1
|
Liu MW, Duan SX, Zhao XY, Wang QF, Yang SL, Ma N, Li X. Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). Int J Mol Med 2025; 55:94. [PMID: 40242975 PMCID: PMC12045470 DOI: 10.3892/ijmm.2025.5535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Sepsis‑induced organ dysfunction syndrome (ODS) arises from a dysregulated response to infection, leading to multiple life‑threatening organ dysfunctions, and is a common complication in critically ill patients. Sepsis results in varying degrees of injury to the brain, lungs, kidneys and liver, culminating in immune dysfunction and multiple ODS (MODS). Current evidence indicates a direct correlation between the severity of organ injury and the prognosis of septic patients. Understanding the mechanisms of MODS in sepsis and developing effective management strategies are vital research areas. The protective effects of dexmedetomidine (DEX) on sepsis are well established, demonstrating its capacity to mitigate injuries to the brain, lungs, kidneys, liver and immune system. The present study reviews recent research progress on the role and mechanisms of action of DEX in the treatment of sepsis.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency Medicine, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Shao-Xin Duan
- Department of Trauma, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Xue-Yan Zhao
- Department of Endocrinology, First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Qiong-Fen Wang
- Department of Gastroenterology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Shan-Lan Yang
- Department of Oncology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Ni Ma
- Department of Pharmacy, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Xuan Li
- Department of Rehabilitation, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
2
|
Zhou Q, Ali S, Shi X, Cao G, Feng J, Yang C, Zhang R. Protective impacts of bamboo leaf flavonoids in stressed broilers induced by diquat: Insight of antioxidant, immune response and intestinal barrier function. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:158-170. [PMID: 39967701 PMCID: PMC11834062 DOI: 10.1016/j.aninu.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 02/20/2025]
Abstract
This research explored the protective impact of bamboo leaf flavonoids (BLF) in diquat (DQ) stressed broilers; providing insight of antioxidant, immune response and intestinal barrier function. This experiment consisted of two parts. In the first, 240 chicks were allotted to 2 groups with 8 replicates and 15 chicks per replicate. Treatments consisted of a basic feed (control group, CON) and the basic feed plus 1000 mg/kg BLF (BLF group, BLF) for 28 d, respectively. Then, following the conclusion of the first part, 16 healthy broilers were selected from the CON group and the BLF group. They formed the second part of the experiment, and were allotted to 4 treatments with 8 broilers each: CON-no stress (CON-NS) group, CON-DQ group, BLF-NS group and the BLF-DQ group. Broilers were separately injected intraperitoneally with DQ solution at 40 mg/kg body weight or the same dose of phosphate buffer saline. The results revealed adding BLF to diet reduced the ratio of feed to weight gain of broilers compared to the basic feed group (P = 0.021). In comparison to the CON-NS group, BLF improved the levels of serum and jejunal mucosa total antioxidant capacity, immunoglobulin M, serum catalase, immunoglobulin A, interleukin 10, jejunal mucosa interleukin 4, cecal butyric acid, valeric acid, isobutyric acid, isovaleric acid, upregulated zonula occludens-1 (ZO-1), occludin (OCLN) and claudin-1 (CLDN1) expressions, and reduced the levels of jejunal mucosa malondialdehyde (MDA), interleukin 1β, interleukin 6 and serum diamine oxidase (P < 0.05). Diquat stress elevated the contents of serum MDA, D-lactate, jejunal mucosa tumor necrosis factor α, reactive oxygen species and unclassified_f_Lachnospiraceae relative abundance, downregulated ZO-1, OCLN and CLDN1 expressions, and reduced Sobs, Chao and Ace indices (P < 0.05). Compared with CON-NS group, the concentration of isovaleric acid in the BLF-DQ group was higher (P < 0.05). In conclusion, by establishing a DQ stress injury model, it was elucidated that BLF may enhance antioxidant capacity, strengthen immunity, regulate volatile fatty acid contents, improve intestinal morphology, microbiota and other intestinal barrier functions, so as to mitigate the injury induced by oxidative stress in broilers.
Collapse
Affiliation(s)
- Qilu Zhou
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, Zhejiang, China
| | - Sikandar Ali
- Zhejiang Vegamax Biotechnology Co., Ltd., Anji 313300, Zhejiang, China
| | - Xueyan Shi
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, Zhejiang, China
| | - Guangtian Cao
- College of Quality and Standardization, China Jiliang University, Hangzhou 310018, Zhejiang, China
| | - Jie Feng
- College of Animal Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Caimei Yang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, Zhejiang, China
| | - Ruiqiang Zhang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, Zhejiang, China
| |
Collapse
|
3
|
Liu R, Chen Y, Zhang X, Cai Y, Xu S, Xu Q, Li X, Li W, Liu P, Liu W. Pharmacological efficacy study of the cardio-cerebral stasis transforming medicines on cerebral ischemia and myocardial infarction in rats. Heliyon 2024; 10:e39162. [PMID: 39640627 PMCID: PMC11620065 DOI: 10.1016/j.heliyon.2024.e39162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
The purpose of this study was to investigate the efficacy and mechanisms of cardio-cerebral stasis transforming medicines (CCSTM) against cerebral infarction (CI) and myocardial infarction (MI). CI modeling was conducted using the refined Longa suture-occluded technique, while MI modeling was accomplished through the occlusion of the anterior descending branch of the left coronary artery. We found that compared with the model groups, CCSTM decreased the infarct size in models of CI and MI in a dose-dependent manner. After brain ischemia, CCSTM decreased the level of myeloperoxidase (MPO) and malondialdehyde (MDA), and increased the level of superoxide dismutase (SOD). Besides, CCSTM reduced the concentrations of lactate dehydrogenase (LDH), malondialdehyde MDA, and endothelin (ET) in the plasma of rats injured with MI. Histological examination of brain sections revealed that CCSTM alleviated cerebral damage after ischemia compared with the model group. CCSTM can reduce myocardial and cerebral infarction injury, and the oxidation level after myocardial and cerebral infarction in rats.
Collapse
Affiliation(s)
- Ruilian Liu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
- The Hospital Affiliated to Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Yangchu Chen
- Beijing Jianhua Research Institute of Medicine, Beijing, 100000, PR China
| | - Xili Zhang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Yuhan Cai
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Shuang Xu
- The Hospital Affiliated to Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan Province, PR China
| | - Qian Xu
- The Hospital Affiliated to Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan Province, PR China
| | - Xin Li
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Wenjiao Li
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Pingan Liu
- Hunan Academy of Chinese Medicine, Changsha, 410017, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| | - Wenlong Liu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
- Hunan Key Laboratory of Druggability and Preparation Modification for Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, PR China
| |
Collapse
|
4
|
Ren C, Zeng Y, Qiu L, Luo D, Wang J, Chen X, Yan Y. Study on the effects of intraoperative administration of dexmedetomidine on the prognosis and survival outcomes of patients with colorectal cancer. Heliyon 2024; 10:e38241. [PMID: 39381090 PMCID: PMC11456851 DOI: 10.1016/j.heliyon.2024.e38241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Background The perioperative period of tumor surgery commonly utilizes dexmedetomidine as an adjuvant analgesic for anesthesia. Nevertheless, there is a paucity of research investigating its influence on the prognosis of colorectal cancer (CRC). This article primarily aims to examine the correlation between the intraoperative administration of dexmedetomidine and recurrence-free survival (RFS) and overall survival (OS) of colorectal cancer patients, as well as its prognostic implications on survival. Methods According to the exclusion criteria, 76 patients undergoing laparoscopic radical resection of CRC under general anesthesia were enrolled at Huizhou Central People's Hospital in 2014. Kaplan-Meier method was used for univariate survival analysis of clinical prognostic factors, RFS, and OS in patients with CRC Cox regression analysis was used for multivariate survival analysis. Results A total of 76 patients with CRC were enrolled in this study. Among them, 36 patients were treated with dexmedetomidine (group D), and 40 patients were not treated with dexmedetomidine (group C) during the operation. Survival analysis showed that the RFS and OS of patients in group D were significantly higher than those in group C (P = 0.046 and P = 0.021, respectively). Multivariate regression analysis demonstrated that the intraoperative administration of dexmedetomidine independently predicted a protective effect on OS (P = 0.025). Conclusions The intraoperative application of dexmedetomidine as an adjuvant analgesic has a protective effect on the prognosis and survival of patients with CRC and can improve the overall survival rate. Additionally, it influences the recurrence status of patients to a certain extent. These results suggest that dexmedetomidine significantly benefits on the long-term prognosis of patients with CRC.
Collapse
Affiliation(s)
- Chu Ren
- Shantou University Medical College, Shantou, 515041, Guangdong Province, China
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Ying Zeng
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Liuji Qiu
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Dexing Luo
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Junfang Wang
- Central Laboratory, Medical College of Jiaying University, Meizhou, 514031, Guangdong Province, China
| | - Xin Chen
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Yan Yan
- Shantou University Medical College, Shantou, 515041, Guangdong Province, China
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| |
Collapse
|
5
|
Zhang X, Xu H, Gong L, Wang J, Fu J, Lv Z, Zhou L, Li X, Liu Q, Xia P, Guo Y. Mannanase improves the growth performance of broilers by alleviating inflammation of the intestinal epithelium and improving intestinal microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:376-394. [PMID: 38371477 PMCID: PMC10874740 DOI: 10.1016/j.aninu.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/26/2023] [Accepted: 06/10/2023] [Indexed: 02/20/2024]
Abstract
This experiment aimed to discuss and reveal the effect and mechanism of mannanase on intestinal inflammation in broilers triggered by a soybean meal diet. In this experiment, 384 Arbor Acres broilers at 1 d old were randomly divided into 3 treatment groups. The broilers were fed a corn-soybean meal basal diet, a low-energy diet (metabolizable energy reduced by 50 kcal/kg), and a low-energy diet supplemented with 100 mg/kg mannanase for 42 d. The low-energy diet increased feed conversion ratio from 0 to 42 d, reduced ileal villus height and villus height-to-crypt depth ratio and upregulated the expression of nuclear factor kappa B (NF-κB) in the ileum (P < 0.05). It also reduced cecal short-chain fatty acids (SCFA), such as acetic acid (P < 0.05). Compared with low-energy diets, the addition of mannanase increased body weight at 42 d, promoted the digestibility of nutrients, and maintained the morphology and integrity of the intestinal epithelium of broilers (P < 0.05). In addition, mannanase upregulated the expression of claudin-1 (CLDN1) and zonula occludens-1 (ZO-1) in the jejunum at 21 d, downregulated the expression of ileal NF-κB, and increased the content of isobutyric acid in the cecum of broilers (P < 0.05). The results for the ileal microbiota showed that a low-energy diet led to a decrease in the relative abundance of Lactobacillus reuteri in the ileum of broilers. The addition of mannanase increased the relative abundance of Lactobacillus-KC45b and Lactobacillus johnsonii in broilers. Furthermore, a low-energy diet reduced the relative abundance of Butyricicoccus in the intestine of broilers and inhibited oxidative phosphorylation and phosphoinositol metabolism. Mannanase increased the relative abundance of Odoribacter, promoted energy metabolism and N-glycan biosynthesis, and increased the activities of GH3 and GH18. It is concluded that mannanase could improve the growth performance of broilers by reducing the expression of NF-κB in the ileum, increasing the production of SCFA in the cecum, suppressing intestinal inflammation, balancing the intestinal microbiota, reducing damage to the intestinal barrier, and improving the efficiency of nutrient utilization to alleviate the adverse effects caused by the decrease in dietary energy level.
Collapse
Affiliation(s)
- Xiaodan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huiping Xu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Lu Gong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiao Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jianyang Fu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Liangjuan Zhou
- Beijing Strowin Biotechnology Co., Ltd., Beijing, 100094, China
| | - Xuejun Li
- Beijing Strowin Biotechnology Co., Ltd., Beijing, 100094, China
| | - Qiong Liu
- Beijing Strowin Biotechnology Co., Ltd., Beijing, 100094, China
| | - Pingyu Xia
- Beijing Strowin Biotechnology Co., Ltd., Beijing, 100094, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
6
|
Di Franco C, Evangelista F, Briganti A. Multiple uses of dexmedetomidine in small animals: a mini review. Front Vet Sci 2023; 10:1135124. [PMID: 37342619 PMCID: PMC10278766 DOI: 10.3389/fvets.2023.1135124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Dexmedetomidine is an alpha-2 adrenergic agonist, which use had an exponential increase in human and veterinary medicine in the last 10 years. The aim of this mini review is to summarize the various uses of dexmedetomidine underlining its new applications and capabilities in the small animals' clinical activity. While this drug was born as sedative in veterinary medicine, some studies demonstrated to be effective as an analgesic both in single administration and in continuous infusion. Recent studies have also shown the role of dexmedetomidine as an adjuvant during locoregional anesthesia, increasing the duration of the sensitive block and consequently decreasing the demand for systemic analgesics. The various analgesic properties make dexmedetomidine an interesting drug for opioid-free analgesia. Some studies highlighted a potential neuroprotective, cardioprotective and vasculoprotective role of dexmedetomidine, thus conferring it a place in critical care medicine, such as trauma and septic patients. Dexmedetomidine has demonstrated to be a multitasking molecule and it is ready to face new challenges.
Collapse
Affiliation(s)
- Chiara Di Franco
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - Flavia Evangelista
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
- Vet Hospital H24, Firenze, Italy
| | - Angela Briganti
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| |
Collapse
|
7
|
Dexmedetomidine Protects against Airway Inflammation and Airway Remodeling in a Murine Model of Chronic Asthma through TLR4/NF- κB Signaling Pathway. Mediators Inflamm 2023; 2023:3695469. [PMID: 36846195 PMCID: PMC9946744 DOI: 10.1155/2023/3695469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
Asthma is a common respiratory disease characterized by chronic airway inflammation. Dexmedetomidine (DEX), a highly selective α2 adrenergic receptor agonist, has been shown to participate in regulating inflammatory states and thus exert organ protective actions. However, the potential of DEX in asthma is still unknown. This study is aimed at investigating the role of DEX in a mouse model of house dust mite- (HDM-) induced asthma and exploring its underlying mechanism. Here, we found that DEX treatment significantly ameliorated airway hyperresponsiveness, airway inflammation, and airway remodeling in the asthmatic mice, which were similar to the efficacy of the reference anti-inflammatory drug dexamethasone. In addition, DEX reversed the increased expression of toll-like receptor 4 (TLR4) and its downstream signaling adaptor molecule nuclear factor-κB (NF-κB) in the lung tissue of asthmatic mice. Furthermore, these protective effects of DEX were abolished by yohimbine, an α2 adrenergic receptor antagonist. These results indicate that DEX is capable of ameliorating airway inflammation and remodeling in asthmatic mice, and this protective effect is associated with the inhibition of the TLR4/NF-κB signaling pathway.
Collapse
|
8
|
Bacillus amyloliquefaciens 40 regulates piglet performance, antioxidant capacity, immune status and gut microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 12:116-127. [PMID: 36632621 PMCID: PMC9826887 DOI: 10.1016/j.aninu.2022.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 08/17/2022] [Accepted: 09/16/2022] [Indexed: 01/06/2023]
Abstract
Probiotics can improve animal growth performance and intestinal health. Bacillus species, Lactobacillus species, Bifidobacterium species, yeast etc. are the common types of probiotics. However, understanding the effects of probiotics on the immune status and gut microbiota of weaning piglets and how the probiotics exert their impact are still limited. This study aimed to investigate the effects of Bacillus amyloliquefaciens 40 (BA40) on the performance, immune status and gut microbiota of piglets. A total of 12 litters of newborn piglets were randomly divided into 3 groups. Piglets in control group were orally dosed with phosphate buffered saline; BA40 group and probiotics group were orally gavaged with resuspension BA40 and a probiotics product, respectively. The results showed that BA40 treatment significantly decreased (P < 0.05) the diarrhea incidence (from d 5 to 40), diamine oxidase, D-lactate, interleukin (IL)-1β and interferon-γ concentrations compared with control group and probiotics group. Meanwhile BA40 dramatically increased the total antioxidant capacity, IL-10 and secretory immunoglobulin-A concentrations in contrast to control group. For the microbial composition, BA40 modulated the microbiota by improving the abundance of Bacteroides, Phascolarctobacterium (producing short-chain fatty acids) and Desulfovibrio and reducing the proliferation of pathogens (Streptococcus, Tyzzerella, Vellionella and paraeggerthella). Meanwhile, a metabolic function prediction explained that carbohydrate metabolism and amino acid metabolism enriched in BA40 group in contrast to control group and probiotics group. For correlation analysis, the results demonstrated that BA40-enriched Phascolarctobacterium and Desulfovibrio provide insights into strategies for elevating the health status and performance of weaned piglets. Altogether, BA40 exerted stronger ability in decreasing diarrhea incidence and improved antioxidant activity, gut barrier function and immune status of piglets than the other treatments. Our study provided the experimental and theoretical basis for the application of BA40 in pig production.
Collapse
|
9
|
Liu X, Li Y, Kang L, Wang Q. Recent Advances in the Clinical Value and Potential of Dexmedetomidine. J Inflamm Res 2022; 14:7507-7527. [PMID: 35002284 PMCID: PMC8724687 DOI: 10.2147/jir.s346089] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has sedative, anxiolytic, analgesic, sympatholytic, and opioid-sparing properties and induces a unique sedative response which shows an easy transition from sleep to wakefulness, thus allowing a patient to be cooperative and communicative when stimulated. Recent studies indicate several emerging clinical applications via different routes. We review recent data on dexmedetomidine studies, particularly exploring the varying routes of administration, experimental implications, clinical effects, and comparative advantages over other drugs. A search was conducted on the PubMed and Web of Science libraries for recent studies using different combinations of the words “dexmedetomidine”, “route of administration”, and pharmacological effect. The current routes, pharmacological effects, and application categories of dexmedetomidine are presented. It functions by stimulating pre- and post-synaptic α2-adrenoreceptors within the central nervous system, leading to hyperpolarization of noradrenergic neurons, induction of an inhibitory feedback loop, and reduction of norepinephrine secretion, causing a sympatholytic effect, in addition to its anti-inflammation, sleep induction, bowel recovery, and sore throat reduction effects. Compared with similar α2-adrenoceptor agonists, dexmedetomidine has both pharmacodynamics advantage of a significantly greater α2:α1-adrenoceptor affinity ratio and a pharmacokinetic advantage of having a significantly shorter elimination half-life. In its clinical application, dexmedetomidine has been reported to present a significant number of benefits including safe sedation for various surgical interventions, improvement of intraoperative and postoperative analgesia, sedation for compromised airways without respiratory depression, nephroprotection and stability of hypotensive hemodynamics, reduction of postoperative nausea and vomiting and postoperative shivering incidence, and decrease of intraoperative blood loss. Although the clinical application of dexmedetomidine is promising, it is still limited and further research is required to enhance understanding of its pharmacological properties, patient selection, dosage, and adverse effects.
Collapse
Affiliation(s)
- Xiaotian Liu
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yueqin Li
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Li Kang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Jia T, Xing Z, Wang And H, Li G. Protective effect of dexmedetomidine on intestinal mucosal barrier function in rats after cardiopulmonary bypass. Exp Biol Med (Maywood) 2021; 247:498-508. [PMID: 34878923 DOI: 10.1177/15353702211062509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cardiopulmonary bypass can result in damage to the intestines, leading to the occurrence of systemic inflammatory response syndrome. Dexmedetomidine is reported to confer anti-inflammatory properties. Here, the purpose of this study is to investigate the effect of dexmedetomidine on the intestinal mucosa barrier damage in a rat model of cardiopulmonary bypass. It was observed that cardiopulmonary bypass greatly decreased the levels of hemodynamic parameters than SHAM group, whereas dexmedetomidine pretreatment in a cardiopulmonary bypass model rat prevented this reduction. Also, it showed that compared with control animals, cardiopulmonary bypass caused obvious mucosal damage, which was attenuated in dexmedetomidine + cardiopulmonary bypass group. The above findings were in line with that of dexmedetomidine pretreatment, which increased the expression of tight junction proteins, but it decreased the levels of DAO, D-LA, FABP2, and endotoxin. Moreover, the results demonstrated that due to pre-administration of dexmedetomidine, the level of pro-inflammatory factors was decreased, while the level of anti-inflammatory cytokine was increased. Also, it showed that dexmedetomidine suppressed TLR4/JAK2/STAT3 pathway that was activated by cardiopulmonary bypass. Together, these results revealed that dexmedetomidine pretreatment relieves intestinal microcirculation, attenuates intestinal damage, and inhibits the inflammatory response of cardiopulmonary bypass model rats, demonstrating that in CPB-induced damage of intestinal mucosal barrier function, dexmedetomidine pretreatment plays a protective role by inactivating TLR4/JAK2/STAT3-mediated inflammatory pathway.
Collapse
Affiliation(s)
- Tong Jia
- Anesthesiology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, P. R. China
| | - Zhen Xing
- Anesthesiology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, P. R. China
| | - Huijuan Wang And
- Anesthesiology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, P. R. China
| | - Guoli Li
- Anesthesiology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, P. R. China
| |
Collapse
|
11
|
Baserga M, DuPont TL, Ostrander B, Minton S, Sheffield M, Balch AH, Bahr TM, Watt KM. Dexmedetomidine Use in Infants Undergoing Cooling Due to Neonatal Encephalopathy (DICE Trial): A Randomized Controlled Trial: Background, Aims and Study Protocol. FRONTIERS IN PAIN RESEARCH 2021; 2:770511. [PMID: 35295519 PMCID: PMC8915736 DOI: 10.3389/fpain.2021.770511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Neonatal hypoxia-ischemia encephalopathy (HIE) is the leading cause of neonatal death and poor neurodevelopmental outcomes worldwide. Therapeutic hypothermia (TH), while beneficial, still leaves many HIE treated infants with lifelong disabilities. Furthermore, infants undergoing TH often require treatment for pain and agitation which may lead to further brain injury. For instance, morphine use in animal models has been shown to induce neuronal apoptosis. Dexmedetomidine is a potent α2-adrenergic receptor agonist that may be a better alternative to morphine for newborns with HIE treated with TH. Dexmedetomidine provides sedation, analgesia, and prevents shivering but does not suppress ventilation. Importantly, there is increasing evidence that dexmedetomidine has neuroprotective properties. Even though there are limited data on pharmacokinetics (PK), safety and efficacy of dexmedetomidine in infants with HIE, it has been increasingly administered in many centers. Objectives: To review the current approach to treatment of pain, sedation and shivering in infants with HIE undergoing TH, and to describe a new phase II safety and pharmacokinetics randomized controlled trial that proposes the use of dexmedetomidine vs. morphine in this population. Methods: This article presents an overview of the current management of pain and sedation in critically ill infants diagnosed with HIE and undergoing TH for 72 h. The article describes the design and methodology of a randomized, controlled, unmasked multicenter trial of dexmedetomidine vs. morphine administration enrolling 50 (25 per arm) neonates ≥36 weeks of gestation with moderate or severe HIE undergoing TH and that require pain/sedation treatment. Results and Conclusions: Dexmedetomidine may be a better alternative to morphine for the treatment of pain and sedation in newborns with HIE treated with TH. There is increasing evidence that dexmedetomidine has neuroprotective properties in several preclinical studies of injury models including ischemia-reperfusion, inflammation, and traumatic brain injury as well as adult clinical trials of brain trauma. The Dexmedetomidine Use in Infants undergoing Cooling due to Neonatal Encephalopathy (DICE) trial will evaluate whether administration of dexmedetomidine vs. morphine is safe, establish dexmedetomidine optimal dosing by collecting opportunistic PK data, and obtain preliminary neurodevelopmental data to inform a large Phase III efficacy trial with long term neurodevelopment impairment as the primary outcome.
Collapse
Affiliation(s)
- Mariana Baserga
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
- *Correspondence: Mariana Baserga
| | - Tara L. DuPont
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Betsy Ostrander
- Division of Neurology, Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | | | | | - Alfred H. Balch
- Division of Pediatric Clinical Pharmacology, University of Utah, Salt Lake City, UT, United States
| | | | - Kevin M. Watt
- Division of Pediatric Clinical Pharmacology and Division of Critical Care, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
12
|
Zhang H, Liu Y, Fang X, Gu L, Luo C, Chen L, Wang Q. Vitamin D 3 Protects Mice from Diquat-Induced Oxidative Stress through the NF- κB/Nrf2/HO-1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6776956. [PMID: 34824670 PMCID: PMC8610683 DOI: 10.1155/2021/6776956] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
Vitamin D3, as an indispensable and fat-soluble micronutrient, plays an important role in the health of humans and animals. At present, studies are focusing on the calcium absorption and immunoregulation function of vitamin D3; this study was aimed at exploring the antioxidative stress ability of vitamin D3 on diquat-induced intestinal dysfunction of ICR mice and the underlying mechanism. The results showed that oral gavage of vitamin D3 daily significantly improved the body weight gain and immune organ index and significantly reverted the abnormal changes of ALT, AST, SOD, GSH-Px, T-AOC, and MDA in the serum and jejunum induced by diquat. The addition of vitamin D3 also significantly reduced the concentration of DAO, D-LA, and certain proinflammatory cytokines in serum. Moreover, vitamin D3 improved the pathological morphology of the duodenum, jejunum, colon, liver, and kidney tissues, and it also largely attenuated the degree of inflammatory infiltration of macrophages and cell apoptotic index of jejunal epithelial tissue induced by diquat. The results demonstrated that vitamin D3 significantly recovered the intestinal barrier injury by enhancing the expression of mucins and tight junction proteins in the jejunum. In addition, the results indicated that vitamin D3 could significantly reduce the phosphorylation level of NF-κB (p65) and enhance the expression of Nrf2 and HO-1 in the jejunum compared with the diquat-induced group. This study suggested that oral administration of vitamin D3 can protect mice against oxidative damage by inhibiting the phosphorylation level of NF-κB (p65) and activating Nrf2-related signaling pathways.
Collapse
Affiliation(s)
- Haiwen Zhang
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| | - Youming Liu
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| | - Xin Fang
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| | - Lihong Gu
- Hainan Academy of Agricultural Sciences, Haikou, Hainan 570228, China
| | - Caiwei Luo
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| | - Lu Chen
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| | - Qian Wang
- College of Animal Science and Technology, Hainan University, Haikou, Hainan 570228, China
| |
Collapse
|
13
|
Jiang Z, Li W, Su W, Wen C, Gong T, Zhang Y, Wang Y, Jin M, Lu Z. Protective Effects of Bacillus amyloliquefaciens 40 Against Clostridium perfringens Infection in Mice. Front Nutr 2021; 8:733591. [PMID: 34746206 PMCID: PMC8566672 DOI: 10.3389/fnut.2021.733591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to investigate the protective effects of Bacillus amyloliquefaciens (BA40) against Clostridium perfringens (C. perfringens) infection in mice. Bacillus subtilis PB6 was utilized as a positive control to compare the protective effects of BA40. In general, a total of 24 5-week-old male C57BL/6 mice were randomly divided into four groups, with six mice each. The BA40 and PB6 groups were orally dosed with resuspension bacteria (1 × 109 CFU/ml) once a day, from day 1 to 13, respectively. In the control and infected groups, the mice were orally pre-treated with phosphate-buffered saline (PBS) (200 μl/day). The mice in the infected groups, PB6 + infected group and BA40 + infected group, were orally challenged with C. perfringens type A (1 × 109 CFU/ml) on day 11, whereas the control group was orally dosed with PBS (200 μl/day). The results showed that the BA40 group ameliorated intestinal structure damage caused by the C. perfringens infection. Furthermore, the inflammatory responses detected in the infected groups which include the concentrations of IL-1β, TNF-α, IL-6, and immunoglobulin G (IgG) in the serum and secretory immunoglobulin (SigA) in the colon, and nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) activity in the jejunum, were also alleviated (P < 0.05) by BA40 treatment. Similarly, cytokines were also detected by quantitative PCR (qPCR) in the messenger RNA (mRNA) levels, and the results were consistent with the enzyme-linked immunosorbent assay (ELISA) kits. Additionally, in the infected group, the mRNA expression of Bax and p53 was increasing and the Bcl-2 expression was decreasing, which was reversed by BA40 and PB6 treatment (P < 0.05). Moreover, the intestinal microbiota imbalance induced by the C. perfringens infection was restored by the BA40 pre-treatment, especially by improving the relative abundance of Verrucomicrobiota (P < 0.05) and decreasing the relative abundance of Bacteroidetes (P < 0.05) in the phyla level, and the infected group increased the relative abundance of some pathogens, such as Bacteroides and Staphylococcus (P < 0.05) in the genus level. The gut microbiota alterations in the BA40 group also influenced the metabolic pathways, and the results were also compared. The purine metabolism, 2-oxocarboxylic acid metabolism, and starch and sucrose metabolism were significantly changed (P < 0.05). In conclusion, our results demonstrated that BA40 can effectively protect mice from C. perfringens infection.
Collapse
Affiliation(s)
- Zipeng Jiang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Wentao Li
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Weifa Su
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Chaoyue Wen
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Tao Gong
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Mingliang Jin
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Zeqing Lu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Institute of Feed Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
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: 37] [Impact Index Per Article: 9.3] [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
|
15
|
Pediococcus pentosaceus ZJUAF-4 relieves oxidative stress and restores the gut microbiota in diquat-induced intestinal injury. Appl Microbiol Biotechnol 2021; 105:1657-1668. [PMID: 33475796 DOI: 10.1007/s00253-021-11111-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 01/23/2023]
Abstract
Lactic acid bacteria (LAB) play a key role in promoting health and preventing diseases because of their beneficial effects, such as antimicrobial activities, modulating immune responses, maintaining the gut epithelial barrier and antioxidant capacity. However, the mechanisms with which LAB relieve oxidative stress and intestinal injury induced by diquat in vivo are poorly understood. In the present study, Pediococcus pentosaceus ZJUAF-4 (LAB, ZJUAF-4), a selected probiotics strain with strong antioxidant capacities, was appointed to evaluate the efficiency against oxidative stress in diquat-induced intestinal injury of mice. Alanine transaminase (ALT) and aspartate aminotransferase (AST) were analyzed to estimate the liver injury. The intestinal permeability was evaluated by 4 kDa fluorescein isothiocyanate (FITC)-dextran (FD4), D-lactate (DLA), and diamine oxidase (DAO) levels. Jejunum reactive oxygen species (ROS) production was examined by dihydroethidium (DHE) staining. Western blotting was used to detect the expression of nuclear factor (erythroid-derived-2)-like 2 (Nrf2) and its downstream genes in jejunum. The gut microbiota was analyzed by high-throughput sequencing method based on the 16S rRNA genes. The results showed that ZJUAF-4 pretreatment was found to protect the intestinal barrier function and maintain intestinal redox homeostasis under diquat stimulation. Moreover, oral administration of ZJUAF-4 increased the expression of Nrf2 and its downstream genes. High-throughput sequencing analysis indicated that ZJUAF-4 contributed to restoring the gut microbiota influenced by diquat. Our results suggested that ZJUAF-4 protected the intestinal barrier from oxidative stress-induced damage by modulating the Nrf2 pathway and gut microbiota, indicating that ZJUAF-4 may have potential applications in preventing and treating oxidative stress-related intestinal diseases. KEY POINTS: • ZJUAF-4 exerted protective effects against diquat-induced intestinal injury. • Activation of Nrf2 and its downstream targets towards oxidative stress. • ZJUAF-4 administration restoring gut microbiota.
Collapse
|
16
|
Dexmedetomidine: What's New for Pediatrics? A Narrative Review. J Clin Med 2020; 9:jcm9092724. [PMID: 32846947 PMCID: PMC7565844 DOI: 10.3390/jcm9092724] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past few years, despite the lack of approved pediatric labelling, dexmedetomidine’s (DEX) use has become more prevalent in pediatric clinical practice as well as in research trials. Its respiratory-sparing effects and bioavailability by various routes are only some of the valued features of DEX. In recent years the potential organ-protective effects of DEX, with the possibility for preserving neurocognitive function, has put it in the forefront of clinical and bench research. This comprehensive review focused on the pediatric literature but presents relevant, supporting adult and animal studies in order to detail the recent growing body of literature around the pharmacology, end-organ effects, organ-protective effects, alternative routes of administration, synergetic effects, and clinical applications, with considerations for the future.
Collapse
|
17
|
Kim H, Kwon H, Jeon S, Choi EK. The effect of dexmedetomidine and remifentanil on the postoperative sore throat after thyroidectomy. Medicine (Baltimore) 2020; 99:e21060. [PMID: 32702848 PMCID: PMC7373553 DOI: 10.1097/md.0000000000021060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Postoperative sore throat (POST) is an important concern in surgical patients undergoing endotracheal intubation. Its prevalence after thyroidectomy is up to 80%. The current study aimed to assess the effect of dexmedetomidine and remifentanil on postoperative sore throat. METHODS Seventy-four patients who underwent thyroidectomy were randomized to receive either dexmedetomidine (group D) or remifentanil (group R). At anesthesia induction, group D received dexmedetomidine 1 μg/kg over 10 minutes, followed by continuous dexmedetomidine infusion at 0.3 to 0.6 μg/kg/hour during surgery. Group R received remifentanil of 3 to 4 ng/ml during induction, followed by 1.5 to 2.5 ng/ml remifentanil infusion during surgery. POST at rest and swallowing was assessed during the first 24 hours in serial time periods (0-1, 1-6, and 6-24 hours). Hoarseness and postoperative pain score were also assessed. RESULTS POST incidence at rest (0-1, 1-6, and 6-24 hours) and swallowing (1-6 and 6-24 hours) was lower in group D than in group R. POST severity was significantly lower in group D than in group R during each time period. The incidence of postoperative hoarseness was also lower in group D than in group R at 1 to 6 and 6 to 24 hours. The postoperative pain score was lower in group D than in group R during each time period. CONCLUSION Intraoperative dexmedetomidine infusion reduced the incidence and severity of POST for 24 hours after thyroidectomy.
Collapse
|
18
|
Meng Q, Guo P, Jiang Z, Bo L, Bian J. Dexmedetomidine inhibits LPS-induced proinflammatory responses via suppressing HIF1α-dependent glycolysis in macrophages. Aging (Albany NY) 2020; 12:9534-9548. [PMID: 32433037 PMCID: PMC7288940 DOI: 10.18632/aging.103226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has been reported to exert an anti-inflammatory effect in several animal models, but the mechanism remains unclear. Previous studies have shown that hypoxia inducible factor 1α-induced glycolysis is essential for the activation of inflammatory macrophages. However, whether dexmedetomidine influences hypoxia inducible factor 1α-induced glycolysis and thus exerts an anti-inflammatory effect has been poorly investigated. This study aims to elucidate the anti-inflammatory mechanism of dexmedetomidine involving the hypoxia inducible factor 1α-dependent glycolytic pathway. We showed that dexmedetomidine could suppress lipopolysaccharide-induced inflammatory cytokine production; inhibit the extracellular acidification rate, glucose consumption and lactate production; and decrease the expression of glycolytic genes in macrophages. The enhancement of glycolysis by the granulocyte-macrophage colony-stimulating factor or higher concentration of glucose could reverse the anti-inflammatory effect of dexmedetomidine on lipopolysaccharide-treated macrophages. Moreover, dexmedetomidine significantly inhibited the upregulation of hypoxia inducible factor 1α at the mRNA and protein levels. Genetic inhibition of hypoxia inducible factor 1α expression could reverse the anti-inflammatory effect of dexmedetomidine. Taken together, our results indicate that dexmedetomidine attenuates lipopolysaccharide-induced proinflammatory responses partially by suppressing hypoxia inducible factor 1α-dependent glycolysis in macrophages.
Collapse
Affiliation(s)
- Qingyuan Meng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Pinhao Guo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
19
|
Perioperative Dexmedetomidine Fails to Improve Postoperative Analgesic Consumption and Postoperative Recovery in Patients Undergoing Lateral Thoracotomy for Thoracic Esophageal Cancer: A Randomized, Double-Blind, Placebo-Controlled Trial. Pain Res Manag 2020; 2020:4145893. [PMID: 32454920 PMCID: PMC7229565 DOI: 10.1155/2020/4145893] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/09/2020] [Accepted: 03/18/2020] [Indexed: 12/16/2022]
Abstract
Objectives Dexmedetomidine is widely used as an adjunct to general anesthesia. In this study, we evaluated the effects of perioperative dexmedetomidine infusion on postoperative analgesia in patients undergoing lateral thoracotomy for thoracic esophageal cancer. Methods A total of 62 patients undergoing lateral thoracotomy for thoracic esophageal cancer were randomized to receive adjuvant therapy with either dexmedetomidine (0.5 μg/kg intravenous bolus injection for 10 min before induction of anesthesia, followed by continuous infusion of 0.2-0.4 μg/kg/h until the end of surgery, and 0.06 μg/kg/h for 5 days after surgery) or equal volumes of saline. Acute postoperative pain was treated with patient-controlled intravenous sufentanil and flurbiprofen axetil. The primary outcomes of this study were the numbers of analgesic requirements in the first postoperative 72 h. Results Perioperative dexmedetomidine did not decrease the numbers of analgesic requirements in the first postoperative 72 h (dexmedetomidine group: 12.14 ± 4.76, saline group: 10.89 ± 5.66; p=0.367). Likewise, the groups did not differ with respect to total postoperative analgesic requirements, postoperative pain, perioperative inflammation, blood cell count, incidence of adverse events, surgical recovery (assessed at postoperative days 2 and 5 using the surgical recovery scale), length of hospital stay, hospital cost, incidence of chronic pain, or quality of life. Notably, dexmedetomidine had beneficial effects on decreasing intraoperative opioid consumption and improving postoperative sleep quality. Discussion. Perioperative dexmedetomidine has limited analgesic benefits in lateral thoracotomy for esophageal cancer when added to an opioid-based multimodal anesthetic regimen but can reduce opioid consumption.
Collapse
|
20
|
Flanders CA, Rocke AS, Edwardson SA, Baillie JK, Walsh TS. The effect of dexmedetomidine and clonidine on the inflammatory response in critical illness: a systematic review of animal and human studies. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:402. [PMID: 31829277 PMCID: PMC6907244 DOI: 10.1186/s13054-019-2690-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/28/2019] [Indexed: 02/08/2023]
Abstract
Background The α2 agonists, dexmedetomidine and clonidine, are used as sedative drugs during critical illness. These drugs may have anti-inflammatory effects, which might be relevant to critical illness, but a systematic review of published literature has not been published. We reviewed animal and human studies relevant to critical illness to summarise the evidence for an anti-inflammatory effect from α2 agonists. Methods We searched PubMed, the Cochrane library, and Medline. Animal and human studies published in English were included. Broad search terms were used: dexmedetomidine or clonidine, sepsis, and inflammation. Reference lists were screened for additional publications. Titles and abstracts were screened independently by two reviewers and full-text articles obtained for potentially eligible studies. Data extraction used a bespoke template given study diversity, and quality assessment was qualitative. Results Study diversity meant meta-analysis was not feasible so descriptive synthesis was undertaken. We identified 30 animal studies (caecal ligation/puncture (9), lipopolysaccharide (14), acute lung injury (5), and ischaemia-reperfusion syndrome (5)), and 9 human studies. Most animal (26 dexmedetomidine, 4 clonidine) and all human studies used dexmedetomidine. In animal studies, α2 agonists reduced serum and/or tissue TNFα (20 studies), IL-6 (17 studies), IL-1β (7 studies), NFκB (6 studies), TLR4 (6 studies), and a range of other mediators. Timing and doses varied widely, but in many cases were not directly relevant to human sedation use. In human studies, dexmedetomidine reduced CRP (4 studies), TNFα (5 studies), IL-6 (6 studies), IL-1β (3 studies), and altered several other mediators. Most studies were small and low quality. No studies related effects to clinical outcomes. Conclusion Evidence supports potential anti-inflammatory effects from α2 agonists, but the relevance to clinically important outcomes is uncertain. Further work should explore whether dose relationships with inflammation and clinical outcomes are present which might be separate from sedation-mediated effects.
Collapse
Affiliation(s)
| | - Alistair S Rocke
- Critical Care Department, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Stuart A Edwardson
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, UK
| | - J Kenneth Baillie
- Critical Care Department, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Timothy S Walsh
- Critical Care Department, Royal Infirmary of Edinburgh, Edinburgh, UK. .,Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, UK. .,The Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK.
| |
Collapse
|
21
|
Wieruszewski PM, Wittwer ED. Is Dexmedetomidine the Key for Reducing Acute Kidney Injury After Cardiac Surgery? J Cardiothorac Vasc Anesth 2019; 34:614-615. [PMID: 31791854 DOI: 10.1053/j.jvca.2019.10.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Patrick M Wieruszewski
- Department of Pharmacy, Mayo Clinic, Rochester, MN; Multidisciplinary Epidemiology and Translational Research in Intensive Care (METRIC) Group, Mayo Clinic, Rochester, MN
| | - Erica D Wittwer
- Multidisciplinary Epidemiology and Translational Research in Intensive Care (METRIC) Group, Mayo Clinic, Rochester, MN; Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
22
|
Dexmedetomidine Prevents Cognitive Decline by Enhancing Resolution of High Mobility Group Box 1 Protein-induced Inflammation through a Vagomimetic Action in Mice. Anesthesiology 2019; 128:921-931. [PMID: 29252509 DOI: 10.1097/aln.0000000000002038] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inflammation initiated by damage-associated molecular patterns has been implicated for the cognitive decline associated with surgical trauma and serious illness. We determined whether resolution of inflammation mediates dexmedetomidine-induced reduction of damage-associated molecular pattern-induced cognitive decline. METHODS Cognitive decline (assessed by trace fear conditioning) was induced with high molecular group box 1 protein, a damage-associated molecular pattern, in mice that also received blockers of neural (vagal) and humoral inflammation-resolving pathways. Systemic and neuroinflammation was assessed by proinflammatory cytokines. RESULTS Damage-associated molecular pattern-induced cognitive decline and inflammation (mean ± SD) was reversed by dexmedetomidine (trace fear conditioning: 58.77 ± 8.69% vs. 41.45 ± 7.64%, P < 0.0001; plasma interleukin [IL]-1β: 7.0 ± 2.2 pg/ml vs. 49.8 ± 6.0 pg/ml, P < 0.0001; plasma IL-6: 3.2 ± 1.6 pg/ml vs. 19.5 ± 1.7 pg/ml, P < 0.0001; hippocampal IL-1β: 4.1 ± 3.0 pg/mg vs. 41.6 ± 8.0 pg/mg, P < 0.0001; hippocampal IL-6: 3.4 ± 1.3 pg/mg vs. 16.2 ± 2.7 pg/mg, P < 0.0001). Reversal by dexmedetomidine was prevented by blockade of vagomimetic imidazoline and α7 nicotinic acetylcholine receptors but not by α2 adrenoceptor blockade. Netrin-1, the orchestrator of inflammation-resolution, was upregulated (fold-change) by dexmedetomidine (lung: 1.5 ± 0.1 vs. 0.7 ± 0.1, P < 0.0001; spleen: 1.5 ± 0.2 vs. 0.6 ± 0.2, P < 0.0001), resulting in upregulation of proresolving (lipoxin-A4: 1.7 ± 0.2 vs. 0.9 ± 0.2, P < 0.0001) and downregulation of proinflammatory (leukotriene-B4: 1.0 ± 0.2 vs. 3.0 ± 0.3, P < 0.0001) humoral mediators that was prevented by α7 nicotinic acetylcholine receptor blockade. CONCLUSIONS Dexmedetomidine resolves inflammation through vagomimetic (neural) and humoral pathways, thereby preventing damage-associated molecular pattern-mediated cognitive decline.
Collapse
|
23
|
Lv P, Chen T, Liu P, Zheng L, Tian J, Tan F, Chen J, Deng Y, Li J, Cai J, Chi X. Dexmedetomidine Attenuates Orthotopic Liver Transplantation-Induced Acute Gut Injury via α 2-Adrenergic Receptor-Dependent Suppression of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9426368. [PMID: 31827710 PMCID: PMC6885230 DOI: 10.1155/2019/9426368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/04/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Patients with orthotopic liver transplantation (OLT) frequently develop acute gut injury (AGI), and dexmedetomidine (Dex) has been reported to exert a protective effect against AGI. We investigated whether Dex protects against AGI through antioxidative stress effects by the Nrf2/HO-1 antioxidative signaling pathway. Rats were randomly allocated into a sham group and six orthotopic autologous liver transplantation (OALT) groups receiving different doses of Dex together with/without α 2-adrenergic receptor (AR) blockers. Intestinal tissues were collected to visualize the barrier damage and to measure the indexes of oxidative stress. For in vitro studies, rat intestinal recess epithelial cells (IEC-6) underwent hypoxia/reoxygenation (H/R), and the protective role of Dex was evaluated after α 2A-AR siRNA silencing. OALT resulted in increased oxidative stress, significant intestinal injury, and barrier dysfunction. Dex attenuated OALT-induced oxidative stress and intestinal injury, which was abolished by the pretreatment with the nonspecific α 2A-AR siRNA blocker atipamezole and the specific α 2A-AR siRNA blocker BRL-44408, but not by the specific 2B/C-AR siRNA blocker ARC239. Silencing of α 2A-AR siRNA also attenuated the protective role of Dex on alleviating oxidative stress in IEC-6 cells subjected to H/R. Dex exerted its protective effects by activating Nrf2/HO-1 antioxidative signaling. Collectively, Dex attenuates OALT-induced AGI via α 2A-AR-dependent suppression of oxidative stress, which might be a novel potential therapeutic target for OALT-induced AGI.
Collapse
Affiliation(s)
- Peibiao Lv
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Yuedong Hospital, Meizhou, Guangdong 514700, China
| | - Tufeng Chen
- Department of Gastroenterological Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Peibin Liu
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Lei Zheng
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Jingling Tian
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Fan Tan
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Jiaxin Chen
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Yingqing Deng
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
| | - Jun Li
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Yuedong Hospital, Meizhou, Guangdong 514700, China
| | - Jun Cai
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xinjin Chi
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518017, China
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
24
|
Chen H, Sun X, Yang X, Hou Y, Yu X, Wang Y, Wu J, Liu D, Wang H, Yu J, Yi W. Dexmedetomidine reduces ventilator-induced lung injury (VILI) by inhibiting Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. Bosn J Basic Med Sci 2018; 18:162-169. [PMID: 29510084 DOI: 10.17305/bjbms.2018.2400] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/14/2017] [Accepted: 11/14/2017] [Indexed: 12/30/2022] Open
Abstract
Mechanical ventilation (MV) may lead to ventilator-induced lung injury (VILI). Previous research has shown that dexmedetomidine attenuates pulmonary inflammation caused by MV, but the underlying mechanisms remain unclear. Our study aims to test whether dexmedetomidine has a protective effect against VILI and to explore the possible molecular mechanisms using the rat model. Thirty adult male Wistar rats weighing 200-250 g were randomly assigned to 5 groups (n = 6): control, low tidal volume MV (LMV), high tidal volume (HVT) MV (HMV), HVT MV + dexmedetomidine (DEX), HVT MV + dexmedetomidine + yohimbine (DEX+Y). Rats were euthanized after being ventilated for 4 hours. Pathological changes, lung wet/dry (W/D) weight ratio, lung myeloperoxidase (MPO) activity, levels of inflammatory cytokines (i.e., interleukin [IL]-1β, tumor necrosis factor alpha [TNF-α], and IL-6) in the bronchoalveolar lavage fluid (BALF) and lung tissues, expression of Toll-like receptor 4 (TLR4) and nuclear factor (NF)-κB, and activation of NF-κB in lung tissues were measured. Compared with HMV, DEX group showed fewer pathological changes, lower W/D ratios and decreased MPO activity of the lung tissues and lower concentrations of the inflammatory cytokines in the BALF and lung tissues. Dexmedetomidine significantly inhibited the expression of TLR4 and NF-κB and activation of NF-κB. Yohimbine partly alleviated the effects of dexmedetomidine. Dexmedetomidine reduced the inflammatory response to HVT-MV and had a protective effect against VILI, with the inhibition of the TLR4/NF-κB signaling pathway, at least partly via α2-adrenoceptors.
Collapse
Affiliation(s)
- Hongli Chen
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Qiu R, Yao W, Ji H, Yuan D, Gao X, Sha W, Wang F, Huang P, Hei Z. Dexmedetomidine restores septic renal function via promoting inflammation resolution in a rat sepsis model. Life Sci 2018; 204:1-8. [PMID: 29733849 DOI: 10.1016/j.lfs.2018.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Acute kidney injury occurred after sepsis, resulting in high mortality. This research aims to elucidate the mechanistic effect of DEX on the renal inflammation resolution during sepsis in rats. METHODS The rats were randomly divided into a sham group and the other three cecal ligation and puncture (CLP) model groups, based on different treatments: placebo, DEX and 2-adrenergic receptor (AR) inhibitor atipamezole (AT) treatment (DEX + AT) groups. The survival of septic rats within 24 h was recorded. Tissue pathology, plasma IL-1β, IL-6, TNF-α, lipoxygenase-5 and lipoxin A4 were evaluated. Western blotting and immunostaining was used to determine expression of TLR4, IκB, IKK, NF-κB p65 and pp65 in kidney tissue. Then qPCR was used to analyze the mRNA expression of renal α2A-AR, α2B-AR and α2C-AR. RESULTS Rat mortality and kidney inflammation were significantly increased in septic rats. Specifically, IL-1β, IL-6 and TNF-α plasma levels, NF-κB activity, and TLR4 expression in rat kidney tissues were increased after CLP. In the DEX treatment group, mortality was reduced, histology changes were minor, and lipoxygenase-5, and lipoxin A4 expression were increased. The expression of IL-1β, IL-6 and TNF-α, NF-κB activity and TLR4 expression in rat kidney tissues were also decreased. These results indicated that DEX treatment alleviates acute kidney injury induced by CLP. However, the effects of DEX were apparently suppressed by atipamezole in the DEX + AT group. CONCLUSION The current study demonstrated the protective effect of DEX on CLP-induced kidney injury, which may be effective by attenuating NF-κB pathway activation with lipoxin A4.
Collapse
Affiliation(s)
- Rongzong Qiu
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China; Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Weifeng Yao
- Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| | - Haocong Ji
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China
| | - Dongdong Yuan
- Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiaofeng Gao
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China
| | - Weiping Sha
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China
| | - Fei Wang
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China
| | - Pinjie Huang
- Department of Anesthesiology, Huizhou First Hospital, Huizhou, Guangdong 516000, China
| | - Ziqing Hei
- Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
26
|
Dexmedetomidine Protects Against Multi-Organ Dysfunction Induced by Heatstroke via Sustaining The Intestinal Integrity. Shock 2018; 48:260-269. [PMID: 28709158 DOI: 10.1097/shk.0000000000000826] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Previous studies have indicated that gut-derived endotoxin played a pivotal role for aggravating systemic inflammatory response to multi-organ dysfunction under heatstroke. Dexmedetomidine (DEX) could protect against inflammation and multi-organ injury in various scenarios. The aim of this study was to explore the protective effect of DEX on heatstroke and the mechanism involved. Male C57BL/6 mice were placed in a controlled climate chamber (40 ± 1°C) until the maximum core temperature (Tc, Max) of 42.7°C, the received criterion of heatstroke, was attained, DEX (25 μg/kg) or 0.9% saline was injected intraperitoneally immediately. The results showed that DEX could significantly attenuate multi-organ injury induced by heatstroke, simultaneously decrease levels of serum inflammatory cytokines through inhibiting the intestinal nuclear factor-κB activation. Furthermore, to assess the effects of DEX on intestine mucosal barrier under heatstroke, the levels of plasma endotoxin, FD4, and D-lactate were detected and the expression of tight junction proteins occludin and ZO-1 was analyzed by western blot and immunohistochemistry. Meanwhile, transmission electron microscopy was employed to confirm the ultrastructure of intestine. Interestingly, we found that DEX decreased the intestinal permeability and sustained the integrity of intestinal barrier. Finally, to evaluate the anti-apoptosis effect of DEX, the pro-apoptotic protein Bax and anti-apoptotic protein Bcl-2 were analyzed by western blot, and terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling (TUNEL) staining was conducted. The results showed that DEX decreased TUNEL-positive cells induced by heatstroke in a Bax/Bcl-2-related manner. Taken together, our results indicate that DEX could protect against inflammation and multi-organ injury induced by heatstroke via sustaining the intestinal integrity.
Collapse
|
27
|
Hu H, Shi D, Hu C, Yuan X, Zhang J, Sun H. Dexmedetomidine mitigates CLP-stimulated acute lung injury via restraining the RAGE pathway. Am J Transl Res 2017; 9:5245-5258. [PMID: 29312480 PMCID: PMC5752878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/17/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE RAGE pathway plays crucial effects in causing acute lung injury (ALI). Dexmedetomidine (DEX) is showed to mitigate sepsis-stimulated ALI. However, its mechanisms have not been verified. The study was to evaluate whether the RAGE pathway participated in the actions of DEX on sepsis-stimulated ALI in rats. METHODS Male rats were administrated with intravenously DEX 30 min after sepsis. At 24 h of sepsis, lung myeloperoxidase (MPO) and macrophages in the bronchoalveolarlavage fluid (BALF) were observed. The actions of DEX on pro-inflammatory molecules and related mechanisms were determined by immunological methods. RESULTS It was indicated that DEX markedly attenuated CLP-stimulated augment of lung inflammatory cells infiltration, along with significantly mitigated MPO activity. Besides, DEX obviously reduced lung wet/dry weight ratio and the levels of HMGB1 and RAGE in BALF and lung tissue. Moreover, DEX post-treatment apparently attenuated the histopathological lung injury compared with CLP model group. Furthermore, western blot analysis revealed that DEX efficiently restrained the activation of IκB-α, NF-κB p65, and MAPK. CONCLUSION Our studies demonstrated that DEX attenuates the aggravation of sepsis-stimulated ALI via down regulation of RAGE pathway, which has a potential value in the clinical therapy.
Collapse
Affiliation(s)
- Hongyi Hu
- Department of Anesthesiology, Zhejiang Provincial Hospital of TCMHangzhou 310006, Zhejiang, China
| | - Dongsheng Shi
- Department of Anesthesiology, Zhejiang Provincial Hospital of TCMHangzhou 310006, Zhejiang, China
| | - Chenlu Hu
- Department of General Surgery, The Second Affiliated Hospital Zhejiang University School of MedicineZhejiang, China
| | - Xiao Yuan
- Department of Endocrinology, Zhejiang Provincial Hospital of TCMHangzhou 310006, Zhejiang, China
| | - Juan Zhang
- Department of Anesthesiology, Zhejiang Provincial Hospital of TCMHangzhou 310006, Zhejiang, China
| | - Huaqin Sun
- Department of Anesthesiology, Zhejiang Provincial Hospital of TCMHangzhou 310006, Zhejiang, China
| |
Collapse
|
28
|
Gallego-Ligorit L, Vives M, Vallés-Torres J, Sanjuán-Villarreal TA, Pajares A, Iglesias M. Use of Dexmedetomidine in Cardiothoracic and Vascular Anesthesia. J Cardiothorac Vasc Anesth 2017; 32:1426-1438. [PMID: 29325842 DOI: 10.1053/j.jvca.2017.11.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Indexed: 12/16/2022]
Abstract
Dexmedetomidine is a highly selective α2-adrenergic agonist with analgesic and sedative properties. In the United States, the Food and Drug Administration approved the use of the drug for short-lasting sedation (24 h) in intensive care units (ICUs) in patients undergoing mechanical ventilation and endotracheal intubation. In October 2008, the Food and Drug Administration extended use of the drug for the sedation of nonintubated patients before and during surgical and nonsurgical procedures. In the European Union, the European Medicine Agency approved the use of dexmedetomidine in September 2011 with a single recognized indication: ICU adult patients requiring mild sedation and awakening in response to verbal stimulus. At present, the use of dexmedetomidine for sedation outside the ICU remains an off-label indication. The benefits of dexmedetomidine in critically ill patients and in cardiac, electrophysiology-related, vascular, and thoracic procedures are discussed.
Collapse
Affiliation(s)
- Lucía Gallego-Ligorit
- Department of Anesthesiology and Critical Care Medicine, Cardiovascular and Thoracic Anesthesia Section,Hospital Universitario Miguel Servet, Zaragoza, Spain.
| | - Marc Vives
- Department of Anesthesiology and Critical Care Medicine, Hospital de Bellvitge, Barcelona, Spain
| | - Jorge Vallés-Torres
- Department of Anesthesiology and Critical Care Medicine, Cardiovascular and Thoracic Anesthesia Section,Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - T Alberto Sanjuán-Villarreal
- Department of Anesthesiology and Critical Care Medicine, Cardiovascular and Thoracic Anesthesia Section,Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Azucena Pajares
- Department of Anesthesiology and Critical Care Medicine, Cardiovascular and Thoracic Anesthesia Section,Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Mario Iglesias
- Department of Anesthesiology and Reanimation, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), Madrid, Spain
| |
Collapse
|
29
|
Kong W, Kang K, Gao Y, Liu H, Meng X, Yang S, Yu K, Zhao M. Dexmedetomidine alleviates LPS-induced septic cardiomyopathy via the cholinergic anti-inflammatory pathway in mice. Am J Transl Res 2017; 9:5040-5047. [PMID: 29218102 PMCID: PMC5714788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/04/2017] [Indexed: 06/07/2023]
Abstract
This study was conducted to investigate the role of the cholinergic anti-inflammatory pathway in LPS-induced septic cardiomyopathy in mice. C57BL/6 mice were used to construct septic cardiomyopathy models. The optimal duration of lipopolysaccharide (LPS) treatment was determined by HE staining and TUNEL assay. Blank controls were intraperitoneally injected with saline and models were injected with LPS (10 mg/kg) (LPS), α-bungarotoxin (BT-LPS), BT and dexmedetomidine (BT-DEX-LPS). The pathological examinations were performed on HE- stained myocardium tissues, apoptosis was determined using TUNEL assay, mRNA expression of NF-κB p65, Caspase-3, Caspase-8, Bcl-2, Bax, p53 and α7nACh was quantified using qRT-PCR, protein levels of IL-6, IL-1β, TNF-α and phosphorylated STAT3 (p-STAT3) were analyzed using Western blot analysis. HE staining and TUNEL assays showed that the optimal LPS treatment time for septic cardiomyopathy induction was 16 h. Compared with the blank control, mice in LPS group had significantly higher apoptosis, while DEX and BT reduced apoptosis when they were used separately and increased apoptosis when they were used jointly. In the LPS-treated mice, the levels of NF-κb p65, Caspase-3, Caspase-8, Bax, p53, IL-6, IL-1β, TNF-α and p-STAT3 were significantly increased, while α7nAChR level was decreased significantly (P < 0.01); DEX alone had no impact on the expression of these proteins but significantly up-regulated the expression of these genes except α7nAChR when used jointly with BT (P < 0.01). It is clear that DEX can alleviate heart injury, while α7nAChR-specific blocker BT is antagonistic against the anti-inflammatory effect of DEX on sepsis in mice.
Collapse
Affiliation(s)
- Weilan Kong
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Yang Gao
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Haitao Liu
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Songliu Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
- Institute of Critical Care Medicine in Sino Russian Medical Research Center of Harbin Medical UniversityHarbin 150081, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| |
Collapse
|
30
|
Perez-Zoghbi J, Zhu W, Grafe M, Brambrink A. Dexmedetomidine-mediated neuroprotection against sevoflurane-induced neurotoxicity extends to several brain regions in neonatal rats. Br J Anaesth 2017; 119:506-516. [DOI: 10.1093/bja/aex222] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2017] [Indexed: 12/21/2022] Open
|
31
|
Paeschke N, von Haefen C, Endesfelder S, Sifringer M, Spies CD. Dexmedetomidine Prevents Lipopolysaccharide-Induced MicroRNA Expression in the Adult Rat Brain. Int J Mol Sci 2017; 18:ijms18091830. [PMID: 28832497 PMCID: PMC5618479 DOI: 10.3390/ijms18091830] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/14/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022] Open
Abstract
During surgery or infection, peripheral inflammation can lead to neuroinflammation, which is associated with cognitive impairment, neurodegeneration, and several neurodegenerative diseases. Dexmedetomidine, an α-2-adrenoceptor agonist, is known to exert anti-inflammatory and neuroprotective properties and reduces the incidence of postoperative cognitive impairments. However, on the whole the molecular mechanisms are poorly understood. This study aims to explore whether dexmedetomidine influences microRNAs (miRNAs) in a rat model of lipopolysaccharide (LPS)-induced neuroinflammation. Adult Wistar rats were injected with 1 mg/kg LPS intraperitoneal (i.p.) in the presence or absence of 5 µg/kg dexmedetomidine. After 6 h, 24 h, and 7 days, gene expressions of interleukin 1-β (IL1-β), tumor necrosis factor-α (TNF-α), and microRNA expressions of miR 124, 132, 134, and 155 were measured in the hippocampus, cortex, and plasma. Dexmedetomidine decreased the LPS-induced neuroinflammation in the hippocampus and cortex via significant reduction of the IL1-β and TNF-α gene expressions after 24 h. Moreover, the LPS-mediated increased expressions of miR 124, 132, 134, and 155 were significantly decreased after dexmedetomidine treatment in both brain regions. In plasma, dexmedetomidine significantly reduced LPS-induced miR 155 after 6 h. Furthermore, there is evidence that miR 132 and 134 may be suitable as potential biomarkers for the detection of neuroinflammation.
Collapse
Affiliation(s)
- Nadine Paeschke
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Marco Sifringer
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Claudia D Spies
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
32
|
Inada T, Sumi C, Hirota K, Shingu K, Okamoto A, Matsuo Y, Kamibayashi T. Mitigation of inflammation using the intravenous anesthetic dexmedetomidine in the mouse air pouch model. Immunopharmacol Immunotoxicol 2017; 39:225-232. [PMID: 28555511 DOI: 10.1080/08923973.2017.1327964] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Dexmedetomidine, an α2-adrenergic/imidazoline receptor agonist, is a widely used intravenous anesthetic. Its primary current usage is for sedation of patients in the intensive care unit. The mouse air pouch model is versatile in studying the anti-inflammatory effect of a drug on a local inflammation, which is induced by a variety of substances. In the present study, using the carrageenan-induced air pouch inflammation model, we tested whether dexmedetomidine mitigates inflammation occurring locally in the mouse air pouch. We found that dexmedetomidine dose-dependently inhibited the production of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in the pouch and decreased the number of white blood cells (WBC) recruited into the pouch. Dexmedetomidine also dose-dependently inhibited the production of neutrophil chemokines, cxcl1 and cxcl2. Furthermore, the dexmedetomidine-induced decreased recruitment of WBC into the pouch was successfully reversed with intra-pouch administration of cxcl1/cxcl2, but not TNF-α or IL-6. Lastly, the inhibition of the production of the cytokines and chemokines with dexmedetomidine was reversed by the treatment of yohimbine, suggesting that dexmedetomidine's anti-inflammatory effect is primarily via the stimulation of the α2-adrenergic receptor. We conclude that dexmedetomidine has an anti-inflammatory property in the carrageenan-induced mouse air pouch inflammation model, and that the dexmedetomidine-induced inhibition of production of the neutrophil chemokines, cxcl1 and cxcl2, may be related, at least in part, to the inhibition of WBC intra-pouch recruitment.
Collapse
Affiliation(s)
- Takefumi Inada
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | - Chisato Sumi
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | - Kiichi Hirota
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | - Koh Shingu
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | - Akihisa Okamoto
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | - Yoshiyuki Matsuo
- a Department of Anesthesiology , Kansai Medical University , Osaka , Japan
| | | |
Collapse
|
33
|
Song D, Cheng Y, Li X, Wang F, Lu Z, Xiao X, Wang Y. Biogenic Nanoselenium Particles Effectively Attenuate Oxidative Stress-Induced Intestinal Epithelial Barrier Injury by Activating the Nrf2 Antioxidant Pathway. ACS APPLIED MATERIALS & INTERFACES 2017; 9:14724-14740. [PMID: 28406025 DOI: 10.1021/acsami.7b03377] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In the present study, a new form of selenium nanoparticle (biogenic nanoselenium (BNS) particles) was synthesized using bacteria. The protection of BNS particles against oxidative stress-induced intestinal barrier dysfunction and the inherent mechanisms of this process were investigated, and selenomethionine (SeMet) and chemically synthesized nanoselenium (Nano-Se) particles were used for comparison. Characterization of BNS particles revealed that they were monodispersed and homogeneous spheres, with an average size of 139.43 ± 7.44 nm. In the mouse model of intestinal oxidative stress, BNS particles were found to protect the mouse intestinal barrier function and preserve intestinal redox homeostasis more efficiently than SeMet and Nano-Se. In vitro experiments with porcine jejunum epithelial (IPEC-J2) cells verified the stronger epithelial barrier-protecting effect of BNS particles against oxidative stress, with reduced cell apoptosis and an improved cell redox state. BNS activated the nuclear factor (erythroid-derived-2)-like 2 (Nrf2) and increased the expression of its downstream genes, including thioredoxin reductase (TXNRD)-1, NADPH dehydrogenase (NQO)-1, heme oxygenase (HO)-1, and thioredoxin (Trx), in dose- and time-dependent manners. In contrast, SeMet and Nano-Se merely enhanced the activity of the selenoenzymes TXNRD-1 and glutathione peroxidase (GPx)-1, indicating the role of selenium donors. Moreover, the knock down of Nrf2 significantly blocked the antioxidative effect of BNS, confirming that BNS protects the intestinal barrier from oxidative stress-induced damage by activating Nrf2 and its downstream genes. Our results suggest that BNS is a promising selenium species with potential application in treating oxidative stress-related intestinal diseases.
Collapse
Affiliation(s)
- Deguang Song
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Yuanzhi Cheng
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Xiaoxiao Li
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Fengqin Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Zeqing Lu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Xiao Xiao
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key laboratory of Molecular Animal Nutrition, Ministry of Education, Institute of Feed Science, Zhejiang University , 866 Yuhang Tang Road, Hangzhou 310058, China
| |
Collapse
|
34
|
El-Tanbouly GS, El-Awady MS, Megahed NA, Salem HA, El-Kashef HA. The NF-κB inhibitor celastrol attenuates acute hepatic dysfunction induced by cecal ligation and puncture in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 50:175-182. [PMID: 28189063 DOI: 10.1016/j.etap.2017.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/12/2017] [Accepted: 02/03/2017] [Indexed: 06/06/2023]
Abstract
Acute hepatic dysfunction associating sepsis is mediated mainly by toll-like receptor-4 (TLR-4)/nuclear factor kappa-B (NF-κB) inflammatory pathway. This study explores potential hepatoprotective effect of the NF-κB inhibitor celastrol in cecal ligation and puncture (CLP) model in rats. Protective effect of celastrol (1mg/kg, i.p., 1h before CLP) was illustrated after 24h by preventing CLP-induced hepatic histopathological changes and elevation in serum hepatic biomarkers [alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TB) and gamma aminotransferase (γ-GT)] without affecting mortality. Celastrol anti-inflammatory effect was illustrated by inhibiting increased serum and hepatic mRNA expression of interleukin-6 (IL-6) without affecting IL-10 elevation. Furthermore, celastrol inhibited CLP-induced elevations in hepatic mRNA expression of nuclear factor inhibitory protein kappa-B alpha (NFκBia), TLR-4, 5-lipoxygenase (5-LOX) and prevented NF-κB/p65 nuclear translocation and activation. In conclusion, celastrol prevented CLP-induced acute hepatic dysfunction through its anti-inflammatory effect by attenuating NF-κB activation, TLR-4 and 5-LOX expression with subsequent reduction in pro-inflammatory IL-6.
Collapse
Affiliation(s)
- Ghada S El-Tanbouly
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for science and technology, Gamasa, Egypt
| | - Mohammed S El-Awady
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Nermeen A Megahed
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hatem A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Hassan A El-Kashef
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for science and technology, Gamasa, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
35
|
Liu SZ, He XM, Zhang X, Zeng FC, Wang F, Zhou XY. Ischemic Preconditioning-Induced SOCS-1 Protects Rat Intestinal Ischemia Reperfusion Injury via Degradation of TRAF6. Dig Dis Sci 2017; 62:105-114. [PMID: 27538408 DOI: 10.1007/s10620-016-4277-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/10/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND The inflammatory immune response plays an important role in mesenteric ischemia and ischemia-reperfusion injury. Toll-like receptor 4 (TLR4) is a critical receptor in transduction of the inflammatory response and plays an important role in intestinal homeostasis. Tumor necrosis factor receptor-associated factor 6 (TRAF6), known as a key adaptor protein downstream of TLR4, is involved in the inflammatory response by activating multiple apoptotic signaling pathways. However, mechanisms of the suppressor of cytokine signaling-1 (SOCS-1) in regulating cell inflammation and apoptosis are still obscure. OBJECTIVES To investigate the TLR4-TRAF6 signaling pathway in intestinal ischemia and reperfusion injury, as well as SOCS-1 expression after ischemic preconditioning in the rat intestine. METHODS The small bowel ischemia, ischemia-reperfusion, and preconditioning models were induced using ligation of the superior mesenteric artery in male Sprague-Dawley rats; then, the mRNA and protein levels of TLR4, TRAF6, and SOCS-1 were analyzed using real-time PCR, Western blot, and immunohistochemistry, respectively. RESULTS The expression of TLR4 and TRAF6 was gradually increased with increasing intestinal ischemia duration, but increased substantially after ischemia-reperfusion injury. After ischemic preconditioning, TLR4 and TRAF6 expressions decreased; however, expression of SOCS-1 and the TLR4-TRAF6 pathway inhibitor was increased. CONCLUSION These data show that ischemic preconditioning may induce the activation of SOCS-1 to inhibit the TLR4-TRAF6 signaling pathway, thereby playing a protective role in ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Sheng-Zhi Liu
- SouthWest Medical University Clinical Medicine, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China.,Department of Vascular Surgery, The Second People's Hospital of Yibin, 96 North Street, Yi Bin, 644000, Sichuan Province, People's Republic of China
| | - Xue-Mei He
- Medical Research Center, The Affiliated Hospital of SouthWest Medical University, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China
| | - Xu Zhang
- Department of Pathology, The Affiliated Hospital of SouthWest Medical University, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China
| | - Fan-Cai Zeng
- Department of Biochemistry, SouthWest Medical University, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China
| | - Fang Wang
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of SouthWest Medical University, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China
| | - Xiang-Yu Zhou
- Department of Vascular and Thyroid Surgery, The Affiliated Hospital of SouthWest Medical University, 25 Tai Ping Street, Lu Zhou, 646000, Sichuan Province, People's Republic of China.
| |
Collapse
|
36
|
Pan W, Lin L, Zhang N, Yuan F, Hua X, Wang Y, Mo L. Neuroprotective Effects of Dexmedetomidine Against Hypoxia-Induced Nervous System Injury are Related to Inhibition of NF-κB/COX-2 Pathways. Cell Mol Neurobiol 2016; 36:1179-88. [PMID: 26683659 PMCID: PMC11482469 DOI: 10.1007/s10571-015-0315-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 12/15/2022]
Abstract
Dexmedetomidine has been reported to provide neuroprotection against hypoxia-induced damage. However, the underlying mechanisms remain unclear. We examined whether dexmedetomidine's neuroprotective effects were mediated by the NF-κB/COX-2 pathways. Adult male C57BL/6 mice were subjected to a 30-min hypoxic treatment followed by recovery to normal conditions. They received dexmedetomidine (16 or 160 μg/kg) or 25 mg/kg atipamezole, an α2-adrenoreceptor antagonist, intraperitoneally before exposure to hypoxia. The whole brain was harvested 6, 18, or 36 h after the hypoxia to determine the histopathological outcome and cleaved caspase-3, Bax/Bcl, NF-κB, and COX-2 levels. Hypoxia treatment induced significant neurotoxicity, including destruction of the tissue structure and upregulation of the protein levels of caspase-3, the ratio of Bax/Bcl-2, NF-κB, and COX-2. Dexmedetomidine pretreatment effectively improved histological outcome and restored levels of caspase-3, the Bax/Bcl-2 ratio, NF-κB, and COX-2. Atipamezole reversed the neuroprotection induced by dexmedetomidine. Neuroprotection was achieved by PDTC and NS-398, inhibitors of NF-κB and COX-2, respectively. Dexmedetomidine use before hypoxia provides neuroprotection. Inhibition of NF-κB/COX-2 pathways activation may contribute to the neuroprotection of dexmedetomidine.
Collapse
Affiliation(s)
- Wanying Pan
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Lin Lin
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Nan Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Fuli Yuan
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xiaoxiao Hua
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yueting Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Liqiu Mo
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|