tectorigenin (4), calycosin (5), 7-hydroxy-2',4',5'-trimethoxyisoflavone (6), orobol (7) were isolated from the heartwood of Dalbergia odorifera T.Chen. The structure of compounds was characterised by NMR spectroscopic data and comparisons with relevant literature data. The absolute structural configuration of compound 1 was determined through X-ray single crystal diffraction. Moreover, compounds 1-7 have no significant cytotoxic effects on H9c2 cells (IC50 > 200 μM). Compound 1-7 exhibit a significant protective effect against H/R (hypoxia/reoxygenation) induced H9c2 cell damage at 10~40, 5~40, 5~40, 5~40, 5~40, 5~40 and 5~10 μM (P≩0.05). [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Effect of tubastatin A on NLRP3 inflammasome activation in macrophages under hypoxia/ reoxygenation conditions.
- Author
-
Hao Li, Chang Liu, Ying Cui, Panpan Chang, and Wei Chong
- Subjects
- *
HEAT shock proteins , *NITRIC-oxide synthases , *ENZYME-linked immunosorbent assay , *HISTONE deacetylase , *REACTIVE oxygen species - Abstract
BACKGROUND: There are currently no effective drugs to mitigate the ischemia/reperfusion injury caused by fluid resuscitation after hemorrhagic shock (HS). The aim of this study was to explore the potential of the histone deacetylase 6 (HDAC6)-specific inhibitor tubastatin A (TubA) to suppress nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation in macrophages under hypoxia/reoxygenation (H/R) conditions. METHODS: The viability of RAW264.7 cells subjected to H/R after treatment with different concentrations of TubA was assessed using a cell-counting kit-8 (CCK8) assay. Briefly, 2.5 µmol/L TubA was used with RAW264.7 cells under H/R condition. RAW264.7 cells were divided into three groups, namely the control, H/R, and TubA groups. The levels of reactive oxygen species (ROS) in the cells were detected using fluorescence microscopy. The protein expression of HDAC6, heat shock protein 90 (Hsp90), inducible nitric oxide synthase (iNOS), NLRP3, gasdermin-D (GSDMD), Caspase-1, GSDMD-N, and Caspase-1 p20 was detected by western blotting. The levels of interleukin-1β (IL-1β) and IL-18 in the supernatants were detected using enzyme-linked immunosorbent assay (ELISA). RESULTS: HDAC6, Hsp90, and iNOS expression levels were significantly higher (P<0.01) in the H/R group than in the control group, but lower in the TubA group than in the H/R group (P<0.05). When comparing the H/R group to the control group, ROS levels were significantly higher (P<0.01), but significantly reduced in the TubA group (P<0.05). The H/R group had higher NLRP3, GSDMD, Caspase-1, GSDMD-N, and Caspase-1 p20 expression levels than the control group (P<0.05), however, the TubA group had significantly lower expression levels than the H/R group (P<0.05). IL-1β and IL-18 levels in the supernatants were significantly higher in the H/R group compared to the control group (P<0.01), but significantly lower in the TubA group compared to the H/R group (P<0.01). CONCLUSION: TubA inhibited the expression of HDAC6, Hsp90, and iNOS in macrophages subjected to H/R. This inhibition led to a decrease in the content of ROS in cells, which subsequently inhibited the activation of the NLRP3 inflammasome and the secretion of IL-1β and IL-18. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. miR-652-3p Suppressed the Protective Effects of Isoflurane Against Myocardial Injury in Hypoxia/Reoxygenation by Targeting ISL1.
- Author
-
Qi, Kaikai, Cao, Fang, Wang, Jing, Wang, Yu, and Li, Guohua
- Subjects
MYOCARDIAL reperfusion ,MYOCARDIAL injury ,REVERSE transcriptase polymerase chain reaction ,TUMOR necrosis factors ,HOMEOBOX genes ,HYPOXEMIA ,ENZYME-linked immunosorbent assay - Abstract
This research is concentrated on investigating the role and mechanism of miR-652-3p in the protective effects of isoflurane (ISO) against myocardial ischemia–reperfusion (I/R) injury. H9c2 cells underwent pretreatment with varying concentrations of ISO, and subsequently, a hypoxia/reoxygenation (H/R) model was constructed. The levels of miR-652-3p, ISL LIM homeobox 1 (ISL1), and inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α) were evaluated through reverse transcription polymerase chain reaction (RT-qPCR). Enzyme-linked immunosorbent assay was employed to investigate concentrations of myocardial injury markers, such as creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI). Cell counting kit-8 was used to evaluate cell viability, while flow cytometry was utilized to measure apoptosis. Additionally, a dual luciferase reporter assay was conducted to validate the targeting relationship between ISL1 and miR-652-3p. Herein, we confirmed that the level of miR-652-3p was gradually increased with prolonged hypoxia; nevertheless, this increase was suppressed by ISO pretreatment (P < 0.05). Additionally, ISO pretreatment prevented the decrease in cell viability, increase in apoptosis, and overproduction of IL-6, TNF-α, CK-MB, and cTnI induced by H/R (P < 0.05). However, the inhibitory effects of ISO were counteracted by the increased levels of miR-652-3p (P < 0.05). ISL1 is a potential target of miR-652-3p. H/R induction suppressed ISL1 levels compared to the control, but ISO treatment increased its expression (P < 0.05). Overexpression of ISL1 inhibited the elimination of the protective effect of ISO on myocardial damage induced by the elevation of miR-652-3p (P < 0.05). The findings of this research confirm that miR-652-3p attenuated the protective effect of ISO on cardiomyocytes in myocardial ischemia by targeting ISL1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. A potent and selective activator of large‐conductance Ca2+‐activated K+ channels induces preservation of mitochondrial function after hypoxia and reoxygenation by handling of calcium and transmembrane potential.
- Author
-
de Souza, Itanna Isis Araujo, da Silva Barenco, Thais, Pavarino, Maria Eduarda Maciel Fernandes, Couto, Marcos Tadeu, de Resende, Gabriel Oliveira, de Oliveira, Dahienne Ferreira, Ponte, Cristiano Gonsalves, Nascimento, José Hamilton Matheus, and Maciel, Leonardo
- Subjects
- *
MEMBRANE potential , *MITOCHONDRIA , *HYPOXEMIA , *MYOCARDIAL infarction , *CALCIUM - Abstract
Aims: Ischaemic heart disease remains a significant cause of mortality globally. A pharmacological agent that protects cardiac mitochondria against oxygen deprivation injuries is welcome in therapy against acute myocardial infarction. Here, we evaluate the effect of large‐conductance Ca2+‐activated K+ channels (BKCa) activator, Compound Z, in isolated mitochondria under hypoxia and reoxygenation. Methods: Mitochondria from mice hearts were obtained by differential centrifugation. The isolated mitochondria were incubated with a BKCa channel activator, Compound Z, and subjected to normoxia or hypoxia/reoxygenation. Mitochondrial function was evaluated by measurement of O2 consumption in the complexes I, II, and IV in the respiratory states 1, 2, 3, and by maximal uncoupled O2 uptake, ATP production, ROS production, transmembrane potential, and calcium retention capacity. Results: Incubation of isolated mitochondria with Compound Z under normoxia conditions reduced the mitochondrial functions and induced the production of a significant amount of ROS. However, under hypoxia/reoxygenation, the Compound Z prevented a profound reduction in mitochondrial functions, including reducing ROS production over the hypoxia/reoxygenation group. Furthermore, hypoxia/reoxygenation induced a large mitochondria depolarization, which Compound Z incubation prevented, but, even so, Compound Z created a small depolarization. The mitochondrial calcium uptake was prevented by the BKCa activator, extruding the mitochondrial calcium present before Compound Z incubation. Conclusion: The Compound Z acts as a mitochondrial BKCa channel activator and can protect mitochondria function against hypoxia/reoxygenation injury, by handling mitochondrial calcium and transmembrane potential. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. In Vitro Hypoxia/Reoxygenation Induces Mitochondrial Cardiolipin Remodeling in Human Kidney Cells.
- Author
-
Strazdauskas, Arvydas, Trumbeckaite, Sonata, Jakstas, Valdas, Dambrauskiene, Justina, Mieldazyte, Ausra, Klimkaitis, Kristupas, and Baniene, Rasa
- Subjects
- *
REPERFUSION , *CARDIOLIPIN , *PROXIMAL kidney tubules , *MITOCHONDRIA , *ATMOSPHERIC oxygen , *HYPOXEMIA , *HYPOXIA-inducible factor 1 - Abstract
Renal ischemia/reperfusion is a serious condition that not only causes acute kidney injury, a severe clinical syndrome with high mortality, but is also an inevitable part of kidney transplantation or other kidney surgeries. Alterations of oxygen levels during ischemia/reperfusion, namely hypoxia/reoxygenation, disrupt mitochondrial metabolism and induce structural changes that lead to cell death. A signature mitochondrial phospholipid, cardiolipin, with many vital roles in mitochondrial homeostasis, is one of the key players in hypoxia/reoxygenation-induced mitochondrial damage. In this study, we analyze the effect of hypoxia/reoxygenation on human renal proximal tubule epithelial cell (RPTEC) cardiolipins, as well as their metabolism and mitochondrial functions. RPTEC cells were placed in a hypoxic chamber with a 2% oxygen atmosphere for 24 h to induce hypoxia; then, they were replaced back into regular growth conditions for 24 h of reoxygenation. Surprisingly, after 24 h, hypoxia cardiolipin levels substantially increased and remained higher than control levels after 24 h of reoxygenation. This was explained by significantly elevated levels of cardiolipin synthase and lysocardiolipin acyltransferase 1 (LCLAT1) gene expression and protein levels. Meanwhile, hypoxia/reoxygenation decreased ADP-dependent mitochondrial respiration rates and oxidative phosphorylation capacity and increased reactive oxygen species generation. Our findings suggest that hypoxia/reoxygenation induces cardiolipin remodeling in response to reduced mitochondrial oxidative phosphorylation in a way that protects mitochondrial function. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Enhancing Heart Transplantation: Utilizing Gas-Loaded Nanocarriers to Mitigate Cold/Hypoxia Stress.
- Author
-
Rubeo, Chiara, Hoti, Gjylije, Giordano, Magalì, Molinar, Chiara, Aragno, Manuela, Mantuano, Beatrice, Comità, Stefano, Femminò, Saveria, Cavalli, Roberta, Trotta, Francesco, Penna, Claudia, and Pagliaro, Pasquale
- Subjects
- *
HEART transplantation , *HEART , *NANOCARRIERS , *HYPOXEMIA , *LABORATORY rats , *COLD storage - Abstract
Gas-loaded nanocarriers (G-LN) show promise in improving heart transplantation (HTx) outcomes. Given their success in reducing cell death during normothermic hypoxia/reoxygenation (H/R) in vitro, we tested their integration into cardioplegic solutions and static cold storage (SCS) during simulated HTx. Wistar rat hearts underwent four hours of SCS with four G-LN variants: O2- or N2-cyclic-nigerosyl-nigerose-nanomonomers (CNN), and O2- or N2-cyclic-nigerosyl-nigerose-nanosponges (CNN-NS). We monitored physiological-hemodynamic parameters and molecular markers during reperfusion to assess cell damage/protection. Hearts treated with nanomonomers (N2-CNN or O2-CNN) showed improvements in left ventricular developed pressure (LVDP) and a trend towards faster recovery of the rate pressure product (RPP) compared to controls. However, nanosponges (N2-CNN-NS or O2-CNN-NS) did not show similar improvements. None of the groups exhibited an increase in diastolic left ventricular pressure (contracture index) during reperfusion. Redox markers and apoptosis/autophagy pathways indicated an increase in Beclin 1 for O2-CNN and in p22phox for N2-CNN, suggesting alterations in autophagy and the redox environment during late reperfusion, which might explain the gradual decline in heart performance. The study highlights the potential of nanomonomers to improve early cardiac performance and mitigate cold/H/R-induced stunning in HTx. These early improvements suggest a promising avenue for increasing HTx success. Nevertheless, further research and optimization are needed before clinical application. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. 缺氧后处理通过 piRNA-005854 调控衰老心肌细胞自噬发挥保护心肌作用.
- Author
-
迟宏扬, 杨慧霞, 郝银菊, 杨安宁, 白志刚, 焦 运, 熊建团, 马胜超, and 姜怡邓
- Subjects
- *
LACTATE dehydrogenase , *ISCHEMIC postconditioning , *REPERFUSION injury , *MYOCARDIAL injury , *WESTERN immunoblotting , *GALACTOSE , *CREATINE kinase - Abstract
BACKGROUND: Ischemic postconditioning is one of the effective ways to reduce ischemia-reperfusion injury and has been more and more widely used in clinical practice in recent years, but its specific molecular mechanism has yet to be studied. OBJECTIVE: To investigate the role and mechanism of piRNA-005854 in the aging cardiomyocytes caused by hypoxic postconditioning. METHODS: In vitro, cardiomyocytes were administered 8 mg/mL D-galactose for 9 days to induce their aging. β-Galactosidase staining was used to observe the aging of cardiomyocytes. Senescent cells were treated with hypoxia/reoxygenation and hypoxic postconditioning. ELISA was utilized to detect changes in myocardial injury markers creatine kinase isoenzyme MB and lactate dehydrogenase levels. Western blot assay was applied to detect the expression changes of autophagy-related proteins LC3II, p62, ULK1 and phosphorylated ULK1 in aging cardiomyocytes. qRT-PCR was employed to determine the expression level of piRNA-005854. piRNA-005854 inhibitor and piRNA-005854 mimics were transferred into aging cardiomyocytes and followed with hypoxic postconditioning. Western blot assay was used to examine the expression of LC3II, p62, ULK1 and phosphorylated ULK1. RESULTS AND CONCLUSION: (1) D-galactose induced obvious senescence of cardiomyocytes 9 days later. (2) Compared with the normoxia group, creatine kinase isoenzyme MB and lactate dehydrogenase levels increased in the hypoxia/reoxygenation group (P < 0.01); LC3 II/I expression was increased; p62 expression was decreased; ULK1 phosphorylation level was increased, and piRNA-005854 expression was increased (P < 0.01). (3) Compared with the hypoxia/ reoxygenation group, creatine kinase isoenzyme MB and lactate dehydrogenase levels significantly reduced in the hypoxic postconditioning group (P < 0.01); LC3 II/I expression significantly decreased (P < 0.05); p62 expression increased (P < 0.01); ULK1 phosphorylation level decreased (P < 0.05), and piRNA-005854 expression decreased (P < 0.01). (4) After transfection of piRNA-005854 inhibitor, LC3II/I expression was decreased (P < 0.01); the expression of p62 was increased significantly (P < 0.05); the phosphorylation level of ULK1 was decreased significantly (P < 0.01). After transfection of piRNA-005854 mimics, LC3II/ I expression was increased significantly; the expression of p62 was decreased, and the phosphorylation level of ULK1 was increased significantly (P < 0.01). (5) The results show that piRNA-005854-mediated reduction of ULK1-dependent autophagy level is a possible mechanism that hypoxic postconditioning exerts its protective effect on aging cardiomyocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. The m6A methylation and expression profiles of mouse neural stem cells after hypoxia/reoxygenation
- Author
-
Shaoqiong Zhang, Kaile Cui, Yuanyuan Li, Yiting Fan, Dongxu Wang, Xingen Yao, and Bo Fang
- Subjects
Neural stem cell ,Hypoxia/reoxygenation ,N6-methyladenosine methylation ,Proliferation ,Migration ,Differentiation ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Ischemia–reperfusion injury to the central nervous system often causes severe complications. The activation of endogenous neural stem cells (NSCs) is considered a promising therapeutic strategy for nerve repair. However, the specific biological processes and molecular mechanisms of NSC activation remain unclear, and the role of N6-methyladenosine (m6A) methylation modification in this process has not been explored. Methods NSCs were subjected to hypoxia/reoxygenation (H/R) to simulate ischemia–reperfusion in vivo. m6A RNA methylation quantitative kit was used to measure the total RNA m6A methylation level. Quantitative real-time PCR was used to detect methyltransferase and demethylase mRNA expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were conducted for NSCs in control and H/R groups, and the sequencing results were analyzed using bioinformatics. Finally, the migration ability of NSCs was identified by wound healing assays, and the proliferative capacity of NSCs was assessed using the cell counting kit-8, EdU assays and cell spheroidization assays. Results Overall of m6A modification level and Mettl14 mRNA expression increased in NSCs after H/R treatment. The m6A methylation and expression profiles of mRNAs in NSCs after H/R are described for the first time. Through the joint analysis of MeRIP-seq and RNA-seq results, we verified the proliferation of NSCs after H/R, which was regulated by m6A methylation modification. Seven hub genes were identified to play key roles in the regulatory process. Knockdown of Mettl14 significantly inhibited the proliferation of NSCs. In addition, separate analysis of the MeRIP-seq results suggested that m6A methylation regulates cell migration and differentiation in ways other than affecting mRNA expression. Subsequent experiments confirmed the migration ability of NSCs was suppressed by knockdown of Mettl14. Conclusion The biological behaviors of NSCs after H/R are closely related to m6A methylation of mRNAs, and Mettl14 was confirmed to be involved in cell proliferation and migration.
- Published
- 2024
- Full Text
- View/download PDF
20. The m6A methylation and expression profiles of mouse neural stem cells after hypoxia/reoxygenation
- Author
-
Zhang, Shaoqiong, Cui, Kaile, Li, Yuanyuan, Fan, Yiting, Wang, Dongxu, Yao, Xingen, and Fang, Bo
- Published
- 2024
- Full Text
- View/download PDF
21. GPR43 protects human A16 cardiomyocytes against hypoxia/reoxygenation injury by regulating nesfatin1.
- Author
-
Jie Yu, Qingquan Li, Chang Guan, Ming Yuan, Ding Yu, Yaqin Wang, Mengyao Liu, and Ying Lv
- Abstract
Background: The purpose of this study was to investigate the regulatory role of G coupled-protein receptor 43 (GPR43) during myocardial ischaemia/reperfusion (I/R) injury and to explore the relevant molecular mechanism. Materials and methods: AC16 hypoxia/reoxygenation (H/R) model was established to simulate I/R injury in vitro. Gain- and loss-of-function experiments were conducted to regulate GPR43 or nesfatin1 expression. Cell viability and apoptosis was examined adopting CCK-8 and TUNEL assays. Commercial kits were applied for detecting ROS and inflammatory cytokines. Quantitative real-time PCR (qRT-PCR) and western blotting were conducted to measure the expression level of critical genes and proteins. Results: GPR43 was downregulated in H/R-mediated AC16 cells. GPR43 over-expression or the GPR43 agonist greatly inhibited H/R-induced cell viability loss, cell apoptosis, and excessive production of ROS and pro-inflammatory cytokines in AC16 cardiomyocytes. Co-immunoprecipitation (Co-IP) assay identified an interaction between GPR43 and nesfatin1, and GPR43 could positively regulate nesfatin1. In addition, the protective role of GPR43 against H/R injury was partly abolished upon nesfatin1 knockdown. Eventually, GPR43 could inhibit H/R-stimulated JNK/P38 MAPK signalling in AC16 cells, which was also hindered by nesfatin1 knockdown. Conclusions: Our findings demonstrated the protective role of GPR43 against H/R-mediated cardiomyocytes injury through up-regulating nesfatin1, providing a novel target for the prevention and treatment of myocardial I/R injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Downregulation of miR-337-3p in hypoxia/reoxygenation neuroblastoma cells increases KCTD11 expression.
- Author
-
Lin Zhu, Yi-Juan Xin, Mu He, Jun Bian, Xiao-Li Cheng, Rui Li, Jin-Jie Li, Juan Wang, Jia-Yun Liu, and Liu Yang
- Subjects
GENE expression ,HYPOXEMIA ,NEURONS ,NEUROBLASTOMA ,POTASSIUM channels - Abstract
Neurodegeneration is linked to the progressive loss of neural function and is associated with several diseases. Hypoxia is a hallmark in many of these diseases, and several therapies have been developed to treat this disease, including gene expression therapies that should be tightly controlled to avoid side effects. Cells experiencing hypoxia undergo a series of physiological responses that are induced by the activation of various transcription factors. Modulation of microRNA (miRNA) expression to alter transcriptional regulation has been demonstrated to be beneficial in treating multiple diseases, and in this study, we therefore explored potential miRNA candidates that could influence hypoxia-induced nerve cell death. Our data suggest that in mouse neuroblasts Neuro-2a cells with hypoxia/reoxygenation (H/R), miR-337-3p is downregulated to increase the expression of Potassium channel tetramerization domain containing 11 (KCTD11) and subsequently promote apoptosis. Here, we demonstrate for the first time that KCTD11 plays a role in the cellular response to hypoxia, and we also provide a possible regulatory mechanism by identifying the axis of miR-337-3p/KCTD11 as a promising candidate modulator of nerve cell survival after H/R exposure. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. The m6A methylation and expression profiles of mouse neural stem cells after hypoxia/reoxygenation.
- Author
-
Zhang, Shaoqiong, Cui, Kaile, Li, Yuanyuan, Fan, Yiting, Wang, Dongxu, Yao, Xingen, and Fang, Bo
- Abstract
Background: Ischemia–reperfusion injury to the central nervous system often causes severe complications. The activation of endogenous neural stem cells (NSCs) is considered a promising therapeutic strategy for nerve repair. However, the specific biological processes and molecular mechanisms of NSC activation remain unclear, and the role of N6-methyladenosine (m6A) methylation modification in this process has not been explored. Methods: NSCs were subjected to hypoxia/reoxygenation (H/R) to simulate ischemia–reperfusion in vivo. m6A RNA methylation quantitative kit was used to measure the total RNA m6A methylation level. Quantitative real-time PCR was used to detect methyltransferase and demethylase mRNA expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were conducted for NSCs in control and H/R groups, and the sequencing results were analyzed using bioinformatics. Finally, the migration ability of NSCs was identified by wound healing assays, and the proliferative capacity of NSCs was assessed using the cell counting kit-8, EdU assays and cell spheroidization assays. Results: Overall of m6A modification level and Mettl14 mRNA expression increased in NSCs after H/R treatment. The m6A methylation and expression profiles of mRNAs in NSCs after H/R are described for the first time. Through the joint analysis of MeRIP-seq and RNA-seq results, we verified the proliferation of NSCs after H/R, which was regulated by m6A methylation modification. Seven hub genes were identified to play key roles in the regulatory process. Knockdown of Mettl14 significantly inhibited the proliferation of NSCs. In addition, separate analysis of the MeRIP-seq results suggested that m6A methylation regulates cell migration and differentiation in ways other than affecting mRNA expression. Subsequent experiments confirmed the migration ability of NSCs was suppressed by knockdown of Mettl14. Conclusion: The biological behaviors of NSCs after H/R are closely related to m6A methylation of mRNAs, and Mettl14 was confirmed to be involved in cell proliferation and migration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. The Role of δ2-Opioid Receptors in the Regulation of Tolerance of Isolated Cardiomyocytes to Hypoxia and Reoxygenation.
- Author
-
Mukhomedzyanov, A. V., Popov, S. V., Naryzhnaya, N. V., Azev, V. N., and Maslov, L. N.
- Subjects
- *
OPIOID receptors , *HYPOXEMIA , *LACTATE dehydrogenase , *NALOXONE - Abstract
Hypoxia (20 min) and reoxygenation (30 min) were simulated on isolated rat cardiomyocytes to evaluate the cytoprotective effect of selective δ2-opioid receptor agonist deltorphin II, opioid receptor antagonist naloxone methiodide, μ-opioid receptor antagonist CTAP, κ-opioid receptor antagonist nor-binaltorphimine, ε1-opioid receptor antagonist BNTX, and δ2-opioid receptors naltriben. Deltorphin II was administered 5 min before reoxygenation, antagonists were administered 10 min before reoxygenation. The cytoprotective effect of deltorphin II was assessed by the number of cardiomyocytes survived after hypoxia/reoxygenation, as well as by the lactate dehydrogenase content in the incubation medium. It has been established that the cytoprotective effect of deltorphin II occurs at a concentration of 64 nmol/liter and is associated with activation of δ2-opioid receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Cav3.2 channel regulates cerebral ischemia/reperfusion injury: a promising target for intervention
- Author
-
Feibiao Dai, Chengyun Hu, Xue Li, Zhetao Zhang, Hongtao Wang, Wanjun Zhou, Jiawu Wang, Qingtian Geng, Yongfei Dong, and Chaoliang Tang
- Subjects
calcineurin ,cav3.2 channel ,cerebral ischemia/reperfusion ,hippocampus ,hypoxia/reoxygenation ,inflammatory response ,nuclear factor of activated t cells 3 ,oxidative stress ,primary hippocampal neurons ,stroke ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Calcium influx into neurons triggers neuronal death during cerebral ischemia/reperfusion injury. Various calcium channels are involved in cerebral ischemia/reperfusion injury. Cav3.2 channel is a main subtype of T-type calcium channels. T-type calcium channel blockers, such as pimozide and mibefradil, have been shown to prevent cerebral ischemia/reperfusion injury-induced brain injury. However, the role of Cav3.2 channels in cerebral ischemia/reperfusion injury remains unclear. Here, in vitro and in vivo models of cerebral ischemia/reperfusion injury were established using middle cerebral artery occlusion in mice and high glucose hypoxia/reoxygenation exposure in primary hippocampal neurons. The results showed that Cav3.2 expression was significantly upregulated in injured hippocampal tissue and primary hippocampal neurons. We further established a Cav3.2 gene-knockout mouse model of cerebral ischemia/reperfusion injury. Cav3.2 knockout markedly reduced infarct volume and brain water content, and alleviated neurological dysfunction after cerebral ischemia/reperfusion injury. Additionally, Cav3.2 knockout attenuated cerebral ischemia/reperfusion injury-induced oxidative stress, inflammatory response, and neuronal apoptosis. In the hippocampus of Cav3.2-knockout mice, calcineurin overexpression offset the beneficial effect of Cav3.2 knockout after cerebral ischemia/reperfusion injury. These findings suggest that the neuroprotective function of Cav3.2 knockout is mediated by calcineurin/nuclear factor of activated T cells 3 signaling. Findings from this study suggest that Cav3.2 could be a promising target for treatment of cerebral ischemia/reperfusion injury.
- Published
- 2024
- Full Text
- View/download PDF
26. Tongguan capsule for treating myocardial ischemia-reperfusion injury: integrating network pharmacology and mechanism study
- Author
-
Jiantao Liu, Chunping Liu, Huiqi Chen, Huan Cen, Hailong Yang, Peijian Liu, Fang Liu, Liuling Ma, Quanfu Chen, and Lei Wang
- Subjects
Hypoxia/reoxygenation ,apoptosis ,mTOR pathways ,autophagy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
AbstractContext Although Tongguan capsule (TGC) is used in the treatment of coronary atherosclerotic disease, the exact mechanism remains unclear.Objective Network pharmacology and experimental validation were applied to examine the mechanism of TGC for treating myocardial ischemia-reperfusion injury (MIRI).Materials and methods The components and candidate targets were searched based on various databases such as TCMSP, TCMID, BATMAN-TCM. The binding ability was determined by molecular docking. The ischemia-reperfusion (I/R) model was constructed by ligating the left anterior descending (LAD) coronary artery. APOE-/- mice were divided into three groups (n = 6): Sham group, I/R group, and TGC group (1 g/kg/d). To further verification, HCAEC cells were subjected to hypoxia-reoxygenation (H/R) to establish in vitro model.Results The compounds, such as quercetin, luteolin, tanshinone IIA, kaempferol and bifendate, were obtained after screening. The affinity values of the components with GSK-3β, mTOR, Beclin-1, and LC3 were all
- Published
- 2023
- Full Text
- View/download PDF
27. Comparison of Cardioprotective Potential of Cannabidiol and β-Adrenergic Stimulation Against Hypoxia/Reoxygenation Injury in Rat Atria and Ventricular Papillary Muscles
- Author
-
Anna Pędzińska-Betiuk, Ulrich Gergs, Jolanta Weresa, Patryk Remiszewski, Ewa Harasim-Symbor, and Barbara Malinowska
- Subjects
hypoxia/reoxygenation ,isolated atria ,isolated ventricular papillary muscles ,cannabidiol ,isoprenaline ,hypertension ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Background: Hypoxia is one of the most significant pathogenic factors in cardiovascular diseases. Preclinical studies suggest that nonpsychoactive cannabidiol (CBD) and β-adrenoceptor stimulation might possess cardioprotective potential against ischemia-reperfusion injury. The current study evaluates the influence of hypoxia-reoxygenation (H/R) on the function of atria and ventricular papillary muscles in the presence of CBD and the nonselective β-adrenoceptor agonist isoprenaline (ISO). Methods: The concentration curves for ISO were constructed in the presence of CBD (1 µM) before or after H/R. In chronic experiments (CBD 10 mg/kg, 14 days), the left atria isolated from spontaneously hypertensive (SHR) and their normotensive control (WKY) rats were subjected to H/R following ISO administration. Results: Hypoxia decreased the rate and force of contractions in all compartments. The right atria were the most resistant to hypoxia regardless of prior β-adrenergic stimulation. Previous β-adrenergic stimulation improved recovery in isolated left atria and right (but not left) papillary muscles. Acute (but not chronic) CBD administration increased the effects of ISO in left atria and right (but not left) papillary muscles. Hypertension accelerates left atrial recovery during reoxygenation. Conclusions: H/R directly modifies the function of particular cardiac compartments in a manner dependent on cardiac region and β-adrenergic prestimulation. The moderate direct cardioprotective potential of CBD and β-adrenergic stimulation against H/R is dependent on the cardiac region, and it is less than in the whole heart with preserved coronary flow. In clinical terms, our research expands the existing knowledge about the impact of cannabidiol on cardiac ischemia, the world′s leading cause of death.
- Published
- 2024
- Full Text
- View/download PDF
28. Hypoxia/reoxygenation attenuates migration and invasion of human extracellular villus trophoblast cells via DNA methyltransferase pathway
- Author
-
HU Sichen, LAN Xi, and DING Yubin
- Subjects
hypoxia/reoxygenation ,extravillous trophoblast ,dna methyltransferase ,epithelial-mesenchymal transition ,migration ,invasion ,Medicine (General) ,R5-920 - Abstract
Objective To explore the regulative mechanism of hypoxia/reoxygen (H/R) for invasion ability of extravillous trophoblast (EVT). Methods Immortalized human EVT cell line HTR-8/SVneo in early pregnancy, primary EVT cells isolated from the villi of placenta in early pregnancy, and villi explants were cultured respectively. A simulation model of H/R environment in EVT cells invading the uterus was established under the conversion conditions of 1% O2 for 24 h to 20% O2 for another 24 h. Western blotting and immunofluorescence assay were used to detect the expression of key factors such as cell invasion, and Transwell chamber test was employed to observe cell migration and invasion. Results After H/R induction, the migration and invasion abilities of HTR-8/SVneo cells were weakened, and the epitaxial ability of cultured villi explants and the migration and invasion abilities of isolated primary EVT cells were also weakened. Under H/R conditions, the expression levels of MMP-2 and MMP-9, integrin-β1 and integrin-α5 were decreased in HTR-8/SVneo cells (P < 0.05), and the protein expression of DNA methyltransferase (DNMTs) was also down-regulated (P < 0.01). In the primary EVT cells under H/R condition, the expression levels of epithelial-mesenchymal transition (EMT) positive regulatory factors, Slug and Snails, were inhibited (P < 0.05), that of epithelial cell marker E-cadherin was increased (P < 0.01), protein levels of MMP-2 and MMP-9, integrin-β1 and integrin-α5 were decreased (P < 0.05), and cell invasion ability was reduced along with decreased DNMT1 and DNMT3A expression (P < 0.05). However, overexpression of DNMT1, DNMT3A and DNMT3B in HTR-8/SVneo cells promoted the up-regulation of interstitial markers N-cadherin, and MMP-2 and MMP-9 (P < 0.05). Conclusion H/R regulates the expression of EMT-related factors such as MMP-2/-9 through DNMTs, and thereby reduces the migration and invasion abilities of EVT cells.
- Published
- 2023
- Full Text
- View/download PDF
29. Short-chain fatty acid sodium acetate reduces hypoxia- reoxygenation induced injury of human renal tubular epithelial cell line HK2
- Author
-
JIANG Luojia, XU Haibo
- Subjects
sodium acetate ,hypoxia/reoxygenation ,hk2 cells ,inflammation ,mitochondria ,Medicine - Abstract
Objective To study the protective effect and related mechanisms of short-chain fatty acid sodium acetate on hypoxia/reoxygenation (H/R)-induced injury of human renal tubular epithelial cell line HK2. Methods The H/R model was established, and HK2 cells were incubated with sodium acetate (SA) for 2 h. The survival rate of HK2 cells was detected by CCK8 assay; Enzyme activity kit was used for detection of inflammatory factors, cellular reactive oxygen species and ATP production; Flow cytometry was used to measure mitochondrial membrane potential (MMP) and mitochondrial oxidative stress products (mitoSOX) levels; Electron microscope was used to observe mitochondrial ultrastructural damage; Western blot was used to detected the expression of inflammatory signaling pathway IκB-α/NF-κB and mitochondrial energy disorder signaling pathway AMPK/PGC-1α. Results Compared with the control group, the survival rate of HK2 cells in the H/R group was significantly decreased(P<0.05); the expression of intracellular ROS, mitoSOX and inflammatory factors was significantly increased(P<0.05); the ultrastructure of mitochondria was severely damaged, and the content of ATP and MMP was significantly decreased(P<0.05); it was further found that the protein expression of p-IκB-α and NF-κB-p-P65 was significantly increased while the protein expression of p-AMPK and PGC-1α was significantly decreased(P<0.05). Compared with H/R group, SA significantly enhanced the survival rate of HK2 cells (P<0.05); SA inhibited the release of intracellular ROS, mitoSOX and inflammatory factors; SA inhibited mitochondrial ultrastructural damage, decreased ATP and MMP (P<0.05); SA promoted the expression of p-AMPK and PGC-1α and inhibited the expression of p-IκB-α and NF-κB-p-P65 (P<0.05). Conclusions Sodium acetate plays a protective role with potential mechanisms of anti-inflammation, anti-oxidative stress, protection of mitochondrial structure and function of HK2 cells induced by H/R through inhibiting IκB-α/ NF-κB and activating AMPK/PGC-1α signal pathway.
- Published
- 2023
- Full Text
- View/download PDF
30. Sevoflurane preconditioning attenuates myocardial cell damage caused by hypoxia and reoxygenation via regulating the NORAD/miR-144-3p axis.
- Author
-
Qian, Duo, Wen, Jie, Yuan, Yawei, Wang, Long, and Feng, Xiaona
- Abstract
Objective: This study aimed to investigate the function and mechanism of lncRNA NORAD in Sevoflurane (Sev) protection against myocardial hypoxia-reoxygenation (H/R). Methods: Preprocess rat cardiomyocytes H9c2 cells with Sev at concentrations of 0.5%, 1.0%, and 1.5%, and subjected them to H/R treatment. qRT-PCR was used to detect levels of NORAD and miR-144-3p. Measure concentrations of the inflammatory cytokines IL-6, TNF-α, and IL-10, as well as cardiac injury markers cTnI, CK-MB, and LDH using ELISA. Assess cell proliferation and apoptosis using CCK-8 and flow cytometry. Perform dual-luciferase reporter assay and RIP assay to validate the targeting relationship between NORAD and miR-144-3p. Results: H/R induced inhibition of cell proliferation, increase in apoptosis, and production of IL-6, TNF-α, CK-MB, LDH, and cTnI were significantly attenuated by Sev. As hypoxic treatment time lengthened, the NORAD levels in myocardial cells showed an increase, with Sev pretreatment being able to suppress the NORAD levels elevation. The overexpression of NORAD notably weakened the cardioprotective effect of Sev. NORAD targetedly binds to miR-144-3p and negatively regulates miR-144-3p. Increased miR-144-3p levels inhibited the antagonistic effect of NORAD on the cardioprotective effects of Sev. Conclusion: The current study confirmed that sevoflurane attenuated H/R-induced cardiomyocyte injury via the NORAD/miR-144-3p axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. 类视黄醇X 受体对缺氧/复氧诱导的大鼠II 型 肺泡上皮细胞氧化应激反应的调控作用
- Author
-
王肖婷, 徐俊鹏, 黄曼, 陈思安, 张淇昊, 曹文傑, 田云娜, 高慧, and 王万铁
- Subjects
- *
RETINOID X receptors , *EPITHELIAL cells , *OXIDATIVE stress , *HYPOXEMIA - Abstract
AIM: To investigate the regulatory role of retinoid X receptor (RXR) in oxidative stress response of rat type II alveolar epithelial cells( AECII) induced by hypoxia/reoxygenation( HR). METHODS: The AECII were divided into control (C) group, HR group, HR+solvent dimethyl sulfoxide (DMSO) group (HD group), HR+RXR agonist 9-cis-retinoic acid (9-RA) group (RA group), and HR+RXR antagonist HX531 group (HX group). Cell Counting Kit-8 (CCK-8) method was used to measure the cell viability. Immunofluorescence staining was used to detect the expression of surfactant protein A (SP-A) and RXRα in AECII. Kits were detected to the levels of superoxide dismutase (SOD) and malondialdehyde (MDA) in cells. Transmission electron microscopy was used to observe the ultrastructural changes of the cells. Western blot was used to detect the protein level of nuclear factor E2-related factor 2 (Nrf2). RT-PCR was used to detect the expression level of Nrf2 mRNA. RESULTS: Compared with C group, the cell viability and SOD activity in HR, HD, RA and HX groups were decreased significantly (P<0. 05), the MDA content were increased significantly (P<0. 05), the Nrf2 mRNA and protein expression levels were decreased significantly (P<0. 05 or P<0. 01), and the immunofluorescence expression of RXRα was significantly increased (P<0. 01). Compared with HR and HX groups, the cells in RA group showed significantly increased cell viability (P<0. 05), increased SOD activity( P<0. 05), decreased MDA content (P<0. 05), increased Nrf2 mRNA and protein expression levels (P<0. 01), and significantly increased immunofluorescence expression of RXRα (P<0. 01). CONCLUSION: Hypoxia/reoxygenation can aggravate the oxidative stress response of rat AECII, and RXR agonist intervention can alleviate HR-induced rat AECII injury by inhibiting oxidative stress. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Allium mongolicum Regel Flavonoids Alleviate Hypoxia/ Reoxygenation-Induced Cardiomyocyte Injury by Downregulating lncRNA TALNEC2.
- Author
-
HUAMING ZHANG and XI FENG
- Subjects
- *
ALLIUM , *FLAVONOIDS , *COBALT chloride , *GLUTATHIONE peroxidase , *SUPEROXIDE dismutase , *MOLECULAR cloning , *LINCRNA - Abstract
To explore the effect and possible mechanism of Allium mongolicum Regel flavonoids on cardiomyocyte injury induced by hypoxia/reoxygenation. H9C2 cardiomyocytes were induced by hypoxia/reoxygenation and treated with different doses of Allium mongolicum Regel flavonoids. H9C2 cells were transfected with si-negative control/ si-TALNEC2 and treated with hypoxia/reoxygenation. Besides, cells transfected with plasmid cloning deoxyribonucleic acid/plasmid cloning deoxyribonucleic acid-TALNEC2 were treated with Allium mongolicum Regel flavonoids and induced with hypoxia/reoxygenation. Malondialdehyde, glutathione peroxidase and superoxide dismutase levels were tested to evaluate oxidative stress. Apoptosis rate was analyzed by flow cytometry. TALNEC2 expression was examined using quantitative reverse transcription-polymerase chain reaction, and cleaved caspase-3 and cleaved caspase-9 protein levels were tested by Western blot. Allium mongolicum Regel flavonoids could reduce malondialdehyde level, apoptosis rate, cleaved caspase-3 level, cleaved caspase-9 level, and TALNEC2 expression, while enhanced glutathione peroxidase and superoxide dismutase levels in hypoxia/reoxygenation-induced H9C2 cells in a dose-dependent manner. After transfection of si-TALNEC2, malondialdehyde level, apoptosis rate, cleaved caspase-3 level, and cleaved caspase-9 level were reduced, while superoxide dismutase and glutathione peroxidase levels were enhanced. Transfection of plasmid cloning deoxyribonucleic acid-TALNEC2 could abolish the effect of Allium mongolicum Regel flavonoids on cardiomyocyte injury. Allium mongolicum Regel flavonoids could inhibit hypoxia/reoxygenation-induced cardiomyocyte apoptosis and oxidative stress via reducing TALNEC2 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. LncRNA SNHG12 调控 miR-138-5p/HIF-1α 轴改善缺氧/复氧人血管内皮 细胞损伤的研究.
- Author
-
魏宗强, 王琳茹, 胡文贤, 张娟子, 黄贤明, 李 林, and 李 强
- Abstract
Objective: To study the effect of Long non-coding RNA (LncRNA) small nucleolar RNA host gene 12 (SNHG12) regulating miR-138-5p/hypoxia inducible factor-1 (HIF-1α) axis on improving the damage of hypoxia/reoxygenation (H/R) human vascular endothelial cells. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured in vitro and randomly divided into control group, H/R model group, H/R+LncRNA SNHG12 overexpression group, H/R+miR-138-5p mimics group, H/R+co-transfection group and H/R+co-transfection negative control group, each transfection group was transfected separately, and except for control group, the remaining groups were given hypoxia for 5 hours and then reoxygenated for 1 hour to induce the cell models, and then the cell viability of each group was detected by CCK-8 experiment; the cell apoptosis in each group was detected by flow cytometry experiment, and the apoptosis rate of each group was compared; the levels of reactive oxygen species (ROS), lactate dehydrogenase (LDH) and inflammatory factors IL-6, IL-17 and IL-18 in each group were measured by the kit; the expressions of miR-138-5p and HIF-1α mRNA in cells of each group were measured by real-time quantitative PCR (qRT-PCR) experiment; the expressions of apoptotic proteins caspase-9, Bcl-2-associated X protein (Bax) and HIF-1α in each group were evaluated by Western blot. Results: Compared with control group, the apoptosis rate, cellular ROS, LDH, IL-6, IL-17 and IL-18 levels, cellular HIF-1α mRNA and protein levels, cellular caspase-9, Bax and HIF-1α protein levels were increased in H/R model group (P<0.05), the cell viability and miR-138-5p level were decreased (P<0.05). Compared with H/R model group and H/R+co-transfection group, the cell viability, cell HIF-1α mRNA and protein levels were increased in H/R+LncRNA SNHG12 overexpression group (P<0.05), the apoptosis rate,cellular ROS, LDH, IL-6, IL-17 and IL-18 levels,cellular caspase-9 and Bax protein levels, and miR-138-5p level were decreased (P<0.05); the cell viability,cellular HIF-1α mRNA and protein levels were decreased in H/R+miR-138-5p mimics group (P<0.05), the apoptosis rate, cellular ROS, LDH, IL-6, IL-17 and IL-18 levels,cellular caspase-9 and Bax protein levels were increased (P<0.05). Compared with H/R model group, there was no significant difference in cell index levels between the H/R+co-transfection negative control group and the H/R+co-transfection group (P>0.05). Conclusion: LncRNA SNHG12 can upregulate HIF-1α expression by downregulating miR-138-5p expression, inhibit H/R-induced inflammation and oxidative stress in HUVECs, and reduce cell damage and apoptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
34. Enhanced protection against hypoxia/reoxygenation-induced apoptosis in H9c2 cells by puerarin-loaded liposomes modified with matrix metalloproteinases-targeting peptide and triphenylphosphonium.
- Author
-
Fengmei Li, Yan Wang, Wenqun Li, Junyong Wu, Shengnan Li, Xiongbin Hu, Tiantian Tang, and Xinyi Liu
- Subjects
- *
PEPTIDES , *MATRIX metalloproteinases , *REACTIVE oxygen species , *MYOCARDIAL ischemia , *REPERFUSION injury , *LIPOSOMES - Abstract
Based on the inhibition of mitochondrial permeability transition pore (mPTP) opening, puerarin (PUE) has a good potential to reduce myocardial ischemia/reperfusion injury (MI/RI). However, the lack of targeting of free PUE makes it difficult to reach the mitochondria. In this paper, we constructed matrix metalloproteinase-targeting peptide (MMP-TP) and triphenylphosphonium (TPP) cation co-modified liposomes loaded with PUE (PUE@T/M-L) for mitochondria-targeted drug delivery. PUE@T/M-L had a favorable particle size of 144.9 ± 0.8 nm, an encapsulation efficiency of 78.9 ± 0.6%, and a sustainedrelease behavior. The results of cytofluorimetric experiments showed that MMP-TP and TPP doublemodified liposomes (T/M-L) enhanced intracellular uptake, escaped lysosomal capture, and promoted drug targeting into mitochondria. In addition, PUE@T/M-L enhanced the viability of hypoxia-reoxygenation (H/R) injured H9c2 cells by inhibiting mPTP opening and reactive oxygen species (ROS) production, reducing Bax expression and increasing Bcl-2 expression. It was inferred that PUE@T/M-L delivered PUE into the mitochondria of H/R injured H9c2 cells, resulting in a significant increase in cellular potency. Based on the ability of MMP-TP to bind the elevated expression of matrix metalloproteinases (MMPs), T/M-L had excellent tropism for Lipopolysaccharide (LPS) -stimulated macrophages and can significantly reduce TNF-a and ROS levels, thus allowing both drug accumulation in ischemic cardiomyocytes and reducing inflammatory stimulation during MI/RI. Fluorescence imaging results of the targeting effect using a DiR probe also indicated that DiR@T/M-L could accumulate and retain in the ischemic myocardium. Taken together, these results demonstrated the promising application of PUE@T/M-L for mitochondria-targeted drug delivery to achieve maximum therapeutic efficacy of PUE. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
35. MG53 alleviates hypoxia/reoxygenation-induced cardiomyocyte injury by succinylation and ubiquitination modification.
- Author
-
Yan Wang, Hongying Zhou, Jin Wu, and Shanshan Ye
- Subjects
- *
UBIQUITINATION , *POST-translational modification , *ENZYME-linked immunosorbent assay , *MYOCARDIAL infarction , *WOUNDS & injuries - Abstract
Background: Mitsugumin 53 (MG53) is a membrane repair factor that is associated with acute myocardial infarction. This study aimed to investigate the effects of MG53 on cardiomyocyte injury and the posttranslational modification of MG53. Methods: Cardiomyocyte injury was evaluated by enzyme-linked immunosorbent assay and flow cytometry. The succinylation and ubiquitination levels of MG53 were examined by immunoprecipitation (IP) and western blot. The relationship between MG53 and KAT3B or SIRT7 was assessed by co-IP and immunofluorescence. Results: The results showed that overexpression of MG53 inhibited inflammation response and apoptosis of cardiomyocytes induced by hypoxia/reoxygenation (H/R). Succinylation and protein levels of MG53 were downregulated in H/R-induced cells, which was inhibited by SIRT7 and promoted by KAT3B. SIRT7 aggravated and KAT3B alleviated MG53-mediated cardiomyocyte injury. Moreover, MG53 was succinylated and ubiquitinated at K130. Conclusion: SIRT7 inhibited/KAT3B promoted succinylation of MG53 at K130 sites, which suppressed ubiquitination of MG53 and upregulated its protein levels, thereby alleviating H/R-induced cardiomyocyte injury. The findings suggested that MG53 may be a potential therapy for myocardial infarction. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
36. Tongguan capsule for treating myocardial ischemia-reperfusion injury: integrating network pharmacology and mechanism study.
- Author
-
Liu, Jiantao, Liu, Chunping, Chen, Huiqi, Cen, Huan, Yang, Hailong, Liu, Peijian, Liu, Fang, Ma, Liuling, Chen, Quanfu, and Wang, Lei
- Subjects
- *
REPERFUSION injury , *MTOR protein , *RANDOMIZED controlled trials , *PHARMACOLOGY , *MOLECULAR docking - Abstract
Although Tongguan capsule (TGC) is used in the treatment of coronary atherosclerotic disease, the exact mechanism remains unclear. Network pharmacology and experimental validation were applied to examine the mechanism of TGC for treating myocardial ischemia-reperfusion injury (MIRI). The components and candidate targets were searched based on various databases such as TCMSP, TCMID, BATMAN-TCM. The binding ability was determined by molecular docking. The ischemia-reperfusion (I/R) model was constructed by ligating the left anterior descending (LAD) coronary artery. APOE-/- mice were divided into three groups (n = 6): Sham group, I/R group, and TGC group (1 g/kg/d). To further verification, HCAEC cells were subjected to hypoxia-reoxygenation (H/R) to establish in vitro model. The compounds, such as quercetin, luteolin, tanshinone IIA, kaempferol and bifendate, were obtained after screening. The affinity values of the components with GSK-3β, mTOR, Beclin-1, and LC3 were all <-5 kcal/mol. In vivo, TGC improved LVEF and FS, reducing infarct size. In vitro, Hoechst 33258 staining result showed TGC inhibited apoptosis. Compare with the H/R model, TGC treatment increased the levels of GSK-3β, LC3, and Beclin1, while decreasing the expression of mTOR and p62 (p < 0.05). The findings revealed that TGC exerted a cardioprotective effect by up regulating autophagy-related proteins through the mTOR pathway, which may be a therapeutic option for MIRI. However, there are still some limitations in this research. It is necessary to search more databases to obtain information and further demonstrated through randomized controlled trials for generalization. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
37. Anisodine hydrobromide alleviates oxidative stress caused by hypoxia/reoxygenation in human cerebral microvascular endothelial cells predominantly via inhibition of muscarinic acetylcholine receptor 4.
- Author
-
WENLI JIANG, JUNYI SHEN, XIAOQIANG DU, YAN QIU, JIAN ZHONG, ZHI OUYANG, FU, BINGMEI M., and YE ZENG
- Subjects
- *
PARASYMPATHOLYTIC agents , *OXIDATIVE stress , *ENDOTHELIAL cells , *MUSCARINIC acetylcholine receptors , *TRANSCRIPTION factors - Abstract
Background: Anisodine hydrobromide (AT3), an anti-cholinergic agent, could be delivered to the brain across the blood-brain barrier and has been used clinically for the treatment of cerebral ischemia/reperfusion injury. Endothelial dysfunction can be caused by hypoxia/reoxygenation (H/R) via oxidative stress and metabolic alterations. The present study investigated whether AT3 regulates the production of nitric oxide (NO) and reactive oxygen species (ROS), and the HIF-1α pathway via regulation of muscarinic acetylcholine receptors (mAChRs) in brain microvascular endothelial cells after H/R exposure. Methods: Under H/R conditions, hCMEC/D3 cerebral microvascular endothelial cells were treated with AT3. Specific inhibitors of M2- and M4- mAChRs were used to explore the mechanism by which AT3 influences oxidative stress in endothelial cells. Then, mAChRs expression was detected by western blotting and NO production was detected by Greiss reaction. The intracellular ROS level was measured using DCFH-DA probes. The expression of hypoxia-inducible transcription factor 1α (HIF-1α) was also detected. Results: While H/R induced the expression of M2- and M4-mAChRs, AT3 suppressed the H/R-upregulated M2- and M4-mAChRs. H/R also induced the production of NO, ROS, and apoptosis. AT3 and M4-mAChR inhibitors inhibited the H/R-induced production of NO and ROS and apoptosis. HIF-1α was induced by H/R, but was suppressed by AT3. Conclusion: Thus, the in vitro evidence shows that AT3 protects against H/R injury in cerebral microvascular endothelial cells via inhibition of HIF-1α, NO and ROS, predominantly through the downregulation of M4-mAChR. The findings offer novel understandings regarding AT3-mediated attenuation of endothelial cell apoptosis and cerebral ischemia/ reperfusion injury. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. Kynurenic Acid: A Novel Player in Cardioprotection against Myocardial Ischemia/Reperfusion Injuries.
- Author
-
Kamel, Rima, Baetz, Delphine, Gueguen, Naïg, Lebeau, Lucie, Barbelivien, Agnès, Guihot, Anne-Laure, Allawa, Louwana, Gallet, Jean, Beaumont, Justine, Ovize, Michel, Henrion, Daniel, Reynier, Pascal, Mirebeau-Prunier, Delphine, Prunier, Fabrice, and Tamareille, Sophie
- Subjects
- *
REPERFUSION injury , *MYOCARDIAL ischemia , *MYOCARDIAL reperfusion , *REPERFUSION , *MYOCARDIAL infarction , *CELL death , *PROPIDIUM iodide - Abstract
Background: Myocardial infarction is one of the leading causes of mortality worldwide; hence, there is an urgent need to discover novel cardioprotective strategies. Kynurenic acid (KYNA), a metabolite of the kynurenine pathway, has been previously reported to have cardioprotective effects. However, the mechanisms by which KYNA may be protective are still unclear. The current study addressed this issue by investigating KYNA's cardioprotective effect in the context of myocardial ischemia/reperfusion. Methods: H9C2 cells and rats were exposed to hypoxia/reoxygenation or myocardial infarction, respectively, in the presence or absence of KYNA. In vitro, cell death was quantified using flow cytometry analysis of propidium iodide staining. In vivo, TTC-Evans Blue staining was performed to evaluate infarct size. Mitochondrial respiratory chain complex activities were measured using spectrophotometry. Protein expression was evaluated by Western blot, and mRNA levels by RT-qPCR. Results: KYNA treatment significantly reduced H9C2-relative cell death as well as infarct size. KYNA did not exhibit any effect on the mitochondrial respiratory chain complex activity. SOD2 mRNA levels were increased by KYNA. A decrease in p62 protein levels together with a trend of increase in PARK2 may mark a stimulation of mitophagy. Additionally, ERK1/2, Akt, and FOXO3α phosphorylation levels were significantly reduced after the KYNA treatment. Altogether, KYNA significantly reduced myocardial ischemia/reperfusion injuries in both in vitro and in vivo models. Conclusion: Here we show that KYNA-mediated cardioprotection was associated with enhanced mitophagy and antioxidant defense. A deeper understanding of KYNA's cardioprotective mechanisms is necessary to identify promising novel therapeutic targets and their translation into the clinical arena. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. 短链脂肪酸乙酸钠减轻低氧 / 复氧诱导人肾小管上皮细胞系 HK2 损伤.
- Author
-
江罗佳 and 许海波
- Abstract
Objective To study the protective effect and related mechanisms of short-chain fatty acid sodium acetate on hypoxia/reoxygenation (H/R) -induced injury of human renal tubular epithelial cell line HK2. Methods The H/R model was established, and HK2 cells were incubated with sodium acetate (SA) for 2 h. The survival rate of HK2 cells was detected by CCK8 assay; Enzyme activity kit was used for detection of inflammatory factors, cellular reactive oxygen species and ATP production; Flow cytometry was used to measure mitochondrial membrane potential (MMP) and mitochondrial oxidative stress products (mitoSOX) levels; Electron microscope was used to observe mitochondrial ultrastructural damage; Western blot was used to detected the expression of inflammatory signaling pathway IκB-α/NF-κB and mitochondrial energy disorder signaling pathway AMPK/PGC-1α. Results Compared with the control group, the survival rate of HK2 cells in the H/R group was significantly decreased (P<0.05) ; the expression of intracellular ROS, mitoSOX and inflammatory factors was significantly increased (P<0.05) ; the ultrastructure of mitochondria was severely damaged, and the content of ATP and MMP was significantly decreased (P<0.05) ; it was further found that the protein expression of p-IκB-α and NF-κB-p-P65 was significantly increased while the protein expression of p-AMPK and PGC-1α was significantly decreased (P<0.05) . Compared with H/R group, SA significantly enhanced the survival rate of HK2 cells (P<0.05) ; SA inhibited the release of intracellular ROS, mitoSOX and inflammatory factors; SA inhibited mitochondrial ultrastructural damage, decreased ATP and MMP (P<0.05) ; SA promoted the expression of p-AMPK and PGC-1α and inhibited the expression of p-IκB-α and NF-κB-p-P65 (P<0.05) . Conclusions Sodium acetate plays a protective role with potential mechanisms of anti-inflammation, anti-oxidative stress, protection of mitochondrial structure and function of HK2 cells induced by H/R through inhibiting IκB-α/NF-κB and activating AMPK/PGC-1α signal pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Thiopental sodium attenuates hypoxia/reoxygenation-induced injury in osteoblasts by modulating AKT signaling.
- Author
-
Hu, Chuan-yu, Li, Guo-yan, and Li, Chun-Tian
- Abstract
Thiopental sodium (TPTS) is a barbiturate general anesthetic, while its effects on hypoxia/reoxygenation (H/R)-induced injury are still unclear. This study aimed to investigate whether TPTS exerts protective effects against the H/R-induced osteoblast cell injury and explore the underlying mechanisms. Osteoblast cell injury model was induced by the H/R condition, which was treated with or without TPTS. Cell viability and lactate dehydrogenase (LDH) release were determined by the corresponding commercial kits. The levels of oxidative stress were determined in the experimental groups. Cell apoptosis and Caspase-3 activities were determined by propidium iodide staining and substrate-based assay, respectively. Western blotting and qRT-PCR were performed to measure the mRNA and protein levels, respectively. Treatment with TPTS was able to increase cell viability and reduce LDH release in H/R-induced osteoblasts. Additionally, TPTS regulated oxidative stress in H/R-induced osteoblasts by suppressing malondialdehyde (MDA) and reactive oxygen species (ROS) as well as boosting superoxide dismutase (SOD). TPTS was able to suppress cell apoptosis by suppressing Caspase-3 activity and cleavage. TPTS exerted protective effects against cell injury and apoptosis induced by the H/R conditions, which were associated with its regulation of Akt signaling. Moreover, TPTS induced osteoblast differentiation under the H/R condition. In summary, TPTS attenuates H/R-induced injury in osteoblasts by regulating AKT signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Neferine protected cardiomyocytes against hypoxia/oxygenation injury through SIRT1/Nrf2/HO‐1 signaling.
- Author
-
Lu, Cheng, Jiang, Bing, Xu, Jie, Zhang, Xuan, and Jiang, Nianxin
- Subjects
NUCLEAR factor E2 related factor ,CELL death ,MYOCARDIAL infarction - Abstract
Acute myocardial infarction is regarded as myocardial necrosis resulting from myocardial ischemia/reperfusion (I/R) damage and retains a major cause of mortality. Neferine, which was extracted from the green embryos of mature seeds of Nelumbo nucifera Gaertn., has been reported to possess a broad range of biological activities. However, its underlying mechanism on the protective effect of I/R has not been fully clarified. A hypoxia/reoxygenation (H/R) model with H9c2 cells closely simulating myocardial I/R injury was used as a cellular model. This study intended to research the effects and mechanism underlying neferine on H9c2 cells in response to H/R stimulation. Cell Counting Kit‐8 and lactate dehydrogenase (LDH) release assays were employed to measure cell viability and LDH, respectively. Apoptosis and reactive oxygen species (ROS) were determined by flow cytometry analysis. Oxidative stress was evaluated by detecting malondialdehyde, superoxide dismutase, and catalase. Mitochondrial function was assessed by mitochondrial membrane potential, ATP content, and mitochondrial ROS. Western blot analysis was performed to examine the expression of related proteins. The results showed that hypoxia/reoxygenation (H/R)‐induced cell damage, all of which were distinctly reversed by neferine. Moreover, we observed that neferine inhibited oxidative stress and mitochondrial dysfunction induced by H/R in H9c2 that were concomitant with increased sirtuin‐1 (SITR1), nuclear factor erythroid 2‐related factor 2 (Nrf2), and heme oxygenase‐1 expression. On the contrary, silencing the SIRT1 gene with its small interferingRNA eliminated the beneficial effects of neferine. It is concluded that neferine preconditioning attenuated H/R‐induced cardiac damage via suppressing apoptosis, oxidative stress, and mitochondrial dysfunction, which may be partially ascribed to the activation of SIRT1/Nrf2 signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. The Role of δ2-Opioid Receptors in the Regulation of Tolerance of Isolated Cardiomyocytes to Hypoxia and Reoxygenation
- Author
-
Mukhomedzyanov, A. V., Popov, S. V., Naryzhnaya, N. V., Azev, V. N., and Maslov, L. N.
- Published
- 2024
- Full Text
- View/download PDF
43. 下调 miR-320a 表达对缺氧/复氧诱导的心肌细胞增殖和 凋亡的影响.
- Author
-
李红英, 王晨燕, 郭世超, 赵友为, 董彦博, and 黄建成
- Subjects
- *
GENE expression , *BCL-2 proteins , *JAK-STAT pathway , *LACTATE dehydrogenase , *REPORTER genes - Abstract
Objective: To discuss the effect of down-regulation of miR-320a expression on the myocardial hypoxia/reoxygenation (H/R)injury model, and to clarify its related mechanism. Methods: Real-time fluorescence quantitative PCR (RT-qPCR)method was used to detect the expression levels of miR-320a in serum of the patients with actue myocardial infarction (AMI)and the myocardial H9C2 cells induced by H/R. The miR-320a inhibitor, inhibitor NC, small interference Janus kinase 2 (si-JAK2), and si-NC plasmids were transfected into the H9C2 cells respectively, and blank control group was set up. After successful transfection, the H/R treatment was performed. The H9C2 cells were divided into control group, H/R group, H/R + inhibitor NC group, H/R + miR-320a inhibitor group, H/R + miR-320a inhibitor + si-NC group and H/R + miR-320a inhibitor + si-JAK2 group. The targeting relationship between miR-320a and Janus kinase 2 (JAK2)was detected by double luciferase reporter gene; the proliferation rate of cells in various groups were detected by CCK-8 assay; the activities of superoxide dismutase (SOD)and levels of malonaldehyde (MDA)in cells and the levels of lactate dehydrogenase (LDH)in cell culture supernanant in various groups were detected by biochemical method; the apoptotic rates of cells in various groups were detected by flow cytometry; the expression levels of miR-320a and JAK2 mRNA in cells in various groups were detected by RT-qPCR method; the expression levels of B-cell lymphoma-2 (Bcl-2), Bcl-2 associated X protein (Bax), cleaved-cysteinyl aspartate specific proteinase-3 (cleaved-caspase-3), JAK2, signal transducers and activator of transcription 3 (STAT3), and phosphorylated STAT3 (p-STAT3)proteins in cells in various groups were detected by Western blotting method. Results: The expression levels of miR-320a in serum of the patients with AMI and the myocardial H9C2 cells in H/R group were significantly higher than those in control group (P<0. 05). The results of double Luciferase reporter gene detection suggested that miR-320a could targetedly bind with JAK2. Compared with control group, the proliferation rate of the cells and SOD activity in the cells in H/R group were decreased significantly (P<0. 05), the apoptotic rate of the cells, MDA level and LDH activity in the cells were significantly increased (P<0. 05), the expression levels of Bcl-2 and JAK2 proteins and ratio of p-STAT3/STAT3 in the cells were significantly decreased (P<0. 05), and the expression levels of Bax and cleaved caspase-3 proteins in the cells were significantly increased (P<0. 05). Compared with H/R group, the proliferation rate of the cells and SOD activity in the cells in H/R+miR-320a inhibitor group were increased (P<0. 05), while the apoptotic rate of the cells, MDA level in the cells, LDH activity in the cell culture supernanant were decreased (P<0. 05), the expression levels of Bcl-2 and JAK2 proteins and ratio of p-STAT3/STAT3 in the cells were significantly increased (P<0. 05), the expression levels of Bax and cleaved- caspase-3 proteins in the cells were significantly decreased (P<0. 05). Compared with H/R+miR-320a inhibitor+ si-NC group, the proliferation rate of the cells and SOD activity in the cells in H/R+miR-320a inhibitor+ si-JAK2 group were decreased (P<0. 05), and the apoptotic rate of the cells, MDA level in the cells, and LDH activity in the cell culture supernanant were increased (P<0. 05). Conclusion: Down-regulation of miR-320a expression can inhibit the apoptosis of the cardiomyocytes induced by H/R and increase the proliferation activity of cells, and its mechanism is related to the targeted regulation of JAK2/STAT3 signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. A direct interaction between RhoGDIα/Tau alleviates hyperphosphorylation of Tau in Alzheimer's disease and vascular dementia.
- Author
-
Zhang, Heping, Lu, Fan, Liu, Panhong, Qiu, Zhaohui, Li, Jianling, Wang, Xiaotong, Xu, Hui, Zhao, Yandong, Li, Xuemin, Wang, Huadong, Lu, Daxiang, and Qi, Renbin
- Abstract
RhoGDIα is an inhibitor of RhoGDP dissociation that involves in Aβ metabolism and NFTs production in Alzheimer's disease (AD) by regulating of RhoGTP enzyme activity. Our previous research revealed that RhoGDIα, as the target of Polygala saponin (Sen), might alleviate apoptosis of the nerve cells caused by hypoxia/reoxygenation (H/R). To further clarify the role of RhoGDIα in the generation of NFTs, we explored the relationship between RhoGDIα and Tau. We found out that RhoGDIα and Tau can bind with each other and interact by using coimmunoprecipitation (Co-IP) and GST pulldown methods in vitro. This RhoGDIα-Tau partnership was further verified by using immunofluorescence colocalization and fluorescence resonance energy transfer (FRET) approaches in PC12 cells. Using the RNA interference (RNAi) technique, we found that the RhoGDIα may be involved in an upstream signaling pathway for Tau. Subsequently, in Aβ
25-35 - and H/R-induced PC12 cells, forced expression of RhoGDIα via cDNA plasmid transfection was found to reduce the hyperphosphorylation of Tau, augment the expression of bcl-2 protein, and inhibit the expression of Bax protein (reducing the Bax/bcl-2 ratio) and the activity of caspase-3. In mouse AD and VaD models, forced expression of RhoGDIα via injection of a viral vector (pAAV-EGFP-RhoGDIα) into the lateral ventricle of the brain alleviated the pathological symptoms of AD and VaD. Finally, GST pulldown confirmed that the binding sites on RhoGDIα for Tau were located in the range of the ΔC33 fragment (aa 1–33). These results indicate that RhoGDIα is involved in the phosphorylation of Tau and apoptosis in AD and VaD. Overexpression of RhoGDIα can inhibit the generation of NFTs and delay the progress of these two types of dementia. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
45. MicroRNA‐17‐3p protects against excessive posthypoxic autophagy in H9C2 cardiomyocytes via PTEN–Akt–mTOR signaling pathway.
- Author
-
He, Yi, Zhang, Dengwen, Zhang, Qingqing, Cai, Yin, Huang, Chongfeng, Xia, Zhengyuan, and Wang, Sheng
- Subjects
- *
LACTATE dehydrogenase , *PROTEIN kinase B , *AUTOPHAGY , *PTEN protein , *CELLULAR signal transduction - Abstract
The activity of phosphatase and tensin homolog (PTEN) can be inhibited by miR‐17‐3p, which results in attenuating myocardial ischemia/reperfusion injury (IRI), however, the mechanism behind this phenomenon is still elusive. Suppression of PTEN leads to augmented protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling strength and constrained autophagy activation, which might be the one mechanism for the ameliorated myocardial IRI. Thus, we tested the hypothesis that miR‐17‐3p attenuated hypoxia/reoxygenation (H/R)‐mediated damage in cardiomyocytes by downregulating excessive autophagy via the PTEN–Akt–mTOR axis. The expression of miR‐17‐3p was remarkably increased after H/R treatment (6‐h hypoxia followed by 6‐h reoxygenation; H6/R6), which was concomitant with the increase of the release of lactic acid dehydrogenase (cell injury marker) and the enhancement LC3II/I ratio (autophagy markers) in H9C2 cardiomyocytes. Ectoexpression of miR‐17‐3p agomir led to remarkable augmentation of miR‐17‐3p expression and evidently attenuated H/R‐mediated cell damage and excessive autophagy. Furthermore, an increase in miR‐17‐3p expression elicited constrained phosphorylation of PTEN (Ser380) while enhanced the phosphorylation of Akt (Thr308, Ser473) and mTOR (Ser536) after H/R stimulation. In addition, pretreatment with LY‐294002 (an Akt selective inhibitor) and rapamycin (an mTOR selective inhibitor) significantly abrogated the protective function of miR‐17‐3p on H/R‐mediated cell damage and autophagy in H9C2 cardiomyocytes. Taken together, these observations indicated that the enhancement of the PTEN/Akt/mTOR axis and the consequent suppression of autophagy overactivation might represent an underlying mechanism by which miR‐17‐3p attenuated H/R‐mediated damage in H9C2 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. DUSP12 ameliorates myocardial ischemia–reperfusion injury through HSPB8‐induced mitophagy.
- Author
-
Cheng, Jing, Ji, Meihua, Jing, Haijuan, and Lin, Hongqi
- Subjects
REPERFUSION injury ,WESTERN immunoblotting ,CREATINE kinase ,TETRAZOLIUM chloride ,LACTATE dehydrogenase - Abstract
This study aimed to explore the role of dual specificity phosphatase 12 (DUSP12) in regulating myocardial ischemia–reperfusion (I/R) injury and the underlying mechanism. The expression of DUSP12 in myocardial tissues and heat‐shock protein beta‐8 (HSPB8) and mitophagy‐related proteins in myocardial tissues and H9c2 cells were detected by western blot analysis. The serum creatine kinase isoenzymes (CK‐MB) and lactate dehydrogenase (LDH), levels of reactive oxygen species and malondialdehyde, superoxide dismutase activity in myocardial tissues and H9c2 cells, and caspase‐3 activity in H9c2 cells were analyzed by corresponding assay kits. The infarct area in the rat's heart was observed by triphenyl tetrazolium chloride staining. The apoptosis of myocardial cells in myocardial tissues and H9c2 cells was detected by terminal‐deoxynucleotidyl transferase dUTP‐biotin nick‐end labeling assay. The interaction between DUSP12 and HSPB8 was clarified by the coimmunoprecipitation assay. The transfection efficacy of si‐HSPB8#1 and si‐HSPB8#2 in H9c2 cells was confirmed by real‐time quantitative‐polymerase chain reaction and western blot analysis. As a result, DUSP12 expression was downregulated in I/R rats, which was promoted by lentivirus‐expressing DUSP12. DUSP12 overexpression reduced the serum creatine kinase isoenzymes (CK‐MB) and LDH, decreased the infarct area in the rat's heart, and suppressed the apoptosis and oxidative stress in myocardial tissues. DUSP12 overexpression also upregulated the expression of HSPB8 to promote mitophagy. The coimmunoprecipitation assay indicated that DUSP12 could be combined with HSPB8. In addition, DUSP12 overexpression could inhibit hypoxia/reoxygenation‐elicited apoptosis as well as oxidative stress in H9c2 cells by upregulating HSPB8 expression to promote mitophagy, which was countervailed by HSPB8 deficiency. In conclusion, DUSP12 overexpression decreased the apoptosis and oxidative stress in myocardial I/R injury through HSPB8‐induced mitophagy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
47. Dexmedetomidine post‐treatment attenuates cardiac ischaemia/reperfusion injury by inhibiting apoptosis through HIF‐1α signalling
- Author
-
Peng, Ke, Chen, Wei‐rong, Xia, Fan, Liu, Hong, Meng, Xiao‐wen, Zhang, Juan, Liu, Hua‐yue, Xia, Zheng‐yuan, and Ji, Fu‐hai
- Subjects
Cardiovascular ,Heart Disease ,Heart Disease - Coronary Heart Disease ,2.1 Biological and endogenous factors ,Aetiology ,Adrenergic alpha-2 Receptor Agonists ,Animals ,Apoptosis ,Dexmedetomidine ,Disease Models ,Animal ,Gene Expression Regulation ,Hypoxia-Inducible Factor 1 ,alpha Subunit ,Male ,Myocardial Ischemia ,Myocardial Reperfusion Injury ,Protective Agents ,Rats ,Rats ,Sprague-Dawley ,cardiac apoptosis ,cardioprotection ,dexmedetomidine ,HIF-1 alpha ,hypoxia ,reoxygenation ,ischaemia ,reperfusion ,HIF-1α ,hypoxia/reoxygenation ,ischaemia/reperfusion ,Medicinal and Biomolecular Chemistry ,Biochemistry and Cell Biology ,Clinical Sciences ,Biochemistry & Molecular Biology - Abstract
Hypoxia-inducible factor 1α (HIF-1α) plays a critical role in the apoptotic process during cardiac ischaemia/reperfusion (I/R) injury. This study aimed to investigate whether post-treatment with dexmedetomidine (DEX) could protect against I/R-induced cardiac apoptosis in vivo and in vitro via regulating HIF-1α signalling pathway. Rat myocardial I/R was induced by occluding the left anterior descending artery for 30 minutes followed by 6-hours reperfusion, and cardiomyocyte hypoxia/reoxygenation (H/R) was induced by oxygen-glucose deprivation for 6 hours followed by 3-hours reoxygenation. Dexmedetomidine administration at the beginning of reperfusion or reoxygenation attenuated I/R-induced myocardial injury or H/R-induced cell death, alleviated mitochondrial dysfunction, reduced the number of apoptotic cardiomyocytes, inhibited the activation of HIF-1α and modulated the expressions of apoptosis-related proteins including BCL-2, BAX, BNIP3, cleaved caspase-3 and cleaved PARP. Conversely, the HIF-1α prolyl hydroxylase-2 inhibitor IOX2 partly blocked DEX-mediated cardioprotection both in vivo and in vitro. Mechanistically, DEX down-regulated HIF-1α expression at the post-transcriptional level and inhibited the transcriptional activation of the target gene BNIP3. Post-treatment with DEX protects against cardiac I/R injury in vivo and H/R injury in vitro. These effects are, at least in part, mediated via the inhibition of cell apoptosis by targeting HIF-1α signalling.
- Published
- 2020
48. c-Jun-mediated miR-19b expression induces endothelial barrier dysfunction in an in vitro model of hemorrhagic shock
- Author
-
Feng Wu, Jian-Ying Wang, Brooke Dorman, Ahmad Zeineddin, and Rosemary Ann Kozar
- Subjects
Lung microvascular endothelial cells ,Hypoxia/reoxygenation ,miR-19b ,c-Jun ,Endothelial barrier ,Therapeutics. Pharmacology ,RM1-950 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Our previous data demonstrated that miR-19b expression was increased in human lung microvascular endothelial cells in-vitro-, in-vivo and in patients with hemorrhagic shock, leading to a decrease in syndecan-1 mRNA and protein and resulting in loss of endothelial barrier function. However, the mechanism underlying increased miR-19b expression remains unclear. The objective of the current study was to determine if c-Jun mediates the early responsive microRNA, miR-19b, to cause endothelial barrier dysfunction. Method Human lung microvascular endothelial cells (HLMEC) or HEK293T cells were transfected with c-Jun overexpressing vector, c-Jun siRNA, miR-19b promoter vector, miR-19b mutated promoter vector, miR-19b oligo inhibitor, then subjected to hypoxia/reoxygenation as in-vitro model of hemorrhagic shock. Levels of protein, miRNA, and luciferase activity were measured. Transwell permeability of endothelial monolayers were also determined. Plasma levels of c-Jun were measured in injured patients with hemorrhagic shock. Result Hypoxia/reoxygenation induced primary (pri-)miR-19b, mature miR-19b, and c-Jun expression over time in a comparable timeframe. c-Jun silencing by transfection with its specific siRNA reduced both pri-miR-19b and mature miR-19b levels. Conversely, c-Jun overexpression enhanced H/R-induced pri-miR-19b. Studies using a luciferase reporter assay revealed that in cells transfected with vectors containing the wild-type miR-19b promoter and luciferase reporter, c-Jun overexpression or hypoxia/ reoxygenation significantly increased luciferase activity. c-Jun knockdown reduced the luciferase activity in these cells, suggesting that the miR-19b promoter is directly activated by c-Jun. Further, chromatin immunoprecipitation assay confirmed that c-Jun directly bound to the promoter DNA of miR-19b and hypoxia/reoxygenation significantly increased this interaction. Additionally, c-Jun silencing prevented cell surface syndecan-1 loss and endothelial barrier dysfunction in HLMECs after hypoxia/reoxygenation. Lastly, c-Jun was significantly elevated in patients with hemorrhagic shock compared to healthy controls. Conclusion Transcription factor c-Jun is inducible by hypoxia/reoxygenation, binds to and activates the miR-19b promoter. Using an in-vitro model of hemorrhagic shock, our findings identified a novel cellular mechanism whereby hypoxia/ reoxygenation increases miR-19b transcription by inducing c-Jun and leads to syndecan-1 decrease and endothelial cell barrier dysfunction. This finding supports that miR-19b could be a potential therapeutic target for hemorrhage shock.
- Published
- 2022
- Full Text
- View/download PDF
49. Botulinum toxin type A activates protective autophagy by modulating endoplasmic reticulum stress in hypoxia/reoxygenation-treated endothelial cells
- Author
-
Yinhua Zhao, Huang Lin, Jiaxing Ma, and Yue Cui
- Subjects
Botulinum toxin type A ,Human dermal microvascular endothelial cells ,Endoplasmic reticulum stress ,Autophagy ,Hypoxia/reoxygenation ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Botulinum toxin type A (BTXA) previously protected endothelial cells in free skin flaps from ischemia/reperfusion injury by inducing autophagy. Endoplasmic reticulum (ER) stress-autophagy activation may have a role in BTXA-antagonized ischemia/reperfusion damage in human dermal microvascular endothelial cells (HDMECs), however, this has yet to be proven. HDMECs were pretreated with BTXA at various concentrations for 12 h before being subjected to hypoxia and reoxygenation (H/R). Cell Count Kit 8 (CCK8) and Western blot (WB) data showed that H/R treatment significantly increased the expression of ER stress (GRP78, CHOP) and autophagy (LC3II/I, Beclin-1) proteins. The optimal BTXA pretreatment concentration was 1.6 U/mL, which resulted in the highest levels of cell survival and expression of ER stress and autophagy. Following pretreatment with 1.6 U/mL BTXA and the addition of the ER stress inducer Thapsigargin (Tg), the ER stress inhibitor 4-phenylbutyrate (4-PBA), and the inhibitor of autophagy Bafilomycin A1 (Baf A1), respectively, HDMECs were then subjected to H/R treatment. Cell survival and the percentage of ethynyldeoxyuridine-labeled cells in the BTXA pretreatment groups were reduced by the addition of Tg, 4-PBA, and Baf A1. While the expression of GRP78, CHOP, and LC3 was upregulated by Tg and Baf A1, it was downregulated by 4-PBA. The findings showed that ER stress produced by BTXA pretreatment triggers protective autophagy and protects HDMECs from H/R damage. There were no cytoprotective effects from either excessive activation or reduction of ER stress. Our results are consistent with the notion that autophagy and moderate ER stress are critical for cellular longevity and, consequently, functional integrity and may represent a potential therapeutic target.
- Published
- 2023
- Full Text
- View/download PDF
50. 藏红花素减轻皮质神经元缺氧复氧损伤与抑制 TLR4/NF-κB 信号通路有关.
- Author
-
胡科, 李晓蕾, and 麻瑞娟
- Subjects
- *
HIGH mobility group proteins , *BAX protein , *PYROPTOSIS , *CROCIN , *GENE expression , *FORKHEAD transcription factors - Abstract
Aim To investigate the effect of Crocin on hypoxia/reoxygenation injury of cortical neurons and its molecular mechanism. Methods The cortical neurons of SD rats were isolated and cultured, and the control group, model group, Crocin (50 mg/L) group, Crocin (50 mg/L)+lipopolysaccharide (TLR4 activator, 100 μg/L) group were set. The cortical neurons in control group were routinely cultured; the cortical neurons in model group were given hypoxia 4 h and reoxygenation 24 h; the cortical neurons in Crocin group and Crocin+lipopolysaccharide group were intervened for 24 h, and then the model was established. The neuronal viability and apoptosis rate were detected by CCK-8 method and Annexin V-FITC/PI double staining method. The levels of inflammatory factors in the culture medium were detected by ELISA method. The expression of toll-like receptor 4 (TLR4), nuclear factor-κB p65 (NF-κB p65), p-NF-κB p65, inhibitor α of NF-κB (IκBα), p-IκBα, high mobility group box protein B1 (HMGB1), cleaved Caspase-3, B lymphoma-2 gene (Bcl-2), Bcl-2-associated X protein (Bax) were detected by Western blot method. Results Compared with the control group, the activity of cortical neurons in the model group decreased significantly (P<0. 05); the apoptosis rate, the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6 in the culture medium, the expressions of TLR4, HMGB1, cleaved Caspase-3 and the ratio of p-NF-κB p65/NF-κB p65, p-IκBα/IκBα, Bax/Bcl-2 increased significantly (P<0. 05). Compared with the model group, the activity of cortical neurons in Crocin group increased by 94. 97% (P<0. 05); the apoptosis rate decreased by 65. 80% (P<0. 05); the levels of TNF-α, IL-1β, IL-6 in the culture medium decreased by 61. 86%, 78. 34%, 63. 42% (P<0. 05); the expressions of TLR4, HMGB1, cleaved Caspase-3 and the ratio of p-NF-κB p65/NF-κB p65, p-IκBα/IκBα, Bax/Bcl-2 decreased by 73. 43%, 52. 13%, 81. 52%, 69. 70%, 60. 55%, 95. 05% (P<0. 05). Lipopolysaccharide significantly reversed the regulatory effects of Crocin on hypoxia/reoxygenation injured cortical neurons (P<0. 05). Conclusion Crocin can inhibit the apoptosis and inflammatory response of hypoxia/reoxygenation injured neurons, and has protective effect on hypoxia/reoxygenation injury of cortical neurons, which mechanism may be related to the inhibition of TLR4/NF-κB signaling pathway activation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.