51
|
Dexmedetomidine: What's New for Pediatrics? A Narrative Review. J Clin Med 2020; 9:jcm9092724. [PMID: 32846947 PMCID: PMC7565844 DOI: 10.3390/jcm9092724] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past few years, despite the lack of approved pediatric labelling, dexmedetomidine’s (DEX) use has become more prevalent in pediatric clinical practice as well as in research trials. Its respiratory-sparing effects and bioavailability by various routes are only some of the valued features of DEX. In recent years the potential organ-protective effects of DEX, with the possibility for preserving neurocognitive function, has put it in the forefront of clinical and bench research. This comprehensive review focused on the pediatric literature but presents relevant, supporting adult and animal studies in order to detail the recent growing body of literature around the pharmacology, end-organ effects, organ-protective effects, alternative routes of administration, synergetic effects, and clinical applications, with considerations for the future.
Collapse
|
52
|
Yuan HX, Chen CY, Li YQ, Ning DS, Li Y, Chen YT, Li SX, Fu MX, Li XD, Ma J, Jian YP, Liu DH, Mo ZW, Peng YM, Xu KQ, Ou ZJ, Ou JS. Circulating extracellular vesicles from patients with valvular heart disease induce neutrophil chemotaxis via FOXO3a and the inhibiting role of dexmedetomidine. Am J Physiol Endocrinol Metab 2020; 319:E217-E231. [PMID: 32516026 DOI: 10.1152/ajpendo.00062.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously demonstrated that circulating extracellular vesicles (EVs) from patients with valvular heart disease (VHD; vEVs) contain inflammatory components and inhibit endothelium-dependent vasodilation. Neutrophil chemotaxis plays a key role in renal dysfunction, and dexmedetomidine (DEX) can reduce renal dysfunction in cardiac surgery. However, the roles of vEVs in neutrophil chemotaxis and effects of DEX on vEVs are unknown. Here, we investigated the impact of vEVs on neutrophil chemotaxis in kidneys and the influence of DEX on vEVs. Circulating EVs were isolated from healthy subjects and patients with VHD. The effects of EVs on chemokine generation, forkhead box protein O3a (FOXO3a) pathway activation and neutrophil chemotaxis on cultured human umbilical vein endothelial cells (HUVECs) and kidneys in mice and the influence of DEX on EVs were detected. vEVs increased FOXO3a expression, decreased phosphorylation of Akt and FOXO3a, promoted FOXO3a nuclear translocation, and activated the FOXO3a signaling pathway in vitro. DEX pretreatment reduced vEV-induced CXCL4 and CCL5 expression and neutrophil chemotaxis in cultured HUVECs via the FOXO3a signaling pathway. vEVs were also found to suppress Akt phosphorylation and activate FOXO3a signaling to increase plasma levels of CXCL4 and CCL5 and neutrophil accumulation in kidney. The overall mechanism was inhibited in vivo with DEX pretreatment. Our data demonstrated that vEVs induced CXCL4-CCL5 to stimulate neutrophil infiltration in kidney, which can be inhibited by DEX via the FOXO3a signaling. Our findings reveal a unique mechanism involving vEVs in inducing neutrophils chemotaxis and may provide a novel basis for using DEX in reducing renal dysfunction in valvular heart surgery.
Collapse
Affiliation(s)
- Hao-Xiang Yuan
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Cai-Yun Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Quan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shang-Xuan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meng-Xia Fu
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiao-Di Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian Ma
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Peng Jian
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dong-Hong Liu
- Department of Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Kang-Qing Xu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Jun Ou
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
53
|
Dexmedetomidine Ameliorates Lung Injury Induced by Intestinal Ischemia/Reperfusion by Upregulating Cannabinoid Receptor 2, Followed by the Activation of the Phosphatidylinositol 3-Kinase/Akt Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6120194. [PMID: 32655771 PMCID: PMC7327571 DOI: 10.1155/2020/6120194] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023]
Abstract
Intestinal ischemia/reperfusion (I/R) is a clinical emergency, which often causes lung injury with high morbidity and mortality. Although dexmedetomidine has been identified to have a protective effect on lung injury caused by intestinal I/R, its specific mechanism is still elucidated. In recent years, the cannabinoid (CB2) receptor pathway has been found to be involved in I/R injury of some organs. In the current study, we investigated whether the CB2 receptor pathway contributes to the protective effect of dexmedetomidine on the intestinal I/R-induced lung injury in rats. Dexmedetomidine treatment upregulated the expression of CB2 receptor and suppressed the I/R-induced increases in lung injury scores, inflammatory cell infiltration, lung wet/dry ratio, MPO activity, MDA level, inflammatory cytokines, and caspase-3 expression while augmenting SOD activity and Bcl-2 expression, indicating attenuation of lung injury. Dexmedetomidine treatment also increased the expression of Akt. The protective effects of dexmedetomidine treatment were reversed by the CB2 receptor antagonist AM630 or the PI3K inhibitor wortmannin. And the CB2 receptor antagonist AM630 also downregulated the expression of Akt. Thus, our findings suggest that treatment with dexmedetomidine provides a protective role against lung injury caused by intestinal I/R in rats, possibly due to the upregulation of the CB2 receptor, followed by the activation of the PI3K/Akt pathway.
Collapse
|
54
|
Chen J, Perez R, de Mattos AM, Wang C, Li Z, Applegate RL, Liu H. Perioperative Dexmedetomidine Improves Outcomes of Kidney Transplant. Clin Transl Sci 2020; 13:1279-1287. [PMID: 32506659 PMCID: PMC7719359 DOI: 10.1111/cts.12826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Graft function is crucial for successful kidney transplantation. Many factors may affect graft function or cause delayed graft function (DGF), which decreases the prognosis for graft survival. This study was designed to evaluate whether the perioperative use of dexmedetomidine (Dex) could improve the incidence of function of graft kidney and complications after kidney transplantation. A total of 780 patients underwent kidney transplantations, 315 received intravenous Dex infusion during surgery, and 465 did not. Data were adjusted with propensity scores and multivariate logistic regression was used. The primary outcomes are major adverse complications, including DGF and acute rejection in the early post‐transplantation phase. The secondary outcomes included length of hospital stay (LOS), infection, overall complication, graft functional status, post‐transplantation serum creatinine values, and estimated glomerular filtration rate (eGFR). Dex use significantly decreased DGF (19.37% vs. 23.66%; adjusted odds ratio, 0.744; 95% confidence interval, 0.564–0.981; P = 0.036), risk of infection, risk of acute rejection in the early post‐transplantation phase, the risk of overall complications, and LOS. However, there were no statistical differences in 90‐day graft functional status or 7‐day, 30‐day, and 90‐day eGFR. Perioperative Dex use reduced incidence of DGF, risk of infection, risk of acute rejection, overall complications, and LOS in patients who underwent kidney transplantation.
Collapse
Affiliation(s)
- Jun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California, USA
| | - Richard Perez
- Department of Surgery, University of California Davis Health, Sacramento, California, USA
| | - Angelo Mario de Mattos
- Department of Internal Medicine, University of California Davis Health, Sacramento, California, USA
| | - Cecilia Wang
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California, USA
| | - Zhongmin Li
- Department of Internal Medicine, University of California Davis Health, Sacramento, California, USA
| | - Richard L Applegate
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California, USA
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, California, USA
| |
Collapse
|
55
|
Effects of Pre-Cardiopulmonary Bypass Administration of Dexmedetomidine on Cardiac Injuries and the Inflammatory Response in Valve Replacement Surgery With a Sevoflurane Postconditioning Protocol: A Pilot Study. J Cardiovasc Pharmacol 2020; 74:91-97. [PMID: 31356535 PMCID: PMC6688713 DOI: 10.1097/fjc.0000000000000698] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is Available in the Text. Background: Preventing myocardial ischemia–reperfusion injury in on-pump cardiac surgeries remains an enormous challenge. Sevoflurane postconditioning has been effective at overcoming this challenge by modulating inflammatory mediators and ameliorating antioxidative stress. Dexmedetomidine (DEX) is a commonly used medication for cardiac patients with organ-protective properties that lead to positive outcomes. Whether DEX also has cardiac-protective properties and the associated mechanism in sevoflurane postconditioning–based valve replacement surgeries are unknown. Objective: This study was conducted to observe the effect of DEX administration before cardiopulmonary bypass (CPB) on myocardial injury, oxidative stress, and inflammatory response indicators in the peripheral blood. Methods: Twenty-eight eligible cardiac patients who underwent valve replacement surgery with standard sevoflurane postconditioning were included in the study. The patients were randomly divided into a DEX group and a non-DEX group according to whether DEX (0.5-µg/kg overload dose for 10 minutes and a 0.5-μg/kg/h maintenance dose) or saline was administered from induction to the beginning of CPB. The primary outcome was the cardiac troponin I concentration (cTnI) in the blood 24 hours after CPB. The levels of malondialdehyde (MDA), superoxide dismutase, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) were also measured. Results: The mean cTnI at 24 hours after CPB was clearly decreased in the DEX group compared with that in the non-DEX group (4.16 ± 1.58 vs. 6.90 ± 3.73, P < 0.05). TNF-α levels were lower in the DEX group after CPB (T1–T5), with a significant difference found at 1–6 hours after CPB (1 hour, 19.03 vs. 28.09; 6 hours, 20.74 vs. 30.94, P < 0.05). The IL-6 and IL-8 concentrations in the DEX group were dramatically increased at 6 hours after CPB (P < 0.05). The MDA content and superoxide dismutase activity were comparable between the 2 groups. A lower proportion of anemia cases were noted after CPB in the DEX group than in the non-DEX group (non-DEX, 10% vs. DEX, 5%, P < 0.05). Conclusions: In valve replacement surgery with sevoflurane postconditioning, pre-CPB administration of DEX can reduce the cTnI level at 24 hours after CPB and brings synergic benefits of the inflammatory response.
Collapse
|
56
|
Liu Y, Zhang L, Wang S, Lu F, Zhen J, Chen W. Dexmedetomidine Reduces Atrial Fibrillation After Adult Cardiac Surgery: A Meta-Analysis of Randomized Controlled Trials. Am J Cardiovasc Drugs 2020; 20:271-281. [PMID: 31724106 DOI: 10.1007/s40256-019-00380-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Dexmedetomidine has been shown to have antiarrhythmic effects by exhibiting sympatholytic properties and activating the vagus nerve in preclinical studies. Results from clinical trials of dexmedetomidine on atrial fibrillation (AF) following adult cardiac surgery are controversial. MATERIALS AND METHODS We searched EMBASE, PubMed and Cochrane CENTRAL databases for randomized controlled trials (RCTs) comparing the antiarrhythmic effect of dexmedetomidine versus placebo or other anesthetic drugs in adult patients undergoing cardiac surgery. The primary outcome was the incidence of AF. The secondary outcomes were ventricular arrhythmias [ventricular fibrillation (VF), ventricular tachycardia (VT)], mechanical ventilation (MV) duration, intensive care unit (ICU) length of stay, and hospital length of stay, and all-cause mortality. RESULTS Thirteen trials with a total of 1684 study patients were selected. Compared with controls, dexmedetomidine significantly reduced the incidence of postoperative AF [odds ratio (OR) 0.75; 95% confidence interval (CI) 0.58-0.97; P = 0.03] and VT (OR 0.23; 95% CI 0.11-0.48; P < 0.0001). No significant difference for the incidence of VF existed (OR 0.80; 95% CI 0.21-3.03; P = 0.74). There was no significant difference between groups in MV duration [weighted mean difference (WMD) - 0.10; 95% CI - 0.42 to 0.21; P = 0.52], postoperative ICU stay (WMD - 0.49; 95% CI - 2.64 to 1.66; P = 0.65), hospital stay (WMD - 0.01; 95% CI - 0.16 to 0.13; P = 0.88) and mortality (OR 0.59; 95% CI 0.15-2.37; P = 0.46). CONCLUSIONS Perioperative administration of dexmedetomidine in adult patients undergoing cardiac surgery reduced the incidence of postoperative AF and VT. But there was no significant difference in incidence of VF, MV duration, ICU stay, hospital stay and mortality.
Collapse
Affiliation(s)
- Yang Liu
- Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
| | - Lei Zhang
- Department of anesthesiology, Beijing Daxing Maternal and Child Care Hospital, Beijing, China
| | - Suozhu Wang
- Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
| | - Feiping Lu
- Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
| | - Jie Zhen
- Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
| | - Wei Chen
- Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China.
| |
Collapse
|
57
|
Yuan T, Yang Z, Xian S, Chen Y, Wang L, Chen W, Long W, Che Y. Dexmedetomidine-mediated regulation of miR-17-3p in H9C2 cells after hypoxia/reoxygenation injury. Exp Ther Med 2020; 20:917-925. [PMID: 32742334 PMCID: PMC7388268 DOI: 10.3892/etm.2020.8775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 10/30/2019] [Indexed: 01/08/2023] Open
Abstract
Patients with heart disease often suffer from ischemia, which can be treated by reperfusion. However, this treatment can lead to the development of ischemia/reperfusion (I/R) injury, an inflammatory condition that can cause further heart damage. Dexmedetomidine (Dex), an α2-adrenoceptor agonist, and the microRNA (miR)-17-3p, have both been suggested to alleviate I/R injury and cardiac tissue inflammation. The aim of the present study was to investigate whether Dex and miR-17-3p could act together to prevent I/R injury. H9C2 cells, a myoblast cell line used as a model of rat cardiomyocytes, were cultured in a hypoxic environment for 3 h, and then reoxygenated for 3 h. This hypoxia/reoxygenation (H/R) was used to model I/R. Cell Counting kit-8 was used to determine cell viability and an annexin V-FITC/propidium iodide apoptosis kit used to analyze cell apoptosis. A dual luciferase reporter assay was used to determine the interaction between miR-17-3p and toll-like receptor 4 (TLR4). Western blotting and reverse transcription-quantitative PCR were used to determine protein levels and mRNA expression of TLR4 and galectin-3. A concentration of 0.1-10 µmol/l Dex attenuated H/R injury, which was accompanied by increased miR-17-3p levels. Additionally, the inhibition of miR-17-3p exacerbated H/R injury and reduced the effect of Dex on H/R injury. H/R led to an increased galectin-3 level compared with that in control cells, and Dex or miR-17-3p inhibitor did not markedly affect the level of galectin-3, indicating that Dex alleviated the effects of I/R injury through other pathways. Inhibition of miR-17-3p in Dex-induced H9C2 cells during H/R increased the expression of inflammatory mediators including tumor necrosis factor-α, interleukin (IL)-6, IL-1β and phosphorylated NFκB subunit p65, while Dex reduced the H/R-induced expression of these inflammatory mediators. Inhibition of TLR4 also attenuated H/R injury. In summary, the findings of the present study indicated that Dex reduced H/R injury in H9C2 cell via the modulation of inflammatory signaling pathways, and these inflammatory factors could be regulated by miR-17-3p.
Collapse
Affiliation(s)
- Tianhui Yuan
- Department of Drug Clinical Trials, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Clinical Research Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Phase I Program, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Zhongqi Yang
- Department of Drug Clinical Trials, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Clinical Research Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Phase I Program, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Shaoxiang Xian
- Department of Chinese Internal Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yang Chen
- School of Pharmaceuticals, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Lingjun Wang
- Lingnan Medical Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Weitao Chen
- Intensive Care Unit, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Wenjie Long
- College of First Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yuanyuan Che
- Department of Cardiology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277102, P.R. China
| |
Collapse
|
58
|
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: 22] [Impact Index Per Article: 4.4] [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
|
59
|
Ranninger E, Bettschart‐Wolfensberger R. Polymorphic tachycardia in an anaesthetised horse with an undiagnosed pheochromocytoma undergoing emergency coeliotomy. VETERINARY RECORD CASE REPORTS 2020. [DOI: 10.1136/vetreccr-2019-001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Elisabeth Ranninger
- Department of Clinical Diagnostics and ServicesSection of AnaesthesiologyVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Regula Bettschart‐Wolfensberger
- Department of Clinical Diagnostics and ServicesSection of AnaesthesiologyVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
60
|
Chou AH, Liao CC, Lee HC, Liou JT, Liu FC. The MAP2K4/JNK/c-Jun Signaling Pathway Plays A Key Role In Dexmedetomidine Protection Against Acetaminophen-Induced Liver Toxicity. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3887-3898. [PMID: 31814709 PMCID: PMC6861533 DOI: 10.2147/dddt.s215473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022]
Abstract
Purpose Dexmedetomidine [DEX; (S)-4-[1-(2,3-dimethylphenyl)ethyl]-3H-imidazole] is a selective α2-adrenergic receptor (α2-AR) agonist that attenuates the liver damage associated with local or systemic inflammation. However, it remains unclear whether DEX has protective effects against acetaminophen (Paracetamol, PARA)-induced liver toxicity (PILT). Methods PILT mice were established by intraperitoneal administration of a hepatotoxic dose of acetaminophen (300 mg/kg). Thirty minutes later, the mice were treated with DEX at a concentration of 0, 5, 25, or 50 μg/kg. Blood and liver samples were obtained for further analysis. Results DEX treatment significantly attenuated PILT in mice, with the strongest beneficial effects at a dose of 25 μg/kg. The levels of hepatic cytokines, tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), in addition to myeloperoxidase (MPO) activity, were significantly decreased following DEX treatment. Moreover, DEX treatment reduced macrophage recruitment around the area of hepatotoxicity and the expression levels of hepatic phosphorylated mitogen-activated protein kinase kinase 4 (MAP2K4), c-jun N-terminal kinase (JNK), and c-Jun expression induced by acetaminophen overdose. Conclusion The data suggest that DEX likely downregulates the JNK signaling pathway and its downstream effectors to promote its hepatoprotective effect, providing a clinical application of DEX for the attenuation of PILT.
Collapse
Affiliation(s)
- An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Anesthesiology, Xiamen Chang Gung Hospital, Taoyuan, Taiwan
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Tarng Liou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
61
|
Zhai M, Liu C, Li Y, Zhang P, Yu Z, Zhu H, Zhang L, Zhang Q, Wang J, Wang J. Dexmedetomidine inhibits neuronal apoptosis by inducing Sigma-1 receptor signaling in cerebral ischemia-reperfusion injury. Aging (Albany NY) 2019; 11:9556-9568. [PMID: 31682592 PMCID: PMC6874446 DOI: 10.18632/aging.102404] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022]
Abstract
Dexmedetomidine is known to alleviate cerebral ischemia-reperfusion injury (CIRI). We established a rat model of CIRI, which exhibited higher neurological deficit scores and a greater number of apoptotic cells in the cerebral ischemic penumbra than controls. Dexmedetomidine reversed the neuronal apoptosis and improved neurological function in this model. We then examined Sigma-1 receptor (Sig-1R) expression on the endoplasmic reticulum (ER) in brain tissues at different reperfusion time points. Sig-1R expression increased with CIRI and decreased with increasing reperfusion times. After 24 hours of reperfusion, dexmedetomidine upregulated Sig-1R expression, and ER stress proteins (GRP78, CHOP, JNK and Caspase-3) were detected in brain tissues with Western blotting. Moreover, GRP78 expression followed a pattern similar to Sig-1R. Dexmedetomidine induced GRP78 expression but inhibited CHOP, Caspase-3 and phosphorylated-JNK expression in brain tissues. A Sig-1R-specific inhibitor reduced GRP78 expression and partially inhibited the upregulation of GRP78 by dexmedetomidine. The inhibitor also increased CHOP and Caspase-3 expression and partially reversed the inhibitory effects of dexmedetomidine on these pro-apoptotic ER stress proteins. These results suggest that dexmedetomidine at least partially inhibits ER stress-induced apoptosis by activating Sig-1R, thereby attenuating brain damage after 24 hours of ischemia-reperfusion.
Collapse
Affiliation(s)
- Meili Zhai
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Chong Liu
- Department of Anesthesiology, Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, Tianjin 300140, China
| | - Yuexiang Li
- Department of Anesthesiology, Tianjin Xiqing Hospital, Tianjin 300380, China
| | - Peijun Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Zhiqiang Yu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - He Zhu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Li Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Qian Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jianbo Wang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jinhua Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province 318000, China
| |
Collapse
|
62
|
Dexmedetomidine alleviates doxorubicin cardiotoxicity by inhibiting mitochondrial reactive oxygen species generation. Hum Cell 2019; 33:47-56. [PMID: 31643023 DOI: 10.1007/s13577-019-00282-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/14/2019] [Indexed: 10/25/2022]
Abstract
Cardiotoxicity largely limits the application of doxorubicin (Dox) for cancer treatment. Dexmedetomidine (Dex), a selective agonist of α2-adrenergic receptor, has been suggested to exert cardioprotection against myocardial injury. However, the effect and underlying mechanisms of Dex on Dox cardiotoxicity remain unknown. In this study, C57BL/6 mice were treated with Dox followed by Dex administration. Cardiomyocytes were co-incubated with Dox and Dex in vitro. The results showed that Dex markedly attenuated cardiac dysfunction induced by Dox. TUNEL staining exhibited that Dex inhibited Dox-induced cardiomyocyte apoptosis in myocardium. Moreover, the expression of anti-apoptotic protein Bcl-2 was increased, whereas the expression of pro-apoptotic protein Bax was decreased by Dex. Dox-induced the increase of reactive oxygen species (ROS), superoxide anion, and mitochondrial ROS (mROS) generation in myocardial tissues were significantly inhibited after Dex administration. In in vitro study, it was further confirmed that Dex prevented Dox-induced cardiomyocyte apoptosis and injury. However, the stimulation of mROS generation reversed the effect of Dex in cardiomyocytes. Mechanically, Dex blocked Dox-induced the ubiquitination of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), leading to the restoration of PGC-1α and downstream oxidative stress-protective molecules uncoupling protein 2 and manganese-dependent superoxide dismutase expression. Taken together, this study demonstrates that Dex exerts cardioprotection against Dox cardiotoxicity by attenuating mitochondrial dysfunction, oxidative stress, and cardiomyocyte apoptosis via inhibiting PGC-1α-signaling pathway inactivation. This suggests that Dex may be a potential therapeutic strategy for Dox cardiotoxicity treatment.
Collapse
|
63
|
Long non-coding RNA MALAT1 sponges microRNA-429 to regulate apoptosis of hippocampal neurons in hypoxic-ischemic brain damage by regulating WNT1. Brain Res Bull 2019; 152:1-10. [DOI: 10.1016/j.brainresbull.2019.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
|
64
|
The Role of Heme Oxygenase-1 in Remote Ischemic and Anesthetic Organ Conditioning. Antioxidants (Basel) 2019; 8:antiox8090403. [PMID: 31527528 PMCID: PMC6770180 DOI: 10.3390/antiox8090403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
The cytoprotective effects of the heme oxygenase (HO) pathway are widely acknowledged. These effects are mainly mediated by degradation of free, pro-oxidant heme and the generation of carbon monoxide (CO) and biliverdin. The underlying mechanisms of protection include anti-oxidant, anti-apoptotic, anti-inflammatory and vasodilatory properties. Upregulation of the inducible isoform HO-1 under stress conditions plays a crucial role in preventing or reducing cell damage. Therefore, modulation of the HO-1 system might provide an efficient strategy for organ protection. Pharmacological agents investigated in the context of organ conditioning include clinically used anesthetics and sedatives. A review from Hoetzel and Schmidt from 2010 nicely summarized the effects of anesthetics on HO-1 expression and their role in disease models. They concluded that HO-1 upregulation by anesthetics might prevent or at least reduce organ injury due to harmful stimuli. Due to its clinical safety, anesthetic conditioning might represent an attractive pharmacological tool for HO-1 modulation in patients. Remote ischemic conditioning (RIC), first described in 1993, represents a similar secure option to induce organ protection, especially in its non-invasive form. The efficacy of RIC has been intensively studied herein, including on patients. Studies on the role of RIC in influencing HO-1 expression to induce organ protection are emerging. In the first part of this review, recently published pre-clinical and clinical studies investigating the effects of anesthetics on HO-1 expression patterns, the underlying signaling pathways mediating modulation and its causative role in organ protection are summarized. The second part of this review sums up the effects of RIC.
Collapse
|
65
|
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: 4.2] [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
|
66
|
Xue X, Luo L. LncRNA HIF1A-AS1 contributes to ventricular remodeling after myocardial ischemia/reperfusion injury by adsorption of microRNA-204 to regulating SOCS2 expression. Cell Cycle 2019; 18:2465-2480. [PMID: 31354024 DOI: 10.1080/15384101.2019.1648960] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objectives: Long non-coding RNAs (lncRNAs) serve pivotal roles in heart disease, while the role of lncRNA hypoxia-inducible factor 1α-antisense RNA 1 (HIF1A-AS1) is rarely mentioned. Therefore, the objective of this study was to investigate the mechanism of lncRNA HIF1A-AS1 regulating suppressor of cytokine signaling 2 (SOCS2) expression by adsorption of microRNA-204 (miR-204) on ventricular remodeling after myocardial ischemia-reperfusion (I/R) injury in mice. Methods: The mouse model of I/R was established by left coronary artery occlusion. The expression of HIF1A-AS1, miR-204 and SOCS2 was determined. The mice were injected with HIF1A-AS1-siRNA, miR-204 mimics or their controls to investigate their effects on cardiac function and ventricular remodeling of mice after I/R injury. The binding relationship between HIF1A-AS1 and miR-204 as well as between miR-204 and SOCS2 were verified. Results: HIF1A-AS1 and SOCS2 were upregulated and miR-204 was downregulated in myocardial tissues in mice after I/R injury. LVEDD, LVEDS, LVEDP, LVMI and RVMI expression reduced while LVEF, LVFS, +dp/dt max and - dp/dt max increased through knockdown HIF1A-AS1 and upregulated miR-204. The expression of BNP, cTnI, LDH, CK, TNF-α, IL-1β, IL-6 and β-MHC reduced, and the expression of α-MHC increased when HIF1A-AS1 was poorly expressed and miR-204 was highly expressed. Silencing HIF1A-AS1 and upregulating miR-204 inhibited apoptosis of cells. LncRNA HIF1A-AS1 could act as ceRNA to adsorb miR-204 to suppress miR-204 expression and elevate SOCS2 expression. Conclusion: Our study provides evidence that downregulation of HIF1A-AS1 and upregulation of miR-204 could alleviate ventricular remodeling and improve cardiac function in mice after myocardial I/R injury via regulating SOCS2.
Collapse
Affiliation(s)
- Xiang Xue
- Cardiovascular Medicine Department, Changzhou No.7 People' s Hospital , Changzhou , Jiangsu , China
| | - Libo Luo
- Cardiovascular Medicine Department, Changzhou No.7 People' s Hospital , Changzhou , Jiangsu , China
| |
Collapse
|
67
|
Zhou HM, Ling XY, Ni YJ, Wu C, Zhu ZP. Pre-cardiopulmonary bypass administration of dexmedetomidine decreases cardiac troponin I level following cardiac surgery with sevoflurane postconditioning. J Int Med Res 2019; 47:3623-3635. [PMID: 31234690 PMCID: PMC6726774 DOI: 10.1177/0300060519856750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022] Open
Abstract
Objective This study was performed to determine the effect of dexmedetomidine (DEX) administration on myocardial damage in cardiac surgery with sevoflurane postconditioning. Methods We retrospectively examined all cardiac valve replacement surgeries from 1 April 2016 to 30 April 2017. Eligible patients were divided into two groups based on whether DEX was infused. DEX infusion was permitted only between intubation and the beginning of cardiopulmonary bypass (CPB). Sevoflurane was inhaled via the standard postconditioning procedure starting at aortic declamping. The cardiac troponin I (cTnI) level was measured at different time points. The postoperative outcomes and complications were also analyzed. Results One hundred patients were included in the study (DEX group, n = 53; non-DEX group, n = 47). Increased cTnI levels were significantly correlated with the New York Heart Association classification, CPB time, and DEX use. DEX use and the CPB time were potential independent factors contributing to changes in the cTnI level. The cTnI level at 6, 12, and 24 hours postoperatively was remarkably lower in the DEX than non-DEX group by 1.14, 7.83, and 5.86 ng/mL, respectively. Conclusions DEX decreased the cTnI level after CPB when sevoflurane postconditioning was used, especially at 6, 12, and 24 hours postoperatively.
Collapse
Affiliation(s)
- Hong-mei Zhou
- Department of Anesthesiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Xiao-yan Ling
- Outpatient-Nursing Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Yun-jian Ni
- Department of Anesthesiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Cheng Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Zhi-peng Zhu
- Department of Anesthesiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| |
Collapse
|
68
|
Yang C, He L, Wang C, Huang Y, Wang A, Li X, Ao J. Dexmedetomidine alleviated lipopolysaccharide/D-galactosamine-induced acute liver injury in mice. Int Immunopharmacol 2019; 72:367-373. [DOI: 10.1016/j.intimp.2019.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 01/29/2023]
|
69
|
Wang YQ, Tang YF, Yang MK, Huang XZ. Dexmedetomidine alleviates cerebral ischemia-reperfusion injury in rats via inhibition of hypoxia-inducible factor-1α. J Cell Biochem 2019; 120:7834-7844. [PMID: 30456861 DOI: 10.1002/jcb.28058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Dexmedetomidine (Dex) was reported to reduce ischemia-reperfusion (I/R) injury in kidney and brain tissues. Thus, we aimed to study the role and mechanism of Dex in cerebral I/R injury by inhibiting hypoxia-inducible factor-1α (HIF-1α) and apoptosis. First, I/R injury models were established. Six groups were assigned after different treatments: sham, I/R, I/R+Dex, I/R+2-methoxyestradiol (2ME2) (HIF-1α inhibitor), I/R+CoCl 2 (HIF-1α activator), and I/R+Dex+CoCl 2 groups. Neurological function, cerebral infarction volume, survival, and apoptosis of brain cells were then analyzed. Besides, immunohistochemistry and Western blot analysis were used to detect the expression of HIF-1α, BCL-2[B-cell leukemia/lymphoma 2] adenovirus E1B interacting protein 3 (BNIP3), B-cell leukemia/lymphoma 2 (BCL2), BCL2[B-cell leukemia/lymphoma 2] associated X (Bax), and cleaved-caspase3 proteins in brain tissues. I/R rats showed cerebral infarction, increased neurological function score, number of terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL)-positive cells and HIF-1α-positive cells as well as decreased neurons. Inhibition of HIF-1α can reduce the apoptosis induced by I/R, and overexpression of HIF-1α can aggravate apoptosis in brain tissue of I/R rats. Furthermore, activation of HIF-1α expression blocks the inhibitory effect of Dex on neuronal apoptosis in I/R rats. Dex may inhibit the neuronal apoptosis of I/R rats by inhibiting the HIF-1α pathway and then improve the cerebral I/R injury in rats.
Collapse
Affiliation(s)
- Yuan-Qing Wang
- Department of Neurology, Rizhao People's Hospital, Rizhao, China
| | - Yu-Feng Tang
- Department of Neurology, Mianyang Central Hospital, Mianyang, China
| | - Ming-Kun Yang
- Department of Neurology, Chiping People's Hospital, Chiping, China
| | - Xi-Zhao Huang
- Department of Anesthesiology, Guangdong Women and Children's Hospital, Guangzhou, China
| |
Collapse
|
70
|
Tang Y, Jia C, He J, Zhao Y, Chen H, Wang S. The Application and Analytical Pathway of Dexmedetomidine in Ischemia/Reperfusion Injury. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2019; 2019:7158142. [PMID: 31949972 PMCID: PMC6948278 DOI: 10.1155/2019/7158142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/05/2019] [Indexed: 05/07/2023]
Abstract
Ischemia/reperfusion cerebral injury can cause serious damage to nerve cells. The injured organelles are cleared by autophagy eventually, which is critical for cell survival. Dexmedetomidine is neuroprotective in various ischemia/reperfusion models. Mitochondrial calcium uniporter (MCU) is the most important channel of mitochondrial Ca2+ influx into mitochondria, where Ca2+ has a potential effect on mitochondrial autophagy. However, the role of MCU in the changes of mitophagy and autophagy caused by dexmedetomidine is unknown. In this study, we constructed an in vitro I/R model by subjecting the oxygen and glucose deprivation/reperfusion model to SH-SY5Y cells to mimic the cerebral I/R injury. We found that postconditioning with dexmedetomidine and 3-methyladenine (3MA, an autophagy inhibitor) increased the cell survival meanwhile reduced the production of autophagic vesicles and the expression of LC3 and Beclin 1. This process also increased the expression of BCL-2, P62, and TOM20. After applied with spermine (MCU-specific agonist), the expression of autophagy proteins by dexmedetomidine was reversed, and the same changes were also observed in immunofluorescence. The results of our study suggested that dexmedetomidine can inhibit MCU and reduce excessive mitophagy and autophagy for conferring protection against I/R injury.
Collapse
Affiliation(s)
- Ying Tang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Anesthesiology, Weifang Yidu Central Hospital, Weifang, China
| | - Changxin Jia
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianshuai He
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Zhao
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huayong Chen
- Department of Anesthesiology, Weifang Yidu Central Hospital, Weifang, China
| | - Shilei Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
71
|
Song Y, Kim DH, Kwon TD, Han DW, Baik SH, Jung HH, Kim JY. Effect of intraoperative dexmedetomidine on renal function after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy: a randomized, placebo-controlled trial. Int J Hyperthermia 2018; 36:1-8. [PMID: 30354794 DOI: 10.1080/02656736.2018.1526416] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) predispose to postoperative renal dysfunction. Dexmedetomidine is an α2 adrenoreceptor agonist, which has renoprotective effects after cardiac surgery. OBJECTIVE To assess the effect of dexmedetomidine on renal function after CRS and HIPEC. MATERIALS Thirty-eight patients undergoing CRS and HIPEC were randomized to receive dexmedetomidine (dexmedetomidine group, n = 19, loading 1 μg/kg over 20 min followed by infusion at 0.5 μg/kg/h) or 0.9% sodium chloride (control group, n = 19) during surgery. Creatinine clearance (CrCl) was assessed daily until postoperative day 7. Urine neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule (KIM)-1 were measured for 24 h after surgery. RESULTS There was no difference in the lowest CrCl value during the first 7 days postoperatively, but the % change from baseline to the lowest value was lower in the dexmedetomidine group than in the control group (p = .037). Urine NGAL and KIM-1 levels were increased over time in both groups, but the increases were significantly less in the dexmedetomidine group (p = .018 and 0.038, respectively). In the dexmedetomidine group, the length of intensive care unit stay was shorter (p = .034). CONCLUSIONS Intraoperative dexmedetomidine infusion did not improve renal function in terms of serum Cr-related indices following CRS and HIPEC. However, as the decrease in CrCl was attenuated and early tubular-injury markers were lower in the dexmedetomidine group, dexmedetomidine may have protective effects against early tubular injury in CRS and HIPEC. Clinical Trials Registry: http://clinicaltrials.gov (NCT02641938).
Collapse
Affiliation(s)
- Young Song
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea.,b Anesthesia and Pain Research Institute , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Do-Hyeong Kim
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea.,b Anesthesia and Pain Research Institute , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Tae Dong Kwon
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea.,b Anesthesia and Pain Research Institute , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Dong Woo Han
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea.,b Anesthesia and Pain Research Institute , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Seung Hyuk Baik
- c Department of Surgery , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Hwan Ho Jung
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea
| | - Ji Young Kim
- a Department of Anesthesiology and Pain Medicine , Yonsei University College of Medicine , Seoul , Republic of Korea.,b Anesthesia and Pain Research Institute , Yonsei University College of Medicine , Seoul , Republic of Korea
| |
Collapse
|
72
|
Dexmedetomidine protects high-glucose induced apoptosis in human retinal pigment epithelial cells through inhibition on p75(NTR). Biomed Pharmacother 2018; 106:466-471. [DOI: 10.1016/j.biopha.2018.06.117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 01/10/2023] Open
|
73
|
Liu C, Fu Q, Mu R, Wang F, Zhou C, Zhang L, Yu B, Zhang Y, Fang T, Tian F. Dexmedetomidine alleviates cerebral ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress dependent apoptosis through the PERK-CHOP-Caspase-11 pathway. Brain Res 2018; 1701:246-254. [PMID: 30201260 DOI: 10.1016/j.brainres.2018.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/14/2018] [Accepted: 09/06/2018] [Indexed: 01/07/2023]
Abstract
Dexmedetomidine (Dex) has the neuroprotective effect on cerebral ischemia-reperfusion injury (CIRI). But the mechanism is not yet clear. In this study, we established a model of middle cerebral artery occlusion (MCAO) and treated primary cortical neurons with oxygen glucose deprivation (OGD), followed by Dex treatment. Neurological protection of Dex was then assessed by neurological deficit score, brain edema, TTC staining, TUNEL assay, Western blot analysis, immunohistochemistry, and RT-PCR. The results showed that Dex significantly reduced the neurological deficit score, brain edema and cerebral infarction area due to CIRI. After Dex treatment, the expression levels of ER stress-related apoptosis pathway proteins (GRP78, p-PERK, CHOP and Cleaved-caspase-3) were significantly decreased and the apoptosis of brain cells was also significantly reduced. Immunohistochemistry showed that expression and nuclear localization of CHOP decreased significantly after the application of Dex. The downstream apoptotic protein caspase-11 mediated by PERK-CHOP was also markedly inhibited by Dex. In conclusion, our results suggested that Dex reduced ER stress-induced apoptosis after CIRI. Its protective mechanism may be related to PERK-CHOP-Caspase-11 dependent signaling pathway.
Collapse
Affiliation(s)
- Chong Liu
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Qiang Fu
- Department of Critical Care Medicine, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China.
| | - Rong Mu
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Fang Wang
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Chunjing Zhou
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Li Zhang
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Baojin Yu
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Yang Zhang
- Department of Anaesthesiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Tao Fang
- Central Laboratory, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Fengshi Tian
- Department of Cardiology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China.
| |
Collapse
|
74
|
Renoprotective effects of dexmedetomidine against ischemia-reperfusion injury in streptozotocin-induced diabetic rats. PLoS One 2018; 13:e0198307. [PMID: 30114208 PMCID: PMC6095484 DOI: 10.1371/journal.pone.0198307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022] Open
Abstract
Background Diabetic patients are susceptible to renal ischemia-reperfusion injury, which leads to perioperative complications. Activation of NOD-like receptor protein 3 (NLRP3) inflammasome participates in the development of diabetes, and contributes to renal ischemia-reperfusion injury. Dexmedetomidine (DEX), a highly selective α2-adrenoreceptor agonist, shows renoprotective effects against ischemia-reperfusion injury. We aimed to elucidate the effects, underlying mechanisms, and optimal timing of DEX treatment in diabetic rats. Methods Male Sprague-Dawley rats (n = 12 per group) were randomly divided into normal-sham, diabetes-sham, diabetes-ischemia-reperfusion-control, diabetes-ischemia-reperfusion-DEX-pre-treatment, and diabetes-ischemia-reperfusion-DEX-post-treatment groups. Renal ischemia-reperfusion injury was induced in diabetic rats by occlusion of both renal arteries for 45 min, followed by reperfusion for 24 h. DEX (10 μg/kg) was administered intraperitoneally 1 h before ischemia (pre-treatment) or upon reperfusion (post-treatment). After reperfusion, renal tissue was biochemically and histopathologically evaluated. Results DEX treatment attenuated ischemia reperfusion-induced increase in NLRP3, caspase-1, IL-1β, phospho-AKT, and phospho-ERK signaling. Moreover, oxidative stress injury, inflammatory reactions, apoptosis, and renal tubular damage were favorably modulated by DEX treatment. Furthermore, post-reperfusion treatment with DEX was significantly more effective than pre-treatment in modulating NLRP3 inflammasome, AKT and ERK signaling, and oxidative stress. Conclusions This study shows that the protective effects of DEX in renal ischemia-reperfusion injury are preserved in diabetic conditions and may potentially provide a basis for the use of DEX in clinical treatment of renal ischemia-reperfusion injury.
Collapse
|
75
|
Cheng J, Zhu P, Qin H, Li X, Yu H, Yu H, Peng X. Dexmedetomidine attenuates cerebral ischemia/reperfusion injury in neonatal rats by inhibiting TLR4 signaling. J Int Med Res 2018; 46:2925-2932. [PMID: 29926753 PMCID: PMC6124261 DOI: 10.1177/0300060518781382] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective The sedative dexmedetomidine plays a role in multi-organ protection by inhibiting toll-like receptor (TLR) 4 expression in ischemia/reperfusion injury. The present study investigated whether the neuroprotective effects of dexmedetomidine could be blocked by the TLR4 agonist lipopolysaccharide. Methods We established a cerebral ischemia/reperfusion model in neonatal Sprague-Dawley rats through bilateral carotid artery occlusion for 20 minutes followed by a 2-hour reperfusion. Rats were assigned to four groups: Sham operation, ischemia/reperfusion, ischemia/reperfusion preceded by dexmedetomidine treatment (10 µg/kg), and ischemia/reperfusion preceded by dexmedetomidine (10 µg/kg) and lipopolysaccharide (500 µg/kg) treatments. Cerebral tissue injury was assessed by hematoxylin and eosin staining, and cerebral TLR4 expression was evaluated by real-time PCR and western blot. Results Pretreatment with dexmedetomidine reduced ischemia-induced morphological changes in the hippocampal CA3 region and downregulated TLR4 expression, but these neuroprotective effects were partially blocked by co-treatment with the TLR4 agonist lipopolysaccharide. Conclusion Our results indicate that inhibition of cerebral TLR4 expression is related to the neuroprotective effects of dexmedetomidine in this neonatal rat cerebral ischemia/reperfusion model.
Collapse
Affiliation(s)
- Jiangxia Cheng
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pengfei Zhu
- 2 Department of Cardiology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Han Qin
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xia Li
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Yu
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Yu
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohong Peng
- 1 Department of Anesthesia, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
76
|
Zhang MH, Zhou XM, Cui JZ, Wang KJ, Feng Y, Zhang HA. Neuroprotective effects of dexmedetomidine on traumatic brain injury: Involvement of neuronal apoptosis and HSP70 expression. Mol Med Rep 2018; 17:8079-8086. [PMID: 29693126 PMCID: PMC5983975 DOI: 10.3892/mmr.2018.8898] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of dexmedetomidine (Dex) on traumatic brain injury (TBI), and further evaluate whether the underlying neuroprotective mechanisms are associated with neurological apoptosis and the expression of 70 kDa heat shock protein (HSP70) in the hippocampus. A total of 90 adult male Sprague-Dawley rats were randomly assigned into 3 groups (n=30/group): Sham, TBI and Dex groups. The rat models of TBI were established using a modified weight-drop device and Dex (15 µg/kg) was intravenously administered immediately following TBI. The brain edema and neurological function outcomes of TBI were assessed using wet-dry weight analysis and the Neurological Severity Score method. The expression levels of B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X protein (Bax) in the rat hippocampus were evaluated using immunohistochemical staining and western blot analysis. The protein levels of HSP70 in the hippocampal region were analyzed using western blot analysis. The results of the present study revealed that administration of Dex post-TBI improved brain edema and neurological outcomes, due to the attenuation of the TBI-induced reduction of Bax expression and increase of Bcl-2 and HSP70 expression. In conclusion, the results of the present study suggested that administration of Dex may serve as a neuroprotective agent against brain injury, at least partially via the inhibition of neuronal apoptosis and upregulation of HSP70 expression in the hippocampus.
Collapse
Affiliation(s)
- Man-He Zhang
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xiu-Min Zhou
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jian-Zhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Kai-Jie Wang
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yan Feng
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Hong-Ao Zhang
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
77
|
Yan X, Cheng X, Zhou L, He X, Zheng W, Chen H. Dexmedetomidine alleviates lipopolysaccharide-induced lung injury in Wistar rats. Oncotarget 2018; 8:44410-44417. [PMID: 28574842 PMCID: PMC5546489 DOI: 10.18632/oncotarget.17899] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
This study aimed to investigate the protective effects of dexmedetomidine on lipopolysaccharide (LPS)-induced lung injury in Wistar rats. 24 female Wistar rats were randomly assigned into 3 groups (n = 8): a control group, a LPS-challenged group, and a LPS plus dexmedetomidine group. Inflammation, oxidative stress, Nrf2/Keap1, and Akt signal were determined. The results showed that LPS caused inflammation and oxidative stress via increasing pro-inflammatory cytokines and oxidative products. Dexmedetomidine treatment alleviated inflammation and oxidative stress in LPS-challenged rats. Nrf2/Keap1 was inhibited and Akt signal was activated in the lung after exposure to LPS, while dexmedetomidine activated Nrf2/Keap1, which further mediated expressions of antioxidant genes. In conclusion, dexmedetomidine alleviated inflammatory response and oxidative stress in LPS-induced lung injury in rats via influencing Nrf2/Keap1 signal.
Collapse
Affiliation(s)
- Xuetao Yan
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, 518100, China
| | - Xiaoli Cheng
- Department of Pharmacy, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, 518100, China
| | - Liwen Zhou
- Department of Anesthesiology, Xiangyang Central Hospital, Xiangyang, 441021, China
| | - Xianghu He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wenzhong Zheng
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, 518100, China
| | - Hu Chen
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, 518100, China
| |
Collapse
|
78
|
Wu J, Vogel T, Gao X, Lin B, Kulwin C, Chen J. Neuroprotective effect of dexmedetomidine in a murine model of traumatic brain injury. Sci Rep 2018; 8:4935. [PMID: 29563509 PMCID: PMC5862953 DOI: 10.1038/s41598-018-23003-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/05/2018] [Indexed: 12/24/2022] Open
Abstract
No FDA approved pharmacological therapy is available that would reduce cell death following traumatic brain injury (TBI). Dexmedetomidine (Dex) is a highly selective agonist of alpha-2 adrenergic receptors and has demonstrated neuroprotective effects in hippocampal slice cultures undergoing direct impact. However, no one has tested whether Dex, in addition to its sedative action, has neuroprotective effects in an animal model of TBI. Thus, in the present study, we investigated the effects of Dex on an animal model of TBI. Mice received different doses of Dex (1, 10, or 100 µg/kg bodyweight, n = 10 each group) or saline as control at 1 hour and 12 hours following TBI. The mice treated with Dex lost less cortical tissue than the control mice. Further analysis found that Dex treatment reduced cell death in the cortex and the hippocampus measured by Fluoro-Jade B (FJB) staining, prevented axonal degeneration detected by immunostaining with antibody against β-amyloid precursor protein (β-APP), and protected synapses from elimination with synaptophysin staining. Taken together, in an in vivo murine model of TBI, Dex at the dose of 100 µg/kg not only prevented tissue lesion and cell death, but also reduced axonal injury and synaptic degeneration caused by TBI.
Collapse
Affiliation(s)
- Jin Wu
- Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Todd Vogel
- Department of Neurosurgery, Indiana University, Indianapolis, IN, USA
| | - Xiang Gao
- Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indianapolis, IN, USA
| | - Bin Lin
- Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Charles Kulwin
- Department of Neurosurgery, Indiana University, Indianapolis, IN, USA
| | - Jinhui Chen
- Department of Neurosurgery, Indiana University, Indianapolis, IN, USA. .,Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indianapolis, IN, USA.
| |
Collapse
|
79
|
Zhang N, Su QP, Zhang WX, Shi NJ, Zhang H, Wang LP, Liu ZK, Li KZ. Neuroprotection of dexmedetomidine against propofol-induced neuroapoptosis partly mediated by PI3K/Akt pathway in hippocampal neurons of fetal rat *. J Zhejiang Univ Sci B 2017; 18:789-796. [PMCID: PMC5611550 DOI: 10.1631/jzus.b1600476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/18/2016] [Indexed: 11/19/2023]
Abstract
The aim was to investigate how the PI3K/Akt pathway is involved in the protection of dexmedetomidine against propofol. The hippocampal neurons from fetal rats were separated and cultured in a neurobasal medium. Cell viability was assayed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Then neurons were pretreated with different concentrations of dexmedetomidine before 100 μmol/L propofol was added. Akt, phospho-Akt (p-Akt), Bad, phospho-Bad (p-Bad), and Bcl-xL were detected by Western blot. Also, neurons were pretreated with dexmedetomidine alone or given the inhibitor LY294002 before dexmedetomidine pretreatment, and then propofol was added for 3 h. The results demonstrated that propofol decreased the cell viability and the expression of p-Akt and p-Bad proteins, increased the level of Bad, and reduced the ratio of Bcl-xL/Bad. Dexmedetomidine pretreatment could reverse these effects. The enhancement of p-Akt and p-Bad induced by dexmedetomidine was prevented by LY294002. These results showed that dexmedetomidine potently protected the developing neuron and this protection may be partly mediated by the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Anesthesiology, the Second Hospital of Shandong University, Jinan 250033, China
- Department of Anesthesiology, Linyi People’s Hospital, Linyi 276003, China
| | - Quan-ping Su
- The Central Laboratory, Linyi Peoples’ Hospital, Linyi 276003, China
| | - Wei-xia Zhang
- Intensive Care Unit, Linyi People’s Hospital, Linyi 276003, China
| | - Nian-jun Shi
- Department of Anesthesiology, Linyi People’s Hospital, Linyi 276003, China
| | - Hao Zhang
- Department of Anesthesiology, Linyi People’s Hospital, Linyi 276003, China
| | - Ling-ping Wang
- Department of Anesthesiology, Linyi People’s Hospital, Linyi 276003, China
| | - Zhong-kai Liu
- Department of Anesthesiology, Linyi People’s Hospital, Linyi 276003, China
| | - Ke-zhong Li
- Department of Anesthesiology, the Second Hospital of Shandong University, Jinan 250033, China
| |
Collapse
|
80
|
Dual/multitargeted xanthone derivatives for Alzheimer's disease: where do we stand? Future Med Chem 2017; 9:1611-1630. [PMID: 28832188 DOI: 10.4155/fmc-2017-0086] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To date, the current therapy for Alzheimer's disease (AD) based on acetylcholinesterase inhibitors is only symptomatic, being its efficacy limited. Hence, the recent research has been focused in the development of different pharmacological approaches. Here we discuss the potential of xanthone derivatives as new anti-Alzheimer agents. The interference of xanthone derivatives with acetylcholinesterase and other molecular targets and cellular mechanisms associated with AD have been recently systematically reported. Therefore, we report xanthones with anticholinesterase, monoamine oxidase and amyloid β aggregation inhibitory activities as well as antioxidant properties, emphasizing xanthone derivatives with dual/multitarget activity as potential agents to treat AD. We also propose the structural features for these activities that may guide the design of new, more effective xanthone derivatives. [Formula: see text].
Collapse
|
81
|
Jiang L, Hu M, Lu Y, Cao Y, Chang Y, Dai Z. The protective effects of dexmedetomidine on ischemic brain injury: A meta-analysis. J Clin Anesth 2017. [DOI: 10.1016/j.jclinane.2017.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
82
|
Luo C, Ouyang MW, Fang YY, Li SJ, Zhou Q, Fan J, Qin ZS, Tao T. Dexmedetomidine Protects Mouse Brain from Ischemia-Reperfusion Injury via Inhibiting Neuronal Autophagy through Up-Regulating HIF-1α. Front Cell Neurosci 2017; 11:197. [PMID: 28729825 PMCID: PMC5498477 DOI: 10.3389/fncel.2017.00197] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/22/2017] [Indexed: 11/14/2022] Open
Abstract
Stroke is the leading cause of death in China and produces a heavy socio-economic burden in the past decades. Previous studies have shown that dexmedetomidine (DEX) is neuroprotective after cerebral ischemia. However, the role of autophagy during DEX-mediated neuroprotection after cerebral ischemia is still unknown. In this study, we found that post-conditioning with DEX and DEX+3-methyladenine (3-MA) (autophagy inhibitor) reduced brain infarct size and improved neurological deficits compared with DEX+RAPA (autophagy inducer) 24 h after transient middle cerebral artery artery occlusion (tMCAO) model in mice. DEX inhibited the neuronal autophagy in the peri-ischemic brain, and increased viability and decreased apoptosis of primary cultured neurons in oxygen-glucose deprivation (OGD) model. DEX induced expression of Bcl-1 and p62, while reduced the expression of microtubule-associated protein 1 light chain 3 (LC3) and Beclin 1 in primary cultured neurons through inhibition of apoptosis and autophagy. Meanwhile, DEX promoted the expression of hypoxia-inducible factor-1α (HIF-1α) both in vivo and in vitro, and 2-Methoxyestradiol (2ME2), an inhibitor of HIF-1α, could reverse DEX-induced autophagic inhibition. In conclusion, our study suggests that post-conditioning with DEX at the beginning of reperfusion protects mouse brain from ischemia-reperfusion injury via inhibition of neuronal autophagy by upregulation of HIF-1α, which provides a potential therapeutic treatment for acute ischemic injury.
Collapse
Affiliation(s)
- Cong Luo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Ming-Wen Ouyang
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical UniversityGuangzhou, China
| | - Ying-Ying Fang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou, China
| | - Shu-Ji Li
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou, China
| | - Quan Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Jun Fan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Zai-Sheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Tao Tao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| |
Collapse
|
83
|
Wu GJ, Chen JT, Tsai HC, Chen TL, Liu SH, Chen RM. Protection of Dexmedetomidine Against Ischemia/Reperfusion-Induced Apoptotic Insults to Neuronal Cells Occurs Via an Intrinsic Mitochondria-Dependent Pathway. J Cell Biochem 2017; 118:2635-2644. [DOI: 10.1002/jcb.25847] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 12/15/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
| | - Jui-Tai Chen
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Hsiao-Chien Tsai
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Ta-Liang Chen
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Ruei-Ming Chen
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
84
|
Akpınar O, Nazıroğlu M, Akpınar H. Different doses of dexmedetomidine reduce plasma cytokine production, brain oxidative injury, PARP and caspase expression levels but increase liver oxidative toxicity in cerebral ischemia-induced rats. Brain Res Bull 2017; 130:1-9. [DOI: 10.1016/j.brainresbull.2016.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/09/2016] [Indexed: 12/22/2022]
|
85
|
Hummitzsch L, Zitta K, Berndt R, Kott M, Schildhauer C, Parczany K, Steinfath M, Albrecht M. Doxycycline protects human intestinal cells from hypoxia/reoxygenation injury: Implications from an in-vitro hypoxia model. Exp Cell Res 2017; 353:109-114. [PMID: 28300560 DOI: 10.1016/j.yexcr.2017.03.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/29/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a grave clinical emergency and associated with high morbidity and mortality rates. Based on the complex underlying mechanisms, a multimodal pharmacological approach seems necessary to prevent intestinal I/R injury. The antibiotic drug doxycycline, which exhibits a wide range of pleiotropic therapeutic properties, might be a promising candidate for also reducing I/R injury in the intestine. To investigate possible protective effects of doxycycline on intestinal I/R injury, human intestinal CaCo-2 cells were exposed to doxycycline at clinically relevant concentrations. In order to mimic I/R injury, CaCo-2 were thereafter subjected to hypoxia/reoxygenation by using our recently described two-enzyme in-vitro hypoxia model. Investigations of cell morphology, cell damage, apoptosis and hydrogen peroxide formation were performed 24h after the hypoxic insult. Hypoxia/reoxygenation injury resulted in morphological signs of cell damage, elevated LDH concentrations in the respective culture media (P<0.001) and increased protein expression of proapoptotic caspase-3 (P<0.05) in the intestinal cultures. These events were associated with increased levels hydrogen peroxide (P<0.001). Preincubation of CaCo-2 cells with different concentrations of doxycycline (5µM, 10µM, 50µM) reduced the hypoxia induced signs of cell damage and LDH release (P<0.001 for all concentrations). The reduction of cellular damage was associated with a reduced expression of caspase-3 (5µM, P<0.01; 10µM, P<0.01; 50µM, P<0.05), while hydrogen peroxide levels remained unchanged. In summary, doxycycline protects human intestinal cells from hypoxia/reoxygenation injury in-vitro. Further animal and clinical studies are required to prove the protective potential of doxycycline on intestinal I/R injury under in-vivo conditions.
Collapse
Affiliation(s)
- Lars Hummitzsch
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Karina Zitta
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Rouven Berndt
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Matthias Kott
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Christin Schildhauer
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Kerstin Parczany
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Markus Steinfath
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Martin Albrecht
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Kiel, Germany.
| |
Collapse
|
86
|
Tang Q, Wu X, Weng W, Li H, Feng J, Mao Q, Gao G, Jiang J. The preventive effect of dexmedetomidine on paroxysmal sympathetic hyperactivity in severe traumatic brain injury patients who have undergone surgery: a retrospective study. PeerJ 2017; 5:e2986. [PMID: 28229021 PMCID: PMC5314954 DOI: 10.7717/peerj.2986] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 01/12/2017] [Indexed: 11/20/2022] Open
Abstract
Background Paroxysmal sympathetic hyperactivity (PSH) results and aggravates in secondary brain injury, which seriously affects the prognosis of severe traumatic brain injury patients. Although several studies have focused on the treatment of PSH, few have concentrated on its prevention. Methods Ninety post-operation (post-op) severe traumatic brain injury (sTBI) patients admitted from October 2014 to April 2016 were chosen to participate in this study. Fifty of the post-op sTBI patients were sedated with dexmedetomidine and were referred as the “dexmedetomidine group” (admitted from May 2015 to April 2016). The other 40 patients (admitted from October 2014 to May 2015) received other sedations and were referred as the “control group.” The two groups were then compared based on their PSH scores and the scores and ratios of those patients who met the criteria of “probable,” “possible” and “unlikely” using the PSH assessment measure (PSH-AM) designed by Baguley et al. (2014). The durations of the neurosurgery intensive care unit (NICU) and hospital stays and the Glasgow outcome scale (GOS) values for the two groups were also compared to evaluate the therapeutic effects and the patients’ prognosis. Results The overall PSH score for the dexmedetomidine group was 5.26 ± 4.66, compared with 8.58 ± 8.09 for the control group. The difference between the two groups’ PSH scores was significant (P = 0.017). The score of the patients who met the criterion of “probable” was 18.33 ± 1.53 in the dexmedetomidine group and 22.63 ± 2.97 in the control group, and the difference was statistically significant (P = 0.045). The ratio of patients who were classified as “unlikely” between the two groups was statistically significant (P = 0.028); that is, 42 (84%) in the dexmedetomidine group and 25 (62.5%) in the control group. The differences in NICU, hospital stays and GOS values between the two groups were not significant. Conclusion Dexmedetomidine has a preventive effect on PSH in sTBI patients who have undergone surgery.
Collapse
Affiliation(s)
- Qilin Tang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Xiang Wu
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Weiji Weng
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Hongpeng Li
- Department of Neurosurgery, Rizhao City Hospital of Traditional Chinese Medicine , Rizhao , Shandong Province , China
| | - Junfeng Feng
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Qing Mao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Guoyi Gao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| | - Jiyao Jiang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Head Trauma, Shanghai, China
| |
Collapse
|
87
|
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: 4.4] [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
|
88
|
Akpınar H, Nazıroğlu M, Övey İS, Çiğ B, Akpınar O. The neuroprotective action of dexmedetomidine on apoptosis, calcium entry and oxidative stress in cerebral ischemia-induced rats: Contribution of TRPM2 and TRPV1 channels. Sci Rep 2016; 6:37196. [PMID: 27872485 PMCID: PMC5131653 DOI: 10.1038/srep37196] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/25/2016] [Indexed: 01/18/2023] Open
Abstract
Dexmedetomidine (DEX) may act as an antioxidant through regulation of TRPM2 and TRPV1 channel activations in the neurons by reducing cerebral ischemia-induced oxidative stress and apoptosis. The neuroprotective roles of DEX were tested on cerebral ischemia (ISC) in the cultures of rat primary hippocampal and DRG neurons. Fifty-six rats were divided into five groups. A placebo was given to control, sham control, and ISC groups, respectively. In the third group, ISC was induced. The DEX and ISC+DEX groups received intraperitoneal DEX (40 μg/kg) 3, 24, and 48 hours after ISC induction. DEX effectively reversed capsaicin and cumene hydroperoxide/ADP-ribose-induced TRPV1 and TRPM2 densities and cytosolic calcium ion accumulation in the neurons, respectively. In addition, DEX completely reduced ISC-induced oxidative toxicity and apoptosis through intracellular reactive oxygen species production and depolarization of mitochondrial membrane. The DEX and ISC+DEX treatments also decreased the expression levels of caspase 3, caspase 9, and poly (ADP-ribose) polymerase in the hippocampus and DRG. In conclusion, the current results are the first to demonstrate the molecular level effects of DEX on TRPM2 and TRPV1 activation. Therefore, DEX can have remarkable neuroprotective impairment effects in the hippocampus and DRG of ISC-induced rats.
Collapse
Affiliation(s)
- Hatice Akpınar
- Unit of Anesthesiology and Reanimation, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Süleyman Demirel, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Neuroscience, Institute of Health Science, University of Süleyman Demirel, Isparta, Turkey.,Center of Neuroscience, University of Süleyman Demirel, Isparta, Turkey
| | - İshak Suat Övey
- Department of Neuroscience, Institute of Health Science, University of Süleyman Demirel, Isparta, Turkey
| | - Bilal Çiğ
- Department of Neuroscience, Institute of Health Science, University of Süleyman Demirel, Isparta, Turkey
| | - Orhan Akpınar
- Unit of Microbiology, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Süleyman Demirel, Isparta, Turkey
| |
Collapse
|
89
|
Souza-Dantas VC, Póvoa P, Bozza F, Soares M, Salluh J. Preventive strategies and potential therapeutic interventions for delirium in sepsis. Hosp Pract (1995) 2016; 44:190-202. [PMID: 27223862 DOI: 10.1080/21548331.2016.1192453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/18/2016] [Indexed: 06/05/2023]
Abstract
Delirium is the most frequent and severe clinical presentation of brain dysfunction in critically ill septic patients with an incidence ranging from 9% to 71%. Delirium represents a significant burden for patients and relatives, as well as to the health care system, resulting in higher costs, long-term cognitive impairment and significant risk of death after 6 months. Current interventions for the prevention of delirium typically involve early recognition and amelioration of modifiable risk factors and treatment of underlying conditions that predisposes the individual to delirium. Several pharmacological interventions to prevent and treat delirium have been tested, although their effectiveness remains uncertain, especially in larger and more homogeneous subgroups of ICU patients, like in patients with sepsis. To date, there is inconsistent and conflicting data regarding the efficacy of any particular pharmacological agent, thus substantial attention has been paid to non-pharmacological interventions and preventive strategies should be applied to every patient admitted in the ICU. Future trials should be designed to evaluate the impact of these pharmacologic interventions on the prevention and treatment of delirium on clinically relevant outcomes such as length of stay, hospital mortality and long-term cognitive function. The role of specific medications like statins in delirium prevention is also yet to be evaluated.
Collapse
Affiliation(s)
| | - Pedro Póvoa
- b Polyvalent Intensive Care Unit, Hospital S. Francisco Xavier , Centro Hospitalar de Lisboa Ocidental (CHLO) , Lisbon , Portugal
- c Nova Medical School , CEDOC, New University of Lisbon , Portugal
| | - Fernando Bozza
- d Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
- e D'Or Institute for Research and Education , Rio de Janeiro , Brazil
| | - Marcio Soares
- e D'Or Institute for Research and Education , Rio de Janeiro , Brazil
| | - Jorge Salluh
- e D'Or Institute for Research and Education , Rio de Janeiro , Brazil
| |
Collapse
|
90
|
Cheng XY, Gu XY, Gao Q, Zong QF, Li XH, Zhang Y. Effects of dexmedetomidine postconditioning on myocardial ischemia and the role of the PI3K/Akt-dependent signaling pathway in reperfusion injury. Mol Med Rep 2016; 14:797-803. [PMID: 27221008 PMCID: PMC4918562 DOI: 10.3892/mmr.2016.5345] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/13/2016] [Indexed: 11/18/2022] Open
Abstract
The present study aimed to determine whether post-ischemic treatment with dexmedetomidine (DEX) protected the heart against acute myocardial ischemia/reperfusion (I/R)-induced injury in rats. The phosphatidylinositol-3 kinase/protein kinase B(PI3K/Akt)-dependent signaling pathway was also investigated. Male Sprague Dawley rats (n=64) were subjected to ligation of the left anterior descending artery (LAD), which produced ischemia for 25 min, followed by reperfusion. Following LAD ligation, rats were treated with DEX (5, 10 and 20 µg/kg) or underwent post-ischemic conditioning, which included three cycles of ischemic insult. In order to determine the role of the PI3K/Akt signaling pathway, wortmannin (Wort), a PI3K inhibitor, was used to treat a group of rats that had also been treated with DEX (20 µg/kg). Post-reperfusion, lactate dehydrogenase (LDH), cardiac troponin I (cTnI), creatine kinase isoenzymes (CK-MB), superoxide dismutase (SOD) and malondialdehyde (MDA) serum levels were measured using an ultraviolet spectrophotometer. The protein expression levels of phosphorylated (p)-Akt, Ser9-p-glycogen synthase kinase-3β (p-GSK-3β) and cleaved caspase-3 were detected in heart tissue by western blotting. The mRNA expression levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were detected using reverse transcription-polymerase chain reaction. At the end of the experiment, the hearts were removed and perfused in an isolated perfusion heart apparatus with Evans blue (1%) in order to determine the non-ischemic areas. The risk and infarct areas of the heart were not dyed. As expected, I/R induced myocardial infarction, as determined by the increased serum levels of cTnI, CK-MB and MDA, and the decreased levels of SOD. Post-ischemic treatment with DEX increased the expression levels of p-Akt and p-GSK-3β, whereas caspase-3 expression was reduced following DEX treatment compared with in the I/R group. Compared with the I/R group, the ratio of Bcl-2/Bax at the mRNA level was elevated in the DEX and ischemic post-conditioning groups, whereas the expression levels of Bax were decreased. Conversely, the effects of DEX were attenuated by Wort. These results indicated that, similar to post-ischemic conditioning, post-ischemic treatment with DEX protects the heart against I/R via the PI3K/Akt-dependent signaling pathway, possibly by activating GSK-3β.
Collapse
Affiliation(s)
- Xiang Yang Cheng
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Xiao Yu Gu
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Qin Gao
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Qiao Feng Zong
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xiao Hong Li
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Ye Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
91
|
Kismet K, Sadic M, Bag YM, Atilgan HI, Koca G, Onalan AK, Senes M, Peker SA, Yumusak N, Korkmaz M. HEPATOPROTECTIVE EFFECT OF DEXMEDETOMIDINE AGAINST RADIOIODINE TOXICITY IN RATS: EVALUATION OF OXIDATIVE STATUS AND HISTOPATHOLOGICAL CHANGES. Int Surg 2016; 101:176-184. [PMID: 27018824 DOI: 10.9738/intsurg-d-15-00325.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE AND BACKGROUND Based on the anti-inflammatory, antioxidant and anti-apoptotic properties of DEX, the present study was conducted to investigate the possible radioprotective effects of DEX against hepatic radioiodine (I-131) toxicity. METHODS Thirty six rats were randomly divided into three groups as untreated control (group 1); oral radioiodine (RAI, 111 MBq) administrated rats (group 2), and DEX group (oral radioiodine and daily intraperitoneal 25 µg/kg DEX administrated rats-group 3). In the third group, DEX administration was started 2 days before and continued for five days after RAI administration. Twenty-four hours after the administration of the last dose of DEX, liver samples were taken for evaluation of oxidative stress parameters and histopathological changes. RESULTS The tissue malondialdehyde and advanced oxidation protein product levels in DEX group were significantly lower than RAI group. The total tissue sulphydryl and catalase levels of DEX group were higher than RAI group and the difference was statistically significant. The histopathological damage in the DEX-treated group was significantly less than the damage in the RAI group (p<0.05 for all pathological parameters). Treatment with DEX decreased the histopathological abnormalities when compared with the RAI group. CONCLUSION It was presented that DEX had radioprotective effect on the liver after I-131 therapy and anti-inflammatory and antioxidant activities are likely to be involved in the mechanism underlying the radioprotective effects of DEX. After further studies, DEX might be used as a hepatoprotective treatment regimen before administering radioactive iodine therapy particularly in patients with hepatic disease.
Collapse
Affiliation(s)
- Kemal Kismet
- 1 Ankara Education and Research Hospital, Department of General Surgery, Ankara, Turkey
| | - Murat Sadic
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Yusuf Murat Bag
- 1 Ankara Education and Research Hospital, Department of General Surgery, Ankara, Turkey
| | - Hasan Ikbal Atilgan
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Gokhan Koca
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Ali Kemal Onalan
- 3 Siirt State Hospital, Department of General Surgery, Siirt, Turkey
| | - Mehmet Senes
- 4 Ankara Education and Research Hospital, Department of Biochemistry, Ankara, Turkey
| | - Seydi Ali Peker
- 4 Ankara Education and Research Hospital, Department of Biochemistry, Ankara, Turkey
| | - Nihat Yumusak
- 5 Harran University, Faculty of Veterinary Medicine, Department of Pathology, Sanliurfa, Turkey
| | - Meliha Korkmaz
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| |
Collapse
|
92
|
Yu Y, Li M, Su N, Zhang Z, Zhao H, Yu H, Xu Y. Honokiol protects against renal ischemia/reperfusion injury via the suppression of oxidative stress, iNOS, inflammation and STAT3 in rats. Mol Med Rep 2015; 13:1353-60. [PMID: 26647858 DOI: 10.3892/mmr.2015.4660] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 10/06/2015] [Indexed: 11/05/2022] Open
Abstract
Honokiol is the predominant active ingredient in the commonly used traditional Chinese medicine, Magnolia, which has been confirmed in previous studies to exhibit anti-oxidation, antimicrobial, antitumor and other pharmacological effects. However, its effects on renal ischemia/reperfusion injury (IRI) remain to be elucidated. The present study aimed to examine the effects of honokiol on renal IRI, and to investigate its potential protective mechanisms in the heart. Male adult Wistar albino rats were induced into a renal IRI model. Subsequently, the levels of serum creatinine, blood urea nitrogen (BUN), alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP), and the levels of serum nitrite and the kidney nitrite were examined in the IRI group. The levels of oxidative stress, inducible nitric oxide synthase (iNOS), inflammatory factors and caspase-3 were evaluated using a series of commercially available kits. The levels of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) and the protein expression levels of STAT3 were determined using western blotting. Pretreatment with honokiol significantly reduced the levels of serum creatinine, BUN, ALT, AST and ALP, and the level of nitrite in the kidney of the IRI group, compared with the control group. The levels of malondialdehyde, the activity of myeloperoxidase, and the gene expression and activity of iNOS were reduced in the IRI rats, compared with the sham-operated rats, whereas the levels of superoxide dismutase and catalase were increased following treatment with honokiol in the IRI rats. In addition, the expression levels of tumor necrosis factor-α and interleukin-6 in the IRI rats were increased by honokiol. Treatment with honokiol suppressed the protein expression levels of p-STAT3 and caspase-3 in the IRI rats. These findings indicated that honokiol protects against renal IRI via the suppression of oxidative stress, iNOS, inflammation and STAT3 in the rat.
Collapse
Affiliation(s)
- Yongwu Yu
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Mingxv Li
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Ning Su
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Zhiyong Zhang
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Haidan Zhao
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Hai Yu
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| | - Yingluan Xu
- Department of Nephrology, Navy General Hospital of Chinese People's Liberation Army, Beijing 100048, P.R. China
| |
Collapse
|
93
|
Dexmedetomidine protects the heart against ischemia-reperfusion injury by an endothelial eNOS/NO dependent mechanism. Pharmacol Res 2015; 103:318-27. [PMID: 26607864 DOI: 10.1016/j.phrs.2015.11.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/27/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023]
Abstract
The alpha2-adrenergic receptor agonist Dexmedetomidine (Dex) is a sedative medication used by anesthesiologists. Dex protects the heart against ischemia-reperfusion (IR) and can also act as a preconditioning mimetic. The mechanisms involved in Dex-dependent cardiac preconditioning, and whether this action occurs directly or indirectly on cardiomyocytes, still remain unclear. The endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway and endothelial cells are known to play key roles in cardioprotection against IR injury. Therefore, the aims of this work were to evaluate whether the eNOS/NO pathway mediates the pharmacological cardiac effect of Dex, and whether endothelial cells are required in this cardioprotective action. Isolated adult rat hearts were treated with Dex (10nM) for 25min and the dimerization of eNOS and production of NO were measured. Hearts were then subjected to global IR (30/120min) and the role of the eNOS/NO pathway was evaluated. Dex promoted the activation of eNOS and production of NO. Dex reduced the infarct size and improved the left ventricle function recovery, but this effect was reversed when Dex was co-administered with inhibitors of the eNOS/NO/PKG pathway. In addition, Dex was unable to reduce cell death in isolated adult rat cardiomyocytes subjected to simulated IR. Cardiomyocyte death was attenuated by co-culturing them with endothelial cells pre-treated with Dex. In summary, our results show that Dex triggers cardiac protection by activating the eNOS/NO signaling pathway. This pharmacological effect of Dex requires its interaction with the endothelium.
Collapse
|
94
|
Kip G, Çelik A, Bilge M, Alkan M, Kiraz HA, Özer A, Şıvgın V, Erdem Ö, Arslan M, Kavutçu M. Dexmedetomidine protects from post-myocardial ischaemia reperfusion lung damage in diabetic rats. Libyan J Med 2015; 10:27828. [PMID: 26387799 PMCID: PMC4576509 DOI: 10.3402/ljm.v10.27828] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/14/2015] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE Diabetic complications and lipid peroxidation are known to have a close association. Lipid peroxidation commonly occurs at sites exposed to ischaemia, but distant organs and tissues also get damaged during ischaemia/reperfusion (I/R). Some of these targets are vital organs, such as the lung, liver, and kidney; the lung is the most frequently affected. The aim of our study was to investigate the effects of dexmedetomidine on I/R damage in lung tissue and on the oxidant/anti-oxidant system in diabetic rats. MATERIAL AND METHODS Diabetes was induced with streptozotocin (55 mg/kg) in 18 Wistar Albino rats, which were then randomly divided into three groups (diabetes control (DC), diabetes plus ischaemia-reperfusion (DIR), and diabetes plus dexmedetomidine-ischaemia/reperfusion (DIRD)) after the effects of diabetes were clearly evident. The rats underwent a left thoracotomy and then ischaemia was produced in the myocardium muscle by a left anterior descending artery ligation for 30 min in the DIR and DIRD groups. I/R was performed for 120 min. The DIRD group received a single intraperitoneal dose of dexmedetomidine (100 µg/kg); the DIR group received no dexmedetomidine. Group DC was evaluated as the diabetic control group and also included six rats (C group) in which diabetes was not induced. These mice underwent only left thoracotomy and were closed without undergoing myocardial ischaemia. Histopathological changes, activities of catalase (CAT) and glutathione-S-transferase anti-oxidant enzymes, and malondialdehyde (MDA) levels were evaluated in the lung tissues of all rats. RESULTS Neutrophil infiltration/aggregation was higher in the DIR group than in the C, DC, and DIRD groups (p=0.001, p=0.013, and p=0.042, respectively). The lung injury score was significantly higher in the DIR group than in the C and DC groups (p<0.0001 and p=0.024, respectively). The levels of MDA were significantly higher in the DIR group than in the C and DIRD groups. CAT activity was significantly higher in the DIR group than in the DIRD and C groups. CONCLUSION Our results confirm that dexmedetomidine has protective effects against the lung damage resulting from I/R in diabetic rats. Future studies conducted to evaluate the effects of the use of dexmedetomidine on damage to various organs following different I/R durations may help understanding possible protective effects of dexmedetomidine and underlying mechanisms in tissue damage related to I/R injury.
Collapse
Affiliation(s)
- Gülay Kip
- Department of Pediatric Dentistry, Faculty of Dentistry, Gazi University, Ankara, Turkey
| | - Ali Çelik
- Department of Thoracic Surgery, School of Medicine, Gazi University, Ankara, Turkey
| | - Mustafa Bilge
- Department of Biochemistry, School of Medicine, Gazi University, Ankara, Turkey
| | - Metin Alkan
- Department of Anestesiology and Reanimation, School of Medicine, Gazi University, Ankara, Turkey
| | - Hasan Ali Kiraz
- Department of Anestesiology and Reanimation, School of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Abdullah Özer
- Department of Cardiovascular Surgery, School of Medicine, Gazi University, Ankara, Turkey
| | - Volkan Şıvgın
- Department of Anestesiology and Reanimation, School of Medicine, Gazi University, Ankara, Turkey
| | - Özlem Erdem
- Department of Pathology, School of Medicine, Gazi University, Ankara, Turkey
| | - Mustafa Arslan
- Department of Anestesiology and Reanimation, School of Medicine, Gazi University, Ankara, Turkey;
| | - Mustafa Kavutçu
- Department of Biochemistry, School of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
95
|
Yang P, Yang N, Zhang X, Xu X. The significance and mechanism of propofol on treatment of ischemia reperfusion induced lung injury in rats. Cell Biochem Biophys 2015; 70:1527-32. [PMID: 25074530 DOI: 10.1007/s12013-014-0088-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This study is aimed to investigate the efficacy and underlying the mechanism of propofol in treatment of ischemia reperfusion (IR)-induced lung injury in rats, providing a novel insight of therapeutic strategy for IR-induced lung injury. 120 healthy SD rats were selected and randomly divided into sham operation group, IR group, and propofol group (40 rats per group). Bronchoalveolar lavage fluid (BALF) protein content, serum protein content, lung permeability index, lung water content rate, methane dicarboxylic aldehyde (MDA) in lung tissue, superoxide dismutase (SOD), nitric oxide (NO), endothelin (ET-1), toll-like receptor 4 (TLR4), nuclear factor (NF-κB), and tumor necrosis factor-α (TNF-α) were examined and compared among different groups to evaluate the therapeutical effects of propofol on IR-induced lung injury and analyze the mechanism. In sham operation group, neither change in lung tissue nor pulmonary interstitial edema or alveolar wall damage was found under microscope; in IR group, marked pulmonary interstitial edema and alveolar wall damage complicated with inflammatory cell infiltration and hemorrhage were found; in propofol group, alveolar wall widening was observed, however, hemorrhage in alveolar cavity, inflammatory infiltration and tissue damage were less significant than in IR group. At 3 h after reperfusion, BALF protein content, lung permeability index, and lung water content rate were all significantly increased in IR group and propofol group, while the serum protein content was significantly lower than sham operation group (p < 0.05). Moreover, we found that the change of above parameters in propofol group was less significant than in IR group (p < 0.05). No statistically significant difference was found in ET-1 levels in different groups (p > 0.05). In contrast, MDA and NO in IR group and propofol group were significantly increased, while SOD activity was significantly decreased (p < 0.05). Furthermore, the change of above parameters in propofol group was less significant than in IR group (p < 0.05). In addition, mRNAs of TLR4, NF-κB, and TNF-α were significantly increased in IR group and propofol group (p < 0.05) with more significant change in IR group compared with propofol group (p < 0.05). Propofol has protective effects against IR-induced lung injury by improving activity of oxygen radical and restoring NO/ET-1 dynamic balance. Besides, regulation of TLR4, NF-κB, and TNF-α by propofol also play important role in alleviating IR-induced lung injury.
Collapse
Affiliation(s)
- Pei Yang
- Department of Anesthesiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | | | | | | |
Collapse
|
96
|
Dexmedetomidine Dose-Dependently Attenuates Ropivacaine-Induced Seizures and Negative Emotions Via Inhibiting Phosphorylation of Amygdala Extracellular Signal-Regulated Kinase in Mice. Mol Neurobiol 2015; 53:2636-46. [PMID: 26099305 DOI: 10.1007/s12035-015-9276-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/01/2015] [Indexed: 02/07/2023]
Abstract
Ropivacaine (Ropi), one of the newest and safest amino amide local anesthetics, is linked to toxicity, including the potential for seizures, changes in behavior, and even cardiovascular collapse. Dexmedetomidine (Dex), an α2-adrenergic receptor agonist, has been widely used in anesthesia and critical care practice. To date, the underlying mechanisms of the effects of Dex premedication on Ropi-induced toxicity have not been clearly identified. In the current study, we investigated the effects of increasing doses of Dex premedication on 50% convulsive dose (CD50) of Ropi. With increasing doses of intraperitoneal (i.p.) Dex 10 min prior to each i.p. RopiCD50, the latency and duration of seizure activity were recorded. Open-field (OF) and elevated plus maze (EPM) test were used to measure negative behavioral emotions such as depression and anxiety. Immunohistochemistry and Western blot were utilized to investigate phosphorylation-extracellular regulated protein kinases (p-ERK) expression in the basolateral amygdala (BLA) on 2 h and in the central amygdala (CeA) on 24 h after convulsion in mice. The results of our investigation demonstrated that Dex dose-dependently increased RopiCD50, prolonged the latency and shortened the duration of each RopiCD50-induced seizure, improved the negative emotions revealed by both OF and EPM test, and inhibited p-ERK expression in the BLA and the CeA.
Collapse
|
97
|
Mason KP, Lönnqvist PA. Bradycardia in perspective-not all reductions in heart rate need immediate intervention. Paediatr Anaesth 2015; 25:44-51. [PMID: 25410284 DOI: 10.1111/pan.12584] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2014] [Indexed: 12/22/2022]
Abstract
According to Wikipedia, the word 'bradycardia' stems from the Greek βραδύς, bradys, 'slow', and καρδία, kardia, 'heart'. Thus, the meaning of bradycardia is slow heart rate but not necessarily too slow heart rate. If looking at top endurance athletes they may have a resting heart rate in the very low thirties without needing emergent intervention with anticholinergics, isoprenaline, epinephrine, chest compressions or the insertion of an emergency pacemaker (Figure 1). In fact, they withstand these episodes without incident, accommodating with a compensatory increase in stroke volume to preserve and maintain cardiac output. With this in mind, it is difficult for the authors to fully understand and agree with the general sentiment amongst many pediatric anesthesiologists that all isolated bradycardia portends impending doom and must be immediately treated with resuscitative measures.
Collapse
Affiliation(s)
- Keira P Mason
- Department of Anaesthesia, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | | |
Collapse
|
98
|
Ai F, Chen M, Li W, Yang Y, Xu G, Gui F, Liu Z, Bai X, Chen Z. Protective role of Klotho on cardiomyocytes upon hypoxia/reoxygenation via downregulation of Akt and FOXO1 phosphorylation. Mol Med Rep 2014; 11:2013-9. [PMID: 25377663 DOI: 10.3892/mmr.2014.2899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/05/2014] [Indexed: 11/05/2022] Open
Abstract
Klotho is a novel anti-aging hormone involved in human coronary artery disease. The present study aimed to detect the effects and mechanism of Klotho on cardiomyocytes in a hypoxia/reoxygenation (H/R) model in vitro. Neonatal Sprague-Dawley rat cardiomyocytes were randomly distributed into experimental groups as follows: Control group; H/R group, 4‑h hypoxia followed by 3‑h reoxygenation; and H/R+Klotho group, incubated with 0.1, 0.2 or 0.4 µg/ml Klotho protein for 16 h and then subjected to 4‑h hypoxia/3‑h reoxygenation. In order to evaluate cardiomyocyte damage, cell viability and lactate dehydrogenase (LDH) levels were measured. Cell apoptosis was measured by flow cytometry. The 2',7'-dichlorofluorescein diacetate reagent was used to estimate the intracellular generation of reactive oxygen species (ROS). Immunofluorescence staining was used to test whether Klotho induced decreased nuclear translocation of forkhead box protein O1 (FOXO1). Western blot analysis was performed to detect protein levels of FOXO1, phospho-FOXO1, Akt, phospho-Akt and superoxide dismutase 2 (SOD2). Cell viability was significantly decreased, levels of LDH in the cardiomyocyte culture medium were significantly increased and the apoptotic rate was enhanced in the H/R group when compared with those of the control group. Compared with the H/R group, cell viability of the H/R+Klotho groups was significantly higher (P<0.05). Treatment with Klotho protein resulted in a significant resistance of cardiomyocytes to apoptosis and the release of LDH was decreased. Intracellular ROS levels in the H/R group were significantly elevated above those of the control group (P<0.05). Following treatment with Klotho, intracellular ROS levels were significantly decreased compared with those of the H/R group (P<0.05). Western blot analysis confirmed that Klotho protein treatment increased FOXO1 levels in the nucleus and decreased FOXO1 levels in the cytoplasm. Furthermore, exogenous Klotho protein promoted translocation of FOXO1 from cytoplasm to nucleus. In addition, the administration of Klotho protein suppressed phosphorylation of FOXO1 and Akt, and markedly increased the protein expression levels of SOD2. In conclusion, treatment with Klotho protein had beneficial effects on cardiomyocytes undergoing H/R injury. The mechanism of this effect may be associated with suppressed apoptosis of cardiomyocytes, inhibition of phosphorylation of FOXO1 and Akt as well as suppression of cytoplasm transfer of FOXO1.
Collapse
Affiliation(s)
- Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Wei Li
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Yang
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Guizhong Xu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhenxing Liu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xiangyan Bai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
99
|
Wang H, Hu B, Zou Y, Bo L, Wang J, Li J, Luo Y. Dexmedetomidine premedication attenuates concanavalin A-induced hepatitis in mice. J Toxicol Sci 2014; 39:755-64. [PMID: 25242406 DOI: 10.2131/jts.39.755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Haibin Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, China
| | - Baoji Hu
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, China
| | - Yun Zou
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Lulong Bo
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Jun Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Jinbao Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, China
| |
Collapse
|