1
|
Raabe J, Wittig I, Laurette P, Stathopoulou K, Brand T, Schulze T, Klampe B, Orthey E, Cabrera-Orefice A, Meisterknecht J, Thiemann E, Laufer SD, Shibamiya A, Reinsch M, Fuchs S, Kaiser J, Yang J, Zehr S, Wrona KM, Lorenz K, Lukowski R, Hansen A, Gilsbach R, Brandes RP, Ulmer BM, Eschenhagen T, Cuello F. Physioxia rewires mitochondrial complex composition to protect stem cell viability. Redox Biol 2024; 77:103352. [PMID: 39341035 PMCID: PMC11466565 DOI: 10.1016/j.redox.2024.103352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are an invaluable tool to study molecular mechanisms on a human background. Culturing stem cells at an oxygen level different from their microenvironmental niche impacts their viability. To understand this mechanistically, dermal skin fibroblasts of 52 probands were reprogrammed into hiPSCs, followed by either hyperoxic (20 % O2) or physioxic (5 % O2) culture and proteomic profiling. Analysis of chromosomal stability by Giemsa-banding revealed that physioxic -cultured hiPSC clones exhibited less pathological karyotypes than hyperoxic (e.g. 6 % vs. 32 % mosaicism), higher pluripotency as evidenced by higher Stage-Specific Embryonic Antigen 3 positivity, higher glucose consumption and lactate production. Global proteomic analysis demonstrated lower abundance of several subunits of NADH:ubiquinone oxidoreductase (complex I) and an underrepresentation of pathways linked to oxidative phosphorylation and cellular senescence. Accordingly, release of the pro-senescent factor IGFBP3 and β-galactosidase staining were lower in physioxic hiPSCs. RNA- and ATAC-seq profiling revealed a distinct hypoxic transcription factor-binding footprint, amongst others higher expression of the HIF1α-regulated target NDUFA4L2 along with increased chromatin accessibility of the NDUFA4L2 gene locus. While mitochondrial DNA content did not differ between groups, physioxic hiPSCs revealed lower polarized mitochondrial membrane potential, altered mitochondrial network appearance and reduced basal respiration and electron transfer capacity. Blue-native polyacrylamide gel electrophoresis coupled to mass spectrometry of the mitochondrial complexes detected higher abundance of NDUFA4L2 and ATP5IF1 and loss of incorporation into complex IV or V, respectively. Taken together, physioxic culture of hiPSCs improved chromosomal stability, which was associated with downregulation of oxidative phosphorylation and senescence and extensive re-wiring of mitochondrial complex composition.
Collapse
Affiliation(s)
- Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ilka Wittig
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Patrick Laurette
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Thomas Schulze
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Birgit Klampe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Orthey
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Alfredo Cabrera-Orefice
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Jana Meisterknecht
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Ellen Thiemann
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sandra D Laufer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Aya Shibamiya
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Marina Reinsch
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sigrid Fuchs
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jennifer Kaiser
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jiaqi Yang
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Simonida Zehr
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Kinga M Wrona
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Ralf P Brandes
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Bärbel M Ulmer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
2
|
Gong C, Zhou X, Fang Y, Zhang Y, Zhu L, Ding Z. Effects of sevoflurane on left ventricular function by speckle-tracking echocardiography in coronary bypass patients: A randomized trial. J Biomed Res 2023; 38:76-86. [PMID: 38044669 PMCID: PMC10818177 DOI: 10.7555/jbr.37.20230173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
The present study aimed to dynamically observe the segmental and global myocardial movements of the left ventricle during coronary artery bypass grafting by transesophageal speckle-tracking echocardiography, and to assess the effect of sevoflurane on cardiac function. Sixty-four patients scheduled for the off-pump coronary artery bypass grafting were randomly divided into a sevoflurane-based anesthesia (AS) group and a propofol-based total intravenous anesthesia (AA) group. The AS group demonstrated a higher absolute value of left ventricular global longitudinal strain than that of the AA group at both T 1 (after harvesting all grafts and before coronary anastomosis) and T 2 (30 min after completing all coronary anastomoses) ( P < 0.05). Moreover, strain improvement in the segment with the highest preoperative strain was significantly reduced in the AS group, compared with the AA group at both T 1 and T 2 ( P < 0.01). The flow of the left internal mammary artery-left anterior descending artery graft was superior, and the postoperative concentration of troponin T decreased rapidly in the AS group, compared with the AA group ( P < 0.05). Compared with total intravenous anesthesia, sevoflurane resulted in a significantly higher global longitudinal strain, stroke volume, and cardiac output. Sevoflurane also led to an amelioration in the condition of the arterial graft. Furthermore, sevoflurane significantly reduced strain improvement in the segmental myocardium with a high preoperative strain value. The findings need to be replicated in larger studies.
Collapse
Affiliation(s)
- Chanjuan Gong
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaokai Zhou
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yin Fang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yanjuan Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Linjia Zhu
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
3
|
Li Y, Liang Z, Lei S, Wu X, Yuan T, Ma K, Chi K. Sevoflurane Preconditioning Downregulates GRIA1 Expression to Attenuate Cerebral Ischemia-Reperfusion-Induced Neuronal Injury. Neurotox Res 2023; 41:29-40. [PMID: 36595163 DOI: 10.1007/s12640-022-00620-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 01/04/2023]
Abstract
Cerebral ischemia/reperfusion (I/R) injury is the main cause of death following trauma. The neuroprotective effect of sevoflurane (Sev) has been implicated in cerebral I/R injury. However, the mechanisms remain elusive. In this study, we aimed to explore its function in PC12 exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) and in rats challenged with I/R. Sev pretreatment reduced the damage of PC12 cells after OGD/R treatment. Moreover, Sev pretreatment ameliorated neurobehavioral deficits induced by I/R treatment, reduced brain infarct volume, and decreased apoptosis of neurons in hippocampal tissues. Sev pretreatment reduced the surface expression of glutamate receptor 1 (GRIA1) in neurons, while GRIA1 reduced the neuroprotective effects of Sev pretreatment in vitro and in vivo. There was no difference in the surface expression of GRIA2 in rats with I/R and PC12 cells exposed to OGD/R. The ratio of GRIA1/GRIA2 surface expression was reduced, and calcium permeable-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (CP-AMPAR) was blocked by Sev. Together, Sev might exert beneficial effects on cerebral I/R-induced neuronal injury through inhibiting the surface expression of GRIA1 and blocking CP-AMPAR.
Collapse
Affiliation(s)
- Ye Li
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, 050000, Hebei, People's Republic of China
| | - Zhi Liang
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, 050000, Hebei, People's Republic of China
| | - Shuyan Lei
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, 050000, Hebei, People's Republic of China
| | - Xiaoning Wu
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, 050000, Hebei, People's Republic of China
| | - Tao Yuan
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, 050000, Hebei, People's Republic of China
| | - Kai Ma
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, 050000, Hebei, People's Republic of China
| | - Kui Chi
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, 050000, Hebei, People's Republic of China.
| |
Collapse
|
4
|
Zhang J, Wang H, Sun X. Sevoflurane Postconditioning Reduces Hypoxia/Reoxygenation Injury in Cardiomyocytes via Upregulation of Heat Shock Protein 70. J Microbiol Biotechnol 2021; 31:1069-1078. [PMID: 34226409 PMCID: PMC9705948 DOI: 10.4014/jmb.2103.03040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022]
Abstract
Sevoflurane postconditioning (SPostC) has been proved effective in cardioprotection against myocardial ischemia/reperfusion injury. It was also reported that heat shock protein 70 (HSP70) could be induced by sevoflurane, which played a crucial role in hypoxic/reoxygenation (HR) injury of cardiomyocytes. However, the mechanism by which sevoflurane protects cardiomyocytes via HSP70 is still not understood. Here, we aimed to investigate the related mechanisms of SPostC inducing HSP70 expression to reduce the HR injury of cardiomyocytes. After the HR cardiomyocytes model was established, the cells transfected with siRNA for HSP70 (siHSP70) or not were treated with sevoflurane during reoxygenation. The lactate dehydrogenase (LDH) level was detected by colorimetry while cell viability and apoptosis were detected by MTT and flow cytometry. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blotting were used to detect HSP70, apoptosis-, cell cycle-associated factors, iNOS, and Cox-2 expressions. Enzyme-linked immuno sorbent assay (ELISA) was used to measure malondialdehyde (MDA) and superoxide dismutase (SOD). SPostC decreased apoptosis, cell injury, oxidative stress and inflammation and increased viability of HR-induced cardiomyocytes. In addition, SPostC downregulated Bax and cleaved caspase-3 levels, while SPostC upregulated Bcl-2, CDK-4, Cyclin D1, and HSP70 levels. SiHSP70 had the opposite effect that SPostC had on HR-induced cardiomyocytes. Moreover, siHSP70 further reversed the effect of SPostC on apoptosis, cell injury, oxidative stress, inflammation, viability and the expressions of HSP70, apoptosis-, and cell cycle-associated factors in HR-induced cardiomyocytes. In conclusion, this study demonstrates that SPostC can reduce the HR injury of cardiomyocytes by inducing HSP70 expression.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, P.R. China
| | - Haiyan Wang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, P.R. China
| | - Xizhi Sun
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, P.R. China,Corresponding author Phone: +86-0535-6691999 E-mail:
| |
Collapse
|
5
|
Mouithys-Mickalad A, Ceusters J, Charif M, El Moualij B, Schoumacher M, Plyte S, Franck T, Bettendorff L, Pirotte B, Serteyn D, de Tullio P. Modulation of mitochondrial respiration rate and calcium-induced swelling by new cromakalim analogues. Chem Biol Interact 2020; 331:109272. [PMID: 33010220 DOI: 10.1016/j.cbi.2020.109272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/07/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
A cellular model of cardiomyocytes (H9c2 cell line) and mitochondria isolated from mouse liver were used to understand the drug action of BPDZ490 and BPDZ711, two benzopyran analogues of the reference potassium channel opener cromakalim, on mitochondrial respiratory parameters and swelling, by comparing their effects with those of the parent compound cromakalim. For these three compounds, the oxygen consumption rate (OCR) was determined by high-resolution respirometry (HRR) and their impact on adenosine triphosphate (ATP) production and calcium-induced mitochondrial swelling was investigated. Cromakalim did not modify neither the OCR of H9c2 cells and the ATP production nor the Ca-induced swelling. By contrast, the cromakalim analogue BPDZ490 (1) induced a strong increase of OCR, while the other benzopyran analogue BPDZ711 (2) caused a marked slowdown. For both compounds, 1 displayed a biphasic behavior while 2 still showed an inhibitory effect. Both compounds 1 and 2 were also found to decrease the ATP synthesis, with pronounced effect for 2, while cromakalim remained without effect. Overall, these results indicate that cromakalim, as parent molecule, does not induce per se any direct effect on mitochondrial respiratory function neither on whole cells nor on isolated mitochondria whereas both benzopyran analogues 1 and 2 display totally opposite behavior profiles, suggesting that compound 1, by increasing the maximal respiration capacity, might behave as a mild uncoupling agent and compound 2 is taken as an inhibitor of the mitochondrial electron-transfer chain.
Collapse
Affiliation(s)
- Ange Mouithys-Mickalad
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium.
| | - Justine Ceusters
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium
| | - Mounia Charif
- Centre de Recherche sur les Protéines Prions (CRPP), ULiège, Quartier Hôpital, 15, Avenue Hippocrate, B-4000, Liège, Belgium
| | - Benaïssa El Moualij
- Centre de Recherche sur les Protéines Prions (CRPP), ULiège, Quartier Hôpital, 15, Avenue Hippocrate, B-4000, Liège, Belgium
| | - Mathieu Schoumacher
- Department of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, 15, Avenue de l'Hospital, B-4000, Liège, Belgium
| | - Simon Plyte
- Merus, Closing in on Cancer, Immuno-Oncology, Yalelaan 62, 3584 CM, Utrecht, the Netherlands
| | - Thierry Franck
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium; Department of Clinical Sciences, Faculty of Veterinary Medicine, Quartier Vallée 2, 5A-5D, Avenue de Cureghem, ULiège, B-4000, Liège, Belgium
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-neurosciences, ULiège, Quartier Hôpital, 15, Avenue Hippocrate, B-4000, Liège, Belgium
| | - Bernard Pirotte
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium; Department of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, 15, Avenue de l'Hospital, B-4000, Liège, Belgium
| | - Didier Serteyn
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium
| | - Pascal de Tullio
- Center for Oxygen, Research & Development (CORD) & Center for Interdisciplinary Research on Medicines (CIRM), Institute of Chemistry, B6a, ULiège, Allée du six Août, 11, B-4000, Liège, Belgium; Department of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), ULiège, Quartier Hôpital, 15, Avenue de l'Hospital, B-4000, Liège, Belgium
| |
Collapse
|
6
|
Zhang L, Huang L, Wang J, Zhang M, Zhang Y, Hu X. Sevoflurane postconditioning improves spatial learning and memory ability involving mitochondrial permeability transition pore in hemorrhagic shock and resuscitation rats. Brain Behav 2020; 10:e01501. [PMID: 31833229 PMCID: PMC6955830 DOI: 10.1002/brb3.1501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Hemorrhagic shock induces the cognitive deficiency. Sevoflurane postconditioning has been documented to provide neuroprotection against ischemic-reperfusion injury by suppressing apoptosis. Mitochondrial permeability transition pore (mPTP) plays an important role in apoptosis, but it is unknown if the protective effect of sevoflurane postconditioning on hemorrhagic shock and resuscitation is associated with the change of mPTP opening. Hence, the aim of the study was to find out the precise mechanism of the sevoflurane postconditioning. METHODS Sprague Dawley rats were subjected to hemorrhage shock for 60 min and then exposed to 2.4% sevoflurane for 30 min at the instant of reperfusion. Additionally, an opener (atractyloside) or an inhibitor (cyclosporine A) of mPTP was used in the animal model before sevoflurane postconditioning. Rats were randomly assigned into groups of Sham, Shock, Shock+Sevoflurane, Shock+Atractyloside, Shock+Sevoflurane+Atractyloside, Shock+Cyclosporin A, and Shock+Sevoflurane+Cyclosporin A treatment. Rat behavior was assessed for 5 days by Morris water maze 72 hr after surgery, and then hippocampus CA1 region was immunohistochemically stained. Brains were harvested 24 hr after surgery to detect the protein expression levels of Bcl-2, Bax, and cytochrome C by Western blot, the changes of mPTP opening, and mitochondrial membrane potential (MMP). RESULTS We found that sevoflurane postconditioning significantly improved rats' spatial learning and memory ability, down-regulated the expression of Bax, cytochrome C, and caspase-3, up-regulated the expression of Bcl-2, decreased the mPTP opening, and increased the MMP. The neuroprotective effect of sevoflurane postconditioning was abolished by atractyloside, but cyclosporin A played the similar protective role as sevoflurane postconditioning. CONCLUSION These findings proved that sevoflurane postconditioning improved spatial learning and memory ability in hemorrhage shock and resuscitation rats, the mechanism of which may be related to block mPTP opening, increase MMP, and reduce neuron apoptosis in the hippocampus.
Collapse
Affiliation(s)
- Li Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Li Huang
- Department of Anesthesiology, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an, China
| | - Jingxian Wang
- Department of Anesthesiology, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an, China
| | - Muchun Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xianwen Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Sevoflurane Promotes Regeneration of the Endothelial Glycocalyx by Upregulating Sialyltransferase. J Surg Res 2019; 241:40-47. [DOI: 10.1016/j.jss.2019.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/21/2019] [Accepted: 03/20/2019] [Indexed: 01/04/2023]
|
8
|
Yang L, Wu J, Xie P, Yu J, Li X, Wang J, Zheng H. Sevoflurane postconditioning alleviates hypoxia-reoxygenation injury of cardiomyocytes by promoting mitochondrial autophagy through the HIF-1/BNIP3 signaling pathway. PeerJ 2019; 7:e7165. [PMID: 31275755 PMCID: PMC6596409 DOI: 10.7717/peerj.7165] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/22/2019] [Indexed: 12/14/2022] Open
Abstract
Background Sevoflurane postconditioning (SpostC) can alleviate hypoxia-reoxygenation injury of cardiomyocytes; however, the specific mechanism remains unclear. This study aimed to investigate whether SpostC promotes mitochondrial autophagy through the hypoxia-inducible factor-1 (HIF-1)/BCL2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3) signaling pathway to attenuate hypoxia-reoxygenation injury in cardiomyocytes. Methods The H9C2 cardiomyocyte hypoxia/reoxygenation model was established and treated with 2.4% sevoflurane at the beginning of reoxygenation. Cell damage was determined by measuring cell viability, lactate dehydrogenase activity, and apoptosis. Mitochondrial ultrastructural and autophagosomes were observed by transmission electron microscope. Western blotting was used to examine the expression of HIF-1, BNIP3, and Beclin-1 proteins. The effects of BNIP3 on promoting autophagy were determined using interfering RNA technology to silence BNIP3. Results Hypoxia-reoxygenation injury led to accumulation of autophagosomes in cardiomyocytes, and cell viability was significantly reduced, which seriously damaged cells. Sevoflurane postconditioning could upregulate HIF-1α and BNIP3 protein expression, promote autophagosome clearance, and reduce cell damage. However, these protective effects were inhibited by 2-methoxyestradiol or sinBNIP3. Conclusion Sevoflurane postconditioning can alleviate hypoxia-reoxygenation injury in cardiomyocytes, and this effect may be achieved by promoting mitochondrial autophagy through the HIF-1/BNIP3 signaling pathway.
Collapse
Affiliation(s)
- Long Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jianjiang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Peng Xie
- Department of Anesthesiology, Zunyi Medical College, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi, Guizhou, China
| | - Jin Yu
- Chongqing Health Center for Women and Children, Department of Anesthesiology, Chongqing, Chongqing, China
| | - Xin Li
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hong Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
9
|
Sevoflurane Preconditioning plus Postconditioning Decreases Inflammatory Response with Hemodynamic Recovery in Experimental Liver Ischemia Reperfusion. Gastroenterol Res Pract 2019; 2019:5758984. [PMID: 31093276 PMCID: PMC6476030 DOI: 10.1155/2019/5758984] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/13/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Objective The inhalation anesthetic sevoflurane has presented numerous biological activities, including anti-inflammatory properties and protective effects against tissue ischemic injury. This study investigated the metabolic, hemodynamic, and inflammatory effects of sevoflurane pre- and postconditioning for short periods in the rescue of liver ischemia-reperfusion (IR) injury using a rat model. Materials and Methods Twenty Wistar rats were divided into four groups: sham group, control ischemia group (partial warm liver ischemia for 45 min followed by 4 h of reperfusion), SPC group (administration of sevoflurane 2.5% for 15 min with 5 min of washout before liver IR), and SPPoC group (administration of sevoflurane 2.5% for 15 min before ischemia and 20 min during reperfusion). Results All animals showed a decrease in the mean arterial pressure (MAP) and portal vein blood flow during ischemia. After 4 h of reperfusion, only the SPPoC group had MAP recovery. In both the SPC and SPPoC groups, there was a decrease in the ALT level and an increase in the bicarbonate and potassium serum levels. Only the SPPoC group showed an increase in the arterial blood ionized calcium level and a decrease in the IL-6 level after liver reperfusion. Therefore, this study demonstrated that sevoflurane preconditioning reduces hepatocellular injury and acid-base imbalance in liver ischemia. Furthermore, sevoflurane postconditioning promoted systemic hemodynamic recovery with a decrease in inflammatory response.
Collapse
|
10
|
Randhawa PK, Bali A, Virdi JK, Jaggi AS. Conditioning-induced cardioprotection: Aging as a confounding factor. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:467-479. [PMID: 30181694 PMCID: PMC6115349 DOI: 10.4196/kjpp.2018.22.5.467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 05/15/2018] [Indexed: 01/15/2023]
Abstract
The aging process induces a plethora of changes in the body including alterations in hormonal regulation and metabolism in various organs including the heart. Aging is associated with marked increase in the vulnerability of the heart to ischemia-reperfusion injury. Furthermore, it significantly hampers the development of adaptive response to various forms of conditioning stimuli (pre/post/remote conditioning). Aging significantly impairs the activation of signaling pathways that mediate preconditioning-induced cardioprotection. It possibly impairs the uptake and release of adenosine, decreases the number of adenosine transporter sites and down-regulates the transcription of adenosine receptors in the myocardium to attenuate adenosine-mediated cardioprotection. Furthermore, aging decreases the expression of peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α) and subsequent transcription of catalase enzyme which subsequently increases the oxidative stress and decreases the responsiveness to preconditioning stimuli in the senescent diabetic hearts. In addition, in the aged rat hearts, the conditioning stimulus fails to phosphorylate Akt kinase that is required for mediating cardioprotective signaling in the heart. Moreover, aging increases the concentration of Na+ and K+, connexin expression and caveolin abundance in the myocardium and increases the susceptibility to ischemia-reperfusion injury. In addition, aging also reduces the responsiveness to conditioning stimuli possibly due to reduced kinase signaling and reduced STAT-3 phosphorylation. However, aging is associated with an increase in MKP-1 phosphorylation, which dephosphorylates (deactivates) mitogen activated protein kinase that is involved in cardioprotective signaling. The present review describes aging as one of the major confounding factors in attenuating remote ischemic preconditioning-induced cardioprotection along with the possible mechanisms.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | - Anjana Bali
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur 148002, India
| | - Jasleen Kaur Virdi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| |
Collapse
|
11
|
Liu TJ, Zhang JC, Gao XZ, Tan ZB, Wang JJ, Zhang PP, Cheng AB, Zhang SB. Effect of sevoflurane on the ATPase activity of hippocampal neurons in a rat model of cerebral ischemia-reperfusion injury via the cAMP-PKA signaling pathway. Kaohsiung J Med Sci 2017; 34:22-33. [PMID: 29310813 DOI: 10.1016/j.kjms.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/05/2017] [Accepted: 09/13/2017] [Indexed: 01/01/2023] Open
Abstract
We aim to investigate the effects of sevoflurane on the ATPase activity of the hippocampal neurons in rats with cerebral ischemia-reperfusion injury (IRI) via the cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) signaling pathway. Sixty rats were assigned into the normal, model and sevoflurane groups (n = 20, the latter two groups were established as focal cerebral IRI models). The ATPase activity was detected using an ultramicro Na (+)-K (+)-ATP enzyme kit. Immunohistochemical staining was used to detect the positive protein expression of cAMP and PKA. The hippocampal neurons were assigned to the normal, IRI, IRI + sevoflurane, IRI + forskolin, IRI + H89 and IRI + sevoflurane + H89 groups. qRT-PCR and Western blotting were performed for the expressions of cAMP, PKA, cAMP-responsive element-binding protein (CREB) and brain derived neurotrophic factor (BDNF). The normal and sevoflurane groups exhibited a greater positive protein expression of cAMP and PKA than the model group. Compared with the normal group, the expressions of cAMP, PKA, CREB and BDNF all reduced in the IRI, model and IRI + H89 groups. The sevoflurane group showed higher cAMP, PKA, CREB and BDNF expressions than the model group. Compared with the IRI group, ATPase activity and expressions of cAMP, PKA, CREB and BDNF all increased in the normal, IRI + sevoflurane and IRI + forskolin groups but decreased in the IRI + H89 group. It suggests that sevoflurane could enhance ATPase activity in hippocampal neurons of cerebral IRI rats through activating cAMP-PKA signaling pathway.
Collapse
Affiliation(s)
- Tie-Jun Liu
- Department of Anesthesia, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Jin-Cun Zhang
- Department of Urology Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Xiao-Zeng Gao
- Department of Anesthesia, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Zhi-Bin Tan
- Department of Anesthesia, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Jian-Jun Wang
- Department of Critical Care Medicine, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Pan-Pan Zhang
- Department of Respiratory Medicine, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Ai-Bin Cheng
- Department of Critical Care Medicine, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China
| | - Shu-Bo Zhang
- Department of Anesthesia, The Affiliated Hospital of North China University of Science and Technology, Tangshan, PR China.
| |
Collapse
|