1
|
Pan P, Wang X, Chen Y, Chen Q, Yang Y, Wei C, Cheng T, Wan H, Yu D. Effect of Hcp Iron Ion Regulation on the Interaction Between Acinetobacter baumannii With Human Pulmonary Alveolar Epithelial Cells and Biofilm Formation. Front Cell Infect Microbiol 2022; 12:761604. [PMID: 35281445 PMCID: PMC8905654 DOI: 10.3389/fcimb.2022.761604] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
Acinetobacter baumannii is a type of bacterial nosocomial infection with severe drug resistance. Hemolysin co-regulated protein (Hcp) is a marker of activated type VI secretion system (T6SS), a key secretory system that promotes Gram-negative bacteria colonization, adhesion, and invasion of host cells. Hcp is also regulated by iron ions (Fe). In this study, an ATCC17978 hcp deletion strain (ATCC17978Δhcp), an hcp complement strain (ATCC17978Δhcp+), and an A. baumannii–green fluorescent protein (GFP) strain were constructed and used to investigate the role of hcp in bacterial adhesion to cells (human pulmonary alveolar epithelial cells (HPAEpiC)) and biofilm formation. Our results indicate that the inhibitory concentrations of the three A. baumannii strains (ATCC17978 wild type, ATCC17978Δhcp, and ATCC17978Δhcp+) were drug-sensitive strains. A. baumannii hcp gene and iron ions might be involved in promoting the formation of a biofilm and host–bacteria interaction. Iron ions affected the ability of A. baumannii to adhere to cells, as there was no significant difference in the bacterial numbers when assessing the adhesion of the three strains to HPAEpiC in the presence of iron ion concentrations of 0 μM (F = 3.1800, p = 0.1144), 25 μM (F = 2.067, p = 0.2075), 100 μM (F = 30.52, p = 0.0007), and 400 μM (F = 17.57, p = 0.0031). The three strains showed significant differences in their ability to adhere to HPAEpiC. The numbers of bacteria adhesion to HPAEpiC were ATCC17978Δhcp>ATCC17978Δhcp+>ATCC17978 in descending order. Hcp gene was positively regulated by iron ions in the bacteria–cells’ co-culture. It is speculated that the effect of iron ions on the interaction between A. baumannii and HPAEpiC might be related to the transport function of hcp and bacterial immune escape mechanisms.
Collapse
Affiliation(s)
- Ping Pan
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Medical Laboratory, Hangzhou Women’s Hospital, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolei Wang
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Chen
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Medical Laboratory, Zhejiang Hospital, Hangzhou, China
| | - Qiong Chen
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yunxing Yang
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxing Wei
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tongtong Cheng
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Haitong Wan, ; Daojun Yu,
| | - Daojun Yu
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Haitong Wan, ; Daojun Yu,
| |
Collapse
|
2
|
Huang Y, Sun X, Juan Z, Zhang R, Wang R, Meng S, Zhou J, Li Y, Xu K, Xie K. Dexmedetomidine attenuates myocardial ischemia-reperfusion injury in vitro by inhibiting NLRP3 Inflammasome activation. BMC Anesthesiol 2021; 21:104. [PMID: 33823789 PMCID: PMC8022424 DOI: 10.1186/s12871-021-01334-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background Myocardial ischemia-reperfusion injury (MIRI) is the most common cause of death worldwide. The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome plays an important role in the inflammatory response to MIRI. Dexmedetomidine (DEX), a specific agonist of α2-adrenergic receptor, is commonly used for sedation and analgesia in anesthesia and critically ill patients. Several studies have shown that dexmedetomidine has a strong anti-inflammatory effect in many diseases. Here, we investigated whether dexmedetomidine protects against MIRI by inhibiting the activation of the NLRP3 inflammasome in vitro. Methods We established an MIRI model in cardiomyocytes (CMs) alone and in coculture with cardiac fibroblasts (CFs) by hypoxia/reoxygenation (H/R) in vitro. The cells were treated with dexmedetomidine with or without MCC950 (a potent selective NLRP3 inhibitor). The beating rate and cell viability of cardiomyocytes, NLRP3 localization, the expression of inflammatory cytokines and NLRP3 inflammasome-related proteins, and the expression of apoptosis-related proteins, including Bcl2 and BAX, were determined. Results Dexmedetomidine treatment increased the beating rates and viability of cardiomyocytes cocultured with cardiac fibroblasts. The expression of the NLRP3 protein was significantly upregulated in cardiac fibroblasts but not in cardiomyocytes after H/R and was significantly attenuated by dexmedetomidine treatment. Expression of the inflammatory cytokines IL-1β, IL-18 and TNF-α was significantly increased in cardiac fibroblasts after H/R and was attenuated by dexmedetomidine treatment. NLRP3 inflammasome activation induced the increased expression of cleaved caspase1, mature IL-1β and IL-18, while dexmedetomidine suppressed H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. In addition, dexmedetomidine reduced the expression of Bcl2 and BAX in cocultured cardiomyocytes by suppressing H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. Conclusion Dexmedetomidine treatment can suppress H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts, thereby alleviating MIRI by inhibiting the inflammatory response. Supplementary Information The online version contains supplementary material available at 10.1186/s12871-021-01334-5.
Collapse
Affiliation(s)
- Yaru Huang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Xiaotong Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Zhaodong Juan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China.
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Ruoguo Wang
- Department of Pain, Affiliated Hospital of Weifang Medical University, Weifang, 261000, China
| | - Shuqi Meng
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Jiajia Zhou
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Yan Li
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Keyou Xu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Keliang Xie
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China.
| |
Collapse
|
3
|
Chen W, Wang Y, Pan Z, Chen X, Luo D, Wang H. Protective effects of dexmedetomidine on the ischemic myocardium in patients undergoing rheumatic heart valve replacement surgery. Exp Ther Med 2021; 21:427. [PMID: 33747166 PMCID: PMC7967827 DOI: 10.3892/etm.2021.9844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to compare the effects of two methods of dexmedetomidine (Dex) administration on myocardial injury, inflammation and stress in ischemic myocardium during rheumatic heart valve replacement. In total, 90 patients were included in the present study and were divided into the following three groups: i) Dex group (1.0 µg/kg Dex pre-administered 10 min prior to anesthesia, then 0.5 µg/kg/h Dex for maintenance); ii) Dex pre-conditioning group (Pre-Dex; 1.0 µg/kg Dex administered 10 min prior to anesthesia, then saline for maintenance); and iii) control group (saline 10 min prior to anesthesia and saline during maintenance), with 30 patients in each group. Heart rate (HR) and mean artery pressure (MAP) were recorded at eight time-points: i) T1, pre-medication; ii) T2, 10 min post-medication; iii) T3, immediately post-intubation; iv) T4, upon skin incision; v) T5, upon sawing the sternum; vi) T6, immediately post-cardiopulmonary bypass; vii) T7, immediately post-operation; and viii) T8, 24 h post-operation. The serum cardiac troponin I (cTnI), interleukin (IL)-8, IL-10 and malondialdehyde (MDA) levels were also detected at T1, T6, T7 and T8. Blood glucose levels were detected at T1, T5, T6 and T7. In comparison with the control group, patients in the Dex group exhibited a significant increase in cardiac function, as indicated by an increase in HR, MAP and IL-10 levels, and a significant decrease in cTnI, IL-8, MDA and glucose levels. Both Dex perfusion and Dex preconditioning were able to reduce myocardial injury, inflammation, oxidative stress and stress response in rheumatic heart valve replacement surgery. However, Dex perfusion during the whole surgery was more effective than Dex preconditioning treatment. The study was registered with the Chinese Clinical Trial Registry (ChiCTR; no. ChiCTR-INR-17011955).
Collapse
Affiliation(s)
- Wei Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yan Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhiguo Pan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xiyuan Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Dihuan Luo
- Department of Anesthesiology, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
4
|
Zhu Z, Ling X, Zhou H, Zhang C. Dexmedetomidine at a dose of 1 µM attenuates H9c2 cardiomyocyte injury under 3 h of hypoxia exposure and 3 h of reoxygenation through the inhibition of endoplasmic reticulum stress. Exp Ther Med 2020; 21:132. [PMID: 33376514 PMCID: PMC7751463 DOI: 10.3892/etm.2020.9564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) has been confirmed to induce endoplasmic reticulum stress (ERS) during downstream cascade reactions after the sufficient deterioration of cardiomyocyte function. However, clinically outcomes have been inconsistent with experimental findings because the mechanism has not been entirely elucidated. Dexmedetomidine (DEX), an α2 adrenergic receptor agonist with anti-inflammatory and organ-protective activity, has been shown to attenuate IRI in the heart. The present study aimed to determine whether DEX is able to protect injured cardiomyocytes under in vitro hypoxia/reoxygenation (H/R) conditions and evaluate the conditions under which ERS is efficiently ameliorated. The cytotoxicity of DEX in H9c2 cells was evaluated 24 h after treatment with several different concentrations of DEX. The most appropriate H/R model parameters were determined by the assessment of cell viability and injury with Cell Counting Kit-8 and lactate dehydrogenase (LDH) release assays after incubation under hypoxic conditions for 3 h and reoxygenation conditions for 3, 6, 12 and 24 h. Additionally, the aforementioned methods were used to assess cardiomyocytes cultured with various concentrations of DEX under H/R conditions. Furthermore, the degree of apoptosis and the mRNA and protein expression levels of glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and caspase-12 were evaluated in all groups. The addition of 1, 5 and 10 µM DEX to the cell culture significantly increased the proliferation of H9c2 cells by >80% under normal culture conditions. In the H/R model assessment, following 3 h of anoxia exposure, H9c2 cell viability decreased to 62.67% with 3 h of reoxygenation and to 36% with 6 h of reoxygenation compared with the control. The viability of H9c2 cells subjected to hypoxia for 3 h and reoxygenation for 3 h increased by 61.3% when pretreated with 1 µM DEX, and the LDH concentration in the supernatant was effectively decreased by 13.7%. H/R significantly increased the percentage of apoptotic cells, as detected by flow cytometry, and increased the expression levels of GRP78, CHOP and caspase-12, while treatment with either DEX or 4-phenylbutyric acid (4-PBA) significantly attenuated these effects. Additionally, despite the protective effect of DEX against H/R injury, 4-PBA attenuated the changes induced by DEX and H/R. In conclusion, treatment with 1 µM DEX alleviated cell injury, apoptosis and the increases in GRP78, CHOP and caspase-12 expression levels in H9c2 cells induced by 3 h of hypoxia and 3 h of reoxygenation.
Collapse
Affiliation(s)
- Zhipeng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaoyan Ling
- Outpatient Nursing Department, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Hongmei Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Caijun Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
5
|
Shi X, Liu Z, Li J. Protective effects of dexmedetomidine on hypoxia/reoxygenation injury in cardiomyocytes by regulating the CHOP signaling pathway. Mol Med Rep 2020; 22:3307-3315. [PMID: 32945482 PMCID: PMC7453597 DOI: 10.3892/mmr.2020.11442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Hypoxia/reoxygenation (H/R) injury in myocardial cells occurs frequently during cardiac surgery and affects the prognosis of patients. The present study aimed to investigate the protective effects of dexmedetomidine (Dex) on H/R injury and its association with the C/EBP-homologous protein (CHOP) signaling pathway. An H/R model was constructed in H9C2 cells to investigate the effects of Dex on H/R injury. Cell viability, apoptosis and lactate dehydrogenase (LDH) levels were determined by MTT, flow cytometry and 2,4-dinitrophenylhydrazine colorimetric assays, respectively. The expression levels of inflammatory factors were measured by reverse transcription-quantitative PCR (RT-qPCR), and CHOP and glucose-regulated protein-78 (Grp78) expression levels were detected by RT-qPCR and western blotting. CHOP was overexpressed or knocked down to detect the cell viability, apoptosis, LDH level and the expression levels of inflammatory factors and Grp78. The results demonstrated that in the H/R group, cell viability was lower and apoptosis was higher, and that higher levels of LDH and inflammatory factors were present compared with those in the Dex+H/R group. Silencing of CHOP significantly reversed the H/R-reduced cell viability, high apoptotic rate and LDH levels, as well as the elevated expression levels of inflammatory factors and Grp78 caused by H/R injury, whereas the overexpression of CHOP inhibited cell viability and promoted apoptosis, elevated LDH level and expression of inflammatory factors and Grp78 compared with the negative control. Additionally, pretreatment with Dex significantly alleviated the H/R injury; thus, Dex may protect H9C2 cells against H/R induced cell injury, possibly by suppressing the CHOP signaling pathway.
Collapse
Affiliation(s)
- Xiaoqiao Shi
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhiwen Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Junwei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
6
|
Qin SQ, Zhang ZS, Wang XY, Shi JZ, Yang XB. MiR-24 Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Injury Through Regulating Mitogen-Activated Protein Kinase 14. Int Heart J 2020; 61:806-814. [PMID: 32728001 DOI: 10.1536/ihj.19-496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study aimed to explore the function of miR-24 in hypoxia/reoxygenation (H/R) -induced cardiomyocyte injury.We constructed a cardiomyocyte model of H/R using the primary cardiomyocytes isolated from Sprague-Dawley rats. To explore the role of miR-24, cells were transfected with a miR-24 mimic or miR-24 inhibitor. The RNA expression levels of miR-24 and Mapk14 were determined using qRT-PCR. The proliferation and apoptosis of cells were determined using a CCK8 assay and a flow cytometer. The TargetScan website was used to predict the targets of miR-24. A dual-luciferase reporter gene assay was conducted to verify whether Mapk14 is indeed a target of miR-24. A Western blot was applied for protein detection.H/R exposure decreased the expression of miR-24 in rat cardiomyocytes. Transfection of the miR-24 mimic into cardiomyocytes reduced H/R-induced injury as evidenced by an increase in proliferation and a decrease in the apoptotic rate. By contrast, transfection of the miR-24 inhibitor aggravated H/R-induced injury. The expression of Bcl-2 was increased while the levels of Bax and Active-caspase 3 were reduced in the H/R+miR-24 mimic group compared to those in the H/R group. H/R+miR-24 inhibitor group showed the opposite results. Mapk14 was identified as a target of miR-24. The mRNA level of Mapk14 and its protein (p38 MAPK) level were negatively affected by miR-24. Furthermore, we discovered that depletion of Mapk14 reduced the promoting effect of the miR-24 inhibitor on cell apoptosis.Overall, our results illustrated that miR-24 could attenuate H/R-induced injury partly by regulating Mapk14.
Collapse
Affiliation(s)
- Shao-Qiang Qin
- Department of Cardiology, First Affiliated Hospital of Hebei North University
| | - Zhan-Shuai Zhang
- Department of Cardiology, First Affiliated Hospital of Hebei North University
| | - Xiao-Yuan Wang
- Department of Cardiology, First Affiliated Hospital of Hebei North University
| | - Jin-Zheng Shi
- Department of Cardiology, First Affiliated Hospital of Hebei North University
| | | |
Collapse
|
7
|
Xue Y, Xu T, Jiang W. Dexmedetomidine protects PC12 cells from ropivacaine injury through miR-381/LRRC4 /SDF-1/CXCR4 signaling pathway. Regen Ther 2020; 14:322-329. [PMID: 32467829 PMCID: PMC7243045 DOI: 10.1016/j.reth.2020.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/16/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction Ropivacaine has been regularly used because of its good anesthetic and analgesic effects, but it may exert neurotoxic effects on neurocyte. Dexmedetomidine has presented special advantages in the fields of neuroprotection, and it also could improve peripheral nerve block combining with ropivacaine. However, if dexmedetomidine could repair neurocyte injury induced by ropivacaine, and the specific mechanism remain unclear. Methods Western blotting and qRT-PCR were applied for measuring expression of protein and mRNA, respectively. Flow cytometry was used for assessing apoptosis. Cell proliferation was detected using Cell Counting Kit-8 (CCK-8) and colony formation assays. Transwell assay was applied to measure the migration and invasion of cells. Dual luciferase reporter assay was applied for confirming the binding site between microRNA-381 (miR-381) and Leucine-rich repeat C4 protein (LRRC4). Results The viability of PC12 cells increased with raising the concentration of dexmedetomidine (0 μM, 10 μM, 50 μM, 100 μM). Dexmedetomidine reversed role of ropivacaine (0 mM, 0.1 mM, 0.5 mM, 1 mM) by upragulating the expression of miR-381 and suppressing the expression of LRRC4 in PC12 cells. miR-381 can directly interact with target gene LRRC4 and negatively regulate its expression. Dexmedetomidine promoted the proliferation, migration, and invasion and inhibited apoptosis of PC12 cells by suppressing LRRC4 via up-regulating the expressions of miR-381 and further activated SDF-1/CXCR4 signaling pathway. Conclusions Dexmedetomidine could protect PC12 cells from ropivacaine injury through miR-381/LRRC4/SDF-1/CXCR4 signaling pathway. This study may provide new therapeutic strategy targeting miR-381/LRRC4/SDF-1/CXCR4 signaling pathway about the prevention of ropivacaine induced neurocyte injury.
Collapse
Affiliation(s)
- Ying Xue
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, PR China
| | - Tao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, PR China
| | - Wei Jiang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, PR China
| |
Collapse
|
8
|
Yin W, Wang C, Peng Y, Yuan W, Zhang Z, Liu H, Xia Z, Ren C, Qian J. Dexmedetomidine alleviates H 2O 2-induced oxidative stress and cell necroptosis through activating of α2-adrenoceptor in H9C2 cells. Mol Biol Rep 2020; 47:3629-3639. [PMID: 32342432 DOI: 10.1007/s11033-020-05456-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/10/2020] [Indexed: 12/26/2022]
Abstract
Oxidative stress induced necroptosis is important in myocardial ischemia/reperfusion injury. Dexmedetomidine (Dex), an α2-adrenoceptor (α2-AR) agonist, has protective effect on oxidative stress induced cell apoptosis, but effects of Dex and Dex-mediated α2-AR activation on oxidant induced necroptosis was unclear. H9C2 cardiomyocytes were pre-treated with or without Dex and α2-AR antagonist yohimbine hydrochloride (YOH) before being exposed to H2O2 to induce oxidative cellular damage. Cell viability and lactate dehydrogenase (LDH) were detected by ELISA kits, protein expressions of Heme Oxygenase 1(HO-1), receptor interacting protein kinase 1 (RIPK1) and receptor interacting protein kinase 3 (RIPK3) were observed by WB, and TUNEL was used to detected cell apoptosis. H2O2 significantly decreased cell viability and increased LDH release and necroptotic and apoptotic cell deaths (all p < 0.05, H2O2 vs. Control). Dex preconditioning alleviated these injuries induced by H2O2. Dex preconditioning significantly increased expression of protein HO-1 and decreased expressions of proteins RIPK1 and RIPK3 induced by H2O2, while all these protective effects of Dex were reversed by YOH (all p < 0.05, Dex + H2O2 vs. H2O2; and YOH + Dex + H2O2 vs. Dex + H2O2). However, YOH did not prevent this protective effect of Dex against H2O2 induced apoptosis (YOH + Dex + H2O2 vs. Dex + H2O2, p > 0.05). These findings indicated that Dex attenuates H2O2 induced cardiomyocyte necroptotic and apoptotic cell death respectively dependently and independently of α2-AR activation.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
- Department of Anesthesiology, Sichuan Provincial Orthopedic Hospital, Chengdu, 610041, Sichuan, China
| | - Chunyan Wang
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Yue Peng
- Department of Anesthesiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518034, China
| | - Wenlin Yuan
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Zhongjun Zhang
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Hong Liu
- Department of Anaesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA, USA
| | - Zhengyuan Xia
- Department of Anaesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA, USA
| | - Congcai Ren
- Department of Anesthesiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518034, China.
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
9
|
Bunte S, Behmenburg F, Majewski N, Stroethoff M, Raupach A, Mathes A, Heinen A, Hollmann MW, Huhn R. Characteristics of Dexmedetomidine Postconditioning in the Field of Myocardial Ischemia-Reperfusion Injury. Anesth Analg 2020; 130:90-98. [PMID: 31633505 DOI: 10.1213/ane.0000000000004417] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Timing and onset of myocardial ischemia are mostly unpredictable. Therefore, postconditioning could be an effective cardioprotective intervention. Because ischemic postconditioning is an invasive and not practicable treatment, pharmacological postconditioning would be a more suitable alternative cardioprotective measure. For the α2-adrenoreceptor agonist, dexmedetomidine postconditioning has been shown. However, data on a concentration-dependent effect of dexmedetomidine are lacking. Furthermore, it is unclear whether the time point and/or duration of dexmedetomidine administration in the reperfusion period is of relevance. We set out to determine whether infarct size reduction by dexmedetomidine is concentration dependent and whether time point and/or duration of dexmedetomidine application has an impact on the effect size of cardio protection. METHODS Hearts of male Wistar rats were randomized and placed on a Langendorff system perfused with Krebs-Henseleit buffer at a constant pressure of 80 mm Hg. All hearts were subjected to 33 minutes of global ischemia and 60 minutes of reperfusion. In part I of the study, a concentration-response effect was determined by perfusing hearts with various concentrations of dexmedetomidine (0.3-100 nM) at the onset of reperfusion. Based on these results, part II of the study was conducted with 3 nM dexmedetomidine. Application of dexmedetomidine started directly at the onset of reperfusion (Dex60) and 15 minutes (Dex15), 30 minutes (Dex30), or 45 minutes (Dex45) after the start of reperfusion and lasted always until the end of the reperfusion period. Infarct size was determined by triphenyltetrazolium chloride staining. RESULTS In part I, infarct size in control (Con) hearts was 62% ± 4%. Three-nanometer dexmedetomidine was the lowest most effective cardioprotective concentration and reduced infarct size to 24% ± 7% (P < .0001 versus Con). Higher concentrations did not confer stronger protection. Infarct size in control hearts from part II was 66% ± 6%. Different starting times and/or durations of application resulted in similar infarct size reduction (all P < .0001 versus Con). CONCLUSIONS Postconditioning by dexmedetomidine is concentration dependent in ranges between 0.3 and 3 nM. Increased concentrations above 3 nM do not further enhance this cardioprotective effect. This cardioprotective effect is independent of time point and length of application in the reperfusion period.
Collapse
Affiliation(s)
- Sebastian Bunte
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Friederike Behmenburg
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Nicole Majewski
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Martin Stroethoff
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Annika Raupach
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Alexander Mathes
- Department of Anesthesiology, University Hospital Cologne, Cologne, Germany
| | - André Heinen
- Institute of Cardiovascular Physiology, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Markus W Hollmann
- Department of Anesthesiology, Amsterdam Universitair Medische Centra (UMC), University of Amsterdam, the Netherlands
| | - Ragnar Huhn
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
10
|
Abstract
Experimental models of cardiac disease play a key role in understanding the pathophysiology of the disease and developing new therapies. The features of the experimental models should reflect the clinical phenotype, which can have a wide spectrum of underlying mechanisms. We review characteristics of commonly used experimental models of cardiac physiology and pathophysiology in all translational steps including in vitro, small animal, and large animal models. Understanding their characteristics and relevance to clinical disease is the key for successful translation to effective therapies.
Collapse
|
11
|
Peng K, Chen WR, Xia F, Liu H, Meng XW, Zhang J, Liu HY, Xia ZY, Ji FH. Dexmedetomidine post-treatment attenuates cardiac ischaemia/reperfusion injury by inhibiting apoptosis through HIF-1α signalling. J Cell Mol Med 2019; 24:850-861. [PMID: 31680420 PMCID: PMC6933328 DOI: 10.1111/jcmm.14795] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/21/2019] [Accepted: 09/26/2019] [Indexed: 12/18/2022] Open
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.
Collapse
Affiliation(s)
- Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei-Rong Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Anesthesiology, Soochow University Affiliated Children's Hospital, Suzhou, China
| | - Fan Xia
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Yuan M, Meng XW, Ma J, Liu H, Song SY, Chen QC, Liu HY, Zhang J, Song N, Ji FH, Peng K. Dexmedetomidine protects H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation-induced intracellular calcium overload and apoptosis through regulating FKBP12.6/RyR2 signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3137-3149. [PMID: 31564830 PMCID: PMC6730549 DOI: 10.2147/dddt.s219533] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022]
Abstract
Purpose Intracellular calcium ([Ca2+]i) overload is a major cause of cell injury during myocardial ischemia/reperfusion (I/R). Dexmedetomidine (DEX) has been shown to exert anti-inflammatory and organ protective effects. This study aimed to investigate whether pretreatment with DEX could protect H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation (OGD/R) injury through regulating the Ca2+ signaling. Methods H9c2 cardiomyocytes were subjected to OGD for 12 h, followed by 3 h of reoxygenation. DEX was administered 1 h prior to OGD/R. Cell viability, lactate dehydrogenase (LDH) release, level of [Ca2+]i, cell apoptosis, and the expression of 12.6-kd FK506-binding protein/ryanodine receptor 2 (FKBP12.6/RyR2) and caspase-3 were assessed. Results Cells exposed to OGD/R had decreased cell viability, increased LDH release, elevated [Ca2+]i level and apoptosis rate, down-regulated expression of FKBP12.6, and up-regulated expression of phosphorylated-Ser2814-RyR2 and cleaved caspase-3. Pretreatment with DEX significantly blocked the above-mentioned changes, alleviating the OGD/R-induced injury in H9c2 cells. Moreover, knockdown of FKBP12.6 by small interfering RNA abolished the protective effects of DEX. Conclusion This study indicates that DEX pretreatment protects the cardiomyocytes against OGD/R-induced injury by inhibiting [Ca2+]i overload and cell apoptosis via regulating the FKBP12.6/RyR2 signaling. DEX may be used for preventing cardiac I/R injury in the clinical settings.
Collapse
Affiliation(s)
- Mei Yuan
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China.,Department of Anesthesiology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, People's Republic of China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Jiao Ma
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA 95817, USA
| | - Shao-Yong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Qing-Cai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Nan Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| |
Collapse
|
13
|
Chen J, Jiang Z, Zhou X, Sun X, Cao J, Liu Y, Wang X. Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation. Cardiovasc Drugs Ther 2019; 33:45-54. [PMID: 30675709 DOI: 10.1007/s10557-019-06857-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a serious threat to the health of people around the world. Recent evidence has indicated that high-mobility group box-1 (HMGB1) is involved in I/R-induced inflammation, and inflammation can cause necroptosis of cells. Interestingly, dexmedetomidine (DEX) has anti-inflammatory properties. Therefore, we speculated that DEX preconditioning may suppress H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. We found that hypoxia/reoxygenation (H/R) significantly increased cellular damage, as measured by cell viability (100 ± 3.26% vs. 53.33 ± 3.29, p < 0.01), CK-MB (1 vs. 3.25 ± 0.26, p < 0.01), cTnI (1 vs. 2.69 ± 0.31, p < 0.01), inflammation as indicated by TNF-α (1 ± 0.09 vs. 2.57 ± 0.12, p < 0.01), IL-1β (1 ± 0.33 vs. 3.87 ± 0.41, p < 0.01) and IL-6 (1 ± 0.36 vs. 3.60 ± 0.45, p < 0.01), and necroptosis, which were accompanied by significantly increased protein levels of HMGB1. These changes [cellular damage as measured by cell viability (53.33 ± 3.29% vs. 67.59 ± 2.69%, p < 0.01), CK-MB (3.25 ± 0.26 vs. 2.27 ± 0.22, p < 0.01), cTnI (2.69 ± 0.31 vs. 1.90 ± 0.25, p < 0.01), inflammation as indicated by TNF-α (2.57 ± 0.12 vs. 1.75 ± 0.15, p < 0.01), IL-1β (3.87 ± 0.41 vs. 2.09 ± 0.36, p < 0.01) and IL-6 (3.60 ± 0.45 vs. 2.21 ± 0.39, p < 0.01), and necroptosis proteins] were inhibited by DEX preconditioning. We also found that silencing expression of HMGB1 reinforced the protective effects of DEX preconditioning and overexpression of HMGB1 counteracted the protective effects of DEX preconditioning. Thus, we concluded that DEX preconditioning inhibits H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes.
Collapse
Affiliation(s)
- Jingyi Chen
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - Zhenzhen Jiang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xing Zhou
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xingxing Sun
- Department of Ultrasonography Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Jianwei Cao
- Department of Orthopedics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Yongpan Liu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xianyu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| |
Collapse
|
14
|
Peng K, Ji FH, Liu HY, Zhang J, Chen QC, Jiang YH. Effects of Perioperative Dexmedetomidine on Postoperative Mortality and Morbidity: A Systematic Review and Meta-analysis. Clin Ther 2019; 41:138-154.e4. [DOI: 10.1016/j.clinthera.2018.10.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/08/2018] [Accepted: 10/31/2018] [Indexed: 01/11/2023]
|
15
|
Weng X, Zhang X, Lu X, Wu J, Li S. Reduced mitochondrial response sensitivity is involved in the anti‑apoptotic effect of dexmedetomidine pretreatment in cardiomyocytes. Int J Mol Med 2018; 41:2328-2338. [PMID: 29328437 DOI: 10.3892/ijmm.2018.3384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 01/10/2018] [Indexed: 11/05/2022] Open
Abstract
Dexmedetomidine is a commonly used α2-adreno-ceptor agonist, which affects various organs, including providing beneficial effects on the heart. However, the mechanism underlying the cardiac benefit remains to be fully elucidated. In the present study, it was demonstrated that dexmedetomidine pretreatment on primary cultured rat cardiomyocytes protected against reactive oxygen species (ROS)‑induced apoptosis. In terms of the potential mechanism, it was demonstrated that dexmedetomidine inhibited mitochondrial biogenesis and mitochondrial respiratory complexes, but with increased coupling efficiency. However, dexmedetomidine upregulated mitochondrial membrane potential (Δψm) and resisted against the loss of Δψm induced by carbonilcyanide p‑triflouromethoxyphenylhydrazone. Due to the importance of mitochondria affecting ROS, the present study investigated the dexmedetomidine‑suppressed mitochondrial response to H2O2 stimulation, which was explained by suppressed ROS levels and the suppression of the increased oxygen consumption rate. Results demonstrated for the first time, to the best of our knowledge, a novel protective mechanism for dexmedetomidine on cardiomyocytes through the attenuated response of mitochondria towards H2O2, which had a protective effect against ROS‑induced apoptosis.
Collapse
Affiliation(s)
- Xiaojian Weng
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xiaodan Zhang
- Department of Intensive Care Unit, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xiaofei Lu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Jin Wu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Shitong Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
16
|
Abstract
Cardiovascular disease is one of the most common causes of deaths in clinics. Experimental models of cardiovascular diseases are essential to understand disease mechanism, to provide accurate diagnoses, and to develop new therapies. Large numbers of experimental models have been proposed and replicated by many laboratories in the past. Models with significant advantages are chosen and became more popular. Particularly, feasibility, reproducibility, and human disease resemblance are the common key factors for frequently used cardiovascular disease models. In this chapter, we provide a brief overview of these experimental models used for in vitro, in vivo, and in silico studies of cardiovascular diseases.
Collapse
Affiliation(s)
- Jae Gyun Oh
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
Gao JM, Meng XW, Zhang J, Chen WR, Xia F, Peng K, Ji FH. Dexmedetomidine Protects Cardiomyocytes against Hypoxia/Reoxygenation Injury by Suppressing TLR4-MyD88-NF- κB Signaling. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1674613. [PMID: 29359143 PMCID: PMC5735617 DOI: 10.1155/2017/1674613] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/30/2017] [Accepted: 11/06/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE We previously reported that dexmedetomidine (DEX) offers cardioprotection against ischemia/reperfusion injury in rats. Here, we evaluated the role of toll-like receptors 4- (TLR4-) myeloid differentiation primary response 88- (MyD88-) nuclear factor-kappa B (NF-κB) signaling in DEX-mediated protection of cardiomyocytes using in vitro models of hypoxia/reoxygenation (H/R). METHODS The experiments were carried out in H9C2 cells and in primary neonatal rat cardiomyocytes. Cells pretreated with vehicle or DEX were exposed to hypoxia for 1 h followed by reoxygenation for 12 h. We analyzed cell viability and lactate dehydrogenase (LDH) activity and measured tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β mRNA levels, TLR4, MyD88, and nuclear NF-κB p65 protein expression and NF-κB p65 nuclear localization. TLR4 knock-down by TLR4 siRNA transfection and overexpression by TLR4 DNA transfection were used to further confirm our findings. RESULTS DEX protected against H/R-induced cell damage and inflammation, as evidenced by increased cell survival rates, decreased LDH activity, and decreased TNF-α, IL-6, and IL-1β mRNA levels, as well as TLR4 and NF-κB protein expression. TLR4 knock-down partially prevented cell damage following H/R injury, while overexpression of TLR4 abolished the DEX-mediated protective effects. CONCLUSIONS DEX pretreatment protects rat cardiomyocytes against H/R injury. This effect is partly mediated by TLR4 suppression via TLR4-MyD88-NF-κB signaling.
Collapse
Affiliation(s)
- Jin-meng Gao
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou 215003, China
| | - Xiao-wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wei-rong Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Fan Xia
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Fu-hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|