1
|
Choudhary S, Kaur R, Waziri A, Garg A, Kadian R, Alam MS. N-type calcium channel blockers: a new approach towards the treatment of chronic neuropathic pain. EXPLORATION OF MEDICINE 2023. [DOI: 10.37349/emed.2023.00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Neuropathic pain (NP) remains maltreated for a wide number of patients by the currently available treatments and little research has been done in finding new drugs for treating NP. Ziconotide (PrialtTM) had been developed as the new drug, which belongs to the class of ω-conotoxin MVIIA. It inhibits N-type calcium channels. Ziconotide is under the last phase of the clinical trial, a new non-narcotic drug for the management of NP. Synthetically it has shown the similarities with ω-conotoxin MVIIA, a constituent of poison found in fish hunting snails (Conus magus). Ziconotide acts by selectively blocking neural N-type voltage-sensitized Ca2+ channels (NVSCCs). Certain herbal drugs also have been studied but no clinical result is there and the study is only limited to preclinical data. This review emphasizes the N-type calcium channel inhibitors, and their mechanisms for blocking calcium channels with their remedial prospects for treating chronic NP.
Collapse
Affiliation(s)
- Shikha Choudhary
- School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram 122103, Haryana, India
| | - Raminderjit Kaur
- School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram 122103, Haryana, India
| | - Aafrin Waziri
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Delhi, 110078, India
| | - Arun Garg
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India
| | - Renu Kadian
- Ram Gopal College of Pharmacy, Gurugram 122506, Haryana, India
| | - Md Sabir Alam
- SGT College of Pharmacy, SGT University, Gurugram 122505, Haryana, India
| |
Collapse
|
2
|
Sun H, Hu H, Xu X, Fang M, Tao T, Liang Z. Protective effect of dexmedetomidine in cecal ligation perforation-induced acute lung injury through HMGB1/RAGE pathway regulation and pyroptosis activation. Bioengineered 2021; 12:10608-10623. [PMID: 34747306 PMCID: PMC8810048 DOI: 10.1080/21655979.2021.2000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
Dexmedetomidine (DEX) has been reported to attenuate cecal ligation perforation (CLP)-stimulated acute lung injury (ALI) by downregulating HMGB1 and RAGE. This study aimed to further investigate the specific mechanisms of RAGE and its potential-related mechanisms of DEX on ALI models in vitro and in vivo. The in vitro and in vivo ALI models were established by lipopolysaccharide treatment in MLE-12 cells and CLP in mice, respectively. The effect of DEX on pathological alteration was investigated by HE staining. Thereafter, the myeloperoxidase (MPO) activity and inflammatory cytokine levels were respectively detected to assess the lung injury of mice using commercial kits. The expression levels of HMGB1, RAGE, NF-κB, and pyroptosis-related molecules were detected by RT-qPCR and Western blot. HE staining showed that lung injury, increased inflammatory cell infiltration, and lung permeability was found in the ALI mice, and DEX treatment significantly attenuated lung tissue damage induced by CLP. The MPO activity and inflammatory cytokines (TNF-α, IL-1β, and NLRP3) levels were also significantly reduced after DEX treatment compared with those in the ALI mice. Moreover, DEX activated the HMGB1/RAGE/NF-κB pathway and upregulated the pyroptosis-related proteins. However, the protective DEX effect was impaired by RAGE overexpression in ALI mice and MLE-12 cells. Additionally, DEX treatment significantly suppressed HMGB1 translocation from the nucleus region to the cytoplasm, and this effect was reversed by RAGE overexpression. These findings suggested that DEX may be a useful ALI treatment, and the protective effects on ALI mice may be through the inhibition of HMGB1/RAGE/NF-κB pathway and cell pyroptosis.
Collapse
Affiliation(s)
- Huaqin Sun
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Hongyi Hu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoping Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Mingsun Fang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Tao Tao
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhehao Liang
- Department of Ultrasound, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Administration of Low-Dose Dexmedetomidine Did Not Affect Acute Inflammatory Response after Cytoreductive Surgery Combined with Hyperthermic Intraperitoneal Chemotherapy: A Double-Blind Randomized Controlled Trial. J Clin Med 2021; 10:jcm10143145. [PMID: 34300310 PMCID: PMC8303433 DOI: 10.3390/jcm10143145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
During cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC), attenuation of inflammatory responses that increase susceptibility to postoperative complications, morbidity, and mortality is important. We aimed to evaluate whether intraoperative dexmedetomidine infusion impacted inflammatory response in patients undergoing CRS with HIPEC. Fifty-six patients scheduled for CRS with HIPEC were randomly assigned to the control (n = 28) and dexmedetomidine (n = 28) groups. The primary endpoint was the effect of dexmedetomidine on the interleukin-6 (IL-6) level measured at pre-operation (Pre-OP), before HIPEC initiation (Pre-HIPEC), immediately after HIPEC; after the end of the operation; and on postoperative day (POD) 1. In both groups, the IL-6 levels from Pre-HIPEC until POD 1 and the C-reactive protein (CRP) levels on PODs 1, 2, and 3 were significantly higher than the Pre-OP values (all Bonferroni corrected, p < 0.001). However, total differences in IL-6 and CRP levels, based on the mean area under the curve, were not detected between the two groups. The continuous intraoperative infusion of dexmedetomidine (0.4 μg/kg/h) in patients undergoing CRS with HIPEC did not significantly lower the inflammatory indices. Further dose investigative studies are needed to find the dexmedetomidine dose that provides anti-inflammatory and sympatholytic effects during HIPEC.
Collapse
|
4
|
Shehabi Y, Murfin B, James A, Al-Bassam W, Bellomo R. Trials of dexmedetomidine sedation in ventilated critically ill septic patients: Challenges, limitations and opportunities. Anaesth Crit Care Pain Med 2021; 40:100925. [PMID: 34217839 DOI: 10.1016/j.accpm.2021.100925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yahya Shehabi
- Monash Health School of Clinical Sciences, Monash University, Melbourne, Australia; Monash Health, Intensive Care Unit, Clayton, Melbourne, Australia; Prince of Wales Clinical School of Medicine, University New South Wales, Sydney, Australia.
| | - Brendan Murfin
- Monash Health School of Clinical Sciences, Monash University, Melbourne, Australia; Monash Health, Intensive Care Unit, Clayton, Melbourne, Australia
| | - Arthur James
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anaesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France; SFAR Youth Committee, Paris, France
| | - Wisam Al-Bassam
- Monash Health School of Clinical Sciences, Monash University, Melbourne, Australia; Monash Health, Intensive Care Unit, Clayton, Melbourne, Australia
| | - Rinaldo Bellomo
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Department of Intensive Care, Austin Hospital, Melbourne, Australia; Department of Critical Care, The University of Melbourne, Melbourne, Australia; Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
5
|
Yuki K. The immunomodulatory mechanism of dexmedetomidine. Int Immunopharmacol 2021; 97:107709. [PMID: 33933842 DOI: 10.1016/j.intimp.2021.107709] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
Dexmedetomidine has been increasingly introduced into the perioperative care of surgical patients. Because a subset of anesthetics/sedatives are immunomodulatory, it is critical to understand the role of dexmedetomidine in our host immune functions. Here we reviewed the role of dexmedetomidine in different immune cells. We also reviewed published clinical articles that described the role of dexmedetomidine in organ injury, cancer surgery, and infection. In animal studies, dexmedetomidine attenuated organ injury. In clinical studies, dexmedetomidine was associated with an improvement in outcomes in cardiac surgery and transplant surgery. However, there is a paucity in research examining how dexmedetomidine is associated with these outcomes. Further studies are needed to understand its clinical application from immunological standpoints.
Collapse
Affiliation(s)
- Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, USA; Department of Anaesthesia, Harvard Medical School, USA; Department of Immunology, Harvard Medical School, USA.
| |
Collapse
|
6
|
Peripheral Mechanisms of Neuropathic Pain-the Role of Neuronal and Non-Neuronal Interactions and Their Implications for Topical Treatment of Neuropathic Pain. Pharmaceuticals (Basel) 2021; 14:ph14020077. [PMID: 33498496 PMCID: PMC7909513 DOI: 10.3390/ph14020077] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/16/2022] Open
Abstract
Neuropathic pain in humans arises as a consequence of injury or disease of somatosensory nervous system at peripheral or central level. Peripheral neuropathic pain is more common than central neuropathic pain, and is supposed to result from peripheral mechanisms, following nerve injury. The animal models of neuropathic pain show extensive functional and structural changes occurring in neuronal and non-neuronal cells in response to peripheral nerve injury. These pathological changes following damage lead to peripheral sensitization development, and subsequently to central sensitization initiation with spinal and supraspinal mechanism involved. The aim of this narrative review paper is to discuss the mechanisms engaged in peripheral neuropathic pain generation and maintenance, with special focus on the role of glial, immune, and epithelial cells in peripheral nociception. Based on the preclinical and clinical studies, interactions between neuronal and non-neuronal cells have been described, pointing out at the molecular/cellular underlying mechanisms of neuropathic pain, which might be potentially targeted by topical treatments in clinical practice. The modulation of the complex neuro-immuno-cutaneous interactions in the periphery represents a strategy for the development of new topical analgesics and their utilization in clinical settings.
Collapse
|
7
|
Zhai W, Yang L, Sun P, Li Y, Han J, Wang G. Effect of dexmedetomidine on hemodynamic changes and inflammatory responses in patients undergoing off-pump coronary-artery bypass grafting. Exp Ther Med 2020; 20:250. [PMID: 33178348 PMCID: PMC7651881 DOI: 10.3892/etm.2020.9380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to determine the effect of dexmedetomidine on hemodynamic changes and inflammatory responses in patients undergoing off-pump coronary artery bypass grafting (OPCABG). A total of 300 patients about to receive OPCABG were randomized evenly into the control group (n=116) and study group (n=123). Intravenous dexmedetomidine pump infusion was administered to patients in the study group at a rate of 0.4 µg.kg-1.h-1. The control group received physiological saline at the same infusion speed. Changes in hemodynamic parameters and inflammatory indices were compared between the two groups. Hemodynamic parameters, such as the heart rate and mean arterial pressure, were lower in patients from the study group, compared with that in the control group (both P<0.05). The levels of pro-inflammatory factors, such as interleukin (IL)-6, tumor necrosis factor-α and C-reactive protein, were also reduced in the study group (P<0.05). The observed levels of IL-10 were lower in the control group compared with that in the study group, although a statistically significant difference was not achieved. Thus, the administration of dexmedetomidine in patients undergoing OPCABG stabilized hemodynamics and reduced inflammation. The present study was registered at the Chinese Clinical Trial Registry, under the trial registration number ChiCTR-OOC-15005978 (2015).
Collapse
Affiliation(s)
- Wenqian Zhai
- Tianjin Medical University, Tianjin 300070, P.R. China.,Department of Anesthesiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Lieming Yang
- Department of Cardiovascular Surgery, General Hospital of Tianjin Medical University, Tianjin 300052, P.R. China
| | - Peng Sun
- Department of Anesthesiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Yunfei Li
- Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jiange Han
- Department of Anesthesiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Guolin Wang
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin 300052, P.R. China
| |
Collapse
|
8
|
Effect of Dexmedetomidine on Postoperative Lung Injury during One-Lung Ventilation in Thoracoscopic Surgery. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4976205. [PMID: 33083468 PMCID: PMC7557917 DOI: 10.1155/2020/4976205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
Objective To investigate the effect of dexmedetomidine on postoperative lung injury in patients undergoing thoracoscopic surgery. Methods From March 2019 to October 2019, 40 patients were randomly divided into two groups: dexmedetomidine group (group D) and control group (group C). Except recording the general condition of the patients in both groups preoperatively and intraoperatively, the oxygenation index (OI) and alveolar-arterial oxygen partial pressure difference (A-aDO2) were monitored at admission (T0), immediately after one-lung ventilation (T1), 0.5 h after one-lung ventilation (T2), and 15 minutes after inhaling air before leaving the room (T3). The content of IL-8 in arterial blood was measured by enzyme-linked immunosorbent assay (ELISA) at T0 and T2, and the expression of AQP1 protein in isolated lung tissue was measured by immunohistochemistry and Western blot. The incidence of postoperative pulmonary complications (atelectasis, pneumonia, and acute respiratory distress syndrome) was used as the index of lung injury. Results There was no significant difference in the general condition before and during operation between the two groups. There was no significant difference in arterial blood IL-8 content between the two groups at the T0 time point, but the arterial blood IL-8 content at the T2 time point was significantly higher than that at the T0 time point, especially in group C. The results of immunohistochemistry and Western blot showed that the expression level of AQP1 protein in the isolated lung tissue of group D was significantly higher than that of group C (P < 0 05). At T3, the OI of group D was significantly higher than that of group C, and the A-aDO2 of group D was significantly lower than that of group C (P < 0.05). There was no significant difference in the incidence of postoperative PPCs between the two groups. Conclusion Dexmedetomidine can reduce the level of plasma IL-8 and upregulate the expression of AQP1 in the lung tissue of patients undergoing thoracoscopic surgery under one-lung ventilation, but it has no significant effect on the incidence of postoperative PPCs. Dexmedetomidine can be safely used in thoracoscopic surgery and has a certain protective effect on lung injury.
Collapse
|
9
|
Castillo RL, Ibacache M, Cortínez I, Carrasco-Pozo C, Farías JG, Carrasco RA, Vargas-Errázuriz P, Ramos D, Benavente R, Torres DH, Méndez A. Dexmedetomidine Improves Cardiovascular and Ventilatory Outcomes in Critically Ill Patients: Basic and Clinical Approaches. Front Pharmacol 2020; 10:1641. [PMID: 32184718 PMCID: PMC7058802 DOI: 10.3389/fphar.2019.01641] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenergic agonist with sedative and analgesic properties, with minimal respiratory effects. It is used as a sedative in the intensive care unit and the operating room. The opioid-sparing effect and the absence of respiratory effects make dexmedetomidine an attractive adjuvant drug for anesthesia in obese patients who are at an increased risk for postoperative respiratory complications. The pharmacodynamic effects on the cardiovascular system are known; however the mechanisms that induce cardioprotection are still under study. Regarding the pharmacokinetics properties, this drug is extensively metabolized in the liver by the uridine diphosphate glucuronosyltransferases. It has a relatively high hepatic extraction ratio, and therefore, its metabolism is dependent on liver blood flow. This review shows, from a basic clinical approach, the evidence supporting the use of dexmedetomidine in different settings, from its use in animal models of ischemia-reperfusion, and cardioprotective signaling pathways. In addition, pharmacokinetics and pharmacodynamics studies in obese subjects and the management of patients subjected to mechanical ventilation are described. Moreover, the clinical efficacy of delirium incidence in patients with indication of non-invasive ventilation is shown. Finally, the available evidence from DEX is described by a group of Chilean pharmacologists and clinicians who have worked for more than 10 years on DEX.
Collapse
Affiliation(s)
- Rodrigo L Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Unidad de Paciente Crítico, Hospital del Salvador, Santiago, Chile
| | - Mauricio Ibacache
- Programa de Farmacología y Toxicología & División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Cortínez
- Programa de Farmacología y Toxicología & División de Anestesiología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Carrasco-Pozo
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jorge G Farías
- Departmento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Francisco Salazar, Chile
| | - Rodrigo A Carrasco
- Departamento de Cardiología, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Patricio Vargas-Errázuriz
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago, Chile.,Unidad de Paciente Crítico Adulto, Clínica Universidad de Los Andes, Santiago, Chile.,Unidad de Paciente Crítico, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Daniel Ramos
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rafael Benavente
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Henríquez Torres
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Aníbal Méndez
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
10
|
Li J, Yang L, Wang G, Wang Y, Wang C, Shi S. Severe systemic inflammatory response syndrome in patients following Total aortic arch replacement with deep hypothermic circulatory arrest. J Cardiothorac Surg 2019; 14:217. [PMID: 31842939 PMCID: PMC6916067 DOI: 10.1186/s13019-019-1027-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/18/2019] [Indexed: 11/25/2022] Open
Abstract
Background This cohort study aims to retrospectively investigate the incidence of severe systemic inflammatory response syndrome (sSIRS) in patients following total aortic arch replacement (TAR) under deep hypothermic circulatory arrest (DHCA) with selective cerebral perfusion and its effect on clinical outcomes. Methods All patients who underwent TAR with DHCA were consecutively enrolled from January 2013 until December 2015 at our institute. sSIRS was diagnosed between 12 and 48 h postoperatively if patients met all four criteria of the SIRS definition. Results Of the 522 patients undergoing TAR with DHCA, 31.4% developed sSIRS. Patients aged under 60 yr were characterized by a higher prevalence of sSIRS (OR = 2.93; 95% CI 2.01–4.28; P <0.001). Higher baseline serum creatinine (OR = 1.61; 95% CI 1.18–2.20; P = 0.003), concomitant coronary disease (OR = 2.00; 95% CI 1.15–3.48; P = 0.015) and extended cardiopulmonary time (OR = 1.63; 95% CI 1.23–2.18; P = 0.001) independently contributed to a greater likelihood of postoperative sSIRS onset, while the preferred administration of ulinastatin (OR = 0.69; 95% CI 0.51–0.93; P = 0.015) and dexmedetomidine (OR = 0.36; 95% CI 0.23–0.56; P < 0.001) attenuated it. Patients with sSIRS had a greater risk of developing postoperative major adverse complications compared with the no sSIRS group [56.7%(93/164) vs 26.8% (96/358), P < 0.001]. sSIRS was found to be a significant risk factor for major adverse complications (OR, 4.52; 95% CI, 3.40–6.01; P < 0.001). A significant difference was revealed in in-hospital death following TAR between the sSIRS group and the no-sSIRS group [4.88% (8/164) vs 1.12% (4/358), P = 0.019]. The Kaplan-Meier curve indicated that the time to discharge from the intensive care unit was significantly prolonged in the sSIRS group compared with patients without it (log-rank p < 0.001). Conclusions sSIRS occurs commonly in patients following TAR with DHCA. There is an inverse association between age and sSIRS onset, whereby age over 60 yr can lower the risk of it. sSIRS development can increase the likelihood of major postoperative major adverse events.
Collapse
Affiliation(s)
- Jun Li
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Belishi road 167, Xicheng District, Beijing, 100037, China
| | - Lijing Yang
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Belishi road 167, Xicheng District, Beijing, 100037, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yuefu Wang
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Belishi road 167, Xicheng District, Beijing, 100037, China.
| | - Chunrong Wang
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Belishi road 167, Xicheng District, Beijing, 100037, China
| | - Sheng Shi
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Belishi road 167, Xicheng District, Beijing, 100037, China
| |
Collapse
|
11
|
Ding M, Chen Y, Luan H, Zhang X, Zhao Z, Wu Y. Dexmedetomidine reduces inflammation in traumatic brain injury by regulating the inflammatory responses of macrophages and splenocytes. Exp Ther Med 2019; 18:2323-2331. [PMID: 31410183 PMCID: PMC6676199 DOI: 10.3892/etm.2019.7790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) affects people in all demographics, since it is associated with a variety of chronic degenerative diseases, such as Alzheimer's and Parkinson's disease. In TBI, the central nervous system elicits an immune response involving various immune cells that is necessary for healing and defending the body against pathogens, but can also cause secondary damage to the brain if the response is prolonged. In our clinical practice, it has been identified that administration of dexmedetomidine was associated with reduced production of inflammatory cytokines in patients with TBI, which led to the hypothesis that dexmedetomidine may regulate certain inflammatory responses. To test this hypothesis, the roles of dexmedetomidine in the immune system of mice were investigated. Different biological assays were used to assess the influence of dexmedetomidine on the production of inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-8 and IL-1β. To understand how dexmedetomidine affects different types of immune cells, the influence of dexmedetomidine on splenocytes was also investigated. Finally, the effects of dexmedetomidine on macrophage activation and inflammatory functions were studied. In the present study, clinical observations and in vivo results using a mouse model of TBI revealed the regulatory functions of dexmedetomidine in TBI-associated immune response.
Collapse
Affiliation(s)
- Mengyao Ding
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Ying Chen
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Hengfei Luan
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Xiaobao Zhang
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Zhibin Zhao
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Yong Wu
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
12
|
Dardalas I, Stamoula E, Rigopoulos P, Malliou F, Tsaousi G, Aidoni Z, Grosomanidis V, Milonas A, Papazisis G, Kouvelas D, Pourzitaki C. Dexmedetomidine effects in different experimental sepsis in vivo models. Eur J Pharmacol 2019; 856:172401. [PMID: 31108055 DOI: 10.1016/j.ejphar.2019.05.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
Sepsis is a major cause of death and the most common cause of death among critically ill, non-ICU patients. Dexmedetomidine (DEX), an <alpha>2 adrenergic receptor agonist, presents sympatholytic action in certain parts of the brain with anxiolytic, sedative, and pain killing effects. Additionally, through the activation of <alpha>7 nicotinic acetylcholine receptor receptors, DEX reduces cytokine transcription and inhibits inflammation, rendering it beneficial during septic conditions. Moreover, there is a lot of interest in designing experimental sepsis models, where the administration of DEX is evaluated for its impact on multiple systems. This review focuses on experimental studies published between 1999 to March 2019 that were using DEX administration in sepsis in vivo models. From these, 36 articles were selected and summarized. Overall results show evidence that DEX may decrease mortality and inhibit inflammation, as it enhances the activity of the immune system while reducing its systemic reaction and lowering cytokine concentrations. Moreover DEX succeeds to alleviate heart injury during sepsis, acting beneficially for microcirculation and shows a neuroprotective role by inhibiting apoptotic pathways. In addition, DEX appears to have a protective role for liver and spleen as well as a beneficial role for the function of lungs and kidneys as it reduces sepsis-induced injuries and apoptosis in intra-abdominal experimental sepsis models.
Collapse
Affiliation(s)
- Ioannis Dardalas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni Stamoula
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Panagiotis Rigopoulos
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Faye Malliou
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Georgia Tsaousi
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Zoi Aidoni
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Vasileios Grosomanidis
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Antonios Milonas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Georgios Papazisis
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Chryssa Pourzitaki
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
13
|
Ji X, Guo Y, Zhou G, Wang Y, Zhang J, Wang Z, Wang Q. Dexmedetomidine protects against high mobility group box 1-induced cellular injury by inhibiting pyroptosis. Cell Biol Int 2019; 43:651-657. [PMID: 30958608 DOI: 10.1002/cbin.11140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/23/2019] [Indexed: 01/04/2023]
Abstract
Dexmedetomidine (DEX) is a widely used clinical anesthetic with proven anti-inflammatory effects. Both high mobility group box 1 (HMGB1) and pyroptosis play an important role in the inflammatory response to infection and trauma. Thus far, there have been no studies published addressing the effect of DEX on HMGB1 and pyroptosis. In order to fill this gap in the literature, bone marrow-derived macrophages (BMDMs) were exposed to HMGB1 (4 µg/mL) with or without DEX (50 μM) pretreatment. The production of pro-inflammatory cytokines [such as tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and IL-18], phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) and P38, and the activation of caspase-1 were measured by enzyme immunosorbent assay, western blot analysis, confocal microscope, and flow cytometry, respectively. We found that DEX protected against HMGB1-induced cell death of BMDMs. In addition, DEX suppressed the generation of TNF-α, IL-1β, and IL-18 as well as the phosphorylation of ERK1/2 and P38. Moreover, DEX inhibited caspase-1 activation and decreased pyroptosis. Taken together, these findings demonstrate the protective effect of DEX in mediating HMGB1-induced cellular injury, thus indicating that DEX may be a potential therapeutic candidate for the management of infection and trauma-derived inflammation.
Collapse
Affiliation(s)
- Xuexia Ji
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Yuanbo Guo
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Guobin Zhou
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Yan Wang
- Department of Science and Education, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Jianxing Zhang
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Zhipeng Wang
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Qing Wang
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| |
Collapse
|
14
|
Kim SH, Kim DH, Shin S, Kim SJ, Kim TL, Choi YS. Effects of dexmedetomidine on inflammatory mediators after tourniquet-induced ischemia-reperfusion injury: a randomized, double-blinded, controlled study. Minerva Anestesiol 2019; 85:279-287. [DOI: 10.23736/s0375-9393.18.13015-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Jiang Y, Xia M, Huang Q, Ding D, Li Y, Zhang Z, Zhang X. Protective effect of dexmedetomidine against organ dysfunction in a two-hit model of hemorrhage/resuscitation and endotoxemia in rats. ACTA ACUST UNITED AC 2019; 52:e7905. [PMID: 30810621 PMCID: PMC6393854 DOI: 10.1590/1414-431x20187905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 11/11/2018] [Indexed: 11/22/2022]
Abstract
Dexmedetomidine (DEX), a selective agonist of α2-adrenergic receptors, has anti-inflammation properties and potential beneficial effects against trauma, shock, or infection. Therefore, this study aimed to investigate whether DEX might protect against multiple-organ dysfunction in a two-hit model of hemorrhage/resuscitation (HS) and subsequent endotoxemia. Eighty Wistar rats were randomized into four groups: NS (normal saline), HS/L (HS plus lipopolysaccharide), HS/L+D (HS/L plus dexmedetomidine), and HS/L+D+Y (HS/L+D plus yohimbine). Six hours after resuscitation, blood gas (PaO2) and serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urine nitrogen (BUN), creatinine (Cr), TNF-α, IL-β, IL-6, IL-8, IL-10, and nitric oxide (NO) were measured. The histopathology was assayed by staining. Malondialdehyde (MDA) and superoxide dismutase (SOD) levels and heme oxygenase-1 (HO-1) were assayed. The PaO2 levels in HS/L rats were lower whereas the ALT, AST, BUN, Cr, TNF-α, IL-β, IL-6, IL-8, IL-10, and NO levels were higher compared to the control group. The HS/L+D increased PaO2 and further increased IL-10 and decreased ALT, AST, BUN, Cr, TNF-α, IL-β, IL-6, IL-8, and NO levels of the HS/L groups. In addition, the MDA in the HS/L groups increased whereas SOD activity decreased compared to the control group. Moreover, the HO-1 expression levels were increased by DEX administration in lung, liver, and kidney tissues. Lungs, livers, and kidneys of the HS/L group displayed significant damage, but such damage was attenuated in the HS/L+D group. All of the above-mentioned effects of DEX were partly reversed by yohimbine. DEX reduced multiple organ injury caused by HS/L in rats, which may be mediated, at least in part, by α2-adrenergic receptors.
Collapse
Affiliation(s)
- Yuanxu Jiang
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Mingzhu Xia
- Hubei Community Health Service Center, Luohu Hospital group, Shenzhen, China
| | - Qiang Huang
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Dengfeng Ding
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Yali Li
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Zhongjun Zhang
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Xueping Zhang
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, the Second Clinical Medical College, Jinan University, Shenzhen, China
| |
Collapse
|
16
|
Intraoperative use of dexmedetomidine for the prevention of emergence agitation and postoperative delirium in thoracic surgery: a randomized-controlled trial. Can J Anaesth 2019; 66:371-379. [DOI: 10.1007/s12630-019-01299-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/27/2018] [Accepted: 10/10/2018] [Indexed: 12/16/2022] Open
|
17
|
Abad-Gurumeta A, Gómez-Ríos MÁ, Calvo-Vecino JM. Intravenous dexmedetomidine: can it modulate the effects of inflammation, or is it only an antinociceptive agent? Minerva Anestesiol 2018; 85:226-228. [PMID: 30484298 DOI: 10.23736/s0375-9393.18.13217-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Alfredo Abad-Gurumeta
- Department of Anesthesiology and Perioperative Medicine, Infanta Leonor University Hospital, Madrid, Spain -
| | - Manuel Á Gómez-Ríos
- Department of Anesthesiology and Perioperative Medicine, University Hospital of A Coruña, A Coruña, Spain.,Anesthesiology and Pain Management Research Group, Institute for Biomedical Research of A Coruña (INIBIC), A Coruña, Spain.,Spanish Difficult Airway Group (GEVAD), A Coruña, Spain
| | - José M Calvo-Vecino
- Department of Anesthesia and Perioperative Medicine, University Care Complex of Salamanca, Salamanca, Spain
| |
Collapse
|
18
|
Deng Y, Tan F, Gan X, Li X, Ge M, Gong C, Hei Z, Zhu Q, Zhou S. Perioperative application of dexmedetomidine for postoperative systemic inflammatory response syndrome in patients undergoing percutaneous nephrolithotomy lithotripsy: results of a randomised controlled trial. BMJ Open 2018; 8:e019008. [PMID: 30391912 PMCID: PMC6231562 DOI: 10.1136/bmjopen-2017-019008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Our previous retrospective study demonstrated that perioperative dexmedetomidine (Dex) administration was associated with low systemic inflammatory response syndrome (SIRS) incidence. The present study was designed to investigate whether perioperative administration of Dex decreases the incidence of postpercutaneous nephrolithotomy lithotripsy (PCNL) SIRS in patients who undergo PCNL. DESIGN A randomised controlled trial was designed. PARTICIPANTS A total of 190 patients were randomly assigned to receive Dex (DEX group, n=95) or saline control (CON group, n=95) and completed the study. In the DEX group, Dex was loaded (1 µg/kg) before anaesthesia induction and was infused (0.5 µg/kg/h) during surgery. OUTCOMES The incidences of postoperative SIRS were recorded. Serum interleukin-6 (IL-6) and tumour necrosis factor α(TNF-α) were measured. RESULTS The incidence rates of SIRS were significantly lower in the DEX group than in the CON group (35.8% vs 50.5%, p=0.04). No patients developed sepsis in either group. These results might be attributed to inhibition of inflammatory responses and the resulting lower serum levels of IL-6 and TNF-α, caused by Dex administration. However, compared with the CON group, the lower incidence rate of SIRS in the DEX group did not result in better outcomes, such as shorter postoperative hospitalisation stays and lower costs. CONCLUSION The present study showed that Dex administration during PCNL might be beneficial for decreasing the incidence of SIRS through inhibiting the release of inflammatory mediators, but not clinical consequences such as postoperative hospitalisation duration and costs. Further effects of Dex administration on SIRS in patients who are scheduled for PCNL should be explored in future studies. TRIAL REGISTRATION NUMBER ChiCTR-ICR-15006167.
Collapse
Affiliation(s)
- Yingqing Deng
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Fang Tan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaoliang Gan
- Department of Anesthesiology, Zhongshan Ophthalmic Center of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyun Li
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mian Ge
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chulian Gong
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qianqian Zhu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shaoli Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Ma Y, Yu XY, Wang Y. Dose-related effects of dexmedetomidine on immunomodulation and mortality to septic shock in rats. World J Emerg Med 2018; 9:56-63. [PMID: 29290897 DOI: 10.5847/wjem.j.1920-8642.2018.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Dexmedetomidine has already been used in septic patients as a new sedative agent, few studies have examined its effects on immunomodulation. Therefore, the authors have designed a controlled experimental study to characterize the immunomodulation effects of dexmedetomidine in the cecal ligation and puncture (CLP) model in rats. METHODS After CLP, 48 Wistar rats were randomly allocated into four groups: (1) CLP group; (2) small-dose treatment group (2.5 μg·kg-1·h-1); (3) medium-dose treatment group (5.0 μg·kg-1· h-1); and (4) large-dose treatment group (10.0 μg·kg-1·h-1). HLA-DR and plasma cytokine (IL-4, IL-6, IL-10 and TNF-α) levels were measured, and the mean arterial blood pressure (MAP), heart rate (HR), arterial blood gases, lactate concentrations and mortality were also documented. RESULTS The HLA-DR level, inflammatory mediator levels, MAP and HR had no obvious changes among Dexmedetomidine treatment groups (DEX groups). Compared with the CLP group, the DEX groups exhibited decreased HLA-DR levels (Pgroup=0.0202) and increased IL-6 production, which was increased at 3 h (P= 0.0113) and was then attenuated at 5 h; additionally, the DEX groups exhibited decreased HR (P<0.001) while maintaining MAP (Pgroup=0.1238), and remarkably improving lactate (P<0.0001). All of these factors led to a significant decrease in the mortality, with observed rates of 91.7%, 66.7%, 25% and 18% for the CLP, DEX2.5, DEX5.0, DEX10.0 groups, respectively. CONCLUSION Dexmedetomidine treatment in the setting of a CLP sepsis rat model has partially induced immunomodulation that was initiated within 5 h, causing a decreased HR while maintaining MAP, remarkably improving metabolic acidosis and improving mortality dose-dependently.
Collapse
Affiliation(s)
- Yan Ma
- Department of Intensive Care Unit, the First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Xiang-You Yu
- Department of Intensive Care Unit, the First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Yi Wang
- Department of Intensive Care Unit, the First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
20
|
Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M, Marriott A, Moore EM, Morris G, Page RS, Gray L. Post-Operative Cognitive Dysfunction: An exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev 2017; 84:116-133. [PMID: 29180259 DOI: 10.1016/j.neubiorev.2017.11.011] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/16/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Post-Operative Cognitive Dysfunction (POCD) is a highly prevalent condition with significant clinical, social and financial impacts for patients and their communities. The underlying pathophysiology is becoming increasingly understood, with the role of neuroinflammation and oxidative stress secondary to surgery and anaesthesia strongly implicated. This review aims to describe the putative mechanisms by which surgery-induced inflammation produces cognitive sequelae, with a focus on identifying potential novel therapies based upon their ability to modify these pathways.
Collapse
Affiliation(s)
- David R Skvarc
- School of Psychology, Deakin University, Melbourne, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia.
| | - Michael Berk
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| | - Linda K Byrne
- School of Psychology, Deakin University, Melbourne, Australia.
| | - Olivia M Dean
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Seetal Dodd
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia
| | - Matthew Lewis
- School of Psychology, Deakin University, Melbourne, Australia; Aged Psychiatry Service, Caulfield Hospital, Alfred Health, Caulfield, Australia
| | - Andrew Marriott
- Department of Anaesthesia, Perioperative Medicine & Pain Management, Barwon Health, Geelong, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Eileen M Moore
- Department of Anaesthesia, Perioperative Medicine & Pain Management, Barwon Health, Geelong, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia
| | | | - Richard S Page
- Deakin University, School of Medicine, Geelong, Australia; Department of Orthopaedics, Barwon Health, Geelong, Australia
| | - Laura Gray
- Deakin University, School of Medicine, Geelong, Australia.
| |
Collapse
|
21
|
Nie B, Zhang S, Huang Z, Huang J, Chen X, Zheng Y, Bai X, Zeng W, Ouyang H. Synergistic Interaction Between Dexmedetomidine and Ulinastatin Against Vincristine-Induced Neuropathic Pain in Rats. THE JOURNAL OF PAIN 2017; 18:1354-1364. [PMID: 28690001 DOI: 10.1016/j.jpain.2017.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/20/2017] [Accepted: 06/26/2017] [Indexed: 12/29/2022]
Abstract
Antimicrotubulin chemotherapeutic agents such as vincristine (VCR), often induce peripheral neuropathic pain. It is usually permanent and seriously harmful to cancer patients' quality of life and can result in the hampering of clinical treatments. Currently, there is no definitive therapy, and many of the drugs approved for the treatment of other neuropathic pain have shown little or no analgesic effect. It is therefore vital to find new and novel therapeutic strategies for patients suffering from chemotherapeutic agent-induced neuropathic pain to improve patients' quality of life. This study shows that intrathecal injections of dexmedetomidine (DEX), or intraperitoneally administered ulinastatin (UTI) significantly reduces Sprague Dawley rats' mechanical allodynia induced by VCR via upregulation of interleukin-10 expression and activating the α2-adrenergic receptor in dorsal root ganglion (DRG). Moreover, when combined there is a synergistic interaction between DEX and UTI, which acts against VCR-induced neuropathic pain. This synergistic interaction between DEX and UTI may be partly attributed to a common analgesic pathway in which the upregulation of interleukin -10 plays an important role via activating α2-adrenergic receptor in rat dorsal root ganglion. The combined use of DEX and UTI does not affect the rat's blood pressure, heart rate, sedation, motor score, spatial learning, or memory function. All of these show that the combined use of DEX and UTI is an effective method in relieving VCR-induced neuropathic pain in rats. PERSPECTIVE This article documents the synergistic interaction between 2 widely used drugs, DEX and UTI, against VCR-induced neuropathic pain. The results provide a potential target and novel drug administrated method for the clinical treatment of chemotherapy-induced peripheral neuropathic pain.
Collapse
Affiliation(s)
- Bilin Nie
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China; Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Subo Zhang
- Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, Guangzhou, China; Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhuxi Huang
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jingxiu Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaodi Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yaochao Zheng
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaohui Bai
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
22
|
Strahler J, Skoluda N, Rohleder N, Nater UM. Dysregulated stress signal sensitivity and inflammatory disinhibition as a pathophysiological mechanism of stress-related chronic fatigue. Neurosci Biobehav Rev 2016; 68:298-318. [DOI: 10.1016/j.neubiorev.2016.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/03/2016] [Accepted: 05/10/2016] [Indexed: 01/20/2023]
|
23
|
Wu J, Li ST. Dexmedetomidine May Produce Extra Protective Effects on Sepsis-induced Diaphragm Injury. Chin Med J (Engl) 2016; 128:1407-11. [PMID: 25963365 PMCID: PMC4830324 DOI: 10.4103/0366-6999.156808] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The objective was to evaluate the protective effects of dexmedetomidine (DEX), a selective agonist of α2-adrenergic receptor, on sepsis-induced diaphragm injury and the underlying molecular mechanisms. DATA SOURCES The data used in this review were mainly from PubMed articles published in English from 1990 to 2015. STUDY SELECTION Clinical or basic research articles were selected mainly according to their level of relevance to this topic. RESULTS Sepsis could induce severe diaphragm dysfunction and exacerbate respiratory weakness. The mechanism of sepsis-induced diaphragm injury includes the increased inflammatory cytokines and excessive oxidative stress and superfluous production of nitric oxide (NO). DEX can reduce inflammatory cytokines, inhibit nuclear factor-kappaB signaling pathways, suppress the activation of caspase-3, furthermore decrease oxidative stress and inhibit NO synthase. On the basis of these mechanisms, DEX may result in a shorter period of mechanical ventilation in septic patients in clinical practice. CONCLUSIONS Based on this current available evidence, DEX may produce extra protective effects on sepsis-induced diaphragm injury. Further direct evidence and more specific studies are still required to confirm these beneficial effects.
Collapse
Affiliation(s)
| | - Shi-Tong Li
- Department of Anesthesiology, First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| |
Collapse
|
24
|
Tian XH, Jiang WS, Li XL, Li MF, Liu CL, Li XX. Protective effect of fasudil hydrochloride against acute renal injury in septicopyemia rats. ASIAN PAC J TROP MED 2015; 8:1071-1075. [PMID: 26706682 DOI: 10.1016/j.apjtm.2015.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/20/2015] [Accepted: 11/03/2015] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE To observe the protective effect of fasudil hydrochloride against acute renal injury in septicopyemia rats. METHODS A total of 60 Wister rats were included in the study and divided into control group (n = 10), model group (n = 25) and treatment group (n = 25). Model group and treatment group received intraperitoneal injection of endotoxin (ET) to establish acute renal injury models while the control group only received daily intraperitoneal injection of normal saline 1 mL. Five rats were taken out of model group and treatment group respectively at 1 h (T1), 6 h (T2), 12 h (T3), 24 h (T4) and 48 h (T5), for intraperitoneal injection of ET 30 mg/kg. Treatment group received intraperitoneal injection of fasudil hydrochloride 30 mg/kg 1 h before injection of ET. For three groups, 5 mL blood samples were collected from postcava for determination of serum creatinine and urea nitrogen levels at different time points. Concentrations of serum tumor necrosis factor α and ET-1 were determined by using ELISA. The renal pathologic changes were observed under the microscope. RESULTS Serum creatinine levels in both model group and treatment group were significantly higher than control group at T2-T5 (P < 0.05) while the levels in treatment group were significantly lower than control group at T3-T5 (P < 0.05). At T2-T5, blood urea nitrogen levels in model group and treatment group were significantly higher than control group (P < 0.05) while the levels in treatment group were significantly lower than model group at T3-T5 (P < 0.05). Concentrations of serum tumor necrosis factor α in model group and treatment group were significantly higher than control group at T1-T5 (P < 0.05) while the levels in treatment group were significantly lower than model group at T1-T5 (P < 0.05). Serum ET-1 concentrations in model group and treatment group were significantly higher than control group at T1-T5 (P < 0.05) while the levels in treatment group at T1-T4 were significantly lower than model group (P < 0.05). Rats in control group showed no swelling or hyperemia in kidney cells but normal structure and normally arranged renal tubular epithelial cells. Obvious injury was observed in model group at T3 and renal tubular epithelial cells in disorder and at swelling condition, hyperemia and angiectasis in glomerulus, degenerative opacities and vacuolar degeneration, and maximized injury were observed at T4. Injury in renal tissue in treatment group was significantly milder than model group. CONCLUSIONS Fasudil hydrochloride has the significantly protective effect against acute renal injury in septicopyemia rats.
Collapse
Affiliation(s)
- Xing-Han Tian
- Department of Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical College, Yantai, Shandong 264000, China
| | - Wen-Shi Jiang
- Emergency Department, Yangshuo People's Hospital, Yangshuo County, Guilin, Guangxi 541999, China.
| | - Xiao-Li Li
- Department of Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical College, Yantai, Shandong 264000, China
| | - Mei-Feng Li
- Department of Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical College, Yantai, Shandong 264000, China
| | - Chao-Liang Liu
- Emergency Department, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Xiao-Xia Li
- Emergency Department, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| |
Collapse
|
25
|
Li B, Li Y, Tian S, Wang H, Wu H, Zhang A, Gao C. Anti-inflammatory Effects of Perioperative Dexmedetomidine Administered as an Adjunct to General Anesthesia: A Meta-analysis. Sci Rep 2015. [PMID: 26196332 PMCID: PMC4508837 DOI: 10.1038/srep12342] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The aim of this meta-analysis is to examine the effects of dexmedetomidine on serum inflammatory markers when administered perioperatively. We searched multiple electronic databases for relevant research papers, and carried out meta-analyses of weighted mean differences and interpreted in the light of statistical heterogeneity (I2). Fifteen RCTs recruiting 641 patients were included. Dexmedetomidine treatment significantly decreased interleukin-6 (IL-6), IL-8 and tumor necrosis factor-alpha (TNF-α) levels with mean differences [95% CI] in the changes from baseline between dexmedetomidine treated and controls of −25.14 [−35.29, −15.00]; P < 0.00001 (for IL-6), −5.69 [−10.77, −0.60]; P < 0.04 (for IL-8), and −20.30 [−30.93, −9.67]; P < 0.0002 (for TNF-α) immediately after surgery; and −41.55 [−57.41, −25.70]; P < 0.00001 (IL-6), −6.46 [−10.83, −2.08]; P < 0.005 (IL-8), and −14.67 [−22.61, −6.73]; P < 0.0003 (TNF-α) on postoperative day 1 (random effects). IL-10 levels were found to increase significantly a day after surgery (8.33 [3.31, 13.36]; P = 0.001). Subgroup analyses did not reveal significant differences. In conclusion, perioperative adjunctive use of dexmedetomidine substantially decreases serum IL-6, IL-8 and TNF-α levels.
Collapse
Affiliation(s)
- Bo Li
- Department of Anesthesiology, Jinan General Hospital, PLA Jinan Military Area Command, Jinan 250031, Shandong, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China
| | - Shushi Tian
- Department of Radiology, No. 261 Hospital of PLA, Beijing, China
| | - Huixia Wang
- Department of Anesthesiology, Jinan General Hospital, PLA Jinan Military Area Command, Jinan 250031, Shandong, China
| | - Hui Wu
- Department of Anesthesiology, The People's Hospital of Zhangqiu, Zhangqiu 250200, Henan, China
| | - Aihua Zhang
- Department of Anesthesiology, Yancheng City No.1 People's Hospital, Yancheng 224005, Jiangsu, China
| | - Chengjie Gao
- Department of Anesthesiology, Jinan General Hospital, PLA Jinan Military Area Command, Jinan 250031, Shandong, China
| |
Collapse
|
26
|
Das A, Majumdar S, Halder S, Chattopadhyay S, Pal S, Kundu R, Mandal SK, Chattopadhyay S. Effect of dexmedetomidine as adjuvant in ropivacaine-induced supraclavicular brachial plexus block: A prospective, double-blinded and randomized controlled study. Saudi J Anaesth 2014; 8:S72-7. [PMID: 25538527 PMCID: PMC4268534 DOI: 10.4103/1658-354x.144082] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background and Aims: Different additives have been used to prolong brachial plexus block. We evaluated the effect of adding dexmedetomidine to ropivacaine for supraclavicular brachial plexus blockade. The primary endpoints were the onset and duration of sensory and motor block and duration of analgesia. Materials and Methods: A total of 84 patients (20-50 years) posted for elective forearm and hand surgery under supraclavicular brachial plexus block were divided into two equal groups (Group R and RD) in a randomized, double-blind fashion. In group RD (n = 42) 30 ml 0.5% ropivacaine +1 ml (100 μg) of dexmedetomidine and group R (n = 42) 30 ml 0.5% ropivacaine +1 ml normal saline were administered in supraclavicular block. Sensory and motor block onset times and block durations, time to first analgesic use, total analgesic need, postoperative visual analog scale (VAS), hemodynamics and side-effects were recorded for each patient. Results: Though with similar demographic profile in both groups, sensory and motor block in group RD (P < 0.05) was earlier than group R. Sensory and motor block duration and time to first analgesic use were significantly longer and the total need for rescue analgesics was lower in group RD (P < 0.05) than group R. Post-operative VAS value at 12 h were significantly lower in group RD (P < 0.05). Intra-operative hemodynamics were significantly lower in group RD (P < 0.05) without any appreciable side-effects. Conclusion: It can be concluded that adding dexmedetomidine to supraclavicular brachial plexus block increases the sensory and motor block duration and time to first analgesic use, and decreases total analgesic use with no side-effects.
Collapse
Affiliation(s)
- Anjan Das
- Department of Anaesthesiology, College of Medicine & Sagore Dutta Hospital, Kolkata, West Bengal, India
| | - Saikat Majumdar
- Department of Anaesthesiology, NRS Medical College, Kolkata, West Bengal, India
| | - Susanta Halder
- Department of Anaesthesiology, R. G. Kar Medical College, Kolkata, West Bengal, India
| | - Surajit Chattopadhyay
- Department of Anaesthesiology, Bankura Sammilani Medical College, Bankura, West Bengal, India
| | - Saswati Pal
- Department of Anaesthesiology, NRS Medical College, Kolkata, West Bengal, India
| | - Ratul Kundu
- Department of Anaesthesiology, Institute of Post Graduate Medical Education And Research, Kolkata, West Bengal, India
| | - Subrata Kumar Mandal
- Department of Anaesthesiology, College of Medicine & Sagore Dutta Hospital, Kolkata, West Bengal, India
| | - Sandip Chattopadhyay
- Department of G & O, College of Medicine & Sagore Dutta Hospital, Kolkata, West Bengal, India
| |
Collapse
|
27
|
Ueki M, Kawasaki T, Habe K, Hamada K, Kawasaki C, Sata T. The effects of dexmedetomidine on inflammatory mediators after cardiopulmonary bypass. Anaesthesia 2014; 69:693-700. [DOI: 10.1111/anae.12636] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2014] [Indexed: 02/01/2023]
Affiliation(s)
- M. Ueki
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - T. Kawasaki
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - K. Habe
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - K. Hamada
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - C. Kawasaki
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - T. Sata
- University of Occupational and Environmental Health; Kitakyushu Japan
| |
Collapse
|
28
|
Wang H, Hu B, Zou Y, Bo L, Wang J, Li J, Luo Y. Dexmedetomidine premedication attenuates concanavalin A-induced hepatitis in mice. J Toxicol Sci 2014; 39:755-64. [PMID: 25242406 DOI: 10.2131/jts.39.755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Haibin Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, China
| | - Baoji Hu
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, China
| | - Yun Zou
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Lulong Bo
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Jun Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Jinbao Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, China
| |
Collapse
|