1
|
Liang ZK, Xiong W, Wang C, Chen L, Zou X, Mai JW, Dong B, Guo C, Xin WJ, Luo DX, Xu T, Feng X. Resolving neuroinflammatory and social deficits in ASD model mice: Dexmedetomidine downregulates NF-κB/IL-6 pathway via α2AR. Brain Behav Immun 2024; 119:84-95. [PMID: 38552922 DOI: 10.1016/j.bbi.2024.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that severely affects individuals' daily life and social development. Unfortunately, there are currently no effective treatments for ASD. Dexmedetomidine (DEX) is a selective agonist of α2 adrenergic receptor (α2AR) and is widely used as a first-line medication for sedation and hypnosis in clinical practice. In recent years, there have been reports suggesting its potential positive effects on improving emotional and cognitive functions. However, whether dexmedetomidine has therapeutic effects on the core symptoms of ASD, namely social deficits and repetitive behaviors, remains to be investigated. In the present study, we employed various behavioral tests to assess the phenotypes of animals, including the three-chamber, self-grooming, marble burying, open field, and elevated plus maze. Additionally, electrophysiological recordings, western blotting, qPCR were mainly used to investigate and validate the potential mechanisms underlying the role of dexmedetomidine. We found that intraperitoneal injection of dexmedetomidine in ASD model mice-BTBR T+ Itpr3tf/J (BTBR) mice could adaptively improve their social deficits. Further, we observed a significant reduction in c-Fos positive signals and interleukin-6 (IL-6) expression level in the prelimbic cortex (PrL) of the BTBR mice treated with dexmedetomidine. Enhancing or inhibiting the action of IL-6 directly affects the social behavior of BTBR mice. Mechanistically, we have found that NF-κB p65 is a key pathway regulating IL-6 expression in the PrL region. In addition, we have confirmed that the α2AR acts as a receptor switch mediating the beneficial effects of dexmedetomidine in improving social deficits. This study provides the first evidence of the beneficial effects of dexmedetomidine on core symptoms of ASD and offers a theoretical basis and potential therapeutic approach for the clinical treatment of ASD.
Collapse
Affiliation(s)
- Zheng-Kai Liang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Wei Xiong
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Chen Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Li Chen
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Xin Zou
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Jing-Wen Mai
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516000, PR China
| | - Bo Dong
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Chongqi Guo
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Wen-Jun Xin
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China
| | - De-Xing Luo
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516000, PR China.
| | - Ting Xu
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, PR China.
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
2
|
The Role of Dexmedetomidine in Hepatic Ischemia-Reperfusion Injury Via a Nitric Oxide-Dependent Mechanism in Rats. Transplant Proc 2021; 53:2060-2069. [PMID: 34238590 DOI: 10.1016/j.transproceed.2021.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/20/2021] [Accepted: 05/04/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Dexmedetomidine is known to protect against ischemia-reperfusion (IR) in various organs; however, the mechanisms of dexmedetomidine in the liver remain unclear. We investigated whether dexmedetomidine preconditioning leads to hepatic protection and whether nitric oxide was associated with this protective mechanism by employing N-nitro-l-arginine methyl ester (l-NAME), a nitrous oxide synthase inhibitor. METHODS Experiment 1 included 24 rats in 4 groups: sham, IR, 30 μg/kg of dexmedetomidine, and 50 μg/kg of dexmedetomidine. Experiment 2 included 36 rats in 6 groups: IR, 50 μg/kg of dexmedetomidine, 10 mg/kg of l-NAME, 10 mg/kg of l-NAME + 50 μg/kg of dexmedetomidine, 30 of mg/kg l-NAME, and 30 mg/kg of l-NAME + 50 μg/kg of dexmedetomidine. All drugs were administered intraperitoneally. The levels of serum transaminases, malondialdehyde, superoxide dismutase, tumor necrosis factor-α, nuclear factor-κB, and c-Jun N-terminal kinase were measured 6 hours after hepatic surgery. RESULTS Dexmedetomidine demonstrated a dose-dependent decrease in serum transaminase levels. The 50-μg/kg dexmedetomidine group showed a significant decrease in malondialdehyde levels (P = .002), increase in superoxide dismutase levels (P = .002), and a significantly lower level of phosphorylated tumor necrosis factor-α, nuclear factor-κB, and c-Jun N-terminal kinase (P = .002, respectively) compared with the IR injury group. These protective effects of dexmedetomidine were partially reversed by pretreatment with l-NAME (P < .01 for 20 and 30 mg/kg of l-NAME). CONCLUSION In hepatic IR injury, dexmedetomidine might protect the liver via antioxidative and anti-inflammatory responses, and nitric oxide production could play a role in these protective mechanisms.
Collapse
|
3
|
Zhang Y, Tan SL, Du J, Chen Y, Jia J, Feng JG, Liu KX, Zhou J. Dexmedetomidine alleviates neuroinflammation, restores sleep disorders and neurobehavioral abnormalities in rats with minimal hepatic encephalopathy. Int Immunopharmacol 2021; 96:107795. [PMID: 34162157 DOI: 10.1016/j.intimp.2021.107795] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/05/2021] [Accepted: 05/16/2021] [Indexed: 10/21/2022]
Abstract
The occurrence and progress of minimal hepatic encephalopathy (MHE) is closely related to the inflammatory response; however, inflammation contributes to behavioral abnormalities and sleep disorders. Dexmedetomidine has anti-inflammatory effects against various diseases. Whether dexmedetomidine improves MHE and the underlying mechanism is yet unclear. The present study aimed to explore the effects of dexmedetomidine on sleep structure, neurobehavior, and brain morphology of MHE rats and investigate its underlying mechanism. A rat MHE model was established by intraperitoneal injection of thioacetamide (TAA). Dexmedetomidine or yohimbine was administered intraperitoneally to investigate the role of α2 adrenoreceptor in the protection conferred by dexmedetomidine. The 24-h sleep, neurobehavioral changes, the liver function, blood ammonia and morphological changes of the liver and brain were assessed. Also, the microglia, astrocytes, neurons, the expression of pro-inflammatory factors (IL-1β, TNF-α, IL-18), and NLRP3 inflammasomes were detected. The results showed that marked sleep disorders, cognitive impairment, anxiety, abnormal liver function and pathological damage of liver and brain were detected in the MHE rats. The microglia in the prefrontal cortex was highly activated along with the increased expression of pro-inflammatory factors and NLRP3 inflammasomes. Interestingly, dexmedetomidine improved above indicators, however, yohimbine significantly abolished the protection of dexmedetomidine. These findings showed that dexmedetomidine restored the changes in the sleep disorders and neurobehavior in rats and reduced brain damage. The mechanism might be partially related to the activation of α2 adrenergic receptors, reduction of neuroinflammatory response, and inhibition of the activation of microglia and NLRP3/Caspase1 signaling pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Su-Lan Tan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Juan Du
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China.
| |
Collapse
|
4
|
Zhang W, Yu J, Guo M, Ren B, Tian Y, Hu Q, Xie Q, Xu C, Feng Z. Dexmedetomidine Attenuates Glutamate-Induced Cytotoxicity by Inhibiting the Mitochondrial-Mediated Apoptotic Pathway. Med Sci Monit 2020; 26:e922139. [PMID: 32419697 PMCID: PMC7251967 DOI: 10.12659/msm.922139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Glutamate (GLU) is the most excitatory amino acid in the central nervous system and plays an important role in maintaining the normal function of the nervous system. During cerebral ischemia, massive release of GLU leads to neuronal necrosis and apoptosis. It has been reported that dexmedetomidine (DEX) possesses anti-oxidant and anti-apoptotic properties. The objective of this study was to investigate the effects of DEX on GLU-induced neurotoxicity in PC12 cells. Material/Methods PC12 cells were treated with 20 mM GLU to establish an ischemia-induced injury model. Cell viability was accessed by MTT assay. MDA content and SOD activity were analyzed by assay kits. Apoptosis rate, ROS production, intracellular Ca2+ concentration, and MMP were evaluated by flow cytometry. Western blot analysis was performed to analyze expressions of caspase-3, caspase-9, cyt-c, bax, and bcl-2. Results PC12 cells treated with GLU exhibited reduced cell viability and increased apoptosis rates, which were ameliorated by pretreatment with DEX. DEX significantly increased SOD activity, reduced content of MDA, and decreased production of ROS in PC12 cells. In addition, DEX clearly reduced the level of intracellular Ca2+ and attenuated the decline of MMP. Moreover, DEX notably reduced expressions of caspase-3, caspase-9, cyt-c, and bax and increased expression of bcl-2. Conclusions Our findings suggest that DEX can protect PC12 cells against GLU-induced cytotoxicity, which may be attributed to its anti-oxidative property and reduction of intracellular calcium overload, as well as its ability to inhibit the mitochondria-mediated apoptotic pathway.
Collapse
Affiliation(s)
- Weidong Zhang
- Anesthesia and Operation Center, The First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland).,Anesthesia and Operation Center, The Fifth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Jun Yu
- Department of Anesthesiology, The Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Mengzhuo Guo
- Department of Anesthesiology, Beijing Tsinghua Changung Hospital, Beijing, China (mainland)
| | - Bo Ren
- Anesthesia and Operation Center, The Fifth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Yanyan Tian
- Department of Anesthesiology, Air Force Characteristic Medical Center, Beijing, China (mainland)
| | - Qinggang Hu
- Department of Anesthesiology, The Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Qun Xie
- Department of Anesthesiology, The Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Chen Xu
- Anesthesia and Operation Center, The Fifth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Zeguo Feng
- Anesthesia and Operation Center, The First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| |
Collapse
|
5
|
Gumbert SD, Kork F, Jackson ML, Vanga N, Ghebremichael SJ, Wang CY, Eltzschig HK. Perioperative Acute Kidney Injury. Anesthesiology 2020; 132:180-204. [PMID: 31687986 PMCID: PMC10924686 DOI: 10.1097/aln.0000000000002968] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Perioperative organ injury is among the leading causes of morbidity and mortality of surgical patients. Among different types of perioperative organ injury, acute kidney injury occurs particularly frequently and has an exceptionally detrimental effect on surgical outcomes. Currently, acute kidney injury is most commonly diagnosed by assessing increases in serum creatinine concentration or decreased urine output. Recently, novel biomarkers have become a focus of translational research for improving timely detection and prognosis for acute kidney injury. However, specificity and timing of biomarker release continue to present challenges to their integration into existing diagnostic regimens. Despite many clinical trials using various pharmacologic or nonpharmacologic interventions, reliable means to prevent or reverse acute kidney injury are still lacking. Nevertheless, several recent randomized multicenter trials provide new insights into renal replacement strategies, composition of intravenous fluid replacement, goal-directed fluid therapy, or remote ischemic preconditioning in their impact on perioperative acute kidney injury. This review provides an update on the latest progress toward the understanding of disease mechanism, diagnosis, and managing perioperative acute kidney injury, as well as highlights areas of ongoing research efforts for preventing and treating acute kidney injury in surgical patients.
Collapse
Affiliation(s)
- Sam D. Gumbert
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Felix Kork
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Maisie L. Jackson
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Naveen Vanga
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Semhar J. Ghebremichael
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Christy Y. Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
6
|
|
7
|
Gao Y, Yin H, Zhang Y, Dong Y, Yang F, Wu X, Liu H. Dexmedetomidine protects hippocampal neurons against hypoxia/reoxygenation-induced apoptosis through activation HIF-1α/p53 signaling. Life Sci 2019; 232:116611. [PMID: 31260683 DOI: 10.1016/j.lfs.2019.116611] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE To observe the effect of dexmedetomidine (DEX) on mitochondrial apoptosis of hippocampal neurons in hypoxia/reoxygenation (H/R) brain injury in developing rats, and to investigate its regulatory mechanism on HIF-1α/p53 signaling pathway. METHODS Hypoxia/reoxygenation model was used in this study. TUNEL assay was performed to detect cell apoptosis. Immunohistochemical analysis and Western-blotting analysis were conducted to detect Cytochrome-C (Cyt-c), APAF-1, Caspase-3, Neuroglobin (Ngb), HIF-1α and p53 expression. After 28 days, Morris water maze (MWM) was performed. RESULTS 50 μg/kg DEX improved H/R-induced brain injury and inhibited mitochondrial apoptosis in rats. Western-blotting and Immunohistochemical results demonstrated that DEX could up-regulate Ngb through α2 receptor to inhibit H/R-induced mitochondrial apoptosis. In addition, by adding inhibitors yohimbine and 2-methoxyestradiol (2ME2), we found that DEX could activate HIF-1α/p53 signaling pathway. MWM test showed that DEX could enhance long-term learning and memory of H/R brain injury rats. CONCLUSION DEX alleviates H/R-induced brain injury and mitochondrial apoptosis in developing rats through α2 receptor, which may be related to activation of HIF-1α/p53 signaling pathway to up-regulate the expression of Ngb.
Collapse
Affiliation(s)
- Yan Gao
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China; Department of Anesthesiology, The First Affiliated Hospital of Hebei North University Hebei, Zhangjiakou 075000, PR China
| | - Hong Yin
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China
| | - Yongfang Zhang
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China
| | - Yunxia Dong
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China
| | - Fan Yang
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China
| | - Hongtao Liu
- Department of Anesthesiology, Shengjing Hospital, China Medical University Shenyang, 110004, Liaoning, PR China.
| |
Collapse
|
8
|
Ge Y, Li Q, Nie Y, Gao J, Luo K, Fang X, Wang C. Dexmedetomidine improves cognition after carotid endarterectomy by inhibiting cerebral inflammation and enhancing brain-derived neurotrophic factor expression. J Int Med Res 2019; 47:2471-2482. [PMID: 31014147 PMCID: PMC6567697 DOI: 10.1177/0300060519843738] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objectives Carotid endarterectomy (CEA) is efficient in preventing stroke for patients with significant carotid stenosis, but results in mild cognitive dysfunction. Dexmedetomidine is neuroprotective in stroke models. We hypothesized that dexmedetomidine may improve cognition after CEA. Methods Forty-nine patients scheduled for elective CEA were randomly assigned to intravenous dexmedetomidine treatment group (n = 25) and control group C (normal saline, n = 24). Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MOCA), as well as lactate, TNF-α, IL-6, and BDNF levels in blood, were assessed before, during, and after surgery. Results MMSE and MOCA scores showed subtle decline in both groups at 24 hours postoperatively; this decline remained at 48 hours postoperatively in group C. Both scores were higher in group D than in group C at 48 and 72 hours postoperatively. TNF-α and IL-6 were lower from 5 minutes post-clamping through 24 hours postoperatively in group D; lactate was lower at 5 minutes post-clamping in group D. BDNF was higher from 5 minutes post-clamping through 1 hour postoperatively in both groups, and remained high in group D at 24 hours postoperatively. Conclusions Dexmedetomidine improved recovery of cognition after CEA, potentially due to reduced inflammation and enhanced BDNF expression.
Collapse
Affiliation(s)
- Yali Ge
- 1 Department of Anesthesiology, Northern Jiangsu People's Hospital, Yangzhou City, Jiangsu Province, China
| | - Qian Li
- 2 Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yuyan Nie
- 2 Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Ju Gao
- 1 Department of Anesthesiology, Northern Jiangsu People's Hospital, Yangzhou City, Jiangsu Province, China
| | - Ke Luo
- 3 Department of Anesthesiology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiangzhi Fang
- 1 Department of Anesthesiology, Northern Jiangsu People's Hospital, Yangzhou City, Jiangsu Province, China
| | - Cunjing Wang
- 1 Department of Anesthesiology, Northern Jiangsu People's Hospital, Yangzhou City, Jiangsu Province, China
| |
Collapse
|
9
|
Goyagi T. Dexmedetomidine reduced sevoflurane-induced neurodegeneration and long-term memory deficits in neonatal rats. Int J Dev Neurosci 2019; 75:19-26. [PMID: 30959098 DOI: 10.1016/j.ijdevneu.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
Exposure to sevoflurane and other inhalational anesthetics can induce neurodegeneration in the developing brain. Although dexmedetomidine (DEX) has provided neuroprotection against hypoxic ischemic injury, relatively little is known about whether it has the neuroprotective effects against anesthetic-induced neurodegeneration. This study examined whether DEX improves the long-term cognitive dysfunction observed after exposure of neonatal rats to 3% sevoflurane. Seven-day-old rats received intraperitoneal saline (DEX 0) or DEX (6.6, 12.5, 25 μg/kg) 30 min before exposure to 3% sevoflurane with 21% oxygen for 4 h (n = 10 per group). The pups in the control group received only DEX 25 μg/kg without anesthesia. The escape latency in the Morris water maze was significantly increased in the DEX 0 group compared with the sham and control group, and the escape latency, but not the swimming path length, was significantly shorter at post-natal day 47 in the DEX 25 than in the DEX 0 group. The percent time spent in the quadrant was significantly decreased in the DEX 0 group compared with the sham and control group, and the percent time spent in the quadrant was significantly increased in the DEX 25 group compared with the DEX 0 groups. The freezing times of the DEX 0 and 6.6 groups were significantly decreased compared with those in the sham, control and DEX 25 groups. The number of NeuN-positive cells in the CA1 region was significantly decreased in the DEX 0 and 6.6 groups compared with the sham, control and DEX 25 groups. These findings indicate pre-treatment with DEX may improve long-term cognitive function and ameliorate the neuronal degeneration induced by sevoflurane exposure in neonatal rats.
Collapse
Affiliation(s)
- Toru Goyagi
- Department of Anesthesia and Intensive Care Medicine, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita 010-8543, Japan.
| |
Collapse
|
10
|
Davidson JO, Dhillon SK, Wassink G, Zhou KQ, Bennet L, Gunn AJ. Endogenous neuroprotection after perinatal hypoxia-ischaemia: the resilient developing brain. J R Soc N Z 2018. [DOI: 10.1080/03036758.2018.1529685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Joanne O. Davidson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Simerdeep K. Dhillon
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kelly Q. Zhou
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Alistair J. Gunn
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Han XR, Wen X, Wang YJ, Wang S, Shen M, Zhang ZF, Fan SH, Shan Q, Wang L, Li MQ, Hu B, Sun CH, Wu DM, Lu J, Zheng YL. MicroRNA-140-5p elevates cerebral protection of dexmedetomidine against hypoxic-ischaemic brain damage via the Wnt/β-catenin signalling pathway. J Cell Mol Med 2018. [PMID: 29536658 PMCID: PMC5980153 DOI: 10.1111/jcmm.13597] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hypoxia–ischaemia (HI) remains a major cause of foetal brain damage presented a scarcity of effective therapeutic approaches. Dexmedetomidine (DEX) and microRNA‐140‐5p (miR‐140‐5p) have been highlighted due to its potentially significant role in the treatment of cerebral ischaemia. This study was to investigate the role by which miR‐140‐5p provides cerebral protection using DEX to treat hypoxic–ischaemic brain damage (HIBD) in neonatal rats via the Wnt/β‐catenin signalling pathway. The HIBD rat models were established and allocated into various groups with different treatment plans, and eight SD rats into sham group. The learning and memory ability of the rats was assessed. Apoptosis and pathological changes in the hippocampus CA1 region and expressions of the related genes of the Wnt/β‐catenin signalling pathway as well as the genes responsible of apoptosis were detected. Compared with the sham group, the parameters of weight, length growth, weight ratio between hemispheres, the rate of reaching standard, as well as Bcl‐2 expressions, were all increased. Furthermore, observations of increased levels of cerebral infarction volume, total mortality rate, response times, total response duration, expressions of Wnt1, β‐catenin, TCF‐4, E‐cadherin, apoptosis rate of neurons, and Bax expression were elevated. Following DEX treatment, the symptoms exhibited by HIBD rats were ameliorated. miR‐140‐5p and si‐Wnt1 were noted to attenuate the progression of HIBD. Our study demonstrates that miR‐140‐5p promotes the cerebral protective effects of DEX against HIBD in neonatal rats by targeting the Wnt1 gene through via the negative regulation of the Wnt/β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Liang Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Chun-Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
12
|
Lv M, Zeng H, He Y, Zhang J, Tan G. Dexmedetomidine promotes liver regeneration in mice after 70% partial hepatectomy by suppressing NLRP3 inflammasome not TLR4/NFκB. Int Immunopharmacol 2018; 54:46-51. [DOI: 10.1016/j.intimp.2017.10.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/30/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023]
|
13
|
Abd-ellatief R, Mohamed H, Kotb H. Reactive Astrogliosis in an Experimental Model of Fibromyalgia: Effect of Dexmedetomidine. Cells Tissues Organs 2018; 205:105-119. [DOI: 10.1159/000488757] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
To our knowledge, this is the first study which investigates the induction of neuroinflammation in rats using an acidic-saline model of fibromyalgia. It is well known that the hippocampus has a fundamental role in pain perception, and astrocytes play a crucial role in pain signaling. Our aim is to evaluate the ability of dexmedetomidine to attenuate the inflammatory responses induced in astrocytes. In a group of healthy rats, induction of chronic muscle pain by intramuscular injection of 100 µL of acidic saline on days 0 and 5 resulted in peripheral sensitization (measured using the von Frey test) and significant (<i>p</i> < 0.05) increases in IL-1β (160.2 ± 1.1 to 335.2 ± 1.8), IL-6 (100.1 ± 1.4 to 202.4 ± 1.1), and TNF-α (60.0 ± 0.7 to 115.5 ± 1). Light and electron microscopy revealed degenerative changes in the hippocampus and reactive astrogliosis. Immunohistochemistry showed increased expression of glial fibrillary acid protein and inducible nitric oxide synthase. Surprisingly, treatment with a single dose of an α<sub>2</sub>-adrenergic agonist, dexmedetomidine (5 µg/kg i.p.), attenuated these changes. This trial suggests that dexmedetomidine possibly directly acts on astrocytes, and a peripheral action is also suggested.
Collapse
|
14
|
Wang K, Zhu Y. Dexmedetomidine protects against oxygen-glucose deprivation/reoxygenation injury-induced apoptosis via the p38 MAPK/ERK signalling pathway. J Int Med Res 2017; 46:675-686. [PMID: 29210287 PMCID: PMC5971521 DOI: 10.1177/0300060517734460] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Objective To investigate the protective effects of dexmedetomidine (DEX) in
oxygen-glucose deprivation/reoxygenation (OGD/R) injury, which is involved
in a number of ischaemic diseases. Methods An in vitro OGD/R injury model was generated using mouse
Neuro 2A neuroblastoma (N2A) cells. Different concentrations of DEX were
administrated to OGD/R cells. CV-65 was used to inhibit p38 microtubule
associated protein kinase/extracellular signal-regulated kinases (MAPK/ERK)
signalling. Cell proliferation, cell cycle, apoptosis, and the levels of
proteins related to p38 MAPK/ERK signalling and apoptosis were evaluated
using Cell Counting Kit-8, flow cytometry, TdT-UTP nick end labelling and
Western blot analysis, respectively. Results DEX treatment of OGD/R cells promoted cell survival and attenuated
OGD/R-induced cell apoptosis. It also activated the p38 MAPK/ERK signalling
pathway, increased the levels of Bcl-2, and decreased the levels of Bax and
cleaved caspase-3. Treatment with the p38 MAPK/ERK inhibitor CV-65 inhibited
the activation of p38 MAPK/ERK and abrogated the DEX-induced effects on cell
survival and apoptosis. Conclusions DEX protects N2A cells from OGD/R-induced apoptosis via the activation of the
p38 MAPK/ERK signalling pathway. DEX might be an effective agent for the
treatment of ischaemic diseases.
Collapse
Affiliation(s)
- Ke Wang
- Department of Anaesthesiology, Suzhou Wuzhong People's Hospital, Suzhou, Jiangsu Province, China
| | - Yuekun Zhu
- Department of Anaesthesiology, Suzhou Wuzhong People's Hospital, Suzhou, Jiangsu Province, China
| |
Collapse
|
15
|
Huang Z, Liu G, Zeng Q, Gao R, Zhang S, Wang L, Liu B, Yu Y, Zhao A, Li R, Zhou S, Yu W. MiR-29b expression is associated with a dexmedetomidine-mediated protective effect against oxygen-glucose deprivation-induced injury to SK-N-SH cells in vitro. Cell Biol Int 2017; 42:344-352. [PMID: 29087603 DOI: 10.1002/cbin.10906] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/27/2017] [Indexed: 12/18/2022]
Abstract
Ischemic cerebral stroke is a leading cause of death and long-term disability world-wide. Neuronal injury following cerebral ischemia initiates a complex series of signaling cascades that lead to neuronal cell death. MicroRNA 29b (miR-29b) has reported involvement in the pathogenic process of ischemic brain injury. Dexmedetomidine (Dex) is a highly selective α2 adrenergic receptor stimulant that exerts a protective effect on brain tissue. To determine whether Dex might directly influence miR-29b expression after an ischemic injury, human neuroblastoma SK-N-SH cells were subjected to oxygen-glucose deprivation (OGD) for the purpose of creating a neuronal injury model that mimics the effects of brain ischemia in vitro. Next, the association of miR-29b with the protective effect of Dex against ischemic brain injury was studied through the enhancement or inhibition of miR-29b expression by transfection with an miR-29b mimic or inhibitor. We demonstrated that Dex treatment could reduce miR-29b expression, increase cell viability, and inhibit cell apoptosis in the OGD-induced neuronal injury model in vitro. Furthermore, down-regulation of miR-29b expression produced effects on OGD-induced neuronal injuries that were similar to those produced by Dex treatment. Moreover, up-regulation of miR-29b reversed the protective effect of Dex treatment against OGD-induced neuronal injury. Therefore, down-regulation of miR-29b expression might play a role in anti-apoptotic signaling similar to that played by Dex. Elucidation of the role played by miR-29b in ischemia, and identification of a definite association between Dex and miR-29b may lead to the development of new strategies for treating ischemic brain injuries.
Collapse
Affiliation(s)
- Zhi Huang
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, 550005, China.,Key Laboratory of Endemic and Ethnic Diseases, The Key Laboratory of Medical Molecular Biology in Guizhou Medical University, Guiyang, 550002, China
| | - Guoli Liu
- School of Medical Imaging of Guizhou Medical University, Guiyang city Beijing Road 9#, Guiyang, 550002, China
| | - Qingfan Zeng
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, 550005, China
| | - Rui Gao
- Guizhou Entry-Exit Inspection and Quarantine Bureau of the People's Republic of China, Guiyang, 550005, China
| | - Shuai Zhang
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550005, China
| | - Lizhou Wang
- School of Medical Imaging of Guizhou Medical University, Guiyang city Beijing Road 9#, Guiyang, 550002, China
| | - Bingjie Liu
- School of Medical Imaging of Guizhou Medical University, Guiyang city Beijing Road 9#, Guiyang, 550002, China
| | - YanLong Yu
- School of Medical Imaging of Guizhou Medical University, Guiyang city Beijing Road 9#, Guiyang, 550002, China
| | - Ansu Zhao
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550001, China
| | - Rui Li
- Department of Rehabilitation, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Shi Zhou
- School of Medical Imaging of Guizhou Medical University, Guiyang city Beijing Road 9#, Guiyang, 550002, China.,Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550005, China
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases, The Key Laboratory of Medical Molecular Biology in Guizhou Medical University, Guiyang, 550002, China
| |
Collapse
|
16
|
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]
|
17
|
Kim E, Kim HC, Lee S, Ryu HG, Park YH, Kim JH, Lim YJ, Park HP. Dexmedetomidine confers neuroprotection against transient global cerebral ischemia/reperfusion injury in rats by inhibiting inflammation through inactivation of the TLR-4/NF-κB pathway. Neurosci Lett 2017; 649:20-27. [DOI: 10.1016/j.neulet.2017.04.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/10/2017] [Accepted: 04/05/2017] [Indexed: 12/23/2022]
|
18
|
Küçükebe ÖB, Özzeybek D, Abdullayev R, Ustaoğlu A, Tekmen I, Küme T. Efeito de dexmedetomidina sobre lesão pulmonar aguda em modelo experimental de isquemia‐reperfusão. Braz J Anesthesiol 2017; 67:139-146. [DOI: 10.1016/j.bjan.2016.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/20/2015] [Accepted: 08/17/2015] [Indexed: 01/24/2023] Open
|
19
|
Zhang P, Li Y, Han X, Xing Q, Zhao L. Dexmedetomidine Regulates 6-hydroxydopamine-Induced Microglial Polarization. Neurochem Res 2017; 42:1524-1532. [PMID: 28247333 DOI: 10.1007/s11064-017-2209-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/04/2017] [Accepted: 02/15/2017] [Indexed: 12/14/2022]
Abstract
Microglia have undergone extensive characterization and have been shown to present distinct phenotypes, such as the M1 or M2 phenotypes, depending on their stimuli. As a highly specific neurotoxin, 6-hydroxydopamine (6-OHDA) can be used to further our understanding of the immune response in Parkinson's disease (PD). Dexmedetomidine (DEX), a centrally selective α2-adrenoceptor agonist, performs very well as an anti-anxiety medication, sedative and analgesic. In the present study, we investigated the effects of DEX on 6-OHDA-induced microglial polarization. Our results indicate that treatment with 6-OHDA promotes microglial polarization toward the M1 state in BV2 microglia cells by increasing the release of interleukin (IL)-6, IL-1β, or tumor necrosis factor-α, which can be prevented by pretreatment with DEX. In addition, we found that 6-OHDA blocked IL-4-mediated microglial M2 polarization by suppressing expression of the microglial M2 markers arginase-1 (Arg-1), resistin-like α (Retnla/Fizz1), and chitinase 3-like 3 (Chi3l3/Ym1), which could be ameliorated by pretreatment with DEX. Notably, the inhibitory effects of 6-OHDA on IL-4-mediated induction of the anti-inflammatory marker genes IL-10, IL-13, and transforming growth factor-β2 could be significantly alleviated by pretreatment with DEX in a dose-dependent manner (P < 0.01). Mechanistically, alternations in the activation of signal transducer and activator of transcription 6 were involved in this process. These findings suggest that administration of DEX has the potential to interrupt the process of microgliosis in PD.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, 24 Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China
| | - Yu Li
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, 24 Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China
| | - Xuechang Han
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, 24 Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China
| | - Qunzhi Xing
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, 24 Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China.
| | - Lei Zhao
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, 24 Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China
| |
Collapse
|
20
|
Neuroprotection and neurotoxicity in the developing brain: an update on the effects of dexmedetomidine and xenon. Neurotoxicol Teratol 2017; 60:102-116. [PMID: 28065636 DOI: 10.1016/j.ntt.2017.01.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 12/13/2022]
Abstract
Growing and consistent preclinical evidence, combined with early clinical epidemiological observations, suggest potentially neurotoxic effects of commonly used anesthetic agents in the developing brain. This has prompted the FDA to issue a safety warning for all sedatives and anesthetics approved for use in children under three years of age. Recent studies have identified dexmedetomidine, the potent α2-adrenoceptor agonist, and xenon, the noble gas, as effective anesthetic adjuvants that are both less neurotoxic to the developing brain, and also possess neuroprotective properties in neonatal and other settings of acute ongoing neurologic injury. Dexmedetomidine and xenon are effective anesthetic adjuvants that appear to be less neurotoxic than other existing agents and have the potential to be neuroprotective in the neonatal and pediatric settings. Although results from recent clinical trials and case reports have indicated the neuroprotective potential of xenon and dexmedetomidine, additional randomized clinical trials corroborating these studies are necessary. By reviewing both the existing preclinical and clinical evidence on the neuroprotective effects of dexmedetomidine and xenon, we hope to provide insight into the potential clinical efficacy of these agents in the management of pediatric surgical patients.
Collapse
|
21
|
Rashid MR, Najeeb R, Mushtaq S, Habib R. Comparative evaluation of midazolam, dexmedetomidine, and propofol as Intensive Care Unit sedatives in postoperative electively ventilated eclamptic patients. J Anaesthesiol Clin Pharmacol 2017; 33:331-336. [PMID: 29109631 PMCID: PMC5672512 DOI: 10.4103/joacp.joacp_380_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background and Aims: Eclampsia is a common hypertensive disorder of pregnancy and treatment often includes termination of pregnancy with elective postoperative mechanical ventilation. The present study was aimed to compare midazolam, propofol, and dexmedetomidine for sedation and antihypertensive requirements of such patients admitted to Intensive Care Unit (ICU) after termination of pregnancy. Material and Methods: A total of ninety eclamptic patients administered general anesthesia for the termination of pregnancy through cesarean section and who also required postoperative ventilation were taken up for the study and were randomly allocated into three groups. All patients received MgSO4 (loading dose, 4 g intravenous) following first seizure episode followed by a continuous infusion for next 24 h. Midazolam group (GrM) received 0.05 mg/kg loading dose of midazolam, followed by infusion of 0.05–0.3 mg/kg/h, propofol group (GrP) received 1 mg/kg loading dose of propofol followed by infusion of 2–8 mg/kg/h, and dexmedetomidine group (GrD) received dexmedetomidine loading dose at 1 mcg/kg followed by infusion of 0.2–1.2 mcg/kg/h. Postoperatively, patients were assessed for hemodynamic stability, requirement of antihypertensive and analgesics, duration of sedation and stop sedation-discharge, and total time spent in the ICU. Results: Mean heart rate and mean arterial pressure recorded at different time intervals were lowest in GrD. Nearly 70% (n = 21) patients in the GrM required antihypertensive, 50% (n = 15) in GrP, and 36.6% (n = 11) in the GrD (P < 0.05). Duration of stop sedation-discharge from ICU was least in GrD. A number of patients demanding additional analgesics was also least in GrD. Conclusion: Sedation with dexmedetomidine produced better hemodynamic stability in eclamptic patients, and there was a significant reduction in requirement of additional analgesics (P = 0.035) and antihypertensive (P = 0.004). Total duration of ICU stay was also less in this group of patients.
Collapse
Affiliation(s)
- Malik Rameez Rashid
- Department of Anaesthesia and Critical Care, Government Medical College, Srinagar, Jammu and Kashmir, India
| | - Rukhsana Najeeb
- Department of Anaesthesia and Critical Care, Government Medical College, Srinagar, Jammu and Kashmir, India
| | - Saima Mushtaq
- Department of Biochemistry, Government Medical College, Srinagar, Jammu and Kashmir, India
| | - Rizwana Habib
- Department of Obstretrics and Gynecology, Government Medical College, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
22
|
Choi IY, Hwang L, Jin JJ, Ko IG, Kim SE, Shin MS, Shin KM, Kim CJ, Park SW, Han JH, Yi JW. Dexmedetomidine alleviates cerebral ischemia-induced short-term memory impairment by inhibiting the expression of apoptosis-related molecules in the hippocampus of gerbils. Exp Ther Med 2016; 13:107-116. [PMID: 28123477 PMCID: PMC5244889 DOI: 10.3892/etm.2016.3956] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/04/2016] [Indexed: 01/26/2023] Open
Abstract
Cerebral ischemia results from cerebrovascular occlusion, which leads to neuronal cell death and eventually causes neurological impairments. Dexmedetomidine is a potent and highly selective α2-adrenoreceptor agonist with actions such as sedation, anxiolysis, analgesia and anesthetic-sparing effects. We investigated the effect of dexmedetomidine on apoptosis in the hippocampus after transient global ischemia in gerbils. Transient global ischemia was induced by ligation of both common carotid arteries. Dexmedetomidine was administrated intraperitoneally at three respective doses (0.1, 1 and 10 µg/kg) once per day for 14 consecutive days beginning a day after surgery. Short-term memory was assessed by use of a step-down avoidance task. Apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling assay, immunohistochemistry for caspase-3, and western blot analysis of Bcl-2-associated X protein, B-cell lymphoma 2, Bid, cytochrome c, apoptotic protease activating factor-1 and caspase-9 in the hippocampus. Induction of global ischemia deteriorated short-term memory by enhancing the expression of apoptosis-related molecules in the hippocampus. Treatment with dexmedetomidine suppressed the expression of apoptosis-related molecules under ischemic conditions, resulting in short-term memory improvement. Under normal conditions, dexmedetomidine exerted no significant effect on apoptosis in the hippocampus. The present results suggest that the α2-adrenoceptor agonist dexmedetomidine may be a useful therapeutic agent for the treatment of ischemic brain diseases.
Collapse
Affiliation(s)
- In-Young Choi
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Lakkyong Hwang
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Jun-Jang Jin
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Il-Gyu Ko
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Sung-Eun Kim
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Mal-Soon Shin
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Key-Moon Shin
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, Kyung Hee University College of Medicine, Seoul 130-701, Republic of Korea
| | - Sung-Wook Park
- Department of Anesthesiology and Pain Medicine, Kyung Hee Medical Center, Kyung Hee University College of Medicine, Seoul 130-872, Republic of Korea
| | - Jin-Hee Han
- Department of Anesthesiology and Pain Medicine, Kyung Hee Medical Center, Kyung Hee University College of Medicine, Seoul 130-872, Republic of Korea
| | - Jae-Woo Yi
- Department of Anesthesiology and Pain Medicine, Kangdong Kyung Hee Hospital, Kyung Hee University College of Medicine, Seoul 134-727, Republic of Korea
| |
Collapse
|
23
|
Seo H, Ryu HG, Son JD, Kim JS, Ha EJ, Kim JE, Park HP. Intraoperative dexmedetomidine and postoperative cerebral hyperperfusion syndrome in patients who underwent superficial temporal artery-middle cerebral artery anastomosis for moyamoya disease: A retrospective observational study. Medicine (Baltimore) 2016; 95:e5712. [PMID: 28033272 PMCID: PMC5207568 DOI: 10.1097/md.0000000000005712] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Dexmedetomidine, a selective α2-agonist, reduces cerebral blood flow and has neuroprotective effects against cerebral ischemia/reperfusion injury in experimental animals. We examined whether intraoperative dexmedetomidine would reduce the incidence of postoperative cerebral hyperperfusion syndrome (CHS) after superficial temporal artery-middle cerebral artery (STA-MCA) anastomosis in patients with moyamoya disease.The electronic medical records of 117 moyamoya patients who underwent STA-MCA anastomosis were reviewed retrospectively. The patients were divided into 2 groups: 48 patients received intraoperative dexmedetomidine (Group D), while 69 patients did not (Group ND). The incidence (primary outcome), onset, and duration of postoperative CHS were noted.The incidence of postoperative CHS was 45.8% and 40.6% in groups D and ND, respectively (P = 0.708). The duration of postoperative CHS was shorter in group D than in group ND (median [Q1-Q3], 5 [3-7] vs 8 [5-10] days, P = 0.021). There was no significant difference in the onset of CHS between group D and group ND (0 [0-2] vs 1 [0-3] days, P = 0.226).In conclusion, intraoperative dexmedetomidine did not reduce the incidence of postoperative CHS, although it reduced the duration of CHS, in patients who had undergone direct revascularization surgery for moyamoya disease.
Collapse
Affiliation(s)
- Hyungseok Seo
- Department of Anesthesiology and Pain Medicine, Dankook University Hospital, Cheonan
| | - Ho-Geol Ryu
- Department of Anesthesiology and Pain Medicine
| | - Je Do Son
- Department of Anesthesiology and Pain Medicine
| | | | - Eun Jin Ha
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong-Eun Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | | |
Collapse
|
24
|
Regulatory effects of anesthetics on nitric oxide. Life Sci 2016; 151:76-85. [DOI: 10.1016/j.lfs.2016.02.094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/20/2016] [Accepted: 02/26/2016] [Indexed: 12/26/2022]
|
25
|
Effect of dexmedetomidine on acute lung injury in experimental ischemia-reperfusion model. Braz J Anesthesiol 2016; 67:139-146. [PMID: 28236861 DOI: 10.1016/j.bjane.2015.08.002] [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: 03/01/2015] [Revised: 04/20/2015] [Accepted: 08/17/2015] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Ischemia-reperfusion injury is one of the consequences of tourniquet application for extremity surgery. The aim of the study was to establish the effect of dexmedetomidine on the acute lung injury following lower extremity experimental ischemia-reperfusion model in rats. METHODS Twenty-eight Wistar-Albino breed Rats were recruited after Ethics Committee approval and allocated into 4 groups, each with 7 subjects. Group 1 (SHAM) received only anesthesia. Group 2 (IR) had experienced 3h of ischemia and 3h of reperfusion using left lower extremity tourniquet after anesthesia application. Groups 3 (D-50) and 4 (D-100) had undergone the same procedures as in the Group 2, except for receiving 50 and 100mg·kg-1, respectively, dexmedetomidine intraperitoneally 1h before the tourniquet release. Blood samples were obtained for the analysis of tumor necrosing factor-α and interleukin-6. Pulmonary tissue samples were obtained for histological analysis. RESULTS No significant difference regarding blood tumor necrosing factor-α and interleukin-6 values was found among the groups, whereas pulmonary tissue injury scores revealed significant difference. Histological scores obtained from the Group 2 were significantly higher from those in the Groups 1, 3 and 4 with p-values 0.001 for each comparison. Moreover, Group 1 scores were found to be significantly lower than those in the Groups 3 and 4 with p-values 0.001 and 0.011, respectively. No significant difference was observed between the Groups 3 and 4. CONCLUSION Dexmedetomidine is effective in reduction of the experimental ischemia-reperfusion induced pulmonary tissue injury in rats, formed by extremity tourniquet application.
Collapse
|
26
|
Dexmedetomidine Protects against Transient Global Cerebral Ischemia/Reperfusion Induced Oxidative Stress and Inflammation in Diabetic Rats. PLoS One 2016; 11:e0151620. [PMID: 26982373 PMCID: PMC4794239 DOI: 10.1371/journal.pone.0151620] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/01/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Transient global cerebral ischemia/reperfusion (I/R) is a major perioperative complication, and diabetes increases the response of oxidative stress and inflammation induced by I/R. The objective of this study was to determine the protective effect of dexmedetomidine against transient global cerebral ischemia/reperfusion induced oxidative stress and inflammation in diabetic rats. METHODS Sixty-four rats were assigned into four experimental groups: normoglycemia, normoglycemia + dexmedetomidine, hyperglycemia, and hyperglycemia + dexmedetomidine and all subsequent neurological examinations were evaluated by a blinded observer. Damage to the brain was histologically assessed using the TUNEL staining method while western blotting was used to investigate changes in the expression levels of apoptosis-related proteins as well as the microglia marker, ionized calcium-binding adapter molecule 1 (Iba1). Water content in the brain was also analyzed. In addition, hippocampal concentrations of malondialdehyde (MDA) and Nox2 (a member of the Nox family of NADPH oxidases), and the activity of superoxide dismutase and catalase were analyzed. Finally, changes in serum concentrations of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 were detected. RESULTS Results showed that diabetes increased brain water content, the number of apoptotic neurons, early neurological deficit scores, oxidative stress (MDA and Nox2) and inflammation (pro-inflammatory cytokines including TNF-α and IL-6) levels following transient global I/R injury, but that these symptoms were attenuated following administration of dexmedetomidine. CONCLUSIONS These findings suggest that dexmedetomidine can significantly alleviate damage resulting from I/R, and this mechanism may be related to a reduction in both oxidative stress and inflammation which is normally associated with I/R.
Collapse
|
27
|
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
|
28
|
Dexmedetomidine Inhibits TLR4/NF-κB Activation and Reduces Acute Kidney Injury after Orthotopic Autologous Liver Transplantation in Rats. Sci Rep 2015; 5:16849. [PMID: 26585410 PMCID: PMC4653646 DOI: 10.1038/srep16849] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/21/2015] [Indexed: 12/24/2022] Open
Abstract
Patients who undergo orthotopic liver transplantation often sustain acute kidney injury(AKI). The toll-like receptor 4(TLR4)/Nuclear factor-кB(NF-кB) pathway plays a role in AKI. Dexmedetomidine(Dex) has been shown to attenuate AKI. The current study aimed to determine whether liver transplantation-induced AKI is associated with inflammatory response, and to assess the effects of dexmedetomidine pretreatment on kidneys in rats following orthotopic autologous liver transplantation(OALT). Seventy-seven adult male rats were randomized into 11 groups. Kidney tissue histopathology and levels of blood urea nitrogen(BUN) and serum creatinine(SCr) were evaluated. Levels of TLR4, NF-κB, tumor necrosis factor-α, and interleukin-1β levels were measured in kidney tissues. OALT resulted in significant kidney functional impairment and tissue injury. Pre-treatment with dexmedetomidine decreased BUN and SCr levels and reduced kidney pathological injury, TLR4 expression, translocation of NF-κB, and cytokine production. The effects of dexmedetomidine were reversed by pre-treatment with atipamezole and BRL44408, but not ARC239. These results were confirmed by using α2A-adrenergic receptor siRNA which reversed the protective effect of dexmedetomidine on attenuating NRK-52E cells injury induced by hypoxia reoxygenation. In conclusion, Dexmedetomidine-pretreatment attenuates OALT-induced AKI in rats which may be contributable to its inhibition of TLR4/MyD88/NF-κB pathway activation. The renoprotective effects are related to α2A-adrenergic receptor subtypes.
Collapse
|
29
|
Dexmedetomidine Analgesia Effects in Patients Undergoing Dental Implant Surgery and Its Impact on Postoperative Inflammatory and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:186736. [PMID: 26171113 PMCID: PMC4485522 DOI: 10.1155/2015/186736] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/18/2014] [Indexed: 12/31/2022]
Abstract
The aim of the study was to determine whether or not dexmedetomidine- (DEX-) based intravenous infusion in dental implantation can provide better sedation and postoperative analgesia via suppressing postoperative inflammation and oxidative stress. Sixty patients were randomly assigned to receive either DEX (group D) or midazolam (group M). Recorded variables were vital sign (SBP/HR/RPP/SpO2/RR), visual analogue scale (VAS) pain scores, and observer's assessment of alertness/sedation scale (OAAS) scores. The plasma levels of interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), antioxidant superoxide dismutase (SOD), and the lipid peroxidation product malondialdehyde (MDA) were detected at baseline and after 2, 4, and 24 h of drug administration. The VAS pain scores and OAAS scores were significantly lower for patients in group D compared to group M. The plasma levels of TNF-α, IL-6, and MDA were significantly lower in group D patients than those in group M at 2 h and 4 h. In group M, SOD levels decreased as compared to group D at 2 h and 4 h. The plasma levels of TNF-α, IL-6, and MDA were positively correlated with VAS pain scores while SOD negatively correlated with VAS pain scores. Therefore, DEX appears to provide better sedation during office-based artificial tooth implantation. DEX offers better postoperative analgesia via anti-inflammatory and antioxidation pathway.
Collapse
|
30
|
Sun Y, Gao Q, Wu N, Li SDE, Yao JX, Fan WJ. Protective effects of dexmedetomidine on intestinal ischemia-reperfusion injury. Exp Ther Med 2015; 10:647-652. [PMID: 26622369 DOI: 10.3892/etm.2015.2561] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 02/09/2015] [Indexed: 01/17/2023] Open
Abstract
Dexmedetomidine (DEX) has been hypothesized to possess anti-oxidative properties that may mitigate the damage caused by ischemia-reperfusion (IR) injury. The aim of the present study was to examine the effects of DEX on intestinal contractile activity, inflammation and apoptosis following intestinal IR injury. Intestinal IR injury was induced in rats by complete occlusion of the superior mesenteric artery for 60 min, followed by a 60-min reperfusion period. Rats received an intraperitoneal injection of 25 µg/kg DEX at 30 min prior to the mesenteric IR injury. Following reperfusion, segments of the terminal ileum were rapidly extracted and transferred into an isolated organ bath. The contractile responses to receptor-mediated acetylcholine (Ach) and non-receptor-mediated potassium chloride (KCl) were subsequently examined. Nitric oxide (NO) levels were determined and the expression levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, Bax and Bcl-2 were measured using an enzyme-linked immunosorbent assay. The levels of telomerase and caspase-3 were determined using reverse transcription-quantitative polymerase chain reaction. The results indicated that DEX treatment produced a significant reduction in the IR-induced contractile response to Ach and KCl in the intestinal tissue. Furthermore, DEX appeared to significantly ameliorate intestinal IR injury, in addition to reducing the production of NO. Similar reductions were observed in the intestinal expression levels of TNF-α and IL-6. In addition, DEX treatment resulted in a reduction in the expression levels of Bax in the intestinal tissues, while increasing those of Bcl-2, in addition to significantly increasing the mRNA levels of telomerase and caspase-3. Therefore, the present study indicated that NO, TNF-α and IL-6 may partially contribute to the pathogenesis of intestinal IR injury in addition to the increased expression levels of Bax, Bcl-2, telomerase and caspase-3. These findings suggest that DEX possesses beneficial anti-apoptotic and anti-inflammatory effects in intestinal tissue following bowel injury.
Collapse
Affiliation(s)
- Yong Sun
- Department of Surgery, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| | - Qiang Gao
- Department of Surgery, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| | - Nan Wu
- Department of Anesthesia, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| | - Sheng-DE Li
- Department of Anesthesia, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| | - Jing-Xin Yao
- Department of Anesthesia, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| | - Wen-Jie Fan
- Department of Anesthesia, Qingdao Women and Children's Hospital, Qingdao, Shandong 266034, P.R. China
| |
Collapse
|
31
|
Wassink G, Lear CA, Gunn KC, Dean JM, Bennet L, Gunn AJ. Analgesics, sedatives, anticonvulsant drugs, and the cooled brain. Semin Fetal Neonatal Med 2015; 20:109-14. [PMID: 25457080 DOI: 10.1016/j.siny.2014.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Multiple randomized controlled trials have shown that prolonged, moderate cerebral hypothermia initiated within a few hours after severe hypoxia-ischemia and continued until resolution of the acute phase of delayed cell death reduces mortality and improves neurodevelopmental outcome in term infants. The challenge is now to find ways to further improve outcomes. In the present review, we critically examine the evidence that conventional analgesic, sedative, or anticonvulsant agents might improve outcomes, in relation to the known window of opportunity for effective protection with hypothermia. This review strongly indicates that there is insufficient evidence to recommend routine use of these agents during therapeutic hypothermia. Further systematic research into the effects of pain and stress on the injured brain, and their treatment during hypothermia, is essential to guide the rational development of clinical treatment protocols.
Collapse
Affiliation(s)
- Guido Wassink
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Katherine C Gunn
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Justin M Dean
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
32
|
Neuroprotective effect of dexmedetomidine on hyperoxia-induced toxicity in the neonatal rat brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:530371. [PMID: 25653737 PMCID: PMC4310240 DOI: 10.1155/2015/530371] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/10/2014] [Indexed: 11/19/2022]
Abstract
Dexmedetomidine is a highly selective agonist of α2-receptors with sedative, anxiolytic, analgesic, and anesthetic properties. Neuroprotective effects of dexmedetomidine have been reported in various brain injury models. In the present study, we investigated the effects of dexmedetomidine on neurodegeneration, oxidative stress markers, and inflammation following the induction of hyperoxia in neonatal rats. Six-day-old Wistar rats received different concentrations of dexmedetomidine (1, 5, or 10 µg/kg bodyweight) and were exposed to 80% oxygen for 24 h. Sex-matched littermates kept in room air and injected with normal saline or dexmedetomidine served as controls. Dexmedetomidine pretreatment significantly reduced hyperoxia-induced neurodegeneration in different brain regions of the neonatal rat. In addition, dexmedetomidine restored the reduced/oxidized glutathione ratio and attenuated the levels of malondialdehyde, a marker of lipid peroxidation, after exposure to high oxygen concentration. Moreover, administration of dexmedetomidine induced downregulation of IL-1β on mRNA and protein level in the developing rat brain. Dexmedetomidine provides protections against toxic oxygen induced neonatal brain injury which is likely associated with oxidative stress signaling and inflammatory cytokines. Our results suggest that dexmedetomidine may have a therapeutic potential since oxygen administration to neonates is sometimes inevitable.
Collapse
|
33
|
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
|
34
|
Tanabe K, Matsushima-Nishiwaki R, Kozawa O, Iida H. Dexmedetomidine suppresses interleukin-1β-induced interleukin-6 synthesis in rat glial cells. Int J Mol Med 2014; 34:1032-8. [PMID: 25069417 DOI: 10.3892/ijmm.2014.1863] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/18/2014] [Indexed: 11/06/2022] Open
Abstract
Dexmedetomidine, an α2-adrenoceptor agonist, is used as a sedative medication for criticalyl ill patients and is known to exert neuroprotective effects by direct action on neurons and indirect action on neurons through astrocytes. Interleukin (IL)-6 plays a key role in neuroinflammation, which accompanies infection, traumatic brain injury, ischemia, neurodegenerative disorders, as both a pro-inflammatory cytokine and an anti-inflammatory cytokine. Dexmedetomidine suppresses immune function. However, the effects of dexmedetomidine on cytokine synthesis in the central nervous system (CNS) remain elusive. We previously reported that IL-1β stimulates IL-6 synthesis in the rat C6 glioma cell line through the phosphorylation of p38 mitogen-activated protein (MAP) kinase, stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK) and IκB. In the present study, we investigated the effects of dexmedetomidine on the IL-1β-induced IL-6 synthesis in C6 cells. Dexmedetomidine inhibited the IL-1β-stimulated IL-6 release and mRNA expression in C6 cells. 8-Bromo-adenosine-3',5'-cyclic monophosphate, but not 8-bromo-guanosine 3',5'-cyclic monophosphate, significantly enhanced the IL-1β-induced IL-6 release and mRNA expression. However, dexmedetomidine failed to affect cAMP accumulation in the cells treated with IL-1β or forskolin, an activator of adenylyl cyclase. Yohimbine, an α2-adrenoceptor antagonist, did not reverse the suppressive effects of dexmedetomidine on the IL-1β-induced IL-6 release. Dexmedetomidine did not affect the IL-1β-induced phosphorylation of p38 MAP kinase, SAPK/JNK, IκB, nuclear factor (NF)-κB or c-Jun. Our findings strongly suggest that dexmedetomidine inhibits the IL-1β-induced IL-6 synthesis independently of the adenylyl cyclase-cAMP pathway through α2-adrenoceptors in C6 glioma cells. It is possible that dexmedetomidine may affect the immune system in the CNS by regulating the production of IL-6.
Collapse
Affiliation(s)
- Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
35
|
Cai Y, Xu H, Yan J, Zhang L, Lu Y. Molecular targets and mechanism of action of dexmedetomidine in treatment of ischemia/reperfusion injury. Mol Med Rep 2014; 9:1542-50. [PMID: 24627001 DOI: 10.3892/mmr.2014.2034] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 02/17/2014] [Indexed: 01/13/2023] Open
Abstract
Dexmedetomidine (DEX), a highly specific α2-adrenergic agonist, which exhibits anaesthetic-sparing, analgesia and sympatholytic properties. DEX modulates gene expression, channel activation, transmitter release, inflammatory processes and apoptotic and necrotic cell death. It has also been demonstrated to have protective effects in a variety of animal models of ischemia/reperfusion (I/R) injury, including the intestine, myocardial, renal, lung, cerebral and liver. The broad spectrum of biological activities associated with DEX continues to expand, and its diverse effects suggest that it may offer a novel therapeutic approach for the treatment of human diseases with I/R involvement.
Collapse
Affiliation(s)
- Ye Cai
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Hui Xu
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Jia Yan
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Lei Zhang
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yi Lu
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
36
|
Xiong B, Shi QQ, Miao CH. Dexmedetomidine renders a brain protection on hippocampal formation through inhibition of nNOS-NO signalling in endotoxin-induced shock rats. Brain Inj 2014; 28:1003-8. [DOI: 10.3109/02699052.2014.888765] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
37
|
Inflammatory Response in Patients under Coronary Artery Bypass Grafting Surgery and Clinical Implications: A Review of the Relevance of Dexmedetomidine Use. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/905238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the fact that coronary artery bypass grafting surgery (CABG) with cardiopulmonary bypass (CPB) prolongs life and reduces symptoms in patients with severe coronary artery diseases, these benefits are accompanied by increased risks. Morbidity associated with cardiopulmonary bypass can be attributed to the generalized inflammatory response induced by blood-xenosurfaces interactions during extracorporeal circulation and the ischemia/reperfusion implications, including exacerbated inflammatory response resembling the systemic inflammatory response syndrome (SIRS). The use of specific anesthetic agents with anti-inflammatory activity can modulate the deleterious inflammatory response. Consequently, anti-inflammatory anesthetics may accelerate postoperative recovery and better outcomes than classical anesthetics. It is known that the stress response to surgery can be attenuated by sympatholytic effects caused by activation of central (α-)2-adrenergic receptor, leading to reductions in blood pressure and heart rate, and more recently, that they can have anti-inflammatory properties. This paper discusses the clinical significance of the dexmedetomidine use, a selective (α-)2-adrenergic agonist, as a coadjuvant in general anesthesia. Actually, dexmedetomidine use is not in anesthetic routine, but this drug can be considered a particularly promising agent in perioperative multiple organ protection.
Collapse
|
38
|
Dexmedetomidine improves the histological and neurological outcomes 48 h after transient spinal ischemia in rats. Brain Res 2014; 1566:24-30. [PMID: 24755109 DOI: 10.1016/j.brainres.2014.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/18/2014] [Accepted: 04/10/2014] [Indexed: 11/20/2022]
Abstract
Dexmedetomidine, an α2 adrenoceptor agonist, provides neuroprotection against various cerebral ischemia models through its anti-apoptotic effects. Dexmedetomidine also improves paraplegia induced by intrathecal morphine after short-term spinal ischemia. However, there are no reports regarding dexmedetomidine׳s ability to provide neuroprotection solely against transient spinal ischemia. We investigated whether dexmedetomidine would provide spinal protection following transient spinal ischemia in rats. Adult male Sprague Dawley rats were randomly assigned to one of the following five groups: (1) intravenous infusion of 0.9% NaCl at the rate of 0.5 mL/h (control), (2) dexmedetomidine 0.1 µg/kg/h, (3) dexmedetomidine 1 µg/kg/h, (4) dexmedetomidine 10 µg/kg/h, or (5) intravenous infusion of 0.9% NaCl without spinal ischemia (sham). The rats received saline solution or dexmedetomidine from 30 min before spinal cord ischemia to 48 h after ischemia. Spinal cord ischemia was induced by intraaortic balloon occlusion combined with proximal arterial hypotension for 10 min. Ischemic injury was assessed by neurological deficit scores and the number of viable motor nerve cells in the anterior spinal cord at 48 h after reperfusion. Neurological deficit scores in the dexmedetomidine-treated rats were significantly lower than the scores in the control group at 24 and 48 h after ischemia (P<0.05). The number of viable motor nerve cells was significantly larger in the dexmedetomidine-treated rats than in the control rats (P<0.05), but the number of motor nerve cells in the dexmedetomidine group was significantly smaller than the sham group. Our results indicate that the continuous administration of dexmedetomidine improves neurological and histological outcomes 48 h after transient spinal ischemia in rats.
Collapse
|
39
|
Cosar M, Kaner T, Sahin O, Topaloglu N, Guven M, Aras AB, Akman T, Ozkan A, Sen HM, Memi G, Deniz M. The neuroprotective effect of Sulindac after ischemia-reperfusion injury in rats. Acta Cir Bras 2014; 29:268-73. [DOI: 10.1590/s0102-86502014000400008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 03/11/2014] [Indexed: 11/22/2022] Open
|
40
|
Jiang L, Li L, Shen J, Qi Z, Guo L. Effect of dexmedetomidine on lung ischemia‑reperfusion injury. Mol Med Rep 2013; 9:419-26. [PMID: 24345905 PMCID: PMC3896524 DOI: 10.3892/mmr.2013.1867] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 11/15/2013] [Indexed: 01/25/2023] Open
Abstract
Dexmedetomidine, a specific selective α2-adrenergic agonist, does not only have the characteristics of being a sedative and analgesic, but also exhibits a protective role in brain ischemia-reperfusion injury and inhibits the inflammation in animals with sepsis. The objective of the present study was to investigate whether dexmedetomidine is capable of attenuating rat pulmonary damage induced by ischemia-reperfusion injury, which is a type of acute sterile lung injury. Sprague-Dawley rats were randomly assigned into six groups: The sham-operated (sham) group, the lung ischemia-reperfusion (I/R) group, intravenous injection of dexmedetomidine 2.5 μg/kg/h (Dex2.5) or 5 μg/kg/h (Dex5) for 1 h prior to ischemia, combination of α2-adrenergic antagonist yohimbine prior to dexmedetomidine pre-treatment (Dex+Yoh) and pre-administration of yohimbine alone (Yoh) prior to ischemia. Lung injury was assessed by the histopathological changes, arterial blood gas, wet/dry (w/d) weight ratio and myeloperoxidase (MPO) activity of the lung. The concentration of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) in bronchoalveolar lavage fluid (BALF) was measured by an enzyme-linked immunosorbent assay. The expression of toll-like receptor-4 (TLR4) and myeloid differentiation factor 88 (MyD88) mRNA in the lung were determined by quantitative PCR, and phosphorylated levels of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK)1/2 were determined by western blotting. Pre-treatment with dexmedetomidine significantly reduced the lung injury, w/d weight ratio and MPO activity, and decreased the concentration of TNF-α, IL-6 and MCP-1 in BALF compared with the I/R group. The expression of TLR4 and MyD88 mRNA and the levels of phosphorylated JNK and ERK1/2 in the lung tissue were markedly downregulated by intravenous injection of dexmedetomidne for 1 h prior to lung I/R. The protective effects of dexmedetomidine on the lung were not completely reversed by the α2-adrenergic antagonist, yohimbine. Pre-treatment with dexmedetomidine is capable of reducing pulmonary damage and inhibiting sterile inflammation induced by lung I/R injury. TLR4/MyD88/mitogen-activated protein kinase (MAPK) signaling is involved in the protective mechanism of dexmedetomidine through α2-adrenoceptor independence.
Collapse
Affiliation(s)
- Lili Jiang
- Department of Anesthesiology, Second Xiang‑Ya Hospital, Xiang‑Ya Medical College, Central South University, Changsha, Hunan 410011, P.R. China
| | - Li Li
- Department of Anesthesiology, Second Xiang‑Ya Hospital, Xiang‑Ya Medical College, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jinmei Shen
- Department of Anesthesiology, Second Xiang‑Ya Hospital, Xiang‑Ya Medical College, Central South University, Changsha, Hunan 410011, P.R. China
| | - Zeyou Qi
- Department of Anesthesiology, Second Xiang‑Ya Hospital, Xiang‑Ya Medical College, Central South University, Changsha, Hunan 410011, P.R. China
| | - Liang Guo
- Department of Anesthesiology, Second Xiang‑Ya Hospital, Xiang‑Ya Medical College, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
41
|
Yang L, Xu JM, Jiang X, Ruan W, Cui Y, He L. Effect of dexmedetomidine on plasma brain-derived neurotrophic factor: A double-blind, randomized and placebo-controlled study. Ups J Med Sci 2013; 118:235-9. [PMID: 23781882 PMCID: PMC4190894 DOI: 10.3109/03009734.2013.808295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Dexmedetomidine (DEX) has neuro-protective effects, but the clinical mechanism remains unclear. METHOD Forty patients were randomly divided into two groups: group A (control) and group B (treated with DEX). Plasma concentrations of brain-derived neurotrophic factor (BDNF) were determined in blood samples using enzyme-linked immunosorbent assays at five time points: T1 (baseline), T2 (15 minutes after intubation and before the surgery was started), T3 (the end of surgery), T4 (10 minutes after extubation in the post-anesthesia care unit), and T5 (24 hours after the surgery). Changes in bispect (BIS) index, heart rates, and doses of anesthetics used for induction were also recorded. RESULTS Baseline plasma concentrations of BDNF did not differ between group A and group B; 15 minutes after induction, concentrations of plasma BDNF were significantly reduced in group A. Twenty-four hours after surgery, the concentration was still higher in group B than in group A. In contrast, plasma concentrations of BDNF at other time points tested did not differ between the two groups. CONCLUSION It appears that DEX could reverse the reduced plasma concentrations of BDNF caused by anesthetics, and this effect lasted for 24 hours after surgery.
Collapse
Affiliation(s)
- Lin Yang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Jun-Mei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - XingHua Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Wei Ruan
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - YuLong Cui
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Liang He
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University and Anesthesiology Research Institute, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
42
|
Gul M, Kayhan B, Elbe H, Dogan Z, Otlu A. Histological and biochemical effects of dexmedetomidine on liver during an inflammatory bowel disease. Ultrastruct Pathol 2013; 39:6-12. [PMID: 24134660 DOI: 10.3109/01913123.2013.829150] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Inflammation in the liver is an extraintestinal manifestation that is frequently seen during inflammatory bowel diseases (IBD). The authors investigated histopathologycal, ultrastructural and antioxidant effects of dexmedetomidine (Dex) on liver during trinitrobenzene sulfonic acid (TNBS)-induced inflammatory bowel disease. Thirty-two BALB/c mice were divided (n = 8) as follows: control; Dex (dexmedetomidine) (30 μg/kg) for 6 days; TNBS 150 μL, TNBS + ethanol (50% w/v) intrarectally; TNBS + Dex. The histopathological and ultrastructural changes were evaluated. The levels of malondialdehyde (MDA), activity of antioxidant enzymes (GPx and SOD) were measured in liver tissue. Induction of colitis induced histopathological and ultrastructural changes of damage in liver. Those changes were markedly reduced in the TNBS + Dex group and that reduction was even significant in comparison to the TNBS group. MDA levels were significantly higher in the TNBS group and dexmedetomidine significantly elevated SOD levels in the TNBS + Dex group. These results suggest that the administration of dexmedetomidine reduces the histopathological and ultrastructural damage and increases the defense capacity against oxidative damage on liver in this IBD mice model.
Collapse
Affiliation(s)
- Mehmet Gul
- Department of Histology and Embryology, Faculty of Medicine and
| | | | | | | | | |
Collapse
|
43
|
Zhao LH, Shi ZH, Yin NN, Zhou JX. Use of dexmedetomidine for prophylactic analgesia and sedation in delayed extubation patients after craniotomy: a study protocol and statistical analysis plan for a randomized controlled trial. Trials 2013; 14:251. [PMID: 23941549 PMCID: PMC3751309 DOI: 10.1186/1745-6215-14-251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/07/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pain and agitation are common in patients after craniotomy. They can result in tachycardia, hypertension, immunosuppression, increased catecholamine production and increased oxygen consumption. Dexmedetomidine, an alpha-2 agonist, provides adequate sedation without respiratory depression, while facilitating frequent neurological evaluation. METHODS/DESIGN The study is a prospective, randomized, double-blind, controlled, parallel-group design. Consecutive patients are randomly assigned to one of the two treatment study groups, labeled 'Dex group' or 'Saline group.' Dexmedetomidine group patients receive a continuous infusion of 0.6 μg/kg/h (10 ug/ml). Placebo group patients receive a maintenance infusion of 0.9% sodium chloride for injection at a volume and rate equal to that of dexmedetomidine. The mean percentages of time in optimal sedation, vital signs, various and adverse events, the percentage of patients requiring propofol for rescue to achieve/maintain targeted sedation (Sedation-Agitation Scale, SAS 3 to 4) and total dose of propofol required throughout the study drug infusion are collected. The percentage of patients requiring fentanyl for additional rescue to analgesia and total dose of fentanyl required are recorded. The effects of dexmedetomidine on hemodynamic and recovery responses during extubation are measured. Intensive care unit and hospital length of stay also are collected. Plasma levels of epinephrine, norepinephrine, dopamine, cortisol, neuron-specific enolase and S100-B are measured before infusion (T1), at two hours (T2), four hours (T3) and eight hours (T4) after infusion and at the end of infusion (T5) in 20 patients in each group. DISCUSSION The study has been initiated as planned in July 2012. One interim analysis advised continuation of the trial. The study will be completed in July 2013. TRIAL REGISTRATION ClinicalTrials (NCT): ChiCTR-PRC-12002903.
Collapse
|
44
|
Şahin T, Begeç Z, Toprak Hİ, Polat A, Vardi N, Yücel A, Durmuş M, Ersoy MÖ. The effects of dexmedetomidine on liver ischemia–reperfusion injury in rats. J Surg Res 2013; 183:385-90. [DOI: 10.1016/j.jss.2012.11.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 11/09/2012] [Accepted: 11/16/2012] [Indexed: 11/30/2022]
|
45
|
Dexmedetomidine reduced cytokine release during postpartum bleeding-induced multiple organ dysfunction syndrome in rats. Mediators Inflamm 2013; 2013:627831. [PMID: 23840096 PMCID: PMC3693180 DOI: 10.1155/2013/627831] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 03/07/2013] [Accepted: 05/22/2013] [Indexed: 11/18/2022] Open
Abstract
Dexmedetomidine (DEX) is an α2-adrenergic agonist. It decreases the levels of norepinephrine release, resulting in a reduction of postsynaptic adrenergic activity. In the present study, the effects of DEX on postpartum bleeding-induced multiple organ dysfunction syndrome (BMODS) were studied in rats in which BMODS was induced by the combination of hypotension and clamping of the superior mesenteric artery. We evaluated the role of dexmedetomidine (DEX) in cytokine release during postpartum BMODS in rats. In summary, the present study demonstrated that DEX administration reduced IFN-r and IL-4 release and decreased lung injury during postpartum BMODS. It is possible that DEX administration decreased inflammatory cytokine production in BMODS by inhibiting inflammation and free radical release by leukocytes independent of the DEX dose.
Collapse
|
46
|
Postischemic Sevoflurane Offers No Additional Neuroprotective Benefit to Preischemic Dexmedetomidine. J Neurosurg Anesthesiol 2013; 25:184-90. [DOI: 10.1097/ana.0b013e3182764d2a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Celik F, Göçmez C, Kamaşak K, Tufek A, Guzel A, Tokgoz O, Fırat U, Evliyaoğlu O. The comparison of neuroprotective effects of intrathecal dexmedetomidine and metilprednisolone in spinal cord injury. Int J Surg 2013; 11:414-8. [PMID: 23542594 DOI: 10.1016/j.ijsu.2013.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/19/2013] [Accepted: 03/18/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND The purpose of this study is the investigation of the effects of intrathecally injected dexmedetomidine and methylprednisolone and their dominancy over one another in rats with generated Spinal Cord Injury (SCI). METHODS 40, female, adult Wistar Albino rats weighing 220-260 g were included in the study. The rats were fixed with Intrathecal catheter (IT) and divided into four groups. All subjects were applied T7-10 laminectomy after catheter. Group S (n:10) was injected with IT 10 μL isotonic saline; Group C (n:10) with IT 10 μL isotonic saline after SCI; Group D (n:10) with IT one doze 10 μL of dexmedetomidine after SCI; Group M (n:10) IT one dose 10 μL of methylprednisolone. The subjects were sacrificed 72 h after this operation. The damaged area was removed biochemically and histopathologically examined. RESULTS Antioxidant and inflammatory parameters searched for in all damages tissue were statistically different in all groups from group S. They were different in group M and group D than group C (p < 0.001). After the comparison of group D and group M, PON and IL6 values were higher in group D (p = 0.003, p = 0.035) while the other two biochemical parameters were similar in both groups (Table 1). After histopathologic trials, edemas, bleeding and necrosis were found less in group S while at the most in group C (p < 0.001). In group M and group D, however, they were higher than group S and lower than group C (p < 0.001). After the comparison of group D and group M, while there was no difference in terms of edema necrosis, the amount of bleeding was lower in group D (p < 0.001) (Table 2). CONCLUSIONS It has been discovered that intrathecal use of dexmedetomidine caused neuroprotective effects similar to methylprednisolone.
Collapse
Affiliation(s)
- Feyzi Celik
- Department of Anesthesiology, Faculty of Medicine, Dicle University, Diyarbakir 21280, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
HWANG LAKKYONG, CHOI INYOUNG, KIM SUNGEUN, KO ILGYU, SHIN MALSOON, KIM CHANGJU, KIM SANGHOON, JIN JUNJANG, CHUNG JUNYOUNG, YI JAEWOO. Dexmedetomidine ameliorates intracerebral hemorrhage-induced memory impairment by inhibiting apoptosis and enhancing brain-derived neurotrophic factor expression in the rat hippocampus. Int J Mol Med 2013; 31:1047-56. [DOI: 10.3892/ijmm.2013.1301] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 03/08/2013] [Indexed: 11/06/2022] Open
|
49
|
The effects of dexmedetomidine on secondary acute lung and kidney injuries in the rat model of intra-abdominal sepsis. ScientificWorldJournal 2013; 2013:292687. [PMID: 23476127 PMCID: PMC3586481 DOI: 10.1155/2013/292687] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/15/2013] [Indexed: 12/13/2022] Open
Abstract
In the present study, the effects of dexmedetomidine on secondary lung and kidney injuries were studied in the rat model of intra-abdominal sepsis by immunohistological and biochemical examinations. We measured serum creatinine, kidney tissue malondialdehide and plasma neutrophil gelatinase-associated lipocalin levels. In order to evaluate tissue injury we determined kidney tissue mononuclear cell infiltration score, alveolar macrophage count, histological kidney and lung injury scores and kidney and lung tissue immunoreactivity scores. We demonstrated that dexmedetomidine attenuates sepsis-induced lung and kidney injuries and apoptosis in the rat model of sepsis. There is still need for comparative studies in order to determine the effects of dexmedetomidine on organ functions in early human sepsis.
Collapse
|
50
|
Tüfek A, Tokgöz O, Aliosmanoglu I, Alabalik U, Evliyaoglu O, Çiftçi T, Güzel A, Yıldırım ZB. The protective effects of dexmedetomidine on the liver and remote organs against hepatic ischemia reperfusion injury in rats. Int J Surg 2012; 11:96-100. [PMID: 23261946 DOI: 10.1016/j.ijsu.2012.12.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/27/2012] [Accepted: 12/03/2012] [Indexed: 12/14/2022]
Abstract
AIM To investigate the protective effects of dexmedetomidine against hepatic ischemia/reperfusion (IR) injury and hepatic IR induced remote organ injury. METHODS Forty Wistar albino rats were divided into the following four groups: sham, dexmedetomidine, IR, and IR + dexmedetomidine. Hepatic ischemia was created by the Pringle maneuver for 30 min followed by a 30 min reperfusion period in the IR and IR + dexmedetomidine groups. The dexmedetomidine and IR + dexmedetomidine groups were administered dexmedetomidine (100 μg/kg, single dose) intraperitoneally after the anesthesia insult. Blood samples and hepatic, renal, and lung tissue specimens were obtained to measure serum and tissue total oxidative activity (TOA), total antioxidant capacity (TAC), paraoxonase (PON-1), and oxidative stress index (OSI) after 60 min in all groups. RESULTS According to the biochemical analyses of the samples taken from the serum and the liver, lung, and kidney tissues, when comparing the sham group and the IR group, TOA and OSI values were higher in the IR group, while TAC and PON-1 values were lower (p < 0.05). It was observed that TOA and OSI values were significantly lower, while TAC and PON-1 values increased with dexmedetomidine treatment (p < 0.05). In addition, dexmedetomidine ameliorated hepatic histopathological changes inducing IR, but there were no significant histopathological changes in the remote organs. CONCLUSION This study demonstrated that dexmedetomidine markedly reduced the oxidative stress in serum, liver, and remote organs induced by hepatic IR injury, and ameliorated the histopathological damage in the liver.
Collapse
Affiliation(s)
- Adnan Tüfek
- Department of Anesthesiology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|