1
|
Guo S, Zhang C, Zeng H, Xia Y, Weng C, Deng Y, Wang L, Wang H. Glycolysis maintains AMPK activation in sorafenib-induced Warburg effect. Mol Metab 2023; 77:101796. [PMID: 37696356 PMCID: PMC10550717 DOI: 10.1016/j.molmet.2023.101796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/19/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second deadly cancer in the world and still lacks curative treatment. Aerobic glycolysis, or Warburg effect, is a major resistance mechanism induced by first-line treatment of HCC, sorafenib, and is regulated by the master regulator of metabolism, AMPK. Activation of AMPK is required for resistance; however, activation dynamics of AMPK and its regulation is rarely studied. Engineering cells to express an AMPK activity biosensor, we monitor AMPK activation in single HCC cells in a high throughput manner during sorafenib-induced drug resistance. Sorafenib induces transient activation of AMPK, duration of which is dependent on glucose. Inhibiting glycolysis shortens AMPK activation; whereas increasing glycolysis increases its activation duration. Our data highlight that activation duration of AMPK is important for cancer evasion of therapeutic treatment and glycolysis is a key regulator of activation duration of AMPK.
Collapse
Affiliation(s)
- Sijia Guo
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Chenhao Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Haiou Zeng
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, School of Integrated Circuit, Peking University, Beijing, 100871, China
| | - Yantao Xia
- University of California Los Angeles, Department of Chemical and Biomolecular Engineering, California, 90095, USA
| | - Chenghao Weng
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yichen Deng
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Luda Wang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, School of Integrated Circuit, Peking University, Beijing, 100871, China
| | - Huan Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
2
|
Su W, Li J, Jiang L, Lei L, Li H. Hexokinase 2-mediated glycolysis supports inflammatory responses to Porphyromonas gingivalis in gingival fibroblasts. BMC Oral Health 2023; 23:103. [PMID: 36793034 PMCID: PMC9933269 DOI: 10.1186/s12903-023-02807-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND When infected with Porphyromonas gingivalis, gingival fibroblasts undergo metabolic reprogramming, and rely on aerobic glycolysis rather than oxidative phosphorylation for rapid energy replenishment. Hexokinases (HKs) are catalysts for glucose metabolism, and HK2 constitutes the major HK inducible isoform. The objective of this study is to determine whether HK2-mediated glycolysis promotes inflammatory responses in inflamed gingiva. METHODS Levels of glycolysis-related genes were assessed in normal and inflamed gingiva. Human gingival fibroblasts were harvested and infected with Porphyromonas gingivalis in order to mimic periodontal inflammation. 2-deoxy-d-glucose, an analogue of glucose, was used to block HK2-mediated glycolysis, while small interfering RNA was used to knock down HK2 expression. The mRNA and protein levels of genes were analyzed by real-time quantitative PCR and western blotting, respectively. HK2 activity and lactate production were assessed by ELISA. Cell proliferation was assessed by confocal microscopy. The generation of reactive oxygen species was assessed by flow cytometry. RESULTS Elevated expression of HK2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 was observed in the inflamed gingiva. P. gingivalis infection was shown to promote glycolysis in human gingival fibroblasts, as evidenced by increased gene transcription of HK2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, cell glucose consumption, and HK2 activity. Inhibition and knockdown of HK2 resulted in reduced cytokine production, cell proliferation, and reactive oxygen species generation. Furthermore, P. gingivalis infection activated the hypoxia-inducible factor-1α signaling pathway, thus promoting HK2-mediated glycolysis and proinflammatory responses. CONCLUSIONS HK2-mediated glycolysis promotes inflammatory responses in gingival tissues, and therefore glycolysis can be targeted in order to inhibit the progression of periodontal inflammation.
Collapse
Affiliation(s)
- Wenqi Su
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingwen Li
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lishan Jiang
- grid.41156.370000 0001 2314 964XDepartment of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008 Jiangsu China ,grid.41156.370000 0001 2314 964XCentral Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lang Lei
- grid.41156.370000 0001 2314 964XDepartment of Orthodontics, Medical School of Nanjing University, Nanjing Stomatological Hospital, Nanjing, China
| | - Houxuan Li
- Department of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, #30 Zhongyang Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
3
|
Shi X, Zhang W, Gu C, Ren H, Wang C, Yin N, Wang Z, Yu J, Liu F, Zhang H. NAD+ depletion radiosensitizes 2-DG-treated glioma cells by abolishing metabolic adaptation. Free Radic Biol Med 2021; 162:514-522. [PMID: 33197538 DOI: 10.1016/j.freeradbiomed.2020.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/31/2020] [Accepted: 11/08/2020] [Indexed: 11/17/2022]
Abstract
Two-deoxy-d-glucose (2-DG) mediated glucose restriction (GR) has been applied as a potential therapeutic strategy for tumor clinical treatments. However, increasing evidences have indicated that 2-DG alone is inefficient in killing tumor cells, and the effect of 2-DG on modifying tumor radio-responses also remains controversial. In this study, we found that 2-DG triggered metabolic adaption in U87 glioma cells by up-regulating nicotinamide phosphoribosyltransferase (NAMPT) and cellular NAD+ content, which abolished 2-DG-induced potential radiosensitizing effect in glioma cells. Strikingly, NAD+ depletion evoked notable oxidative stress by NADPH reduction and hence re-radiosensitized 2-DG-treated glioma cells. Furthermore, isocitrate dehydrogenase-1 (IDH1) mutant U87 glioma cells with deficiency in the rate-limiting enzyme of Preiss-Handler pathway nicotinate phosphoribosyltransferase (Naprt1) revealed notable 2-DG-induced oxidative stress and radiosensitization. Our findings implied that targeting NAD+ could radiosensitize gliomas with GR, and 2-DG administration could be benefit for tumor patients with IDH1 mutation.
Collapse
Affiliation(s)
- Xiaolin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Gu
- Department of Radiation Oncology, Changzhou No.4 People's Hospital, Soochow University, Changzhou, 213001, China
| | - Huangge Ren
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Chen Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Narui Yin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Zhongmin Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahua Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China
| | - Fenju Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China.
| | - Haowen Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, 215123, China.
| |
Collapse
|
4
|
Zhang X, Wang J, Gong G, Ma R, Xu F, Yan T, Wu B, Jia Y. Spinosin Inhibits Aβ 1-42 Production and Aggregation via Activating Nrf2/HO-1 Pathway. Biomol Ther (Seoul) 2020; 28:259-266. [PMID: 31791116 PMCID: PMC7216747 DOI: 10.4062/biomolther.2019.123] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 11/25/2022] Open
Abstract
The present research work primarily investigated whether spinosin has the potential of improving the pathogenesis of Alzheimer’s disease (AD) driven by β-amyloid (Aβ) overproduction through impacting the procession of amyloid precursor protein (APP). Wild type mouse Neuro-2a cells (N2a/WT) and N2a stably expressing human APP695 (N2a/APP695) cells were treated with spinosin for 24 h. The levels of APP protein and secreted enzymes closely related to APP procession were examined by western blot analysis. Oxidative stress related proteins, such as nuclear factor-erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) were detected by immunofluorescence assay and western blot analysis, respectively. The intracellular reactive oxygen species (ROS) level was analyzed by flow cytometry, the levels of Aβ1-42 were determined by ELISA kit, and Thioflavin T (ThT) assay was used to detect the effect of spinosin on Aβ1-42 aggregation. The results showed that ROS induced the expression of ADAM10 and reduced the expression of BACE1, while spinosin inhibited ROS production by activating Nrf2 and up-regulating the expression of HO-1. Additionally, spinosin reduced Aβ1-42 production by impacting the procession of APP. In addition, spinosin inhibited the aggregation of Aβ1-42. In conclusion, spinosin reduced Aβ1-42 production by activating the Nrf2/HO-1 pathway in N2a/WT and N2a/APP695 cells. Therefore, spinosin is expected to be a promising treatment of AD.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinyu Wang
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Ruixin Ma
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tingxu Yan
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bo Wu
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ying Jia
- Key Laboratory of Active Components of Chinese Medicine Screening and Evaluation, School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
5
|
Bianchi M, D'Oria V, Braghini MR, Petrini S, Manco M. Liraglutide Treatment Ameliorates Neurotoxicity Induced by Stable Silencing of Pin1. Int J Mol Sci 2019; 20:ijms20205064. [PMID: 31614723 PMCID: PMC6829573 DOI: 10.3390/ijms20205064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Post-translational modulation of peptidylprolyl isomerase Pin1 might link impaired glucose metabolism and neurodegeneration, being Pin1 effectors target for the glucagon-Like-Peptide1 analog liraglutide. We tested the hypotheses in Pin1 silenced cells (SH-SY5Y) treated with 2-deoxy-d-glucose (2DG) and methylglyoxal (MG), stressors causing altered glucose trafficking, glucotoxicity and protein glycation. Rescue by liraglutide was investigated. Pin1 silencing caused increased levels of reactive oxygen species, upregulated energy metabolism as suggested by raised levels of total ATP content and mRNA of SIRT1, PGC1α, NRF1; enhanced mitochondrial fission events as supported by raised protein expression of FIS1 and DRP1. 2DG and MG reduced significantly cell viability in all the cell lines. In Pin1 KD clones, 2DG exacerbated altered mitochondrial dynamics causing higher rate of fission events. Liraglutide influenced insulin signaling pathway (GSK3b/Akt); improved cell viability also in cells treated with 2DG; but it did not revert mitochondrial dysfunction in Pin1 KD model. In cells treated with MG, liraglutide enhanced cell viability, reduced ROS levels and cell death (AnnexinV/PI); and trended to reduce anti-apoptotic signals (BAX, BCL2, CASP3). Pin1 silencing mimics neuronal metabolic impairment of patients with impaired glucose metabolism and neurodegeneration. Liraglutide rescues to some extent cellular dysfunctions induced by Pin1 silencing.
Collapse
Affiliation(s)
- Marzia Bianchi
- Research Area for Multi-factorial Diseases, Obesity and Diabetes, Bambino Gesù Children's Research Hospital, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), viale di San Paolo 15, 00146 Rome, Italy.
| | - Valentina D'Oria
- Confocal Microscopy Core Facility, Research Laboratories, Bambino Gesu' Children's Research Hospital, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), viale di San Paolo 15, 00146 Rome, Italy.
| | - Maria Rita Braghini
- Molecular Genetics of Complex Phenotypes Research Unit, Bambino Gesù Children's Research Hospital, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), viale di San Paolo 15, 00146 Rome, Italy.
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Laboratories, Bambino Gesu' Children's Research Hospital, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), viale di San Paolo 15, 00146 Rome, Italy.
| | - Melania Manco
- Research Area for Multi-factorial Diseases, Obesity and Diabetes, Bambino Gesù Children's Research Hospital, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), viale di San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
6
|
Zhou R, Li X, Li L, Zhang H. Theaflavins alleviate sevoflurane-induced neurocytotoxicity via Nrf2 signaling pathway. Int J Neurosci 2019; 130:1-8. [PMID: 31518514 DOI: 10.1080/00207454.2019.1667788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aim: Sevoflurane could induce apoptosis of rat hippocampal neurons, while theaflavins (TFs) have antioxidant and anti-inflammatory properties. This study aims to explore whether TFs could alleviate sevoflurane-induced neuronal cell injury.Materials and methods: Cells were treated by concentration gradient of sevoflurane and TFs. Cell viability, level of reactive oxygen species (ROS) and apoptosis rate were determined by cell counting kit-8 (CCK-8) and flow cytometry, respectively. Quantitative PCR (qPCR) and western blot were performed to determine mRNA and protein expressions.Results: TFs promoted viability of cells under the treatment of sevoflurane, while it suppressed apoptosis and down-regulated ROS level in a concentration-dependent manner. TFs could also down-regulate expression levels of caspase-3 and caspase-9 and cytosol and intranuclear nuclear factor E2-related factor 2 (Nrf2) in rat hippocampal nerve cells, while it up-regulated those of heme oxygenase 1 (HO-1), NADPH quinine oxidoreductase 1 (NQO1), glutamate cysteine ligase (GCL) and peroxiredoxin 1 (Prx1).Conclusions: Our study suggests that TFs exert protective effects on sevoflurane-induced neurocytotoxicity and therefore could be used as a potential drug for treatment of neuronal injury.
Collapse
Affiliation(s)
- Rongsheng Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaogang Li
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lu Li
- Department of the Second Anesthesia, The Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Han Zhang
- Department of the Second Anesthesia, The Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
7
|
Huang XW, Xu Y, Sui X, Lin H, Xu JM, Han D, Ye DD, Lv GF, Liu YX, Qu XB, Duan MH. Scutellarein suppresses Aβ-induced memory impairment via inhibition of the NF-κB pathway in vivo and in vitro. Oncol Lett 2019; 17:5581-5589. [PMID: 31186780 PMCID: PMC6507344 DOI: 10.3892/ol.2019.10274] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 02/22/2019] [Indexed: 12/26/2022] Open
Abstract
The flavonoid compound scutellarin (Scu) is a traditional Chinese medicine used to treat a variety of diseases; however, the use of scutellarein (Scue), the hydrolysate of Scu, and its mechanisms of action in Alzheimer's disease (AD) have not been fully elucidated. In the present study, the effects of Scue on amyloid β (Aβ)-induced AD-like pathology were investigated. An in vitro model of inflammation and an aged rat model were used to confirm the effects of Scue. In vitro MTT assays and flow cytometry were used to assess the effects of Scue on cell viability and apoptosis, respectively. A Morris water maze was used to evaluate spatial learning and memory, and the levels of Aβ deposition, superoxide dismutase, malondialdehyde, apoptosis, neuro-inflammatory factors and nuclear factor-κB (NF-κB) activation in hippocampal tissues in vivo were measured to determine the effect of Scue in AD. Scue may be protective, as it decreased the apoptosis of hippocampal cells in vitro, inhibited Aβ-induced cognitive impairment, suppressed hippocampal neuro-inflammation and suppressed activation of NF-κB in vivo. Therefore, Scue may be a useful agent for the treatment of Aβ-associated pathology in the central nervous system through inhibition of the protein kinase B/NF-κB signaling pathway and thus, future studies are required to investigate the efficacy of Scue in patients with AD.
Collapse
Affiliation(s)
- Xiao-Wei Huang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Yan Xu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Xin Sui
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - He Lin
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Jia-Ming Xu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Dong Han
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Dou-Dan Ye
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Guang-Fu Lv
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Yue-Xin Liu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Xiao-Bo Qu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| | - Ming-Hua Duan
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130107, P.R. China
| |
Collapse
|
8
|
Wang R, Zhang Z, Kumar M, Xu G, Zhang M. Neuroprotective potential of ketamine prevents developing brain structure impairment and alteration of neurocognitive function induced via isoflurane through the PI3K/AKT/GSK-3β pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:501-512. [PMID: 30787593 PMCID: PMC6366353 DOI: 10.2147/dddt.s188636] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The aim of the current experimental study was to scrutinize the neuroprotective effect of ketamine on the isoflurane (iso)-induced cognitive dysfunction in rats via phosphoinositide 3 kinase (PI3K)/protein kinase B (AKT)/glycogen synthase kinase 3β (GSK-3β) pathway. Materials and methods Sprague-Dawley rats were used for the current experimental study. The rats were divided into six groups and rats were treated with ketamine and memantine. For the estimation of cognitive function study, we used the Morris water test. Pro-inflammatory cytokines such as IL-1β, IL-6, tumor necrosis factor-α (TNF-α), and caspase-6; the antioxidant parameters malondialdehyde, glutathione, superoxide dismutase, catalase, and protein carbonyl; acetylcholinesterase, amyloid β, and brain-derived neurotrophic factor were estimated, respectively. The protein expression of AKT, GSK-3β, p21WAF1/CIP1, and p53 was also estimated, respectively. Results Ketamine significantly enhanced cognitive function and showed anti-inflammatory and antioxidant effects, and exhibited the neuroprotective effect of ketamine against the isoflurane-induced cognitive impairment. Additionally, ketamine significantly (P<0.005) suppressed IL-1β, TNF-α, IL-6, caspase-6 and p21WAF1/CIP1, p53 expression and up-regulated the PI3K/AKT/GSK-3β expression in the group of iso-induced rats. Conclusion We can conclude that ketamine prevented the cognitive impairment induced by isoflurane anesthesia through anti-apoptotic, anti-inflammatory, and antioxidant effects via the PI3K/AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Ruiwei Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province 250021, People's Republic of China,
| | - Zihao Zhang
- Department of Clinical Medicine, Nanchang University, Nanchang, Jiangxi Province 330031, People's Republic of China
| | | | - Guangming Xu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province 250021, People's Republic of China,
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province 250021, People's Republic of China,
| |
Collapse
|
9
|
Chai D, Cheng Y, Jiang H. Fundamentals of fetal toxicity relevant to sevoflurane exposures during pregnancy. Int J Dev Neurosci 2018; 72:31-35. [DOI: 10.1016/j.ijdevneu.2018.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/10/2018] [Accepted: 11/12/2018] [Indexed: 02/08/2023] Open
Affiliation(s)
- Dongdong Chai
- Department of Anesthesiology and Critical Care MedicineShanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yanyong Cheng
- Department of Anesthesiology and Critical Care MedicineShanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hong Jiang
- Department of Anesthesiology and Critical Care MedicineShanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Cheng Y, Jiang Y, Zhang L, Wang J, Chai D, Hu R, Li C, Sun Y, Jiang H. Mesenchymal stromal cells attenuate sevoflurane-induced apoptosis in human neuroglioma H4 cells. BMC Anesthesiol 2018; 18:84. [PMID: 30021512 PMCID: PMC6052698 DOI: 10.1186/s12871-018-0553-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 06/27/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Inhalation of sevoflurane can induce neuronal apoptosis, cognitive impairment and abnormal behaviors. Bone marrow mesenchymal stem cells (MSCs) can secret neurotrophic factors and cytokines to protect from oxidative stress-related neuronal apoptosis. However, whether MSCs can protect from sevoflurane-induced neuronal apoptosis and the potential mechanisms are unclear. METHODS A non-contact co-culture of MSCs with human neuroglioma H4 cells (H4 cells) was built. H4 cells were co-cultured with MSCs or without MSCs (control) for 24 h. The co-cultured H4 cells were exposed to 4% sevoflurane for 6 h. The levels of caspase-3, reactive oxygen species (ROS), adenosine triphosphate (ATP), and the release of cytochrome C were determined by Western blot and fluorescence assay. RESULTS Sevoflurane exposure significantly elevated the levels of cleaved caspase 3 and Bax in H4 cells. However, these phenomena were significantly offset by the co-culture with MSCs in H4 cells. Co-culture with MSCs before, but not after, sevoflurane exposure, significantly attenuated sevoflurane-induced ROS production in H4 cells. MSCs prevented sevoflurane-mediated release of cytochrome C from the mitochondria and production of ATP in H4 cells. CONCLUSIONS Our study indicated that soluble factors secreted by MSCs attenuated the sevoflurane-induced oxidative stress and apoptosis of neuronal cells by preserving their mitochondrial function.
Collapse
Affiliation(s)
- Yanyong Cheng
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Yunfeng Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Jiayi Wang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Dongdong Chai
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Rong Hu
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Chunzhu Li
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, 639 Zhizaoju Road, Shanghai, 200011 China
| |
Collapse
|
11
|
Yang Y, Chen X, Min H, Song S, Zhang J, Fan S, Yi L, Wang H, Gu X, Ma Z, Gao Q. Persistent mitoKATP Activation Is Involved in the Isoflurane-induced Cytotoxicity. Mol Neurobiol 2016; 54:1101-1110. [DOI: 10.1007/s12035-016-9710-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 01/11/2016] [Indexed: 01/27/2023]
|
12
|
Duan MH, Wang LN, Jiang YH, Pei YY, Guan DD, Qiu ZD. Angelica sinensis reduced Aβ-induced memory impairment in rats. J Drug Target 2015; 24:340-7. [PMID: 26821843 DOI: 10.3109/1061186x.2015.1077848] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Studies have shown that Angelica sinensis (JiLin AoDong Medicine Industry Groups Co., Ltd., Jilin, China) root (AS) ameliorates various diseases, although its effects in Alzheimer's disease (AD) have not been elucidated. PURPOSE The present study examined the effects of AS in a rat model of AD. METHODS Positional Aβ injections were administered to rats. The behavioral effects of AS administration were examined using the Morris water maze, and the molecular effects on gene and protein expression, and apoptosis, were determined. RESULTS AS reversed the social behavioral impairments observed in this rat model of Aβ-induced memory impairment. Western blot analysis also revealed lower hippocampal levels of Aβ and β-site amyloid precursor protein-cleaving enzyme. Terminal deoxynucleotidyl transferased UTP nick end labeling indicated that AS significantly inhibited apoptosis via effects on nuclear factor kappa B (NF-κB) signaling. Real-time PCR, enzyme-linked immunosorbent assay, and immunohistochemical staining indicated that AS effectively inhibited inflammation and upregulated expression of glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) in the hippocampus of this rat AD model. DISCUSSION AS effectively rescued the symptoms of AD in a rat model by inhibiting inflammation, apoptosis, and NF-κB signaling pathway. CONCLUSION These findings suggested that AS could provide a potential drug for the treatment of AD.
Collapse
Affiliation(s)
- Ming-Hua Duan
- a Changchun University of Chinese Medicine , Changchun , China
| | - Li-Na Wang
- a Changchun University of Chinese Medicine , Changchun , China
| | - Yan-Hong Jiang
- a Changchun University of Chinese Medicine , Changchun , China
| | - Ying-Yuan Pei
- a Changchun University of Chinese Medicine , Changchun , China
| | - Dong-Dong Guan
- a Changchun University of Chinese Medicine , Changchun , China
| | - Zhi-Dong Qiu
- a Changchun University of Chinese Medicine , Changchun , China
| |
Collapse
|
13
|
Wang H, Xu Z, Wu A, Dong Y, Zhang Y, Yue Y, Xie Z. 2-deoxy-D-glucose enhances anesthetic effects in mice. Anesth Analg 2015; 120:312-9. [PMID: 25390277 DOI: 10.1213/ane.0000000000000520] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND The mechanisms of general anesthesia by volatile drugs remain largely unknown. Mitochondrial dysfunction and reduction in energy levels have been suggested to be associated with general anesthesia status. 2-Deoxy-D-glucose (2-DG), an analog of glucose, inhibits hexokinase and reduces cellular levels of adenosine triphosphate (ATP). 3-Nitropropionic acid is another compound which can deplete ATP levels. In contrast, idebenone and L-carnitine could rescue deficits of energy. We therefore sought to determine whether 2-DG and/or 3-nitropropionic acid can enhance the anesthetic effects of isoflurane, and whether idebenone and L-carnitine can reverse the actions of 2-DG. METHODS C57BL/6J mice (8 months old) received different concentrations of isoflurane with and without the treatments of 2-DG, 3-nitropropionic acid, idebenone, and L-carnitine. Isoflurane-induced loss of righting reflex (LORR) was determined in the mice. ATP levels in H4 human neuroglioma cells were assessed after these treatments. Finally, 31P-magnetic resonance spectroscopy was used to determine the effects of isoflurane on brain ATP levels in the mice. RESULTS 2-DG enhanced isoflurane-induced LORR (P = 0.002, N = 15). 3-Nitropropionic acid also enhanced the anesthetic effects of isoflurane (P = 0.005, N = 15). Idebenone (idebenone + saline versus idebenone + 2-DG: P = 0.165, N = 15), but not L-carnitine (L-carnitine + saline versus L-carnitine + 2-DG: P < 0.0001, N = 15), inhibited the effects of 2-DG on enhancing isoflurane-induced LORR in the mice, as evidenced by 2-DG not enhancing isoflurane-induced LORR in the mice pretreated with idebenone. Idebenone (idebenone + saline versus idebenone + 2-DG: P = 0.177, N = 6), but not L-carnitine (L-carnitine + saline versus L-carnitine + 2-DG: P = 0.029, N = 6), also mitigated the effects of 2-DG on reducing ATP levels in cells, as evidenced by 2-DG not decreasing ATP levels in the cells pretreated with idebenone. Finally, isoflurane decreased ATP levels in both cultured cells and mouse brains (β-ATP: P = 0.003, N = 10; β-ATP/phosphocreatine: P = 0.006, N = 10; β-ATP/inorganic phosphate: P = 0.001, N = 10). CONCLUSIONS These results from our pilot studies have established a system and generated a hypothesis that 2-DG enhances anesthetic effects via reducing energy levels. These findings should promote further studies to investigate anesthesia mechanisms.
Collapse
Affiliation(s)
- Hui Wang
- From the *Department of Anesthesia, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; †Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts; and ‡Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Altered metabolomic profiles may be associated with sevoflurane-induced neurotoxicity in neonatal rats. Neurochem Res 2015; 40:788-99. [PMID: 25663300 DOI: 10.1007/s11064-015-1529-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/23/2015] [Accepted: 01/28/2015] [Indexed: 12/25/2022]
Abstract
Experimental studies demonstrate that inhaled anesthetics can cause neurodegeneration and neurobehavioral dysfunctions. Evidence suggests changes in cerebral metabolism following inhaled anesthetics treatment can perturb cerebral homeostasis, which may be associated with their induced neurotoxicity. Seven-day-old rat pups were divided into two groups: control group (Group C) and sevoflurane group (Group S, 3 % sevoflurane exposure for 6 h). Gas chromatography-mass spectrometry (GC-MS) was used for analyzed differential metabolites of cerebral cortex in both groups, Also western blot, flow cytometry, enzymatic methods and electron microscopy were performed in various biochemical and anatomical assays. Sevoflurane exposure significantly elevated caspase-3 activation and ROS levels, decreased mitochondrial cardiolipin contents, and changed cellular ultrastructure in the cerebral cortex. Correspondingly, these results corroborated the GC-MS findings which showed altered metabolic pathways of glucose, amino acids, and lipids, as well as intracellular antioxidants and osmolyte systems in neonatal brain following prolonged exposure to high sevoflurane concentration. Our data indicate that sevoflurane anesthesia causes significant oxidative stress, neuroapoptosis, and cellular ultrastructure damage which is associated with altered brain metabotype in the neonatal rat. Our study also confirmed that GC-MS is a strategic and complementary platform for the metabolomic characterization of sevoflurane-induced neurotoxicity in the developing brain.
Collapse
|
15
|
Schallner N, Ulbrich F, Engelstaedter H, Biermann J, Auwaerter V, Loop T, Goebel U. Isoflurane but Not Sevoflurane or Desflurane Aggravates Injury to Neurons In Vitro and In Vivo via p75NTR-NF-ĸB Activation. Anesth Analg 2014; 119:1429-41. [DOI: 10.1213/ane.0000000000000488] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Wang Q, Zhao Y, Sun M, Liu S, Li B, Zhang L, Yang L. 2-Deoxy-d-glucose attenuates sevoflurane-induced neuroinflammation through nuclear factor-kappa B pathway in vitro. Toxicol In Vitro 2014; 28:1183-9. [PMID: 24907647 DOI: 10.1016/j.tiv.2014.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/03/2014] [Accepted: 05/05/2014] [Indexed: 10/25/2022]
Abstract
OBJECT Sevoflurane, one of the most commonly used anesthetics in clinic, induced neuroinflammation and caused cognitive impairment. 2-deoxy-d-glucose (2-DG) is a synthetic analogue of glucose and is clinically used in medical imaging safely. METHODS We examined the effect of 2-DG on sevoflurane-induced neuroinflammation in the mouse primary microglia cells. Mouse microglia cells were treated with 4.1% sevoflurane for 6h to examine the expression of interleukin (IL)-6 and tumor necrosis factor (TNF-α) and activation of nuclear factor-kappa B (NF-κB). Pyrrolidine dithiocarbamate (PDTC) or 2-DG was used 1h before sevoflurane treatment. RESULTS In the present study, we found that sevoflurane increased level of IL-6 and TNF-α through activating NF-κB signaling, and that 2-DG reduced sevoflurane-induced increase in IL-6 and TNF-α and nuclear NF-κB in microglia cells. CONCLUSION Our data suggests that NF-κB signaling pathway could be a target for sevoflurane-induced neuroinflammation and 2-DG might be a potential therapy to prevent or treat sevoflurane-induced neuroinflammation.
Collapse
Affiliation(s)
- Qingxiu Wang
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, 150#, Jimo Road, Shanghai 200120, PR China
| | - Yupeng Zhao
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, 150#, Jimo Road, Shanghai 200120, PR China
| | - Min Sun
- Medical College of Qingdao University, 16, Jiangsu Road, Shinan District, Qingdao 266071, PR China
| | - Sheng Liu
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, 150#, Jimo Road, Shanghai 200120, PR China
| | - Baolin Li
- Department of Anesthesiology, The First People 's Hospital of Changzhou and The Third Affiliated Hospital of Soochow University, Changzhou, PR China
| | - Lei Zhang
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, 150#, Jimo Road, Shanghai 200120, PR China.
| | - Longqiu Yang
- Department of Anesthesiology, The First People 's Hospital of Changzhou and The Third Affiliated Hospital of Soochow University, Changzhou, PR China.
| |
Collapse
|
17
|
Lei X, Zhang W, Liu T, Xiao H, Liang W, Xia W, Zhang J. Perinatal supplementation with omega-3 polyunsaturated fatty acids improves sevoflurane-induced neurodegeneration and memory impairment in neonatal rats. PLoS One 2013; 8:e70645. [PMID: 23967080 PMCID: PMC3742769 DOI: 10.1371/journal.pone.0070645] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/20/2013] [Indexed: 12/31/2022] Open
Abstract
Objectives To investigate if perinatal Omega-3 polyunsaturated fatty acids (n-3 PUFAs) supplementation can improve sevoflurane-induced neurotoxicity and cognitive impairment in neonatal rats. Methods Female Sprague-Dawley rats (n = 3 each group) were treated with or without an n-3 PUFAs (fish oil) enriched diet from the second day of pregnancy to 14 days after parturition. The offspring rats (P7) were treated with six hours sevoflurane administration (one group without sevoflurane/prenatal n-3 PUFAs supplement as control). The 5-bromodeoxyuridine (Brdu) was injected intraperitoneally during and after sevoflurane anesthesia to assess dentate gyrus (DG) progenitor proliferation. Brain tissues were harvested and subjected to Western blot and immunohistochemistry respectively. Morris water maze spatial reference memory, fear conditioning, and Morris water maze memory consolidation were tested at P35, P63 and P70 (n = 9), respectively. Results Six hours 3% sevoflurane administration increased the cleaved caspase-3 in the thalamus, parietal cortex but not hippocampus of neonatal rat brain. Sevoflurane anesthesia also decreased the neuronal precursor proliferation of DG in rat hippocampus. However, perinatal n-3 PUFAs supplement could decrease the cleaved caspase-3 in the cerebral cortex of neonatal rats, and mitigate the decrease in neuronal proliferation in their hippocampus. In neurobehavioral studies, compared with control and n-3 PUFAs supplement groups, we did not find significant spatial cognitive deficit and early long-term memory impairment in sevoflurane anesthetized neonatal rats at their adulthood. However, sevoflurane could impair the immediate fear response and working memory and short-term memory. And n-3 PUFAs could improve neurocognitive function in later life after neonatal sevoflurane exposure. Conclusion Our study demonstrated that neonatal exposure to prolonged sevoflurane could impair the immediate fear response, working memory and short-term memory of rats at their adulthood, which may through inducing neuronal apoptosis and decreasing neurogenesis. However, these sevoflurane-induced unfavorable neuronal effects can be mitigated by perinatal n-3 PUFAs supplementation.
Collapse
Affiliation(s)
- Xi Lei
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Wenting Zhang
- National Key Laboratory of Medical neurobiology, Fudan University, Shanghai, P. R. China
| | - Tengyuan Liu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Hongyan Xiao
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Weimin Liang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiaotong University, Shanghai, P. R. China
- * E-mail: (WX); (JZ)
| | - Jun Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, P. R. China
- * E-mail: (WX); (JZ)
| |
Collapse
|
18
|
Sevoflurane anesthesia in pregnant mice induces neurotoxicity in fetal and offspring mice. Anesthesiology 2013; 118:516-26. [PMID: 23314109 DOI: 10.1097/aln.0b013e3182834d5d] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Each year, over 75,000 pregnant women in the United States undergo anesthesia care. The authors set out to assess the effects of the anesthetic sevoflurane on neurotoxicity in pregnant mice and on learning and memory in fetal and offspring mice. METHODS Pregnant mice (gestational day 14) and mouse primary neurons were treated with 2.5% sevoflurane for 2 h and 4.1% sevoflurane for 6 h, respectively. Brain tissues of both fetal and offspring mice (P31) and the primary neurons were harvested and subjected to Western blot and immunohistochemistry to assess interleukin-6, the synaptic markers postsynaptic density-95 and synaptophysin, and caspase-3 levels. Separately, learning and memory function in the offspring mice was determined in the Morris water maze. RESULTS Sevoflurane anesthesia in pregnant mice induced caspase-3 activation, increased interleukin-6 levels (256 ± 50.98% [mean ± SD] vs. 100 ± 54.12%, P = 0.026), and reduced postsynaptic density-95 (61 ± 13.53% vs. 100 ± 10.08%, P = 0.036) and synaptophysin levels in fetal and offspring mice. The sevoflurane anesthesia impaired learning and memory in offspring mice at P31. Moreover, interleukin-6 antibody mitigated the sevoflurane-induced reduction in postsynaptic density-95 levels in the neurons. Finally, environmental enrichment attenuated the sevoflurane-induced increases in interleukin-6 levels, reductions of synapse markers, and learning and memory impairment. CONCLUSIONS These results suggest that sevoflurane may induce detrimental effects in fetal and offspring mice, which can be mitigated by environmental enrichment. These findings should promote more studies to determine the neurotoxicity of anesthesia in the developing brain.
Collapse
|
19
|
Zhang L, Zhang J, Yang L, Dong Y, Zhang Y, Xie Z. Isoflurane and sevoflurane increase interleukin-6 levels through the nuclear factor-kappa B pathway in neuroglioma cells. Br J Anaesth 2013; 110 Suppl 1:i82-91. [PMID: 23604542 DOI: 10.1093/bja/aet115] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Isoflurane can increase pro-inflammatory cytokine interleukin (IL)-6 levels. However, the up-stream mechanism remains unknown. Nuclear factor-kappa B (NF-κB) promotes the generation of pro-inflammatory cytokines. We examined the effects of isoflurane and sevoflurane on the NF-κB signalling pathway and its association with IL-6 levels in cultured cells. METHODS H4 human neuroglioma cells (H4 cells), and mouse primary neurones and microglia were treated with 2% isoflurane or 4.1% sevoflurane for 6 h, for analysis of IL-6 and NF-κB. Pyrrolidine dithiocarbamate (an NF-κB inhibitor) or 2-deoxy-d-glucose (2-DG) (an inhibitor of glucose glycolysis) was applied 1 h before anaesthetic treatment. RESULTS Isoflurane or sevoflurane treatment increased the levels of IL-6 [isoflurane: 410% (54); sevoflurane: 290% (24)], the nuclear levels of NF-κB [isoflurane: 170% (36); sevoflurane: 320% (30)], and the transcription activity of NF-κB in H4 cells. Moreover, isoflurane enhanced the transcription activity of NF-κB in mouse microglia, but not primary neurones. Finally, pyrrolidine dithiocarbamate and 2-DG attenuated isoflurane-induced increases in IL-6 and NF-κB, and the transcription activity of NF-κB. CONCLUSIONS These studies in H4 cells suggest that the NF-κB signalling pathway could contribute to isoflurane or sevoflurane-induced neuroinflammation. This could lead to the targeted intervention of anaesthetic-induced neuroinflammation.
Collapse
Affiliation(s)
- L Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | | | | | | | | | | |
Collapse
|
20
|
Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment. Anesthesiology 2013; 118:502-15. [PMID: 23314110 DOI: 10.1097/aln.0b013e3182834d77] [Citation(s) in RCA: 309] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND : Recent population studies have suggested that children with multiple exposures to anesthesia and surgery at an early age are at an increased risk of cognitive impairment. The authors therefore have established an animal model with single versus multiple exposures of anesthetic(s) in young versus adult mice, aiming to distinguish the role of different types of anesthesia in cognitive impairment. METHODS : Six- and 60-day-old mice were exposed to various anesthesia regimens. The authors then determined the effects of the anesthesia on learning and memory function, levels of proinflammatory cytokine interleukin-6 and tumor necrosis factor-α in brain tissues, and the amount of ionized calcium-binding adaptor molecule 1-positive cells, the marker of microglia activation, in the hippocampus. RESULTS : In this article, the authors show that anesthesia with 3% sevoflurane for 2 h daily for 3 days induced cognitive impairment and neuroinflammation (e.g., increased interleukin-6 levels, 151 ± 2.3% [mean ± SD] vs. 100 ± 9.0%, P = 0.035, n = 6) in young but not in adult mice. Anesthesia with 3% sevoflurane for 2 h daily for 1 day and 9% desflurane for 2 h daily for 3 days induced neither cognitive impairment nor neuroinflammation. Finally, an enriched environment and antiinflammatory treatment (ketorolac) ameliorated the sevoflurane-induced cognitive impairment. CONCLUSIONS : Anesthesia-induced cognitive impairment may depend on developmental stage, anesthetic agent, and number of exposures. These findings also suggest the cellular basis and the potential prevention and treatment strategies for anesthesia-induced cognitive impairment, which may ultimately lead to safer anesthesia care and better postoperative outcomes for children.
Collapse
|
21
|
Zhang Y, Dong Y, Xu Z, Xie Z. Propofol and magnesium attenuate isoflurane-induced caspase-3 activation via inhibiting mitochondrial permeability transition pore. Med Gas Res 2012; 2:20. [PMID: 22901676 PMCID: PMC3489514 DOI: 10.1186/2045-9912-2-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 08/03/2012] [Indexed: 11/25/2022] Open
Abstract
Background The inhalation anesthetic isoflurane has been shown to open the mitochondrial permeability transition pore (mPTP) and induce caspase activation and apoptosis, which may lead to learning and memory impairment. Cyclosporine A, a blocker of mPTP opening might attenuate the isoflurane-induced mPTP opening, lessening its ripple effects. Magnesium and anesthetic propofol are also mPTP blockers. We therefore set out to determine whether propofol and magnesium can attenuate the isoflurane-induced caspase activation and mPTP opening. Methods We investigated the effects of magnesium sulfate (Mg2+), propofol, and isoflurane on the opening of mPTP and caspase activation in H4 human neuroglioma cells stably transfected to express full-length human amyloid precursor protein (APP) (H4 APP cells) and in six day-old wild-type mice, employing Western blot analysis and flowcytometry. Results Here we show that Mg2+ and propofol attenuated the isoflurane-induced caspase-3 activation in H4-APP cells and mouse brain tissue. Moreover, Mg2+ and propofol, the blockers of mPTP opening, mitigated the isoflurane-induced mPTP opening in the H4-APP cells. Conclusion These data illustrate that Mg2+ and propofol may ameliorate the isoflurane-induced neurotoxicity by inhibiting its mitochondrial dysfunction. Pending further studies, these findings may suggest the use of Mg2+ and propofol in preventing and treating anesthesia neurotoxicity.
Collapse
Affiliation(s)
- Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th St, Room 4310, Charlestown, MA, 02129-2060, USA.
| | | | | | | |
Collapse
|