1
|
Jiang L, Xiong W, Yang Y, Qian J. Insight into Cardioprotective Effects and Mechanisms of Dexmedetomidine. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07579-9. [PMID: 38869744 DOI: 10.1007/s10557-024-07579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Cardiovascular disease remains the leading cause of death worldwide. Dexmedetomidine is a highly selective α2 adrenergic receptor agonist with sedative, analgesic, anxiolytic, and sympatholytic properties, and several studies have shown its possible protective effects in cardiac injury. The aim of this review is to further elucidate the underlying cardioprotective mechanisms of dexmedetomidine, thus suggesting its potential in the clinical management of cardiac injury. RESULTS AND CONCLUSION Our review summarizes the findings related to the involvement of dexmedetomidine in cardiac injury and discusses the results in the light of different mechanisms. We found that numerous mechanisms may contribute to the cardioprotective effects of dexmedetomidine, including the regulation of programmed cell death, autophagy and fibrosis, alleviation of inflammatory response, endothelial dysfunction and microcirculatory derangements, improvement of mitochondrial dysregulation, hemodynamics, and arrhythmias. Dexmedetomidine may play a promising and beneficial role in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Leyu Jiang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Xiong
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Jia Y, Huo X, Wu L, Zhang H, Xu W, Leng H. circSNTB2 and CUL4A Induces Dysfunction of Nucleus Pulposus Cells by Competitively Binding miR-665. Biochem Genet 2024; 62:968-986. [PMID: 37507642 DOI: 10.1007/s10528-023-10465-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Circular RNA (circRNA) plays important roles in lumbar degenerative diseases. This study aimed to investigate the role of circSNTB2 in regulating the development of lumbar disc herniation (LDH) in vitro and in vivo. The abnormally expressed circSNTB2 in intervertebral disc degeneration (IDD) through bioinformatics analysis was identified, and verified in nucleus pulposus (NP) tissues of patients with LDH. NP cells were treated with TNF-α to mimic the LDH microenvironment. RT-qPCR was applied to determine levels of mRNA and microRNA (miRNA) in clinical samples and cells. We performed CCK-8, EdU, TUNEL and flow cytometric apoptosis assays to evaluate the proliferation and apoptosis of NP cells. The predicted the miRNAs and downstream target genes were verified with the help of luciferase reporter gene and RNA pull-down experiments. Finally, we established an LDH rat model to further verify the role of circSNTB2 in vivo. circSNTB2 was significantly up-regulated in the NP tissues of LDH group and TNF-α -treated NP cells. miR-665 binds to circSNTB2 and cullin 4A (CUL4A) is the downstream target gene of miR-665. Knockdown of circSNTB2 promoted NP cells proliferation and inhibited apoptosis, which was reversed by down-regulation of miR-665. In addition, up-regulated CUL4A reversed the effects of over-expressed miR-665 on proliferation and apoptosis of NP cells. Meanwhile, results of in vivo experiments demonstrated that knocking down circSNTB2 alleviated LDH-induced thermo-mechanical pain and NP injury. In summary, circSNTB2 regulates the proliferation and apoptosis of NP by mediating miR-665 regulation of CUL4A, which provides a reliable idea for targeted therapy of LDH.
Collapse
Affiliation(s)
- Yiming Jia
- Department of Spine Surgery, Chifeng Municipal Hospital, Hongshan District, No. 1, Zhaowuda Road, Chifeng City, 024000, Inner Mongolia, China
| | - Xin Huo
- Department of Orthopaedics, Chifeng Second Hospital, Hongshan District, No.1, Changqing Street, Chifeng City, 024000, Inner Mongolia, China
| | - Leilei Wu
- Department of Spine Surgery, Chifeng Municipal Hospital, Hongshan District, No. 1, Zhaowuda Road, Chifeng City, 024000, Inner Mongolia, China
| | - Haibo Zhang
- Department of Spine Surgery, Chifeng Municipal Hospital, Hongshan District, No. 1, Zhaowuda Road, Chifeng City, 024000, Inner Mongolia, China
| | - Wenda Xu
- Department of Spine Surgery, Chifeng Municipal Hospital, Hongshan District, No. 1, Zhaowuda Road, Chifeng City, 024000, Inner Mongolia, China
| | - Hui Leng
- Department of Spine Surgery, Chifeng Municipal Hospital, Hongshan District, No. 1, Zhaowuda Road, Chifeng City, 024000, Inner Mongolia, China.
| |
Collapse
|
3
|
Bo Y, Zhao X, Li L. Cardiotoxic effects of common and emerging drugs: role of cannabinoid receptors. Clin Sci (Lond) 2024; 138:413-434. [PMID: 38505994 DOI: 10.1042/cs20231156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Drug-induced cardiotoxicity has become one of the most common and detrimental health concerns, which causes significant loss to public health and drug resources. Cannabinoid receptors (CBRs) have recently achieved great attention for their vital roles in the regulation of heart health and disease, with mounting evidence linking CBRs with the pathogenesis and progression of drug-induced cardiotoxicity. This review aims to summarize fundamental characteristics of two well-documented CBRs (CB1R and CB2R) from aspects of molecular structure, signaling and their functions in cardiovascular physiology and pathophysiology. Moreover, we describe the roles of CB1R and CB2R in the occurrence of cardiotoxicity induced by common drugs such as antipsychotics, anti-cancer drugs, marijuana, and some emerging synthetic cannabinoids. We highlight the 'yin-yang' relationship between CB1R and CB2R in drug-induced cardiotoxicity and propose future perspectives for CBR-based translational medicine toward cardiotoxicity curation and clinical monitoring.
Collapse
Affiliation(s)
- Yiming Bo
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xin Zhao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liliang Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Takahashi K, Yoshikawa Y, Kanda M, Hirata N, Yamakage M. Dexmedetomidine as a cardioprotective drug: a narrative review. J Anesth 2023; 37:961-970. [PMID: 37750978 DOI: 10.1007/s00540-023-03261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
Dexmedetomidine (DEX), a highly selective alpha2-adrenoceptors agonist, is not only a sedative drug used during mechanical ventilation in the intensive care unit but also a cardio-protective drug against ischemia-reperfusion injury (IRI). Numerous preclinical in vivo and ex vivo studies, mostly evaluating the effect of DEX pretreatment in healthy rodents, have shown the efficacy of DEX in protecting the hearts from IRI. However, whether DEX can maintain its cardio-protective effect in hearts with comorbidities such as diabetes has not been fully elucidated. Multiple clinical trials have reported promising results, showing that pretreatment with DEX can attenuate cardiac damage in patients undergoing cardiac surgery. However, evidence of the post-treatment effects of DEX in clinical practice remains limited. In this narrative review, we summarize the previously reported evidence of DEX-induced cardio-protection against IRI and clarify the condition of the hearts and the timing of DEX administration that has not been tested. With further investigations evaluating these knowledge gaps, the use of DEX as a cardio-protective drug could be further facilitated in the management of patients undergoing cardiac surgery and might be considered in a broader area of clinical settings beyond cardiac surgery, including patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Kanako Takahashi
- Department of Anesthesiology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-Ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Yusuke Yoshikawa
- Department of Anesthesiology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-Ku, Sapporo, Hokkaido, 060-8556, Japan.
| | - Masatoshi Kanda
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naoyuki Hirata
- Department of Anesthesiology, Kumamoto University, Kumamoto, Japan
| | - Michiaki Yamakage
- Department of Anesthesiology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-Ku, Sapporo, Hokkaido, 060-8556, Japan
| |
Collapse
|
5
|
Yang K, Ma Y, Xie C, He L, Zhao H, Dai Z, Wang X. Dexmedetomidine combined with propofol attenuates myocardial ischemia/reperfusion injury by activating the AMPK signaling pathway. Heliyon 2023; 9:e22054. [PMID: 38034796 PMCID: PMC10682120 DOI: 10.1016/j.heliyon.2023.e22054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Objective Myocardial ischemia/reperfusion (MI/R) injury is a major cause of cardiac tissue damage, with high disability and death rates. Although both dexmedetomidine (Dex) and propofol (PPF) have been indicated to alleviate MI/R injury in rat models, the effects of the combined use of these two drugs remain unclear. This study aimed to investigate the combined effects of Dex and PPF against MI/R injury and related mechanisms. Methods A rat model of MI/R injury was established and used to explore the combined effects of Dex and PPF on MI/R injury. Hematoxylin-eosin (HE) and Masson staining were used for histopathological evaluation. 2,3,5-triphenyltetrazolium chloride (TTC), echocardiography, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining were used to determine myocardial infarction size, cardiac function, and apoptosis, respectively. Enzyme-linked immunosorbent assay (ELISA) was performed to assess myocardial function and oxidative stress (OS). Autophagy was observed through transmission electron microscopy. Moreover, western blotting was conducted to detect autophagy markers and the AMPK pathway. Results The combination of Dex and PPF alleviated histopathological injury, reduced myocardial infarction, and rescued cardiac dysfunction in MI/R rats. Furthermore, Dex combined with PPF decreased the levels of MDA and ROS and increased the SOD level in MI/R rats. Besides, Dex combined with PPF inhibited myocardial apoptosis in MI/R rats. After combined treatment with Dex and PPF, the number of autophagosomes, expression levels of Beclin-1 and LC3II/LC3I were elevated, while the expression levels of p62 were reduced in MI/R rats. The combined use of Dex and PPF activated the AMPK pathway in MI/R rats. Compound C (an AMPK inhibitor) could abolish the combined effects of Dex and PPF on alleviating myocardial injury and enhancing autophagy in MI/R rats. Conclusion The combination of Dex and PPF attenuated MI/R injury in rats, which may be associated with the activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
| | | | - Chunmei Xie
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Lixian He
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Haoxing Zhao
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Zheng Dai
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Xiaoqi Wang
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| |
Collapse
|
6
|
Wang L, Liu J, Wang Z, Qian X, Zhao Y, Wang Q, Dai N, Xie Y, Zeng W, Yang W, Bai X, Yang Y, Qian J. Dexmedetomidine abates myocardial ischemia reperfusion injury through inhibition of pyroptosis via regulation of miR-665/MEF2D/Nrf2 axis. Biomed Pharmacother 2023; 165:115255. [PMID: 37549462 DOI: 10.1016/j.biopha.2023.115255] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023] Open
Abstract
The current study intended to delve into the mechanisms of dexmedetomidine (Dex) in regulating myocardial pyroptosis against myocardial ischemia/reperfusion injury (MIRI). The rat MIRI models were induced by ligation/release of the coronary artery in vivo and Langendorff perfusion ex vivo. Hemodynamic parameters, infarction sizes, and histopathological changes were assessed to understand the effects of Dex on MIRI. We explored the mechanisms through functional experiments on an H9c2 cell hypoxia/reoxygenation (H/R) model. Cell viability and apoptosis were evaluated using cell counting kit 8 (CCK-8) and AV/PI dual staining respectively. The expressions of miR-665 and MEF2D mRNA were detected by qRT-PCR. Western blot was employed to determine the expression levels of pyroptosis- and signaling pathway- related proteins. The interplays between miR-665 and MEF2D were validated by Dual-luciferase reporter assays. Our findings indicated that Dex preconditioning dramatically attenuated hemodynamic derangements, infarct size, and histopathological damage in rats undergoing MIRI. Dex markedly augmented cell viability, while suppressing cell apoptosis and expressions of NLRP3, cleaved-caspase-1, ASC, GSDMD, IL-1β, and IL-18 in H9c2 cells subjected to H/R injury. MiR-665 was significantly upregulated, MEF2D and Nrf2 downregulated following H/R, whereas Dex preconditioning reversed these changes. MEF2D was validated to be a target gene of miR-665. Overexpression of miR-665 decreased the expression of MEF2D and blunted the protective effects of Dex in H9c2 cells. Moreover, the functional rescue experiment further verified that Dex regulated MEF2D/Nrf2 pathway via miR-665. In conclusion, Dex mitigates MIRI through inhibiting pyroptosis via regulating miR-665/MEF2D/Nrf2 axis.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jin Liu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhuoran Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xi Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yu Zhao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Quan Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Na Dai
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuhan Xie
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Weijun Zeng
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wei Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiangfeng Bai
- Department of Cardiac Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
7
|
Li X, Zhang C, Tao H, Yang X, Qian X, Zhou J, Zhang L, Shi Y, Gu C, Geng D, Hao Y. Dexmedetomidine alleviates osteoarthritis inflammation and pain through the CB 2 pathway in rats. Int Immunopharmacol 2023; 119:110134. [PMID: 37044038 DOI: 10.1016/j.intimp.2023.110134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023]
Abstract
As a common joint disease, osteoarthritis (OA) is often associated with chronic pain. Synovial inflammation is correlated with OA progression and pain. Synovial inflammation can produce a series of destructive substances, such as inflammatory factors and pain mediators, which aggravate cartilage injury and further accelerate the progression of OA. Although many studies investigated the effects of synovial inflammation on the onset and progression of OA, there are limited reports regarding slowing the progression of OA and relieving pain by modulating synovial inflammation. Therefore, there is an urgent need to search for safe and effective drugs to alleviate synovial inflammation. Dexmedetomidine, a selective α2 agonist, has been shown to have anti-inflammatory and analgesic effects. However, its role and mechanism in OA remain unclear. Here, the effects and mechanisms of dexmedetomidine in OA synovial inflammation were investigated both in vivo and in vitro. We observed that dexmedetomidine stunted LPS-induced migration and invasion of FLSs and the expression of inflammatory factors by upregulating cannabinoid receptor type 2 (CB2) expression. Surprisingly, the application of AM630 (CB2 antagonist) reversed this therapeutic effect. The results of the animal experiments showed that dexmedetomidine reduced synovial inflammation and increased the pain threshold in an OA rat model. These preliminary results imply that dexmedetomidine may be an effective compound for OA treatment.
Collapse
Affiliation(s)
- Xueyan Li
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Chun Zhang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 999, Xiwang Road, Shanghai, China
| | - Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu 226000, China
| | - Xiaobo Qian
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu 226000, China
| | - Liyuan Zhang
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Yi Shi
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Chengyong Gu
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu 226000, China.
| |
Collapse
|
8
|
Toghiani R, Abolmaali SS, Najafi H, Tamaddon AM. Bioengineering exosomes for treatment of organ ischemia-reperfusion injury. Life Sci 2022; 302:120654. [PMID: 35597547 DOI: 10.1016/j.lfs.2022.120654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a leading cause of death worldwide. It arises from blood reflowing after tissue hypoxia induced by ischemia that causes severe damages due to the accumulation of reactive oxygen species and the activation of inflammatory responses. Exosomes are the smallest members of the extracellular vesicles' family, which originate from nearly all eukaryotic cells. Exosomes have a great potential in the treatment of I/R injury either in native or modified forms. Native exosomes are secreted by different cell types, such as stem cells, and contain components such as specific miRNA molecules with tissue protective properties. On the other hand, exosome bioengineering has recently received increased attention in context of current advances in the purification, manipulation, biological characterization, and pharmacological applications. There are various pre-isolation and post-isolation manipulation approaches that can be utilized to increase the circulation half-life of exosomes or the availability of their bioactive cargos in the target site. In this review, the various therapeutic actions of native exosomes in different I/R injury will be discussed first. Exosome bioengineering approaches will then be explained, including pre- and post-isolation manipulation methods, applicability for delivery of bioactive agents to injured tissue, clinical translation issues, and future perspectives.
Collapse
Affiliation(s)
- Reyhaneh Toghiani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Haniyeh Najafi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Liu Q, Wu J, Lai S, Li G. MiR-665 Participates in the Protective Effect of Dexmedetomidine in Ischemic Stroke by ROCK2/NF-κB Axis. Neurochem Res 2022; 47:2064-2075. [PMID: 35538293 DOI: 10.1007/s11064-022-03597-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/29/2022]
Abstract
Ischemic stroke is a grievous intimidation to the healthiness of sufferers. Previous studies have reported that dexmedetomidine (DEX) has a protective effect on a variety of organs. This paper aimed to explore the regulatory mechanism of DEX in ischemic stroke through miR-665/ROCK2 axis. The mice model of ischemic stroke was constructed by middle cerebral artery occlusion (MCAO). The cell model of ischemic stroke was constructed by oxygen-glucose deprivation (OGD). Cell viability and apoptosis were assessed by CCK-8 assay and flow cytometry. The expression of cytokines was detected by ELISA. Lactate dehydrogenase (LDH) concentration was evaluated by LDH kit. The cerebral infarct volume of MCAO mice was detected by TTC staining, and the apoptosis of brain cells was detected by TUNEL staining. The target relationship between ROCK2 and miR-665 was analyzed by dual-luciferase reporter assay. DEX contributed cell viability from 42 to 66% (1 μM) and restrained cell apoptosis from 26 to 18% in HT22 cells treated with OGD (P < 0.01). Meanwhile, DEX decreased the expression of cytokines and LDH concentration from 184 to 126% (P < 0.001). Moreover, the expression of miR-665 enhanced 2.9 times (P < 0.05) and the expression of ROCK2 (P < 0.05) and NF-κB p65 (P < 0.01) reduced 1.8 times and 2.2 times after DEX treatment in OGD induced HT22. And miR-665 knockdown attenuated the effect of DEX on inflammation damage (the levels of TNF-α, IL-1β and IL-6 increased 1.36 times, 1.31 times, 1.43 time, respectively, and IL-10 decreased 1.68 times) and apoptosis from 17 to 25% (P < 0.01). MiR-665 directly targeted ROCK2 and regulated ROCK2 and NF-κB p65 expression (P < 0.01). Furthermore, ROCK2 overexpression inhibited the protective effect of DEX in HT22 induced by OGD (P < 0.001), while miR-665 overexpression reversed the regulatory of ROCK2 (P < 0.01). In vivo, DEX decreased cerebral infarction volume and inhibited apoptosis of brain cell (P < 0.001). DEX has a protective effect in ischemic stroke by promoting miR-665 expression to downregulate ROCK2/NF-κB axis, suggesting DEX has a beneficial effect on ischemic stroke and miR-665 is a conceivable target for the therapeutics and diagnosis of ischemic stroke.
Collapse
Affiliation(s)
- QiongHua Liu
- Department of Anesthesiology, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330000, Jiangxi, People's Republic of China
| | - JianE Wu
- Department of Anesthesiology, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330000, Jiangxi, People's Republic of China
| | - ShangShu Lai
- Department of Anesthesiology, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330000, Jiangxi, People's Republic of China
| | - Gan Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Zhengjie, Donghu District, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
10
|
Circ_0136474 contributes to the IL-1β-induced chondrocyte injury by binding to miR-665 to induce the FGFR1 upregulation. Transpl Immunol 2022:101615. [DOI: 10.1016/j.trim.2022.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/02/2022] [Indexed: 11/18/2022]
|
11
|
Shi J, Zhao G, Liu P, Bai Y, Chen Y. Effects of Dexmedetomidine Combined with Intravenous Anesthesia on Oxidative Stress Index, Postoperative Sleep Quality, and Brain Function in HICH Patients. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:5463986. [PMID: 35345654 PMCID: PMC8957430 DOI: 10.1155/2022/5463986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 11/17/2022]
Abstract
To investigate the effects of dexmedetomidine combined with intravenous anesthesia on oxidative emergency indicators, postoperative sleep quality, and brain function in patients with hypertensive cerebral hemorrhage (HICH), a total of 285 HICH patients admitted to our hospital from February 2020 to February 2021 were selected. The combined anesthesia group (n = 142) and the control group (n = 143) were established by the random number table method. The control group received conventional intravenous anesthesia, and the combined anesthesia group received dexmedetomidine combined intravenous anesthesia. Two groups of patients before and after operation was observed vital signs, oxidative stress index difference, comparing each time, the change of the two groups of brain function index, adverse reactions occurred between observation group, and the postoperative period of Pittsburgh Sleep Quality Index Scale (PSQI) score as a result, the Pearson correlation coefficient analysis of oxidative stress level and the correlation of HICH patients sleep quality. After operation, the mean arterial pressure (MAP) and heart rate (HR) of patients in both groups decreased significantly. The MAP level in the combined anesthesia group significantly increased compared to the control group, and the HR level decreased significantly than the control group (all P < 0.05). The levels of TNF-α, IL-6 and MDA in both groups increased significantly on day 7 after operation compared with before operation, but the indexes in the combined anesthesia group significantly decreased compared with the control group (P < 0.05). The level of superoxide dismutase (SOD) in both groups significantly decreased compared to that before operation, and the index value in combined anesthesia group significantly increased compared to that in the control group (P < 0.05). After surgery, the levels of central nerve specific protein (S100-β) and neuron specific enolase (NSE) in 2 groups increased with time, and the indexes in the combined anesthesia group significantly decreased compared to the control group (all P < 0.05). The incidence of adr in combined anesthesia group decreased significantly than that in control group (P < 0.05). After surgery, PSQI scores of the two groups showed a downward trend with time extension, and scores of the combined anesthesia group decreased significantly than those of the control group at 24 h, 48 h and 7 d after surgery (all P < 0.05). Pearson's correlation coefficient was used to analyze that TNF-α, IL-6, and MDA levels were positively correlated with PSQI score, while SOD level was negatively correlated with PSQI score (all P < 0.05). Dexmedetomidine combined with intravenous anesthesia can significantly improve the vital signs and oxidative stress response of HICH patients, effectively reduce the risk of adverse reactions, have little impact on the brain function of patients, and can improve the postoperative sleep quality of patients. This operation is worthy of clinical application. In addition, this study further analyzed the influence mechanism of postoperative sleep quality in patients with HICH and showed that TNF-α, IL-6, MDA, and SOD were all correlated with sleep quality in patients with HICH, suggesting that follow-up detection of these indicators has positive significance in improving the prognosis of patients.
Collapse
Affiliation(s)
- Jiong Shi
- Anesthesiology Department, Handan Central Hospital, Handan 056001, China
| | - Guangping Zhao
- Anesthesiology Department, Handan Central Hospital, Handan 056001, China
| | - Panpan Liu
- Anesthesiology Department, Handan Central Hospital, Handan 056001, China
| | - Yuwei Bai
- Anesthesiology Department, Handan Central Hospital, Handan 056001, China
| | - Yongxue Chen
- Anesthesiology Department, Handan Central Hospital, Handan 056001, China
| |
Collapse
|
12
|
Yu Y, Tian T, Tan S, Wu P, Guo Y, Li M, Huang M. MicroRNA-665-3p exacerbates nonalcoholic fatty liver disease in mice. Bioengineered 2022; 13:2927-2942. [PMID: 35038955 PMCID: PMC8973643 DOI: 10.1080/21655979.2021.2017698] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Oxidative stress and chronic inflammation are major culprits of nonalcoholic fatty liver disease (NAFLD). MicroRNA-665-3p (miR-665-3p) is implicated in regulating inflammation and oxidative stress; however, its role and molecular basis in NAFLD remain elusive. Herein, we measured a significant upregulation of miR-665-3p level in the liver and primary hepatocytes upon high fat diet (HFD) or 0.5 mmol/L palmitic acid plus 1.0 mmol/L oleic acid stimulation, and the elevated miR-665-3p expression aggravated oxidative stress, inflammation and NAFLD progression in mice. In contrast, miR-665-3p inhibition by the miR-665-3p antagomir significantly prevented HFD-induced oxidative stress, inflammation and hepatic dysfunction in vivo. Manipulation of miR-665-3p in primary hepatocytes also caused similar phenotypic alterations in vitro. Mechanistically, we demonstrated that miR-665-3p directly bound to the 3'-untranslated region of fibronectin type III domain-containing 5 (FNDC5) to downregulate its expression and inactivated the downstream AMP-activated protein kinase alpha (AMPKα) pathway, thereby facilitating oxidative stress, inflammation and NAFLD progression. Our findings identify miR-665-3p as an endogenous positive regulator of NAFLD via inactivating FNDC5/AMPKα pathway, and inhibiting miR-665-3p may provide novel therapeutic strategies to treat NAFLD.
Collapse
Affiliation(s)
- Yuanjie Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tian Tian
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiyun Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pengbo Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yitian Guo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengjun Huang
- Department of Nutrition, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Wang H, Xia Y, Sun Z. Dexmedetomidine: A potential therapy for myocardial ischemia/reperfusion injury. Int J Cardiol 2021; 335:104. [PMID: 33930513 DOI: 10.1016/j.ijcard.2021.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Hongkun Wang
- Department of Cardiology, The Second People's Hospital of Liaocheng, Liaocheng 252000, PR China
| | - Ying Xia
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, PR China
| | - Zhiqiang Sun
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, PR China.
| |
Collapse
|
14
|
Wang W, Zheng H. Myocardial Infarction: The Protective Role of MiRNAs in Myocardium Pathology. Front Cardiovasc Med 2021; 8:631817. [PMID: 33748196 PMCID: PMC7973051 DOI: 10.3389/fcvm.2021.631817] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases have been regarded as the leading cause of death around the world, with myocardial infarction (MI) being the most severe form. MI leads to myocardial apoptosis, cardiomyocyte fibrosis, and cardiomyocyte hypertrophy, ultimately leading to heart failure, and death. Micro RNAs (miRNAs) participate in the genesis and progression of myocardial pathology after MI by playing an important regulatory role. This review aims to summarize all available knowledge on the role of miRNAs in the myocardial pathological process after MI to uncover potential major target pathways. In addition, the main therapeutic methods and their latest progress are also reviewed. miRNAs can regulate the main signaling pathways as well as pathological processes. Thus, they have the potential to induce therapeutic effects. Hence, the combination of miRNAs with recently developed exosome nanocomplexes may represent the future direction of therapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Graduate School of Bengbu Medical College, Bengbu, China
| | - Hao Zheng
- Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
15
|
Roth S, Torregroza C, Feige K, Preckel B, Hollmann MW, Weber NC, Huhn R. Pharmacological Conditioning of the Heart: An Update on Experimental Developments and Clinical Implications. Int J Mol Sci 2021; 22:ijms22052519. [PMID: 33802308 PMCID: PMC7959135 DOI: 10.3390/ijms22052519] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of pharmacological conditioning is to protect the heart against myocardial ischemia-reperfusion (I/R) injury and its consequences. There is extensive literature that reports a multitude of different cardioprotective signaling molecules and mechanisms in diverse experimental protocols. Several pharmacological agents have been evaluated in terms of myocardial I/R injury. While results from experimental studies are immensely encouraging, translation into the clinical setting remains unsatisfactory. This narrative review wants to focus on two aspects: (1) give a comprehensive update on new developments of pharmacological conditioning in the experimental setting concentrating on recent literature of the last two years and (2) briefly summarize clinical evidence of these cardioprotective substances in the perioperative setting highlighting their clinical implications. By directly opposing each pharmacological agent regarding its recent experimental knowledge and most important available clinical data, a clear overview is given demonstrating the remaining gap between basic research and clinical practice. Finally, future perspectives are given on how we might overcome the limited translatability in the field of pharmacological conditioning.
Collapse
Affiliation(s)
- Sebastian Roth
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| | - Carolin Torregroza
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
- Correspondence:
| | - Katharina Feige
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| | - Benedikt Preckel
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Markus W. Hollmann
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Nina C. Weber
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Ragnar Huhn
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| |
Collapse
|
16
|
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
|
17
|
Chen T, Liang Q, Xu J, Zhang Y, Zhang Y, Mo L, Zhang L. MiR-665 Regulates Vascular Smooth Muscle Cell Senescence by Interacting With LncRNA GAS5/SDC1. Front Cell Dev Biol 2021; 9:700006. [PMID: 34386495 PMCID: PMC8353444 DOI: 10.3389/fcell.2021.700006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Vascular aging is considered a special risk factor for cardiovascular diseases, and vascular smooth muscle cells (VSMCs) play a major role in aging-related vascular remodeling and in the pathological process of atherosclerosis. Recent research has reported that long non-coding RNA/microRNA (lncRNA/miRNA) is a critical regulator of cellular senescence. However, the role and mechanism of lncRNA GAS5/miR-665 axis in VSMC senescence remain incompletely understood. Methods: Cellular senescence was evaluated using senescence-associated β-gal activity, the NAD+/NADH ratio, and by immunofluorescence staining of γH2AX immunofluorescence. Differentially expressed miRNAs (DEMs) were identified by miRNA microarray assays and subsequently validated by quantitative real-time PCR (qRT-PCR). A dual luciferase reporter assay was conducted to confirm the binding of lncRNA GAS5 and miR-665 as well as miR-665 and syndecan 1 (SDC1). Serum levels of miR-665, lncRNA GAS5, and SDC1 in 93 subjects were detected by qRT-PCR. The participants were subdivided into control, aging, and early vascular aging (EVA) groups, and their brachial-ankle pulse wave velocity (baPWV) was measured. Results: A total of 20 overlapping DEMs were identified in young and old VSMCs via microarray analysis. MiR-665 showed a significant alteration and, therefore, was selected for further analysis. Upregulation of miR-665 was found in aging VSMCs, and downregulation of miR-665 caused an inhibition of VSMCs senescence. Subsequently, the dual luciferase reporter assay determined the binding site of miR-665 with the 3'-UTR of lncRNA GAS5 and SDC1. Increased expression of lncRNA GAS5 expression inhibited the miR-665 level and VSMC senescence. However, as shown in rescue experiment results, either miR-665 overexpression or SDC1 knockdown significantly reversed the effects of lncRNA GAS5 on VSMC senescence. Finally, compared with that of the control group, miR-665 was highly expressed in serum samples in the aging and EVA groups, especially in the EVA groups. On the contrary, serum levels of lncRNA GAS5 and SDC1 were lower in these two groups. Collectively, in the aging and EVA groups, miR-665 expression was negatively correlated with lncRNA GAS5 and SDC1 expression. Conclusion: miR-665 inhibition functions as a vital modulator of VSMC senescence by negatively regulating SDC1, which is achieved by lncRNA GAS5 that sponges miR-665. Our findings may provide a new treatment strategy for aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Tianbin Chen
- Functional Experiment Center, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qingyang Liang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jialin Xu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanan Zhang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Yi Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Liping Mo
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Li Zhang
| |
Collapse
|
18
|
Torregroza C, Raupach A, Feige K, Weber NC, Hollmann MW, Huhn R. Perioperative Cardioprotection: General Mechanisms and Pharmacological Approaches. Anesth Analg 2020; 131:1765-1780. [PMID: 33186163 DOI: 10.1213/ane.0000000000005243] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardioprotection encompasses a variety of strategies protecting the heart against myocardial injury that occurs during and after inadequate blood supply to the heart during myocardial infarction. While restoring reperfusion is crucial for salvaging myocardium from further damage, paradoxically, it itself accounts for additional cell death-a phenomenon named ischemia/reperfusion injury. Therefore, therapeutic strategies are necessary to render the heart protected against myocardial infarction. Ischemic pre- and postconditioning, by short periods of sublethal cardiac ischemia and reperfusion, are still the strongest mechanisms to achieve cardioprotection. However, it is highly impractical and far too invasive for clinical use. Fortunately, it can be mimicked pharmacologically, for example, by volatile anesthetics, noble gases, opioids, propofol, dexmedetomidine, and phosphodiesterase inhibitors. These substances are all routinely used in the clinical setting and seem promising candidates for successful translation of cardioprotection from experimental protocols to clinical trials. This review presents the fundamental mechanisms of conditioning strategies and provides an overview of the most recent and relevant findings on different concepts achieving cardioprotection in the experimental setting, specifically emphasizing pharmacological approaches in the perioperative context.
Collapse
Affiliation(s)
- Carolin Torregroza
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany.,Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Annika Raupach
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Katharina Feige
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Nina C Weber
- Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Markus W Hollmann
- Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Ragnar Huhn
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
19
|
He L, Wang Z, Zhou R, Xiong W, Yang Y, Song N, Qian J. Dexmedetomidine exerts cardioprotective effect through miR-146a-3p targeting IRAK1 and TRAF6 via inhibition of the NF-κB pathway. Biomed Pharmacother 2020; 133:110993. [PMID: 33220608 DOI: 10.1016/j.biopha.2020.110993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion (I/R) injury is a common cause of mortality. Cardiac miR-146a is emerging as a potent regulator of myocardial function. Dexmedetomidine preconditioning provides cardioprotective effects, of which mechanisms related to miR-146a-3p are unclear. METHODS A myocardial I/R model in rats and a cellular anoxia/reoxygenation (A/R) model in H9C2 cells were established and preconditioned with dexmedetomidine or not. H9C2 cells were transfected with mimics, inhibitor, or negative controls of miR-146a-3p, and siRNAs of IRAK1 or TRAF6. Relative expressions of miR-146a-3p were determined by quantitative real-time polymerase chain reaction. The apoptosis rates and reactive oxygen species (ROS) levels in H9C2 cells were examined by flow cytometry. Protein expressions of IRAK1, TRAF6, cleaved Caspase-3, BAX, BCL-2, NF-κB p65, phosphorylated NF-κB p65 (p-NF-κB p65), IκBα, and phosphorylated IκBα (p-IκBα) in H9C2 cells were detected by Western blot. RESULTS Dexmedetomidine decreased myocardial infarction size and apoptosis rates of H9C2 cells. Dexmedetomidine upregulated expression of miR-146a-3p. Dexmedetomidine significantly decreased protein expressions of IRAK1, TRAF6, cleaved Caspase-3, BAX, and NF-κB p65, but increased expressions of BCL-2 in H9C2 cells. miR-146a-3p overexpression strengthened the anti-apoptotic effect induced by dexmedetomidine in H9C2 cells via decreasing protein levels of IRAK1, TRAF6, cleaved Caspase-3, BAX, NF-κB p65, p-NF-κB p65, and p-IκBα and increasing protein level of BCL-2. Downregulation of miR-146a-3p reversed the changes in these proteins in H9C2 cells. Expressions of NF-κB p65 and p-NF-κB p65 were further decreased following knockdown of IRAK1 or TRAF6. ROS emission was significantly increased after A/R, while significantly decreased following dexmedetomidine preconditioning in H9C2 cells transfected with siIRAK1 or siTRAF6. CONCLUSION miR-146a-3p targeting IRAK1 and TRAF6 through inhibition of NF-κB signaling pathway and ROS emission is involved in cardioprotection induced by dexmedetomidine pretreatment.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China; Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming Medical University, Kunming, Yunnan Province, 650051, China
| | - Zhuoran Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Rui Zhou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Ning Song
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
20
|
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
|
21
|
Zhong Y, Li YP, Yin YQ, Hu BL, Gao H. Dexmedetomidine inhibits pyroptosis by down-regulating miR-29b in myocardial ischemia reperfusion injury in rats. Int Immunopharmacol 2020; 86:106768. [PMID: 32679539 DOI: 10.1016/j.intimp.2020.106768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Dexmedetomidine (DEX) was reported to protect heart against ischemic-reperfusion (IR) but the mechanism herein remains elusive. This study aims to explore the mechanism of DEX on pyroptosis induced by myocardial ischemic reperfusion (MIR). METHODS MIR rat models were established and injected DEX or miR-29b agomir/antagomir separately. The possible effect of DEX or miR-29b on myocardial cells was assessed according to measurement on creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), interleukin-1β (IL-1β) and interleukin-18 (IL-18), myocardial infarction size, myocardial injury and apoptosis. Western blot determined the expression levels of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC) and cleaved-caspase-1. Hypoxia/reoxygenation (H/R) cell model was established. The lactate dehydrogenase (LDH) content released by myocardial cells was examined. The relation between miR-29b and FoxO3a was confirmed by dual luciferase reporter gene assay. FoxO3a or ARC level was elevated in H/R myocardial cells to detect its effect on pyroptosis. RESULTS MIR rat models were successfully established, in which cell pyroptosis was triggered as evidenced by increased expression levels of NLRP3, ASC and cleaved-caspase-1. Rats with DEX precondition had attenuated cell pyroptosis and ameliorated inflammatory response. FoxO3a was a target of miR-29b. MiR-29b agomir or miR-29b antagomir could inhibit or promote the protective effect of DEX on MIR. Overexpression of FoxO3a/ARC axis could suppress myocardial pyroptosis induced by H/R. CONCLUSION DEX could ameliorate MIR injury (MIRI) and H/R injury in rats and inhibit H/R induced pyroptosis in myocardial cells via down-regulating miR-29b to activate FoxO3a/ARC axis.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yi-Ping Li
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yong-Qiang Yin
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Bai-Long Hu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Hong Gao
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| |
Collapse
|
22
|
MicroRNA-665-3p attenuates oxygen-glucose deprivation-evoked microglial cell apoptosis and inflammatory response by inhibiting NF-κB signaling via targeting TRIM8. Int Immunopharmacol 2020; 85:106650. [PMID: 32512270 DOI: 10.1016/j.intimp.2020.106650] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/09/2020] [Accepted: 05/28/2020] [Indexed: 12/21/2022]
Abstract
Microglial inflammation induced by ischemic stroke aggravates brain damage. MicroRNAs (miRNAs) have emerged as pivotal regulators in ischemic stroke-induced inflammation in microglial cells. miR-665-3p has been reported as a critical inflammation-associated miRNA. However, whether miR-665-3p participates in regulating microglial inflammation during ischemic stroke is underdetermined. This study investigated the potential role of miR-665-3p in stroke-induced inflammation in microglial cells using a cellular model of oxygen-glucose deprivation (OGD)-stimulated microglial cells in vitro. We found that miR-665-3p expression was decreased in microglial cells exposed to OGD treatment. Functional experiments demonstrated that the overexpression of miR-665-3p attenuated OGD-induced apoptosis and inflammation in microglial cells. Notably, tripartite motif 8 (TRIM8) was identified as a target gene of miR-665-3p. TRIM8 expression was induced by OGD treatment in microglial cells and the knockdown of TRIM8 protected microglial cells from OGD -induced cytotoxicity and inflammation. Moreover, TRIM8 knockdown or miR-665-3p overexpression blocked OGD-induced activation of nuclear factor (NF)-κB signaling in microglial cells. In addition, TRIM8 overexpression partially reversed the miR-665-3p overexpression-mediated inhibitory effect on OGD-induced inflammation in microglial cells. Taken together, these results indicate that miR-665-3p up-regulation protects microglial cells from OGD-induced apoptosis and inflammatory response by targeting TRIM8 to inhibit NF-κB signaling.
Collapse
|
23
|
Nazari-Shafti TZ, Exarchos V, Biefer HRC, Cesarovic N, Meyborg H, Falk V, Emmert MY. MicroRNA Mediated Cardioprotection - Is There a Path to Clinical Translation? Front Bioeng Biotechnol 2020; 8:149. [PMID: 32266222 PMCID: PMC7099408 DOI: 10.3389/fbioe.2020.00149] [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: 10/21/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
In the past 20 years, there have been several approaches to achieve cardioprotection or cardiac regeneration using a vast variety of cell therapies and remote ischemic pre-conditioning (RIPC). To date, substantial proof that either cell therapy or RIPC has the potential for clinically relevant cardiac repair or regeneration of cardiac tissue is still pending. Preclinical trials indicate that the secretome of cells in situ (during RIPC) as well as of transplanted cells may exhibit cardioprotective properties in the acute setting of cardiac injury. The secretome generally consists of cell-specific cytokines and extracellular vesicles (EVs) containing microRNAs (miRNAs). It is currently hypothesized that a subset of known miRNAs play a crucial part in the facilitation of cardioprotective effects. miRNAs are small non-coding RNA molecules that inhibit post-transcriptional translation of messenger RNAs (mRNAs) and play an important role in gene translation regulation. It is also known that one miRNAs usually targets multiple mRNAs. This makes predictability of pharmacokinetics and mechanism of action very difficult and could in part explain the inferior performance of various progenitor cells in clinical studies. Identification of miRNAs involved in cardioprotection and remodeling, the composition of miRNA profiles, and the exact mechanism of action are important to the design of future cell-based but also cell-free cardioprotective therapeutics. This review will give a description of miRNA with cardioprotective properties and a current overview on known mechanism of action and potential missing links. Additionally, we will give an outlook on the potential for clinical translation of miRNAs in the setting of myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany
| | - Vasileios Exarchos
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Héctor Rodriguez Cetina Biefer
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nikola Cesarovic
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Heike Meyborg
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany
| | - Volkmar Falk
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian Y Emmert
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Wang Z, Yang Y, Xiong W, Zhou R, Song N, Liu L, Qian J. Dexmedetomidine protects H9C2 against hypoxia/reoxygenation injury through miR-208b-3p/Med13/Wnt signaling pathway axis. Biomed Pharmacother 2020; 125:110001. [PMID: 32070878 DOI: 10.1016/j.biopha.2020.110001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 01/22/2023] Open
Abstract
Dexmedetomidine (Dex) has been reported to be cardioprotective. Differential expression of miR-208b-3p is associated with myocardial injury. But it is unknown that aberrant expression of miR-208b-3p is implicated in myocardial protection of Dex. Hypoxia/reoxygenation (HR) model was established in H9C2 cells. qRT-PCR was performed to detect expression levels of miR-208b-3p in H9C2 undergoing HR, Dex preconditioning, overexpression of miR-208b-3p or inhibition, and to assess expression of Med13 in H9C2 following knockdown of Med13 mRNA. CCK8 and, flow cytometry and Western blot were conducted respectively to examine viability, apoptosis rate and protein expressions of H9C2 subjected to a variety of treatments. Dex preconditioning reduced expression of miR-208b-3p and apoptosis of H9C2 cells caused by HR, while Dex preconditioning increased viability of H9C2. Dex preconditioning increased expression of Med13, which was reduced after knockdown of Med13 mRNA in H9C2. Overexpression of miR-208b-3p attenuated Dex exerted protective effects of myocardial cells, which was reversed by inhibition of miR-208b-3p. Increased expression of Med13 or/and decreased expression of miR-208b-3p decreased expression levels of Wnt/β-catenin signaling pathway-related proteins (Wnt3a, Wnt5a and β-catenin), while knockdown of Med13 mRNA or increased expression of miR-208b-3p increased the expression levels of those proteins. Dex protects H9C2 cells against HR injury through miR-208b-3p/Med13/Wnt/β-catenin signaling pathway axis.
Collapse
Affiliation(s)
- Zhuoran Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Rui Zhou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Ning Song
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Lan Liu
- Department of Pathology, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
25
|
The Epigenetics of the Endocannabinoid System. Int J Mol Sci 2020; 21:ijms21031113. [PMID: 32046164 PMCID: PMC7037698 DOI: 10.3390/ijms21031113] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
The endocannabinoid system (ES) is a cell-signalling system widely distributed in biological tissues that includes endogenous ligands, receptors, and biosynthetic and hydrolysing machineries. The impairment of the ES has been associated to several pathological conditions like behavioural, neurological, or metabolic disorders and infertility, suggesting that the modulation of this system may be critical for the maintenance of health status and disease treatment. Lifestyle and environmental factors can exert long-term effects on gene expression without any change in the nucleotide sequence of DNA, affecting health maintenance and influencing both disease load and resistance. This potentially reversible "epigenetic" modulation of gene expression occurs through the chemical modification of DNA and histone protein tails or the specific production of regulatory non-coding RNA (ncRNA). Recent findings demonstrate the epigenetic modulation of the ES in biological tissues; in the same way, endocannabinoids, phytocannabinoids, and cannabinoid receptor agonists and antagonists induce widespread or gene-specific epigenetic changes with the possibility of trans-generational epigenetic inheritance in the offspring explained by the transmission of deregulated epigenetic marks in the gametes. Therefore, this review provides an update on the epigenetics of the ES, with particular attention on the emerging role in reproduction and fertility.
Collapse
|
26
|
Wang YG, Liu CZ, Li YZ, Peng Y, Yan S. Cotreatments with Dex and Na2SeO3further improved antioxidant and anti-inflammatory protection of myocardial cells from I/R injury compared to their individual treatments. Free Radic Res 2020; 54:76-90. [PMID: 31909644 DOI: 10.1080/10715762.2019.1707198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yan-Gui Wang
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Cui-Zhong Liu
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Ying-Zhao Li
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - You Peng
- Department of Geriatrics, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Sulan Yan
- Cardiovascular Department, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
27
|
Overexpression of miR-375 Protects Cardiomyocyte Injury following Hypoxic-Reoxygenation Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7164069. [PMID: 31976033 PMCID: PMC6961604 DOI: 10.1155/2020/7164069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022]
Abstract
The aim of the study was to evaluate the clinical significance of microRNA-375 in acute myocardial infarction patients and its mimic action in hypoxia/reoxygenation- (H/R-) induced ventricular cardiomyocyte H9c2 injury. In the current study, 90 ST-elevated acute MI patients (STEMI), 75 non-ST-elevated acute MI patients (NSTEMI), 90 healthy subjects, 14 weeks old mice, and ventricular cardiomyocyte H9c2 were included. The expressions of plasma microRNA-375 in patients with STEMI and NSTEMI and AMI mouse models were remarkably decreased than in controls (P < 0.001). The areas under the curve (AUC) of plasma microRNA-375 were revealed 0.939 in STEMI and 0.935 in NSTEMI subjects. Moreover, microRNA-375 levels in H/R-exposed cardiac H9c2 cells were evidently downregulated and significantly increased apoptosis rate and caspase-3 activity levels, while overexpression of miR-375 remarkably reduced apoptosis percentage and caspase-3 levels as compared with normal cells. Furthermore, this study also demonstrated that Nemo-like kinase (NLK), NLK mRNA, and protein expression levels were significantly downregulated in H/R-injured H9c2 cells, on the contrary, H9c2 cells transfected with mimic-miR-375 greatly upregulated NLK mRNA and protein expression. Plasma microRNA-375 may serve as an essential clinical biomarker for diagnosis of early-stage AMI. Mimic expression of miR-375 significantly prevented H/R-induced cardiomyocyte injury by decreasing caspase-3 activity through upregulation of the NLK gene, recommended as a new therapeutic option for AMI patient.
Collapse
|
28
|
Wang L, Tang S, Wang Z, Chen H, Rajcha SS, Qian J. The administration of dexmedetomidine changes microRNA expression profiling of rat hearts. Biomed Pharmacother 2019; 120:109463. [PMID: 31541882 DOI: 10.1016/j.biopha.2019.109463] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dexmedetomidine is widely used for perioperative and ICU patients. microRNAs (miRNAs) function as regulators of gene expression. The aim of the study was to assay expression profiling of microRNA in rat hearts following administration of dexmedetomidine. METHODS In this study 6 rats were randomly divided into two groups (n = 3): dexmedetomidine group and control group. The rats of dexmedetomidine group were intraperitoneally given dexmedetomidine in a dose of 100 μg/kg whereas the rats in control group were administered normal saline intraperitoneally. The hearts were excised 30 min after the administration of dexmedetomidine or normal saline under anesthesia. The samples were analyzed for differentially expressed microRNAs with Exiqon miRNA Array. The differentially expressed microRNAs were confirmed by using qRT-PCR. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to find the target genes and signaling pathways of the aberrantly expressed miRNAs. RESULTS Six microRNAs were identified to be significantly expressed, among of which, five microRNAs (miRNA-434-3p, miRNA-3596d, miRNA-496-5p, miRNA-7a-2-3p and miRNA-702-3p) were up-regulated and 1 microRNA (miRNA-208b-3p) down-regulated compared to those of control group. The aberrantly expressed microRNAs were further validated by Quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). GO and KEGG analyses were used to identify target genes and the signaling pathways. CONCLUSIONS The use of dexmedetomidine is associated with differentially expressed microRNAs which may be involved in cardioprotection following administration of dexmedetomidine.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Shumiao Tang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhuoran Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Angel Women's & Children's Hospital, Kunming, Yunnan Province, China
| | - Shiva Sunder Rajcha
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|