1
|
Zhang X, Su C, Zhao S, Li J, Yu F. Combination therapy of Ulinastatin with Thrombomodulin alleviates endotoxin (LPS) - induced liver and kidney injury via inhibiting apoptosis, oxidative stress and HMGB1/TLR4/NF-κB pathway. Bioengineered 2022; 13:2951-2970. [PMID: 35148668 PMCID: PMC8973693 DOI: 10.1080/21655979.2021.2024686] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Sepsis is a type of systemic inflammation response syndrome that leads to organ function disorders. Currently, there is no specific medicine for sepsis in clinical practice. Lipopolysaccharide (LPS) is an important endotoxin that causes sepsis. Here, we report an effective two-drug combination therapy to treat LPS-induced liver and kidney injury in endotoxic rats. Ulinastatin (UTI) and Thrombomodulin (TM) are biological macromolecules extracted from urine. In our study, combination therapy significantly improved LPS-induced liver and kidney pathological structure and functional injury, and significantly improved the survival rate of endotoxic rats. Results of TUNEL staining and Western blot showed that UTI combined with TM inhibited the excessive apoptosis of liver and kidney cells caused by LPS. The drug combination also promoted the proliferation of liver and kidney cells, reduced the levels of pro-inflammatory factors interleukin (IL)-6, IL-1β, tumor or necrosis factor (TNF)-α and nitric oxide, and down-regulated the expression of High Mobility Group Box 1 (HMGB1), Toll-like receptor (TLR) 4 and Nuclear Factor (NF)-κB phosphorylation to inhibit inflammation. In addition, the combination of UTI and TM also promoted the production of a variety of antioxidant enzymes in the tissues and inhibited the production of lipid peroxidation malondialdehyde (MDA) to enhance antioxidant defenses. Our experiments also proved that UTI combined with TM did not reduce the anticoagulant effect of TM. These results suggested that UTI combined with TM can improve endotoxin-induced liver and kidney damage and mortality by inhibiting liver and kidney cell apoptosis, promoting proliferation, and inhibiting inflammation and oxidative injury.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangning, PR China
| | - Chenlin Su
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangning, PR China
| | - Shuxin Zhao
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangning, PR China
| | - Ji Li
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangning, PR China
| | - Feng Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangning, PR China
| |
Collapse
|
2
|
Özdemir Z, Karakurt A, Taşlidere E, Vardi N, Alagöz MA, Parlakpinar H, Uslu H, Bağ HG. Histological assessment of liver and stomach damage caused by pyridazinone derivative antidepressant agents. Biotech Histochem 2021; 97:355-362. [PMID: 34842474 DOI: 10.1080/10520295.2021.1999499] [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: 10/19/2022] Open
Abstract
Depression is a serious psychological disorder that affects a significant population. We investigated the antidepressant activities of four pyridazinone derivatives that contain the hydrazide moiety using the forced swimming test (FST). The compounds tested exhibited good antidepressant activity compared to duloxetine. The most promising compound was compound 2, which reduced the duration of immobility during FST. The toxic effects of the four compounds on the histomorphology of the liver and stomach tissue also was evaluated.
Collapse
Affiliation(s)
- Zeynep Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Arzu Karakurt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Elif Taşlidere
- Department of Embryology and Histology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Nigar Vardi
- Department of Embryology and Histology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Mehmet Abdullah Alagöz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Harun Uslu
- Department of Anesthesiology, Vocational School of Health Services, Fırat University, Elazığ, Turkey
| | - Harika Gözükara Bağ
- Department of Biostatistics, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
3
|
Kölükçü E, Parlaktaş BS, Kölükçü V, Firat F, Deresoy FA, Katar M, Kuyucu YE, Unsal V. Protective effects of dexmedetomidine on ischaemia-reperfusion injury in an experimental rat model of priapism. Andrologia 2021; 53:e13985. [PMID: 33474739 DOI: 10.1111/and.13985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
The study aimed to investigate the effects of dexmedetomidine against ischaemia-reperfusion injury occurring after priapism in a model of induced-priapism in rats. A total of 18 male rats were randomised into three groups. Group 1 was the control group. A priapism model was performed rats in Group 2 and then ischaemia-reperfusion injury was evaluated. Group 3 had similar procedures to the rats in Group 2. Rats in Group 3 additionally had 100 μg/kg dexmedetomidine administered intraperitoneally immediately after reperfusion. Blood and tissue samples were analysed. Biochemical analysis of blood samples revealed a decrease in the levels of the pro-inflammatory cytokines including interleukin-1 beta (IL-1 Beta), interleukin-6 (IL-6), and tumour necrosis factor-alpha (TNF-alpha) in Group 3 compared to Group 2 (p:.04, p:.009 and p:.009, respectively). Similarly, the highest malondialdehyde (MDA) level was in Group 2 (p:.002). The levels of antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities were significantly higher in Group 3 than that of Group 2 (p:.037 and p:.045, respectively). Direct microscopic examinations revealed positive changes in desquamation, oedema, inflammation and vasocongestion scores in Group 3 compared to Group 2 (p:.007, p:.008, p:.007 and p:.006, respectively). Dexmedetomidine has a protective effect against ischaemia-reperfusion injury in penile tissue.
Collapse
Affiliation(s)
- Engin Kölükçü
- Department of Urology, Gaziosmanpasa University, Tokat, Turkey
| | | | - Vildan Kölükçü
- Department of Anesthesia and Reanimation, Tokat State Hospital, Tokat, Turkey
| | - Fatih Firat
- Department of Urology, Tokat State Hospital, Tokat, Turkey
| | - Faik A Deresoy
- Department of Pathology, Gaziosmanpasa University, Tokat, Turkey
| | - Muzaffer Katar
- Department of Biochemistry, Gaziosmanpasa University, Tokat, Turkey
| | - Yunus Emre Kuyucu
- Department of Biostatistics, Gaziosmanpasa University, Tokat, Turkey
| | - Velid Unsal
- Faculty of Health Sciences and Central Research Laboratory, Mardin Artuklu University, Mardin, Turkey
| |
Collapse
|
4
|
Huang LM, Hu Q, Huang X, Qian Y, Lai XH. Preconditioning rats with three lipid emulsions prior to acute lung injury affects cytokine production and cell apoptosis in the lung and liver. Lipids Health Dis 2020; 19:19. [PMID: 32024527 PMCID: PMC7003422 DOI: 10.1186/s12944-019-1137-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Critically ill patients are at higher risk having acute lung injury (ALI) and more often in need of parenteral nutrition. We sought to study whether preconditioning with representative of lipid emulsions for one week could benefit rats from ALI. METHODS Using a lipopolysaccharide (LPS)-induced ALI rat model and techniques such as polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. RESULTS PGE2 production in the serum was highest in the LPS group, followed with Intralipid group, and the PGE2 level of these two groups was significantly (P < 0.05) higher than the rest. Intralipid conditioning caused significantly less production of LTB4 than the LPS, Clinoleic, or Omegaven group. In contrast to Intralipid, rats pretreated with Clinoleic or Omegaven significantly decreased their production of inflammatory mediators (IL-1 β, IL-6 and TNF-α), had less apoptosis in the lung tissues, and Omegaven greatly improved liver function upon lipopolysaccharide (LPS) challenge. CONCLUSIONS In an ALI setting, preconditioning with Omegaven or Clinoleic was better than Intralipid in decreasing the intensity of the cytokine storm and apoptosis caused by LPS challenge, and Omegaven in addition had the potential to improve liver function. The results from the present study set a basis for further investigation of the molecular mechanisms of ALI, including the up- and downstream pathways of proinflammatory factor production, in search of (small) molecules intervening with the pathogenesis of ALI in order to translate relevant research findings into clinical benefit for patients with ALI. The use of Omegaven or Clinoleic, particularly in patients with ALI, is still characterized by uncertainty due to a lack of relevant studies. Future investigations must specifically focus on the route of administration and mode of application (enteral vs. parenteral/bolus vs. continuous), determining an optimal dose of Omegaven or Clinoleic, and the defining the best timepoint(s) for administration. Critically ill patients are at higher risk having acute lung injury (ALI) and more often in need of parenteral nutrition. The effect of lipid emulsion via parenteral nutrition on liver function was first time evaluated in rats in an ALI setting. The comparison of three forms of lipid emulsion in a rat model of acute lung injury was first time studied. The fish oil-based lipid emulsion decrease in PGE 2 and increase in LTB 4 was first time reported.
Collapse
Affiliation(s)
- Li-Mi Huang
- Department of Pediatrics, The First affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang province, China
| | - Qingqing Hu
- Department of Pediatrics, The First affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang province, China
| | - Xiaoxia Huang
- Department of Pediatrics, The First affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang province, China
| | - Yan Qian
- Department of Pediatrics, The First affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang province, China.
| | - Xin-He Lai
- Department of Pediatrics, The First affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang province, China.
| |
Collapse
|
5
|
Li SP, Zhou XL, Zhao Y. Sedation with midazolam worsens the diaphragm function than dexmedetomidine and propofol during mechanical ventilation in rats. Biomed Pharmacother 2019; 121:109405. [PMID: 31810122 DOI: 10.1016/j.biopha.2019.109405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/25/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mechanical ventilation (MV) is identified as an independent contributor to diaphragmatic atrophy and contractile dysfunction. Appropriate sedation is also essential during MV, and anesthetics may have direct adverse effects on the diaphragm. However, there is a lack of research into the effects of different anesthetics on diaphragm function during MV. OBJECTIVES In the present study, we aim to examine the effect of midazolam, dexmedetomidine, and propofol on diaphragm function during MV. DESIGN Animal study. SETTING University research laboratory. SUBJECTS Male Wistar rats. INTERVENTIONS Animals were experienced 12 h of MV or spontaneous breathing (SB) with continuous anesthetics infusion. Diaphragm contractile properties, cross-sectional areas, microcirculation, oxidative stress, and proteolysis were examined. MEASUREMENTS AND MAIN RESULTS Diaphragmatic specific force was markedly reduced in the midazolam group compared with the dexmedetomidine (-60.4 ± 3.01%, p < 0.001) and propofol group (-58.3 ± 2.60%, p < 0.001) after MV. MV sedated with midazolam induced more atrophy of type II fibers compared with dexmedetomidine (-21.8 ± 2.11%, p = 0.0001) and propofol (-8.2 ± 1.53%, p = 0.003). No significant differences of these indices were found in the midazolam, dexmedetomidine, and propofol groups under SB condition (all p > 0.05, respectively). Twelve hours of MV resulted in a time dependent reduction in diaphragmatic functional capillary density (PB -25.1%, p = 0.0001; MZ -21.6%, p = 0.0003; DD -15.2%, p = 0.022; PP -24.8%, p = 0.0001, respectively), which did not occur in the gastrocnemius muscle. The diaphragmatic lipid peroxidation adducts 4-HNE and HIF-1α levels were significantly lower in dexmedetomidine group and propofol group compared to midazolam group (p < 0.05, respectively). Meanwhile, the catalase and SOD levels were also relatively lower (p < 0.05, respectively) in midazolam group compared to dexmedetomidine group and propofol group. CONCLUSIONS Twelve hours of mechanical ventilation during midazolam sedation led to a more severe diaphragm dysfunction than dexmedetomidine and propofol, possibly caused by its relative weaker antioxidant capacity.
Collapse
Affiliation(s)
- Shao-Ping Li
- 169 Donghu Road, Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Xian-Long Zhou
- 169 Donghu Road, Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Yan Zhao
- 169 Donghu Road, Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| |
Collapse
|
6
|
Sha J, Zhang H, Zhao Y, Feng X, Hu X, Wang C, Song M, Fan H. Dexmedetomidine attenuates lipopolysaccharide-induced liver oxidative stress and cell apoptosis in rats by increasing GSK-3β/MKP-1/Nrf2 pathway activity via the α2 adrenergic receptor. Toxicol Appl Pharmacol 2019; 364:144-152. [DOI: 10.1016/j.taap.2018.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/19/2018] [Accepted: 12/24/2018] [Indexed: 12/28/2022]
|
7
|
Sha J, Feng X, Chen Y, Zhang H, Li B, Hu X, Fan H. Dexmedetomidine improves acute stress‐induced liver injury in rats by regulating MKP‐1, inhibiting NF‐κB pathway and cell apoptosis. J Cell Physiol 2019; 234:14068-14078. [DOI: 10.1002/jcp.28096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Jichen Sha
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Xiujing Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Yongping Chen
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Huayun Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Bei Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Xueyuan Hu
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine College of Veterinary Medicine Northeast Agricultural University Harbin Heilongjiang China
| |
Collapse
|
8
|
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
|
9
|
Ding D, Xu S, Zhang H, Zhao W, Zhang X, Jiang Y, Wang P, Dai Z, Zhang J. 3-Methyladenine and dexmedetomidine reverse lipopolysaccharide-induced acute lung injury through the inhibition of inflammation and autophagy. Exp Ther Med 2018; 15:3516-3522. [PMID: 29545877 DOI: 10.3892/etm.2018.5832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 03/31/2017] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to investigate the effects of 3-methyladenine (3-MA) and dexmedetomidine (DEX) pretreatment on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and the potential mechanism underlying the effects. LPS was instilled into the trachea of BALB/c mice to induce the ALI model. Solutions of 3-MA or DEX were intravenously injected into the mice 1 h later to establish the 3-MA and DEX groups. On days 1, 3 and 5 after the injections, arterial blood gas analysis was conducted, and the lung wet-dry weight ratio (W/D) was determined. In addition, albumin, cytokine and myeloperoxidase (MPO) contents were evaluated using ELISAs, and hematoxylin and eosin (H&E) staining was conducted. Furthermore, western blot analysis was used to evaluate the protein expression levels of microtubule-associated protein 1A/1B-light chain 3 (LC3)-I, LC3-II, autophagy protein 5 (ATG5), Rab7 and lysosome-associated membrane protein 1 (LAMP1), and reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to detect the mRNA expression levels of nuclear factor-κB (NF-κB) and Toll-like receptor 4 (TLR4). Treatment with 3-MA or DEX increased the blood partial pressure of oxygen level compared with that in the model group, and restored the W/D and blood partial pressure of carbon dioxide to normal levels. The content of tumor necrosis factor-α, interleukin-6 and albumin in bronchoalveolar fluid and MPO in lung tissue was significantly decreased in the 3-MA and DEX groups compared with the model group (P<0.05). H&E staining demonstrated that 3-MA and DEX each reversed the ALI. In addition, 3-MA and DEX reduced the protein expression levels of LC3-I, LC3-II, ATG5, Rab7 and LAMP1. Also, RT-qPCR results revealed that NF-κB and TLR4 mRNA expression levels were clearly decreased in the 3-MA and DEX groups compared with the model group. In conclusion, LPS-induced ALI was effectively reversed by treatment with 3-MA and DEX through the reduction of inflammation and autophagy and inhibition of the TLR4-NF-κB pathway.
Collapse
Affiliation(s)
- Dengfeng Ding
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China.,Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Shiyuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Hongfei Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Wei Zhao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xueping Zhang
- Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Yuanxu Jiang
- Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Ping Wang
- Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Zhongliang Dai
- Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Junzhi Zhang
- Department of Anesthesiology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
10
|
Güzel A, Doğan E, Türkçü G, Kuyumcu M, Kaplan İ, Çelik F, Yıldırım ZB. Dexmedetomidine and Magnesium Sulfate: A Good Combination Treatment for Acute Lung Injury? J INVEST SURG 2018; 32:331-342. [PMID: 29359990 DOI: 10.1080/08941939.2017.1422575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Objectives: In this study, we aimed to investigate the therapeutic effects of magnesium sulfate (MgSO4) and dexmedetomidine (dex) in a model of acute lung injury (ALI). We determined whether concomitant administration decreased the inflammatory effects of hydrochloric acid (HCl)-induced ALI in a synergistic manner. Materials and Methods: In this study, 42 Sprague-Dawley rats were randomized into six groups: Group S (saline), Group SV (saline + mechanical ventilation), Group HCl (HCl), Group Dex (Dex), Group Mag (MgSO4), and Group DM (Dex + MgSO4). All groups except Group S were mechanically ventilated prior to HCl-induced ALI. Saline or HCl was administered via tracheostomy. Prior to treatment, HCl was administered to Group HCl, Group Dex, Group Mag, and Group DM to induce ALI. Dex and MgSO4 were administered intraperitoneally. The rats were monitored for 4 h after treatment to measure oxidative stress parameters in blood, and prolidase enzyme activity. Lung tissue damage were determined via histopathology. Results: A significant increase in heart rate and rapid desaturation was observed in HCl-administered groups. Treatment administration decreased the pulse values. Increased saturation values and decreased oxidative stress indices were observed in groups that were subsequently administered Dex and MgSO4. Serum prolidase activity increased significantly in Group HCl. Severe pathological findings were detected following HCl-induced ALI. Group Mag showed greater improvement in the pathology of HCl-induced ALI than did Group Dex. Administration of both Dex and MgSO4 did not improve the pathological scores. Conclusions: The antioxidant and anti-inflammatory effects of Dex and MgSO4 ameliorated the detrimental effects of HCI-induced ALI. However, adverse effects on hemodynamics and lung damage were observed when the two drugs were administered together.
Collapse
Affiliation(s)
- Abdulmenap Güzel
- a Department of Anesthesiology , Dicle University , Diyarbakır , Turkey
| | - Erdal Doğan
- a Department of Anesthesiology , Dicle University , Diyarbakır , Turkey
| | - Gül Türkçü
- b Department of Pathology , Dicle University , Diyarbakır , Turkey
| | - Mahir Kuyumcu
- a Department of Anesthesiology , Dicle University , Diyarbakır , Turkey
| | - İbrahim Kaplan
- c Department of Biochemistry , Dicle University , Diyarbakır , Turkey
| | - Feyzi Çelik
- a Department of Anesthesiology , Dicle University , Diyarbakır , Turkey
| | | |
Collapse
|
11
|
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
|
12
|
Fu C, Dai X, Yang Y, Lin M, Cai Y, Cai S. Dexmedetomidine attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress, mitochondrial dysfunction and apoptosis in rats. Mol Med Rep 2016; 15:131-138. [PMID: 27959438 PMCID: PMC5355722 DOI: 10.3892/mmr.2016.6012] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 11/03/2016] [Indexed: 11/13/2022] Open
Abstract
Previous studies have identified that dexmedetomidine (DEX) treatment can ameliorate the acute lung injury (ALI) induced by lipopolysaccharide and ischemia-reperfusion. However, the molecular mechanisms by which DEX ameliorates lung injury remain unclear. The present study investigated whether DEX, which has been reported to exert effects on oxidative stress, mitochondrial permeability transition pores and apoptosis in other disease types, can exert protective effects in lipopolysaccharide (LPS)-induced ALI by inhibiting oxidative stress, mitochondrial dysfunction and mitochondrial-dependent apoptosis. It was revealed that LPS-challenged rats exhibited significant lung injury, characterized by the deterioration of histopathology, vascular hyperpermeability, wet-to-dry weight ratio and oxygenation index (PaO2/FIO2), which was attenuated by DEX treatment. DEX treatment inhibited LPS-induced mitochondrial dysfunction, as evidenced by alleviating the cellular ATP and mitochondrial membrane potential in vitro. In addition, DEX treatment markedly prevented the LPS-induced mitochondrial-dependent apoptotic pathway in vitro (increases of cell apoptotic rate, cytosolic cytochrome c, and caspase 3 activity) and in vivo (increases of |terminal deoxynucleotidyl transferase dUTP nick-end labeling positive cells, cleaved caspase 3, Bax upregulation and Bcl-2 downregulation). Furthermore, DEX treatment markedly attenuated LPS-induced oxidative stress, as evidenced by downregulation of cellular reactive oxygen species in vitro and lipid peroxides in serum. Collectively, the present results demonstrated that DEX ameliorates LPS-induced ALI by reducing oxidative stress, mitochondrial dysfunction and mitochondrial-dependent apoptosis.
Collapse
Affiliation(s)
- Chunlai Fu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xingui Dai
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Institute of Translation Medicine, Chenzhou, Hunan 423000, P.R. China
| | - You Yang
- Medical Imaging Center, The First People's Hospital of Chenzhou, Institute of Translation Medicine, Chenzhou, Hunan 423000, P.R. China
| | - Mengxiang Lin
- Department of Critical Care Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yeping Cai
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Institute of Translation Medicine, Chenzhou, Hunan 423000, P.R. China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
13
|
Nakamichi M, Akishima-Fukasawa Y, Fujisawa C, Mikami T, Onishi K, Akasaka Y. Basic Fibroblast Growth Factor Induces Angiogenic Properties of Fibrocytes to Stimulate Vascular Formation during Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3203-3216. [PMID: 27773739 DOI: 10.1016/j.ajpath.2016.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 11/17/2022]
Abstract
The role of fibrocytes in wound angiogenesis remains unclear. We therefore demonstrated the specific changes in fibrocyte accumulation for angiogesis in basic fibroblast growth factor (bFGF)-treated wounds. bFGF-treated wounds exhibited marked formation of arterioles and inhibition of podoplanin+ lymph vessels that were lacking in vascular endothelial growth factor-A-treated wounds. Real-time PCR in bFGF-treated wounds manifested enhanced expression of CD34, CD31, and bFGF mRNA and reduced expression of podoplanin and collagen type I, III, and IV mRNA. Double immunofluorescence staining focusing on fibrocyte detection in bFGF-treated wounds showed increased formation of capillary-like structures composed of CD34+/procollagen I+ fibrocytes, with a lack of capillary-like structures formed by CD45+/procollagen I+ or CD11b+/procollagen I+ fibrocytes. However, vascular endothelial growth factor-A-treated wounds lacked capillary-like structures composed of CD34+/procollagen I+ fibrocytes, with increased numbers of CD34+/fetal liver kinase-1+ endothelial progenitor cells. Furthermore, fibroblast growth factor receptor 1 siRNA injection into wounds, followed by bFGF, inhibited the formation of capillary-like structures composed of CD34+/procollagen I+ fibrocytes, together with inhibited mRNA expression of CD34 and CD31 and enhanced mRNA expression of collagen type I, indicating the requirements of bFGF/fibroblast growth factor receptor 1 system for capillary structure formation. This study highlights the angiogenic properties of CD34+/procollagen I+ fibrocytes specifically induced by bFGF, providing new insight into the active contribution of fibrocytes for vascular formation during wound healing.
Collapse
Affiliation(s)
- Miho Nakamichi
- Department of Plastic and Reconstructive Surgery, Toho University Omori Medical Center, Tokyo, Japan
| | | | - Chie Fujisawa
- Division of Research Promotion and Development, Advanced Research Center, Toho University, Tokyo, Japan
| | - Tetuo Mikami
- Department of Pathology, School of Medicine, Toho University, Tokyo, Japan
| | - Kiyoshi Onishi
- Department of Plastic and Reconstructive Surgery, Toho University Omori Medical Center, Tokyo, Japan
| | - Yoshikiyo Akasaka
- Department of Pathology, School of Medicine, Toho University, Tokyo, Japan; Regenerative Disease Research Unit, Advanced Research Center, Toho University, Tokyo, Japan.
| |
Collapse
|
14
|
Kismet K, Sadic M, Bag YM, Atilgan HI, Koca G, Onalan AK, Senes M, Peker SA, Yumusak N, Korkmaz M. HEPATOPROTECTIVE EFFECT OF DEXMEDETOMIDINE AGAINST RADIOIODINE TOXICITY IN RATS: EVALUATION OF OXIDATIVE STATUS AND HISTOPATHOLOGICAL CHANGES. Int Surg 2016; 101:176-184. [PMID: 27018824 DOI: 10.9738/intsurg-d-15-00325.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE AND BACKGROUND Based on the anti-inflammatory, antioxidant and anti-apoptotic properties of DEX, the present study was conducted to investigate the possible radioprotective effects of DEX against hepatic radioiodine (I-131) toxicity. METHODS Thirty six rats were randomly divided into three groups as untreated control (group 1); oral radioiodine (RAI, 111 MBq) administrated rats (group 2), and DEX group (oral radioiodine and daily intraperitoneal 25 µg/kg DEX administrated rats-group 3). In the third group, DEX administration was started 2 days before and continued for five days after RAI administration. Twenty-four hours after the administration of the last dose of DEX, liver samples were taken for evaluation of oxidative stress parameters and histopathological changes. RESULTS The tissue malondialdehyde and advanced oxidation protein product levels in DEX group were significantly lower than RAI group. The total tissue sulphydryl and catalase levels of DEX group were higher than RAI group and the difference was statistically significant. The histopathological damage in the DEX-treated group was significantly less than the damage in the RAI group (p<0.05 for all pathological parameters). Treatment with DEX decreased the histopathological abnormalities when compared with the RAI group. CONCLUSION It was presented that DEX had radioprotective effect on the liver after I-131 therapy and anti-inflammatory and antioxidant activities are likely to be involved in the mechanism underlying the radioprotective effects of DEX. After further studies, DEX might be used as a hepatoprotective treatment regimen before administering radioactive iodine therapy particularly in patients with hepatic disease.
Collapse
Affiliation(s)
- Kemal Kismet
- 1 Ankara Education and Research Hospital, Department of General Surgery, Ankara, Turkey
| | - Murat Sadic
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Yusuf Murat Bag
- 1 Ankara Education and Research Hospital, Department of General Surgery, Ankara, Turkey
| | - Hasan Ikbal Atilgan
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Gokhan Koca
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Ali Kemal Onalan
- 3 Siirt State Hospital, Department of General Surgery, Siirt, Turkey
| | - Mehmet Senes
- 4 Ankara Education and Research Hospital, Department of Biochemistry, Ankara, Turkey
| | - Seydi Ali Peker
- 4 Ankara Education and Research Hospital, Department of Biochemistry, Ankara, Turkey
| | - Nihat Yumusak
- 5 Harran University, Faculty of Veterinary Medicine, Department of Pathology, Sanliurfa, Turkey
| | - Meliha Korkmaz
- 2 Ankara Education and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| |
Collapse
|
15
|
Wu J, Li ST. Dexmedetomidine May Produce Extra Protective Effects on Sepsis-induced Diaphragm Injury. Chin Med J (Engl) 2016; 128:1407-11. [PMID: 25963365 PMCID: PMC4830324 DOI: 10.4103/0366-6999.156808] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The objective was to evaluate the protective effects of dexmedetomidine (DEX), a selective agonist of α2-adrenergic receptor, on sepsis-induced diaphragm injury and the underlying molecular mechanisms. DATA SOURCES The data used in this review were mainly from PubMed articles published in English from 1990 to 2015. STUDY SELECTION Clinical or basic research articles were selected mainly according to their level of relevance to this topic. RESULTS Sepsis could induce severe diaphragm dysfunction and exacerbate respiratory weakness. The mechanism of sepsis-induced diaphragm injury includes the increased inflammatory cytokines and excessive oxidative stress and superfluous production of nitric oxide (NO). DEX can reduce inflammatory cytokines, inhibit nuclear factor-kappaB signaling pathways, suppress the activation of caspase-3, furthermore decrease oxidative stress and inhibit NO synthase. On the basis of these mechanisms, DEX may result in a shorter period of mechanical ventilation in septic patients in clinical practice. CONCLUSIONS Based on this current available evidence, DEX may produce extra protective effects on sepsis-induced diaphragm injury. Further direct evidence and more specific studies are still required to confirm these beneficial effects.
Collapse
Affiliation(s)
| | - Shi-Tong Li
- Department of Anesthesiology, First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| |
Collapse
|
16
|
Margaritelis NV, Veskoukis AS, Paschalis V, Vrabas IS, Dipla K, Zafeiridis A, Kyparos A, Nikolaidis MG. Blood reflects tissue oxidative stress: a systematic review. Biomarkers 2015; 20:97-108. [PMID: 25582635 DOI: 10.3109/1354750x.2014.1002807] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We examined whether the levels of oxidative stress biomarkers measured in blood reflect the tissue redox status. Data from studies that measured redox biomarkers in blood, heart, liver, kidney and skeletal muscle were analyzed. In seven out of nine investigated redox biomarkers (malondialdehyde, reduced glutathione, superoxide dismutase, catalase, glutathione peroxidase, vitamin C and E) there was generally good qualitative and quantitative agreement between the blood and tissues. In contrast, oxidized glutathione and the reduced to oxidized glutathione ratio showed poor agreement between the blood and tissues. This study suggests that most redox biomarkers measured in blood adequately reflect tissue redox status.
Collapse
Affiliation(s)
- Nikos V Margaritelis
- School of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki , Serres , Greece
| | | | | | | | | | | | | | | |
Collapse
|