1
|
Ozkul O, Ozkul B, Erdogan MA, Erbas O. Ameliorating Effect of Propofol on Cisplatin-Induced Liver and Kidney Damage in Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1623.1635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
2
|
Zhang S, Rao S, Yang M, Ma C, Hong F, Yang S. Role of Mitochondrial Pathways in Cell Apoptosis during He-Patic Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23042357. [PMID: 35216473 PMCID: PMC8877300 DOI: 10.3390/ijms23042357] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sen Zhang
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Sijing Rao
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Meiwen Yang
- Department of Surgery, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China;
| | - Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Fengfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
3
|
Liu WL, Chiang FT, Kao JTW, Chiou SH, Lin HL. GSK3 modulation in acute lung injury, myocarditis and polycystic kidney disease-related aneurysm. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118798. [PMID: 32693109 PMCID: PMC7368652 DOI: 10.1016/j.bbamcr.2020.118798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/17/2022]
Abstract
GSK3 are involved in different physical and pathological conditions and inflammatory regulated by macrophages contribute to significant mechanism. Infection stimuli may modulate GSK3 activity and influence host cell adaption, immune cells infiltration or cytokine expressions. To further address the role of GSK3 modulation in macrophages, the signal transduction of three major organs challenged by endotoxin, virus and genetic inherited factors are briefly introduced (lung injury, myocarditis and autosomal dominant polycystic kidney disease). As a result of pro-inflammatory and anti-inflammatory functions of GSK3 in different microenvironments and stages of macrophages (M1/M2), the rational resolution should be considered by adequately GSK3.
Collapse
Affiliation(s)
- Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Critical Care Medicine, Department of Emergency and Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Fu-Tien Chiang
- Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan,Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan,Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan,Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Heng-Liang Lin
- Center For Innovation, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan; Division of Fund Managing, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
4
|
Zhang Z, Tian L, Jiang K. Propofol attenuates inflammatory response and apoptosis to protect d-galactosamine/lipopolysaccharide induced acute liver injury via regulating TLR4/NF-κB/NLRP3 pathway. Int Immunopharmacol 2019; 77:105974. [PMID: 31735662 DOI: 10.1016/j.intimp.2019.105974] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Propofol has been reported to be protective against liver injury due to its anti-inflammatory, anti-oxidative and anti-apoptotic activities. The purpose of this study was to examine the protective effects of propofol on d-galactosamine/lipopolysaccharide (d-GalN/LPS) induced acute liver injury. METHODS Mice were given an intraperitoneal injection of propofol before d-GalN/LPS treatment. Liver injury was confirmed by serum biochemical analysis and liver histopathological analysis. Relevant molecular events were determined by ELISA, western blot, and test kits. Cell apoptosis were evaluated by TUNEL assay. RESULTS The results showed that propofol significantly prevented d-GalN/LPS-induced liver damage by preventing associated increases of serum alanine transaminase (ALT) and aspartate transaminase (AST) and restoring liver histopathological changes. Propofol markedly inhibited the production of inflammatory cytokines and oxidative stress-related factors. Propofol markedly reduced hepatocyte apoptosis, decreased Bax, Bad, cleaved caspase-3 and increased Bcl-2 expression. Besides, NLRP3 inflammasome and TLR4/NF-κB pathway were inactivated under the treatment of propofol according to the expression of pathways-related proteins. CONCLUSION Taken together, propofol contributed to liver protection against d-GalN/LPS-induced liver injury in mice by inhibiting inflammation, oxidative stress and hepatocyte apoptosis through regulating TLR4/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Zhaojian Zhang
- Department of Anaesthesiology, The First People's Hospital of Lianyungang, 222000, China
| | - Liang Tian
- Department of Anaesthesiology, The First People's Hospital of Lianyungang, 222000, China
| | - Kai Jiang
- Department of Hepatobiliary and Pancreatic Surgery & Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310058, China.
| |
Collapse
|
5
|
Propofol can suppress renal ischemia-reperfusion injury through the activation of PI3K/AKT/mTOR signal pathway. Gene 2019; 708:14-20. [DOI: 10.1016/j.gene.2019.05.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/04/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
|
6
|
Cox-2 Negatively Affects the Protective Role of Propofol against Hypoxia/Reoxygenation Induced Cardiomyocytes Apoptosis through Suppressing Akt Signaling. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7587451. [PMID: 31380437 PMCID: PMC6662450 DOI: 10.1155/2019/7587451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/12/2019] [Accepted: 06/23/2019] [Indexed: 01/06/2023]
Abstract
Nowadays, the prevention of severe myocardium injury resulting from myocardial ischemia/reperfusion injury (I/R) has been recognized as an important subject in the field of ischemic heart disease. In this study, H9c2 cardiomyocytes were exposed to cycles of hypoxia/reoxygenation (H/R) to mimic myocardial I/R injury. Western blot analysis and qRT-PCR were performed to detect the expression of Cox-2, Akt and p-Akt. Cell viability, LDH release and activity of Caspase-3 were assessed to determine the protective effect of propofol. The results proved that the protective effect of propofol for H/R challenged cardiomyocytes was associated with Akt phosphorylation. We also revealed that treatment of propofol suppressed the expression of Cox-2 in cardiomyocytes which was up-regulated after H/R treatment. Conversely, the over-expression of Cox-2 inhibited Akt phosphorylation while enhancing cardiomyocytes apoptosis. Interestingly, Akt activator exhibited similar protective effect with propofol and could diminish the influences brought by over-expression of Cox-2. Thus, it could be concluded that Cox-2 negatively affects the protective effect of propofol against hypoxia/reoxygenation induced cardiomyocyte apoptosis by suppressing Akt phosphorylation.
Collapse
|
7
|
How does general anaesthesia affect the circadian clock? Sleep Med Rev 2018; 37:35-44. [DOI: 10.1016/j.smrv.2016.12.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 12/20/2022]
|
8
|
Addition of Berberine to Preservation Solution in an Animal Model of Ex Vivo Liver Transplant Preserves Mitochondrial Function and Bioenergetics from the Damage Induced by Ischemia/Reperfusion. Int J Mol Sci 2018; 19:ijms19010284. [PMID: 29351246 PMCID: PMC5796230 DOI: 10.3390/ijms19010284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 02/07/2023] Open
Abstract
Liver transplantation is a therapeutic regimen to treat patients with non-malignant end-stage liver diseases and malignant tumors of hepatic origin. The ischemia/reperfusion (I/R) injury in liver transplantation is associated with disruption of mitochondrial function in the hepatic parenchyma. Several studies have been conducted in animal models to identify pharmacological therapeutic strategies to minimize the injury induced by the cold/warm I/R in liver transplantation. Most of these studies were conducted in unrealistic conditions without the potential to be translated to clinical usage. Berberine (BBR) is a pharmacological compound with a potential protective effect of the mitochondrial function in the context of I/R. For the future clinical application of these pharmacological strategies, it is essential that a close resemblance exists between the methodology used in the animals models and real life. In this study, we have demonstrated that the addition of BBR to the preservation solution in an I/R setting preserves mitochondrial function and bioenergetics, protecting the liver from the deleterious effects caused by I/R. As such, BBR has the potential to be used as a pharmacological therapeutic strategy.
Collapse
|
9
|
Multifaceted Roles of GSK-3 in Cancer and Autophagy-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4629495. [PMID: 29379583 PMCID: PMC5742885 DOI: 10.1155/2017/4629495] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/07/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
GSK-3 is a ubiquitously expressed serine/threonine kinase existing as GSK-3α and GSK-3β isoforms, both active under basal conditions and inactivated upon phosphorylation by different upstream kinases. Initially discovered as a regulator of glycogen synthesis, GSK-3 is also involved in several signaling pathways controlling many different key functions. Here, we discuss recent advances regarding (i) GSK-3 structure, function, regulation, and involvement in several cancers, including hepatocarcinoma, cholangiocarcinoma, breast cancer, prostate cancer, leukemia, and melanoma (active GSK-3 has been shown to induce apoptosis in some cases or inhibit apoptosis in other cases and to induce cancer progression or inhibit tumor cell proliferation, suggesting that different GSK-3 modulators may address different specific targets); (ii) GSK-3 involvement in autophagy modulation, reviewing signaling pathways involved in neurodegenerative and liver diseases; (iii) GSK-3 role in oxidative stress and autophagic cell death, focusing on liver injury; (iv) GSK-3 as a possible therapeutic target of natural substances and synthetic inhibitors in many diseases; and (v) GSK-3 role as modulator of mammalian aging, related to metabolic alterations characterizing senescent cells and age-related diseases. Studies summarized here underline the GSK-3 multifaceted role and indicate such kinase as a molecular target in different pathologies, including diseases associated with autophagy dysregulation.
Collapse
|
10
|
Unacylated ghrelin prevents mitochondrial dysfunction in a model of ischemia/reperfusion liver injury. Cell Death Discov 2017; 3:17077. [PMID: 29354291 PMCID: PMC5712633 DOI: 10.1038/cddiscovery.2017.77] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/02/2017] [Accepted: 09/13/2017] [Indexed: 01/01/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is a common cause of liver dysfunction during hepatectomy, liver transplantation procedures and in generalized shock. Although effort has been dedicated to rescuing tissue damage in these clinical settings, there is still an urgent need for an effective treatment to protect the liver from the burden of I/R injury. In this study, we have investigated the potential clinical impact of unacylated-ghrelin (UnAG) in a liver I/R rat model. Particular attention has been paid to mitochondria. We demonstrate that UnAG was able to reduce the lag-phase time in response to ADP administration and increase oxygen consumption in ex vivo experiments using liver mitochondria recovered from rats subjected to I/R. Moreover, we found that UnAG rescued the expression of a key regulator of mitochondrial morphology and electron transport chain function; the optic atrophy 1 (Opa1) protein. Cytochrome c oxidase (COX), ATP synthase (complex V) activity and mitochondrial permeability transition pore (mPTP) opening were also affected by UnAG administration in vivo. An in vitro, hepatic I/R model was used to validate these data. We demonstrate that UnAG upregulates the expression of Cox subunit IV (CoxIV) and increases cellular ATP content. This results in Bcl-2 upregulation and protection against apoptosis. Opa1 silencing shows that Opa1 is crucial for a UnAG-induced increase in cellular ATP content, apoptosis resistance, Bcl-2 and CoxIV expression. Finally, we show that UnAG improves Opa1's interaction with MIC60 in the I/R setting, hinting at its role in cristae shape regulation. Our results demonstrate that UnAG administration rescues the intrinsic mitochondrial pathway triggered by I/R damage. Opa1's contribution in mediating this effect is also reported. This suggests that UnAG can interfere with mitochondrial dysfunction, via Opa1, in a preclinical liver I/R model. We therefore provide the rationale for exploiting UnAG as an alternative means to rescuing mitochondrial damage and organ dysfunction.
Collapse
|
11
|
Su M, Ren S, Zhong W, Han X. Impact of propofol on renal ischemia/reperfusion endoplasmic reticulum stress. Acta Cir Bras 2017; 32:533-539. [PMID: 28793037 DOI: 10.1590/s0102-865020170070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/26/2017] [Indexed: 01/22/2023] Open
Abstract
Purpose: To investigate the protective mechanisms of propofol (Pro) on renal ischemia/reperfusion (I/R) injury by studying its impact on renal I/R endoplasmic reticulum stress. Methods: Eighteen male Sprague-Dawley rats (SD rats) were randomly divided into three groups: the I/R group, the Pro pretreatment group, and the control group, and corresponding treatments were performed. The levels of serum creatinine (Cr) and blood urea nitrogen (BUN) of each group were detected. The expression levels of CCAAT-enhancer-binding protein (C/EBP) homology protein (CHOP) and caspase-12 protein within renal tissue samples were detected by western blot. Results: The periodic acid-Schiff (PAS) staining was performed to observe the morphological changes within the renal tissues, and the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay was performed to detect the presence of renal apoptosis. The Pro pretreatment significantly reduced the serum Cr and BUN levels, as well as the expressions levels of CHOP and caspase-12 protein inside the kidney of I/R rats, improving renal pathological injury and reducing the I/R-induced renal apoptosis. Conclusion: Propofol could downregulate the expression of stress-apoptotic proteins CHOP and caspase-12 in the endoplasmic reticulum, thus reducing renal I/R injury.
Collapse
Affiliation(s)
- Mengqin Su
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Sueng Ren
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Wei Zhong
- PhD, Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| | - Xueping Han
- Professor, Department of Anesthesiology, First Affiliated Hospital, Zhengzhou University; Institute of Clinical Medical Research, Henan Universities, Zhengzhou, China. Conception, design and intellectual content of the study, supervised all phases of the study
| |
Collapse
|
12
|
Biochemical targets of drugs mitigating oxidative stress via redox-independent mechanisms. Biochem Soc Trans 2017; 45:1225-1252. [PMID: 29101309 DOI: 10.1042/bst20160473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
Acute or chronic oxidative stress plays an important role in many pathologies. Two opposite approaches are typically used to prevent the damage induced by reactive oxygen and nitrogen species (RONS), namely treatment either with antioxidants or with weak oxidants that up-regulate endogenous antioxidant mechanisms. This review discusses options for the third pharmacological approach, namely amelioration of oxidative stress by 'redox-inert' compounds, which do not inactivate RONS but either inhibit the basic mechanisms leading to their formation (i.e. inflammation) or help cells to cope with their toxic action. The present study describes biochemical targets of many drugs mitigating acute oxidative stress in animal models of ischemia-reperfusion injury or N-acetyl-p-aminophenol overdose. In addition to the pro-inflammatory molecules, the targets of mitigating drugs include protein kinases and transcription factors involved in regulation of energy metabolism and cell life/death balance, proteins regulating mitochondrial permeability transition, proteins involved in the endoplasmic reticulum stress and unfolded protein response, nuclear receptors such as peroxisome proliferator-activated receptors, and isoprenoid synthesis. The data may help in identification of oxidative stress mitigators that will be effective in human disease on top of the current standard of care.
Collapse
|
13
|
Hu C, Li L. Pre-conditions for eliminating mitochondrial dysfunction and maintaining liver function after hepatic ischaemia reperfusion. J Cell Mol Med 2017; 21:1719-1731. [PMID: 28301072 PMCID: PMC5571537 DOI: 10.1111/jcmm.13129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The liver, the largest organ with multiple synthesis and secretion functions in mammals, consists of hepatocytes and Kupffer, stem, endothelial, stellate and other parenchymal cells. Because of early and extensive contact with the external environment, hepatic ischaemia reperfusion (IR) may result in mitochondrial dysfunction, autophagy and apoptosis of cells and tissues under various pathological conditions. Because the liver requires a high oxygen supply to maintain normal detoxification and synthesis functions, it is extremely susceptible to ischaemia and subsequent reperfusion with blood. Consequently, hepatic IR leads to acute or chronic liver failure and significantly increases the total rate of morbidity and mortality through multiple regulatory mechanisms. An increasing number of studies indicate that mitochondrial structure and function are impaired after hepatic IR, but that the health of liver tissues or liver grafts can be effectively rescued by attenuation of mitochondrial dysfunction. In this review, we mainly focus on the subsequent therapeutic interventions related to the conservation of mitochondrial function involved in mitigating hepatic IR injury and the potential mechanisms of protection. Because mitochondria are abundant in liver tissue, clarification of the regulatory mechanisms between mitochondrial dysfunction and hepatic IR should shed light on clinical therapies for alleviating hepatic IR‐induced injury.
Collapse
Affiliation(s)
- Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Hao W, Zhao ZH, Meng QT, Tie ME, Lei SQ, Xia ZY. Propofol protects against hepatic ischemia/reperfusion injury via miR-133a-5p regulating the expression of MAPK6. Cell Biol Int 2017; 41:495-504. [PMID: 28198596 DOI: 10.1002/cbin.10745] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Wei Hao
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Zhi-Hui Zhao
- Department of Anesthesiology; Inner Mongolia Autonomous Region People's Hospital; Huhhot 010017 China
| | - Qing-Tao Meng
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Mu-Er Tie
- Department of Anesthesiology; Inner Mongolia Autonomous Region People's Hospital; Huhhot 010017 China
| | - Shao-Qing Lei
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| | - Zhong-Yuan Xia
- Department of Anesthesiology; Renmin Hospital of Wuhan University; Wuhan 430060 China
| |
Collapse
|
15
|
Zhang Y, Chen Z, Feng N, Tang J, Zhao X, Liu C, Xu H, Zhang M. Protective effect of propofol preconditioning on ischemia-reperfusion injury in human hepatocyte. J Thorac Dis 2017; 9:702-710. [PMID: 28449478 DOI: 10.21037/jtd.2017.02.80] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Blood reperfusion after ischemia is the main measure to restore cell function. This study was aimed to explore the effect of propofol on rat and cell models of liver ischemia-reperfusion (I/R) injury, and to investigate its possible mechanism. METHODS Wistar rats were divided into four groups: control group, sham group, I/R group, and propofol group. Human hepatocyte HL7702 was divided into six groups: control group, I/R group and propofol (5, 10, 20 and 40 µmol/L) groups. After the animal and cell models were established, the alanine aminotransferase (ALT), aspartate aminotransferase (AST), malondialdehyde (MDA) and adenosine triphosphate (ATP) levels in liver tissues and hepatocytes were measured. Cell viability and apoptosis of hepatocytes were respectively determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry. Furthermore, the expressions of apoptosis-related proteins in hepatocytes were determined by Western blot analysis. RESULTS ALT, AST and MDA levels were all decreased significantly, and the ATP level was increased significantly in propofol group compared with that in I/R group in both liver tissues and hepatocytes. Additionally, cell viability of hepatocytes in propofol group was higher than that in I/R group, while the percentage of apoptotic cells in propofol group was less than that in I/R group. Moreover, the expression of caspase-3 decreased and the expression of Bcl-2 increased significantly after propofol preconditioning. CONCLUSIONS Our findings suggested that propofol preconditioning might be an effective strategy for protecting the liver from I/R injury, which might provide a scientific basis for clinical application.
Collapse
Affiliation(s)
- Yuzhu Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.,Department of Anesthesiology, Zibo Central Hospital, Zibo 255000, China
| | - Zhenzhen Chen
- Department of Anesthesiology, Zibo Central Hospital, Zibo 255000, China
| | - Nianhai Feng
- Department of Anesthesiology, Zibo Central Hospital, Zibo 255000, China
| | - Junxia Tang
- Department of Anesthesiology, Zibo Central Hospital, Zibo 255000, China
| | - Xingbo Zhao
- Department of Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Chengxiao Liu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Hongyu Xu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.,Department of Anesthesiology, Zibo Central Hospital, Zibo 255000, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| |
Collapse
|
16
|
Bellanti F, Mirabella L, Mitarotonda D, Blonda M, Tamborra R, Cinnella G, Fersini A, Ambrosi A, Dambrosio M, Vendemiale G, Serviddio G. Propofol but not sevoflurane prevents mitochondrial dysfunction and oxidative stress by limiting HIF-1α activation in hepatic ischemia/reperfusion injury. Free Radic Biol Med 2016; 96:323-333. [PMID: 27154980 DOI: 10.1016/j.freeradbiomed.2016.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/16/2016] [Accepted: 05/01/2016] [Indexed: 02/05/2023]
Abstract
Mitochondrial dysfunction, reactive oxygen species (ROS) production and oxidative stress during reperfusion are determinant in hepatic ischemia/reperfusion (I/R) injury but may be impacted by different anesthetic agents. Thus, we aimed at comparing the effects of inhaled sevoflurane or intravenous propofol anesthesia on liver mitochondria in a rodent model of hepatic I/R injury. To this, male Wistar rats underwent I/R surgery using sevoflurane or propofol. In the I/R model, propofol limited the raise in serum aminotransferase levels as compared to sevoflurane. Mitochondrial oxygen uptake, respiratory activity, membrane potential and proton leak were altered in I/R; however, this impairment was significantly prevented by propofol but not sevoflurane. In addition, differently from sevoflurane, propofol limited hepatic I/R-induced mitochondria H2O2 production rate, free radical leak and hydroxynonenal-protein adducts levels. The I/R group anesthetized with propofol also showed a better recovery of hepatic ATP homeostasis and conserved integrity of mitochondrial PTP. Moreover, hypoxia-inducible factor 1 alpha (HIF-1α) expression was limited in such group. By using a cell model of desferoxamine-dependent HIF activation, we demonstrated that propofol was able to inhibit apoptosis and mitochondrial depolarization associated to HIF-1α action. In conclusion, hepatic I/R injury induces mitochondrial dysfunction that is not prevented by inhaled sevoflurane. On the contrary, propofol reduces liver damage and mitochondrial dysfunction by preserving respiratory activity, membrane potential and energy homeostasis, and limiting free radicals production as well as PTP opening. These hepatoprotective effects may involve the inhibition of HIF-1α.
Collapse
Affiliation(s)
- Francesco Bellanti
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Lucia Mirabella
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Domenica Mitarotonda
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maria Blonda
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Tamborra
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gilda Cinnella
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Alberto Fersini
- Department of General Surgery, University of Foggia, Foggia, Italy
| | - Antonio Ambrosi
- Department of General Surgery, University of Foggia, Foggia, Italy
| | - Michele Dambrosio
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Gianluigi Vendemiale
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gaetano Serviddio
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
17
|
The effect of propofol on the expression of rabbit ischemia reperfusion injury-related proteins. Cell Biochem Biophys 2016; 71:1165-70. [PMID: 25384616 DOI: 10.1007/s12013-014-0325-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
To investigate the effect of propofol on the expression of rabbit ischemia-reperfusion injury-related proteins and the mechanism involved. Thirty healthy adult New Zealand rabbit were selected. After establishment of liver I/R model, the rabbits were divided into group A (sham operation group), group B (control group using saline), and group C (propofol group) with ten rabbits in each group. The total protein concentration, differentially expressed protein spots and the difference of apoptotic proteins expression levels among the three groups were compared. The total protein concentrations in group A, B, and C were 0.778, 0.835, and 0.765 μg/μl, respectively, and the protein concentration in group B was significantly higher than group A and C (p < 0.05), with no significant difference between group A and C (p > 0.05); results analyzed by PDQuest software showed that the average number of protein spots and matching ratio had no significant difference among the three groups (p > 0.05); MALDI-TOF-MS mass spectrometry identified 16 differentially expressed protein spots; the numbers of Caspase-3 positive cells in group B and C were significantly higher than those in group A, and the numbers of Bcl-2 and Bax positive cells in group B and C were significantly lower than those in group A (p < 0.05); the number of Capase-3 positive cells in group C was significantly higher than those in group B, and the number of Bcl-2 positive cells in group C was significantly lower than those in group B (p < 0.05). The numbers of Bax positive cells had no significant difference between group B and C (p > 0.05); densities of Caspase-3, Bcl-2 and Bax in group B and C were significantly higher than those in group A (p < 0.05); Western blotting results from the comparison of the number of positive cells between group B and C was in accordance to the result obtained from immunohistochemistry. After I/R injury in rabbit, there was deregulation of various proteins such as Caspase-3, Bcl-2 and Bax, which was an important factor contributing to liver injury even systematic disease. Propofol could regulate the expression of I/R injury-related proteins and inhibit the attack of free radical to liver, having a remarkable advantage in preventing I/R injury and controlling the development of I/R injury. This study provides an effective theoretical basis for the prevention and treatment of I/R injury.
Collapse
|
18
|
Li J, Kandatsu N, Feng GG, Jiang JZ, Huang L, Kinoshita H, Okada S, Fujiwara Y. Propofol reduces liver dysfunction caused by tumor necrosis factor-α production in Kupffer cells. J Anesth 2016; 30:420-6. [PMID: 26882920 DOI: 10.1007/s00540-016-2145-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 01/28/2016] [Indexed: 01/07/2023]
Abstract
PURPOSE The present study, conducted in rats, investigated whether propofol attenuates lipopolysaccharide (LPS)-triggered liver dysfunction via regulation of tumor necrosis factor (TNF)-α production in activated Kupffer cells. METHODS Rats received LPS (500 μg/kg) under Urethane™ sedation (1 g/kg) in combination with propofol (5 mg/kg/h) or Intralipid™ from 1 h before to 6 h after LPS administration. Some rats were treated with 10 mg/kg gadolinium chloride (GdCl3) to induce Kupffer cell depletion. The serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), TNF-α mRNA and protein expression, caspase-3 activation and apoptosis were evaluated in hepatocytes. Immunofluorescence staining revealed expression of the pan-macrophage marker CD68 as well as TNF-α in Kupffer cells. RESULTS ALT and AST serum levels increased approximately four-fold in LPS-exposed rats compared with Intralipid™-treated rats at 6 h after LPS administration, whereas propofol and GdCl3 reduced the LPS-induced increases. LPS simultaneously augmented TNF-α expression in Kupffer cells, followed by increased caspase-3 activity and apoptosis in hepatocytes. Immunofluorescence staining and immunoblotting assay showed that TNF-α expression in Kupffer cells was inhibited by propofol and GdCl3, resulting in a reduction of caspase-3 activity and apoptosis in LPS-treated rat hepatocytes. CONCLUSIONS Propofol (5 mg/kg/h) attenuated LPS-triggered liver dysfunction via inhibition of TNF-α production in activated Kupffer cells. These results suggest that propofol is capable of inhibiting inflammation-induced liver dysfunction in vivo.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Nobuhisa Kandatsu
- Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Guo-Gang Feng
- Department of Pharmacology, Aichi Medical University School of Medicine, 1-1 Karimata Yazako, Nagakute, Aichi, 480-1195, Japan.
| | - Jia-Zhen Jiang
- Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Lei Huang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hiroyuki Kinoshita
- Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Shoshiro Okada
- Department of Pharmacology, Aichi Medical University School of Medicine, 1-1 Karimata Yazako, Nagakute, Aichi, 480-1195, Japan
| | - Yoshihiro Fujiwara
- Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
19
|
Gracia-Sancho J, Casillas-Ramírez A, Peralta C. Molecular pathways in protecting the liver from ischaemia/reperfusion injury: a 2015 update. Clin Sci (Lond) 2015; 129:345-362. [PMID: 26014222 DOI: 10.1042/cs20150223] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ischaemia/reperfusion injury is an important cause of liver damage during surgical procedures such as hepatic resection and liver transplantation, and represents the main cause of graft dysfunction post-transplantation. Molecular processes occurring during hepatic ischaemia/reperfusion are diverse, and continuously include new and complex mechanisms. The present review aims to summarize the newest concepts and hypotheses regarding the pathophysiology of liver ischaemia/reperfusion, making clear distinction between situations of cold and warm ischaemia. Moreover, the most updated therapeutic strategies including pharmacological, genetic and surgical interventions, as well as some of the scientific controversies in the field are described.
Collapse
Affiliation(s)
- Jordi Gracia-Sancho
- *Barcelona Hepatic Hemodynamic Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Araní Casillas-Ramírez
- †Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Carmen Peralta
- †Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| |
Collapse
|
20
|
Lotz C, Kehl F. Volatile Anesthetic-Induced Cardiac Protection: Molecular Mechanisms, Clinical Aspects, and Interactions With Nonvolatile Agents. J Cardiothorac Vasc Anesth 2015; 29:749-60. [DOI: 10.1053/j.jvca.2014.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Indexed: 02/07/2023]
|
21
|
Ge M, Yao W, Wang Y, Yuan D, Chi X, Luo G, Hei Z. Propofol alleviates liver oxidative stress via activating Nrf2 pathway. J Surg Res 2015; 196:373-81. [DOI: 10.1016/j.jss.2015.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/09/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022]
|
22
|
McFadden D, Souba WW. Change is good! The Journal of Surgical Research: 2014-2015. J Surg Res 2015; 197:1-4. [PMID: 25982043 DOI: 10.1016/j.jss.2015.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- David McFadden
- Department of Surgery, University of Connecticut Health Center, Hartford, CT.
| | - Wiley W Souba
- Department of Surgery, Dartmouth College of Medicine, Hanover, NH
| |
Collapse
|
23
|
Pantazi E, Zaouali MA, Bejaoui M, Folch-Puy E, Abdennebi HB, Roselló-Catafau J. Role of sirtuins in ischemia-reperfusion injury. World J Gastroenterol 2013; 19:7594-7602. [PMID: 24616566 PMCID: PMC3837258 DOI: 10.3748/wjg.v19.i43.7594] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/16/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) remains an unresolved and complicated situation in clinical practice, especially in the case of organ transplantation. Several factors contribute to its complexity; the depletion of energy during ischemia and the induction of oxidative stress during reperfusion initiate a cascade of pathways that lead to cell death and finally to severe organ injury. Recently, the sirtuin family of nicotinamide adenine dinucleotide-dependent deacetylases has gained increasing attention from researchers, due to their involvement in the modulation of a wide variety of cellular functions. There are seven mammalian sirtuins and, among them, the nuclear/cytoplasmic sirtuin 1 (SIRT1) and the mitochondrial sirtuin 3 (SIRT3) are ubiquitously expressed in many tissue types. Sirtuins are known to play major roles in protecting against cellular stress and in controlling metabolic pathways, which are key processes during IRI. In this review, we mainly focus on SIRT1 and SIRT3 and examine their role in modulating pathways against energy depletion during ischemia and their involvement in oxidative stress, apoptosis, microcirculatory stress and inflammation during reperfusion. We present evidence of the beneficial effects of sirtuins against IRI and emphasize the importance of developing new strategies by enhancing their action.
Collapse
|