1
|
Yang Y, Song S, Wang H, Ma Z, Gao Q. The antioxidative effect of STAT3 involved in cellular vulnerability to isoflurane. BMC Neurosci 2024; 25:75. [PMID: 39633283 PMCID: PMC11619428 DOI: 10.1186/s12868-024-00911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The vulnerable period to neurotoxicity of isoflurane overlaps with a developmental stage characterized by programmed neuronal death. STAT3 has been identified as a crucial molecule involved in survival pathways during this period. We aimed to investigate the role of STAT3 in cellular vulnerability to isoflurane. METHODS C57/BL6 mice on postnatal day 7 or 21, primary neurons derived from mice embryos at gestational days 14-16 and cultured for 5 or 14 days, as well as human neuroglioma U251 cells were treated with isoflurane. A plasmid containing human wild-type STAT3, STAT3 anti-sense oligonucleotide, STAT3 specific inhibitor STA21, proteasome inhibitor MG-132 and calcineurin inhibitor FK506 were utilized to evaluate the influence of STAT3 levels on isoflurane-induced cytotoxicity. The levels of Western blot results, mRNA, intracellular ROS, apoptotic rate, and calcineurin activity were analyzed using unpaired Student's t-test or one-way ANOVA followed by Bonferroni post hoc test, as appropriate. RESULTS Elevated levels of STAT3, reduced activity of calcineurin, as well as a diminished response to isoflurane-induced calcineurin activation and neuroapoptosis were observed in more mature brain or neurons. Isoflurane accelerated the degradation of ubiquitin-conjugated proteins but did not facilitate ubiquitin conjugation to proteins. STAT3 was of particular importance in the all ubiquitin-conjugated proteins degraded by isoflurane. Knockdown or inhibition of STAT3 nuclear translocation exacerbated isoflurane-induced oxidative injury and apoptosis, while STAT3 overexpression mitigated these effects. Finally, this study demonstrated that FK506 pretreatment mitigated the apoptosis, ROS accumulation, and the impairment of neurite growth in primary neurons after exposed to isoflurane. CONCLUSIONS These findings indicate that specific regulation of STAT3 was closely related with the cellular vulnerability to isoflurane via an antioxidative pathway.
Collapse
Affiliation(s)
- Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| | - Qian Gao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| |
Collapse
|
2
|
Liu WT, Li CQ, Fu AN, Yang HT, Xie YX, Yao H, Yi GH. Therapeutic implication of targeting mitochondrial drugs designed for efferocytosis dysfunction. J Drug Target 2024; 32:1169-1185. [PMID: 39099434 DOI: 10.1080/1061186x.2024.2386620] [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: 05/09/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Efferocytosis refers to the process by which phagocytes remove apoptotic cells and related apoptotic products. It is essential for the growth and development of the body, the repair of damaged or inflamed tissues, and the balance of the immune system. Damaged efferocytosis will cause a variety of chronic inflammation and immune system diseases. Many studies show that efferocytosis is a process mediated by mitochondria. Mitochondrial metabolism, mitochondrial dynamics, and communication between mitochondria and other organelles can all affect phagocytes' clearance of apoptotic cells. Therefore, targeting mitochondria to modulate phagocyte efferocytosis is an anticipated strategy to prevent and treat chronic inflammatory diseases and autoimmune diseases. In this review, we introduced the mechanism of efferocytosis and the pivoted role of mitochondria in efferocytosis. In addition, we focused on the therapeutic implication of drugs targeting mitochondria in diseases related to efferocytosis dysfunction.
Collapse
Affiliation(s)
- Wan-Ting Liu
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Chao-Quan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Ao-Ni Fu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Hao-Tian Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Yu-Xin Xie
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Hui Yao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Guang-Hui Yi
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| |
Collapse
|
3
|
Zhang Z, Yang W, Wang L, Zhu C, Cui S, Wang T, Gu X, Liu Y, Qiu P. Unraveling the role and mechanism of mitochondria in postoperative cognitive dysfunction: a narrative review. J Neuroinflammation 2024; 21:293. [PMID: 39533332 PMCID: PMC11559051 DOI: 10.1186/s12974-024-03285-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a frequent neurological complication encountered during the perioperative period with unclear mechanisms and no effective treatments. Recent research into the pathogenesis of POCD has primarily focused on neuroinflammation, oxidative stress, changes in neural synaptic plasticity and neurotransmitter imbalances. Given the high-energy metabolism of neurons and their critical dependency on mitochondria, mitochondrial dysfunction directly affects neuronal function. Additionally, as the primary organelles generating reactive oxygen species, mitochondria are closely linked to the pathological processes of neuroinflammation. Surgery and anesthesia can induce mitochondrial dysfunction, increase mitochondrial oxidative stress, and disrupt mitochondrial quality-control mechanisms via various pathways, hence serving as key initiators of the POCD pathological process. We conducted a review on the role and potential mechanisms of mitochondria in postoperative cognitive dysfunction by consulting relevant literature from the PubMed and EMBASE databases spanning the past 25 years. Our findings indicate that surgery and anesthesia can inhibit mitochondrial respiration, thereby reducing ATP production, decreasing mitochondrial membrane potential, promoting mitochondrial fission, inducing mitochondrial calcium buffering abnormalities and iron accumulation, inhibiting mitophagy, and increasing mitochondrial oxidative stress. Mitochondrial dysfunction and damage can ultimately lead to impaired neuronal function, abnormal synaptic transmission, impaired synthesis and release of neurotransmitters, and even neuronal death, resulting in cognitive dysfunction. Targeted mitochondrial therapies have shown positive outcomes, holding promise as a novel treatment for POCD.
Collapse
Affiliation(s)
- Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Wei Yang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Lanbo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chengyao Zhu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Shuyan Cui
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Tian Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
4
|
Tashima K, Hayashi M, Oyoshi T, Uemura J, Korematsu S, Hirata N. Anesthesia management for percutaneous mitral valve repair in a patient with mitochondrial cardiomyopathy and low cardiac function: a case report. JA Clin Rep 2024; 10:49. [PMID: 39115707 PMCID: PMC11310374 DOI: 10.1186/s40981-024-00734-z] [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: 07/25/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Mitochondrial cardiomyopathy occurs when impaired mitochondrial energy production leads to myocardial dysfunction. Anesthetic management in such cases is challenging due to risks of circulatory depression associated with anesthesia and mitochondrial dysfunction induced by anesthetics. Although there are reports of anesthetic management for patients with mitochondrial diseases, there are few reports specifically addressing cardiac anesthesia for patients with mitochondrial cardiomyopathy. We present a case where percutaneous mitral valve repair with MitraClip™ was successfully performed under remimazolam anesthesia in a patient with mitochondrial cardiomyopathy who developed functional mitral valve regurgitation due to low cardiac function and cardiomegaly. CASE PRESENTATION A 57-year-old woman was diagnosed with chronic cardiac failure, with a 10-year history of dilated cardiomyopathy. She was diagnosed with mitochondrial cardiomyopathy 8 years ago. Over the past 2 years, her cardiac failure worsened, and mitral valve regurgitation gradually developed. Surgical intervention was considered but deemed too risky due to her low cardiac function, with an ejection fraction of 26%. Therefore, percutaneous MitraClip™ implantation was selected. After securing radial artery and central venous catheterization under sedation with dexmedetomidine, anesthesia was induced with a low dose of remimazolam 4 mg/kg/h. Anesthesia was maintained with remimazolam 0.35-1.0 mg/kg/h and remifentanil 0.1 μg/kg/min. Noradrenaline and dobutamine were administered intraoperatively, and the procedure was completed successfully without circulatory collapse. The patient recovered smoothly from anesthesia and experienced no complications. She was discharged on the eighth day after surgery. CONCLUSION Anesthesia management with remimazolam appears to be a safe and effective for MitraClip™ implantation in patients with mitochondrial cardiomyopathy.
Collapse
Affiliation(s)
- Koichiro Tashima
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan
| | - Masakiyo Hayashi
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan
| | - Takafumi Oyoshi
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan
| | - Jo Uemura
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan
| | - Shinnosuke Korematsu
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan
| | - Naoyuki Hirata
- Department of Anesthesiology, Kumamoto University Hospital, 1-1-1, Honjo, Chuo-Ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
5
|
Zhang P, Shi X, He D, Hu Y, Zhang Y, Zhao Y, Ma S, Cao S, Zhai M, Fan Z. Fer-1 Protects against Isoflurane-Induced Ferroptosis in Astrocytes and Cognitive Impairment in Neonatal Mice. Neurotox Res 2024; 42:27. [PMID: 38819761 DOI: 10.1007/s12640-024-00706-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Early and prolonged exposure to anesthetic agents could cause neurodevelopmental disorders in children. Astrocytes, heavily outnumber neurons in the brain, are crucial regulators of synaptic formation and function during development. However, how general anesthetics act on astrocytes and the impact on cognition are still unclear. In this study, we investigated the role of ferroptosis and GPX4, a major hydroperoxide scavenger playing a pivotal role in suppressing the process of ferroptosis, and their underlying mechanism in isoflurane-induced cytotoxicity in astrocytes and cognitive impairment. Our results showed that early 6 h isoflurane anesthesia induced cognitive impairment in mice. Ferroptosis-relative genes and metabolic changes were involved in the pathological process of isoflurane-induced cytotoxicity in astrocytes. The level of GPX4 was decreased while the expression of 4-HNE and generation of ROS were elevated after isoflurane exposure. Selectively blocking ferroptosis with Fer-1 attenuated the abovementioned cytotoxicity in astrocytes, paralleling with the reverse of the changes in GPX4, ROS and 4-HNE secondary to isoflurane anesthesia. Fer-1 attenuated the cognitive impairment induced by prolonged isoflurane exposure. Thus, ferroptosis conduced towards isoflurane-induced cytotoxicity in astrocytes via suppressing GPX4 and promoting lipid peroxidation. Fer-1 was expected to be an underlying intervention for the neurotoxicity induced by isoflurane in the developing brain, and to alleviate cognitive impairment in neonates.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Anesthesiology, Air Force Hospital of Western Theater Command, PLA, Chengdu, 610011, China
| | - Xiaotong Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Danyi He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Hu
- Department of Anesthesiology, Air Force Hospital of Western Theater Command, PLA, Chengdu, 610011, China
| | - Yongchao Zhang
- Air Force Hospital of Western Theater Command, PLA, Chengdu, 610011, China
| | - Youyi Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Sanxing Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuhui Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Meiting Zhai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ze Fan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Perouansky M, Johnson-Schlitz D, Sedensky MM, Morgan PG. A primordial target: Mitochondria mediate both primary and collateral anesthetic effects of volatile anesthetics. Exp Biol Med (Maywood) 2023; 248:545-552. [PMID: 37208922 PMCID: PMC10350799 DOI: 10.1177/15353702231165025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023] Open
Abstract
One of the unsolved mysteries of medicine is how do volatile anesthetics (VAs) cause a patient to reversibly lose consciousness. In addition, identifying mechanisms for the collateral effects of VAs, including anesthetic-induced neurotoxicity (AiN) and anesthetic preconditioning (AP), has proven challenging. Multiple classes of molecules (lipids, proteins, and water) have been considered as potential VA targets, but recently proteins have received the most attention. Studies targeting neuronal receptors or ion channels had limited success in identifying the critical targets of VAs mediating either the phenotype of "anesthesia" or their collateral effects. Recent studies in both nematodes and fruit flies may provide a paradigm shift by suggesting that mitochondria may harbor the upstream molecular switch activating both primary and collateral effects. The disruption of a specific step of electron transfer within the mitochondrion causes hypersensitivity to VAs, from nematodes to Drosophila and to humans, while also modulating the sensitivity to collateral effects. The downstream effects from mitochondrial inhibition are potentially legion, but inhibition of presynaptic neurotransmitter cycling appears to be specifically sensitive to the mitochondrial effects. These findings are perhaps of even broader interest since two recent reports indicate that mitochondrial damage may well underlie neurotoxic and neuroprotective effects of VAs in the central nervous system (CNS). It is, therefore, important to understand how anesthetics interact with mitochondria to affect CNS function, not just for the desired facets of general anesthesia but also for significant collateral effects, both harmful and beneficial. A tantalizing possibility exists that both the primary (anesthesia) and secondary (AiN, AP) mechanisms may at least partially overlap in the mitochondrial electron transport chain (ETC).
Collapse
Affiliation(s)
- Misha Perouansky
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Laboratory of Genetics, School of Medicine and Public Health and College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dena Johnson-Schlitz
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Margaret M Sedensky
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Philip G Morgan
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA 98101, USA
| |
Collapse
|
7
|
Johnson-Schlitz D, Fischer JA, Schiffman HJ, Scharenbrock AR, Olufs ZPG, Wassarman DA, Perouansky M. Anesthetic Preconditioning of Traumatic Brain Injury Is Ineffective in a Drosophila Model of Obesity. J Pharmacol Exp Ther 2022; 381:229-235. [PMID: 35347062 PMCID: PMC9190232 DOI: 10.1124/jpet.121.000997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/21/2022] [Indexed: 07/09/2024] Open
Abstract
We tested the hypothesis that obesity influences the pharmacodynamics of volatile general anesthetics (VGAs) by comparing effects of anesthetic exposure on mortality from traumatic brain injury (TBI) in lean and obese Drosophila melanogaster We induced TBI with a high-impact trauma device. Starvation-selection over multiple generations resulted in an obese phenotype (SS flies). Fed flies served as lean controls (FC flies). Adult (1-8-day-old) SS and FC flies were exposed to equianesthetic doses of isoflurane or sevoflurane either before or after TBI. The principal outcome was percent mortality 24 hours after injury, expressed as the Mortality Index at 24 hours (MI24). TBI resulted in a lower MI24 in FC than in SS flies [21 (2.35) and 57.8 (2.14), respectively n = 12, P = 0.0001]. Pre-exposure to isoflurane or sevoflurane preconditioned FC flies to TBI, reducing the risk of death to 0.53 (0.25 to 1.13) and 0.82 (0.43 to 1.58), respectively, but had no preconditioning effect in SS flies. Postexposure to isoflurane or sevoflurane increased the risk of death in SS flies, but only postexposure to isoflurane increased the risk in FC flies [1.39 (0.81 to 2.38)]. Thus, obesity affects the pharmacodynamics of VGAs, thwarting the preconditioning effect of isoflurane and sevoflurane in TBI. SIGNIFICANCE STATEMENT: Inadvertent preconditioning in models of traumatic brain injury (TBI) is a recognized confounder. The findings in a fruit fly (Drosophila melanogaster) model of closed-head TBI indicate that anesthetic pharmacodynamics are profoundly affected by obesity. Specifically, obesity thwarts the brain-protective effect of anesthetic preconditioning. This finding is important for experimental studies of TBI and supports the versatility of the fruit fly as a model for the exploration of anesthetic pharmacodynamics in a wide parameter space.
Collapse
Affiliation(s)
- Dena Johnson-Schlitz
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Julie A Fischer
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Hannah J Schiffman
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Amanda R Scharenbrock
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Zachariah P G Olufs
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - David A Wassarman
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Misha Perouansky
- Department of Anesthesiology (D.J.-S., J.A.F., H.J.S., A.R.S., Z.P.G.O., M.P.) and Department of Medical Genetics (D.A.W.), University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
8
|
Cao Y, Cui Y, Liao J, Gao C, Zhao Z, Zhang J. Sevoflurane Preconditioning Increases Stress Resistance via IMB-2/DAF-16 in Caenorhabditis Elegans. Dose Response 2022; 20:15593258221082886. [PMID: 35360453 PMCID: PMC8961365 DOI: 10.1177/15593258221082886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/03/2022] [Indexed: 11/15/2022] Open
Abstract
Sevoflurane preconditioning has been proved to possess therapeutic effects on stress. However, the mechanism by which sevoflurane preconditioning protects against stress remains unclear. In this study, an acute model of heat stress in C.eleans was established. We investigated the dose-response of sevoflurane exposure on coordinated movement in C.elegans and time course for protection against heat stress of sevoflurane preconditioning to determine the optimal concentration and time point in the following experiments. EC99 of sevoflurane is 1.7% (1.3EC50) and sevoflurane preconditioning exerts the maximal protection at 6 hours after incubation, and these 2 parameters were used in our following experiments. We found that sevoflurane preconditioning increased DAF-16 nuclear translocation and enhanced the expression of DAF-16 during heat stress in N2 strain of C.elegans. DAF-16 mutation abolished the sevoflurane preconditioning-induced protection for heat stress. Furthermore, suppression of IMB-2 by RNAi prevented the upregulation of DAF-16 and enhancement of stress resistance caused by sevoflurane preconditioning. Overall, this work reveals that sevoflurane preconditioning increases the expression of DAF-16 via IMB-2 to enhance the stress resistance of C.elegans.
Collapse
Affiliation(s)
- Yue Cao
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yongchen Cui
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Junling Liao
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Chente Gao
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Zhe Zhao
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Junfeng Zhang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
9
|
Benković V, Borojević N, Šikić D, Horvat Knežević A, Milić M. DNA damage assessment in peripheral blood of Swiss albino mice after combined exposure to volatile anesthetics and 1 or 2 Gy radiotherapy in vivo. Int J Radiat Biol 2021; 97:1425-1435. [PMID: 34328801 DOI: 10.1080/09553002.2021.1962565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Patient immobilization by general volatile anesthesia (VA) may be necessary during medical radiology treatment, and its use has increased in recent years. Although ionizing radiation (IR) is a well-known genotoxic and cytotoxic agent, and VA exposure has caused a range of side effects among patients and occupationally exposed personnel, there are no studies to date comparing DNA damage effects from combined VA and single fractional IR dose exposure. MATERIAL AND METHODS We investigate whether there is a difference in white blood cells DNA damage response (by the alkaline comet assay) in vivo in 185 healthy Swiss albino mice divided into 37 groups, anesthetized with isoflurane/sevoflurane/halothane and exposed to 1 or 2 Gy of IR. Blood samples were taken after 0, 2, 6 and 24 h after exposure, and comet parameters were measured: tail length, tail intensity and tail moment. The cellular DNA repair index was calculated to quantify the efficiency of cells in repairing and re-joining DNA strand breaks following different treatments. RESULTS In combined exposures, halothane caused higher DNA damage levels that were dose-dependent; sevoflurane damage increase did not differ significantly from the initial 1 Gy dose, and isoflurane even demonstrated a protective effect, particularly in the 2 Gy dose combined exposure. Nevertheless, none of the exposures reached control levels even after 24 h. CONCLUSION Halothane appears to increase the level of radiation-induced DNA damage, while sevoflurane and isoflurane exhibited a protective effect. DNA damage may have been even greater in target organs such as liver, kidney or even the brain, and this is proposed for future study.
Collapse
Affiliation(s)
- Vesna Benković
- Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Nikola Borojević
- Warrington and Halton Teaching Hospitals, NHS Foundation Trust, Lovely Ln, Warrington, UK
| | - Dunja Šikić
- Faculty of Science, University of Zagreb, Zagreb, Croatia
| | | | - Mirta Milić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| |
Collapse
|
10
|
Isoflurane impairs oogenesis through germ cell apoptosis in C. elegans. Sci Rep 2021; 11:14481. [PMID: 34262087 PMCID: PMC8280124 DOI: 10.1038/s41598-021-93737-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/23/2021] [Indexed: 02/08/2023] Open
Abstract
Anesthetic isoflurane has been reported to induce toxicity. However, the effects of isoflurane on fecundity remain largely unknown. We established a system in C. elegans to investigate the effects of isoflurane on oogenesis. Synchronized L4 stage C. elegans were treated with 7% isoflurane for 4 h. Dead cells, ROS, embryos, and unfertilized eggs laid by hermaphrodites were measured by fluorescence imaging and counting. The C. elegans with losses of ced-3, cep-1, abl-1, male C. elegans, and oxidative stress inhibitor N-acetyl-cysteine were used in the interaction studies. We found that isoflurane decreased the numbers of embryos and unfertilized eggs and increased the levels of dead cells and ROS in C. elegans. The isoflurane-induced impairment of oogenesis was associated with abl-1, ced-3, but not cep-1. N-acetyl-cysteine attenuated the isoflurane-induced impairment of oogenesis in C. elegans. Mating with male C. elegans did not attenuate the isoflurane-induced changes in oogenesis. These findings suggest that isoflurane may impair oogenesis through abl-1- and ced-3-associated, but not cep-1-associated, germ cell apoptosis and oxidative stress, pending further investigation. These studies will promote more research to determine the potential effects of anesthesia on fecundity.
Collapse
|
11
|
Ferroptosis contributes to isoflurane-induced neurotoxicity and learning and memory impairment. Cell Death Dis 2021; 7:72. [PMID: 33828088 PMCID: PMC8027876 DOI: 10.1038/s41420-021-00454-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/21/2021] [Accepted: 03/18/2021] [Indexed: 02/02/2023]
Abstract
Ferroptosis is a novel type of programmed cell death, which is different from apoptosis and autophagic cell death. Recently, ferroptosis has been indicated to contribute to the in vitro neurotoxicity induced by isoflurane, which is one of the most common anesthetics in clinic. However, the in vivo position of ferroptosis in isoflurane-induced neurotoxicity as well as learning and memory impairment remains unclear. In this study, we mainly explored the relationship between ferroptosis and isoflurane-induced learning and memory, as well as the therapeutic methods in mouse model. Our results indicated that isoflurane induced the ferroptosis in a dose-dependent and time-dependent manner in hippocampus, the organ related with learning and memory ability. In addition, the activity of cytochrome c oxidase/Complex IV in mitochondrial electron transport chain (ETC) was increased by isoflurane, which might further contributed to cysteine deprivation-induced ferroptosis caused by isoflurane exposure. More importantly, isoflurane-induced ferroptosis could be rescued by both ferroptosis inhibitor (ferrostatin-1) and mitochondria activator (dimethyl fumarate), which also showed effective therapeutic action against isoflurane-induced learning and memory impairment. Taken together, our data indicate the close association among ferroptosis, mitochondria and isoflurane, and provide a novel insight into the therapy mode against isoflurane-induced learning and memory impairment.
Collapse
|
12
|
Acute Hypobaric and Hypoxic Preconditioning Reduces Myocardial Ischemia-Reperfusion Injury in Rats. Cardiol Res Pract 2021; 2021:6617374. [PMID: 33815836 PMCID: PMC7990552 DOI: 10.1155/2021/6617374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/30/2022] Open
Abstract
Background Chronic and/or intermittent exposure to hypobaric hypoxia reportedly exerts cardioprotective effects against ischemia-reperfusion injury. However, few studies have focused on the cardioprotective effects of acute and/or short-term hypobaric and hypoxic exposures. This study investigated the effects of acute hypobaric hypoxia on myocardial ischemia-reperfusion injury. Materials and Methods Rats were assigned to groups receiving normobaric normoxia (NN group), hypobaric hypoxia (HH group), or normobaric hypoxia (NH group). HH group rats were exposed to 60.8 kPa and 12.6% fraction of inspired oxygen in a hypobaric chamber for 6 h. NH group rats were exposed to hypoxic conditions under normal pressure. After each exposure, 30 min of myocardial ischemia was followed by 60 min of reperfusion. Cardiac function and infarct size were determined after reperfusion. Expression of hypoxia-inducible factor 1 alpha (HIF1α) was also measured. Results Cardiac function was better preserved in the HH and NH groups than in the NN group (p < 0.01 each). Median infarct size/area at risk was significantly lower in the HH group (50%, interquartile range [IQR] 48–54%; p < 0.01 vs. NN group) and NH group (45%, IQR 36–50%; p < 0.01 vs. NN group) than in the NN group (72%, IQR 69–75%). HIF1α expression was significantly higher in the HH group (p < 0.05 vs. NN group) and NH group (p < 0.01 vs. NN group) than in the NN group. Conclusions Exposure to acute and/or short-term hypobaric and hypoxic conditions might exert cardioprotective effects against myocardial ischemia-reperfusion injury via HIF1α modulation.
Collapse
|
13
|
Rozier R, Paul R, Madji Hounoum B, Villa E, Mhaidly R, Chiche J, Verhoeyen E, Marchetti S, Vandenberghe A, Raucoules M, Carles M, Ricci JE. Pharmacological preconditioning protects from ischemia/reperfusion-induced apoptosis by modulating Bcl-xL expression through a ROS-dependent mechanism. FEBS J 2021; 288:3547-3569. [PMID: 33340237 DOI: 10.1111/febs.15675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/02/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a frequent perioperative threat, with numerous strategies developed to limit and/or prevent it. One interesting axis of research is the anesthetic preconditioning (APc) agent's hypothesis (such as sevoflurane, SEV). However, APc's mode of action is still poorly understood and volatile anesthetics used as preconditioning agents are often not well suited in clinical practice. Here, in vitro using H9C2 cells lines (in myeloblast state or differentiated toward cardiomyocytes) and in vivo in mice, we identified that SEV-induced APc is mediated by a mild induction of reactive oxygen species (ROS) that activates Akt and induces the expression of the anti-apoptotic protein B-cell lymphoma-extra large (Bcl-xL), therefore protecting cardiomyocytes from I/R-induced death. Furthermore, we extended these results to human cardiomyocytes (derived from induced pluripotent stem - IPS - cells). Importantly, we demonstrated that this protective signaling pathway induced by SEV could be stimulated using the antidiabetic agent metformin (MET), suggesting the preconditioning properties of MET. Altogether, our study identified a signaling pathway allowing APc of cardiac injuries as well as a rational for the use of MET as a pharmacological preconditioning agent to prevent I/R injuries.
Collapse
Affiliation(s)
- Romain Rozier
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Rachel Paul
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Blandine Madji Hounoum
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Elodie Villa
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Rana Mhaidly
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Johanna Chiche
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Els Verhoeyen
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Sandrine Marchetti
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Ashaina Vandenberghe
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Marc Raucoules
- Anesthésie Réanimation, Centre Hospitalier Universitaire, Nice, France
| | - Michel Carles
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France.,Anesthésie Réanimation, Centre Hospitalier Universitaire, Nice, France.,Réanimation, Faculté des Antilles, Centre Hospitalier Universitaire, Guadeloupe, France
| | - Jean-Ehrland Ricci
- INSERM, C3M, Université Côte d'Azur, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Nice, France
| |
Collapse
|
14
|
Oshima Y, Otsuki A, Endo R, Nakasone M, Harada T, Takahashi S, Inagaki Y. The Effects of Volatile Anesthetics on Lung Ischemia-Reperfusion Injury: Basic to Clinical Studies. J Surg Res 2020; 260:325-344. [PMID: 33373852 DOI: 10.1016/j.jss.2020.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Case reports from as early as the 1970s have shown that intravenous injection of even a small dose of volatile anesthetics result in fatal lung injury. Direct contact between volatile anesthetics and pulmonary vasculature triggers chemical damage in the vessel walls. A wide variety of factors are involved in lung ischemia-reperfusion injury (LIRI), such as pulmonary endothelial cells, alveolar epithelial cells, alveolar macrophages, neutrophils, mast cells, platelets, proinflammatory cytokines, and surfactant. With a constellation of factors involved, the assessment of the protective effect of volatile anesthetics in LIRI is difficult. Multiple animal studies have reported that with regards to LIRI, sevoflurane demonstrates an anti-inflammatory effect in immunocompetent cells and an anti-apoptotic effect on lung tissue. Scattered studies have dismissed a protective effect of desflurane against LIRI. While a single-center randomized controlled trial (RCT) found that volatile anesthetics including desflurane demonstrated a lung-protective effect in thoracic surgery, a multicenter RCT did not demonstrate a lung-protective effect of desflurane. LIRI is common in lung transplantation. One study, although limited due to its small sample size, found that the use of volatile anesthetics in organ procurement surgery involving "death by neurologic criteria" donors did not improve lung graft survival. Future studies on the protective effect of volatile anesthetics against LIRI must examine not only the mechanism of the protective effect but also differences in the effects of different types of volatile anesthetics, their optimal dosage, and the appropriateness of their use in the event of marked alveolar capillary barrier damage.
Collapse
Affiliation(s)
- Yoshiaki Oshima
- Department of Anesthesiology, Yonago Medical Center, Yonago, Tottori, Japan.
| | - Akihiro Otsuki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Ryo Endo
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Masato Nakasone
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomomi Harada
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Shunsaku Takahashi
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshimi Inagaki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
15
|
Berkowitz BA, Podolsky RH, Childers KL, Gow A, Schneider BL, Lloyd SC, Bosse KE, Conti AC, Roberts R, Berri AM, Graffice E, Sinan K, Eliwat W, Shen Y. Age-related murine hippocampal CA1 laminae oxidative stress measured in vivo by QUEnch-assiSTed (QUEST) MRI: impact of isoflurane anesthesia. GeroScience 2020; 42:563-574. [PMID: 31981008 PMCID: PMC7205849 DOI: 10.1007/s11357-020-00162-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related impairments in spatial learning and memory often precede non-familial neurodegenerative disease. Ex vivo studies suggest that physiologic age-related oxidative stress in hippocampus area CA1 may contribute to prodromal spatial disorientation and to morbidity. Yet, conventional blood or cerebrospinal fluid assays appear insufficient for early detection or management of oxidative stress within CA1 sub-regions in vivo. Here, we address this biomarker problem using a non-invasive MRI index of CA1 laminae oxidative stress based on reduction in R1 (= 1/T1) after anti-oxidant administration. An R1 reduction reflects quenching of continuous and excessive production of endogenous paramagnetic free radicals. Careful motion-correction image acquisition, and avoiding repeated exposure to isoflurane, facilitates detection of hippocampus CA1 laminae oxidative stress with QUEnch-assiSTed (QUEST) MRI. Intriguingly, age- and isoflurane-related oxidative stress is localized to the stratum lacunosum of the CA1 region. Our data raise the possibility of using QUEST MRI and FDA-approved anti-oxidants to remediate spatial disorientation and later neurodegeneration with age in animals and humans.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA.
| | - Robert H Podolsky
- Beaumont Research Institute, Beaumont Health, Royal Oak, MI, 48073, USA
| | | | - Alexander Gow
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Brandy L Schneider
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Scott C Lloyd
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Kelly E Bosse
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Alana C Conti
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Ali M Berri
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Emma Graffice
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Kenan Sinan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Waleed Eliwat
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Yimin Shen
- Department of Radiology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
16
|
Wang J, Sun J, Qiao S, Li H, Che T, Wang C, An J. Effects of isoflurane on complex II‑associated mitochondrial respiration and reactive oxygen species production: Roles of nitric oxide and mitochondrial KATP channels. Mol Med Rep 2019; 20:4383-4390. [PMID: 31545457 DOI: 10.3892/mmr.2019.10658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/12/2019] [Indexed: 11/05/2022] Open
Abstract
Volatile anesthetics may protect the heart against ischemia‑reperfusion injury via the direct action on mitochondrial complexes and by regulating the production of reactive oxygen species (ROS). Recently, we reported that isoflurane induced the attenuation of mitochondrial respiration caused by complex I substrates. This process was not associated with endogenous production of mitochondrial nitric oxide (NO). In the present study, we investigated the effects of isoflurane on mitochondrial respiration and ROS production using complex II substrates. The detailed mechanism of these effects was explored with regards to NO production and the expression of mitochondrial ATP‑dependent K+ (mKATP) channels. Mitochondria were isolated from the heart of Sprague‑Dawley rats. The respiratory rates of mitochondria (0.5 mg/ml) were measured via polarography at 28˚C with computer‑controlled Clark‑type O2 electrodes. The complex II substrate succinate (5 mM) was used; 0.25 mM of isoflurane was administered prior to ADP‑initiated state 3 respiration. The mitochondrial membrane potential (ΔΨm) was measured under treatment with the substrate succinate, or succinate in the presence of the complex I inhibitor rotenone. The detection was achieved in a cuvette‑based spectrophotometer operating at wavelengths of 503 nm (excitation) 527 nm (emission) in the presence of 50 nM of the fluorescent dye rhodamine 123. The H2O2 release rates in the mitochondria were measured spectrophotometrically with succinate, or succinate and rotenone using the fluorescent dye Amplex red (12.5‑25 µM). The results indicated that isoflurane increased the state 3 and 4 respiration rates caused by succinate, which were higher than those noted in the control group in the presence of succinate alone. The NOS inhibitor L‑NIO or the NO‑sensitive guanylyl cyclase 1H‑[1,2,4]oxadiazolo[4,3‑a]quinoxalin‑1‑one did not inhibit the increase in the respiration rate (state 3) induced by isoflurane. The ROS scavengers SPBN and manganese (III) tetrakis (4‑benzoic acid) porphyrin chloride inhibited the increase in the respiration rate (state 3 and 4) induced by isoflurane. This effect was not noted for the putative KATP channel blockers 5‑hydroxydecanoic acid and glibenclamide. Isoflurane caused a greater decrease in the concentration of H2O2 during ADP‑initiated state 3 respiration, and L‑N5‑(1‑Iminoethyl)‑ornithine did not inhibit this effect. In conclusion, isoflurane was determined to modulate mitochondrial respiration and ROS production caused by the complex II substrate succinate. These effects were independent of endogenous mitochondrial NO generation and mitochondrial KATP channel opening.
Collapse
Affiliation(s)
- Junan Wang
- Department of Anesthesiology, Pudong New Area People's Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 201299, P.R. China
| | - Jie Sun
- Department of Gastroenterology, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Hua Li
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Tuanjie Che
- Laboratory of Precision Medicine and Translational Medicine, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Chen Wang
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Jianzhong An
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| |
Collapse
|
17
|
Lucchinetti E, Lou PH, Gandhi M, Clanachan AS, Zaugg M. Differential Effects of Anesthetics and Opioid Receptor Activation on Cardioprotection Elicited by Reactive Oxygen Species-Mediated Postconditioning in Sprague-Dawley Rat Hearts. Anesth Analg 2019; 126:1739-1746. [PMID: 29256935 DOI: 10.1213/ane.0000000000002676] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Despite an array of cardioprotective interventions identified in preclinical models of ischemia-reperfusion (IR) injury, successful clinical translation has not been achieved. This study investigated whether drugs routinely used in clinical anesthesia influence cardioprotective effectiveness by reducing effects of reactive oxygen species (ROS), upstream triggers of cardioprotective signaling. Effects of propofol, sevoflurane, or remifentanil were compared on postischemic functional recovery induced by ROS-mediated postconditioning with Intralipid. METHODS Recovery of left ventricular (LV) work, an index of IR injury, was measured in isolated Sprague-Dawley rat hearts subjected to global ischemia (20 minutes) and reperfusion (30 minutes). Hearts were either untreated or were treated with postconditioning with Intralipid (1%, throughout reperfusion). Propofol (10 μM), sevoflurane (2 vol%), remifentanil (3 nM), or combinations thereof were administered peri-ischemically (before and during IR). The effects of anesthetics on ROS production were measured in LV cardiac fibers by Amplex Red assay under phosphorylating and nonphosphorylating conditions. RESULTS Recovery of LV work (expressed as percentage of the preischemic value ± standard deviation) in untreated hearts was poor (20% ± 7%) and was improved by Intralipid postconditioning (58% ± 8%, P = .001). In the absence of Intralipid postconditioning, recovery of LV work was enhanced by propofol (28% ± 9%, P = .049), sevoflurane (49% ± 5%, P < .001), and remifentanil (51% ± 6%, P < .001). The benefit of Intralipid postconditioning was abolished by propofol (33% ± 10%, P < .001), but enhanced by sevoflurane (80% ± 7%, P < .001) or remifentanil (80% ± 9%, P < .001). ROS signaling in LV fibers was abolished by propofol, but unaffected by sevoflurane or remifentanil. We conclude that propofol abolishes ROS-mediated Intralipid postconditioning by acting as a ROS scavenger. Sevoflurane and remifentanil are protective per se and provide additive cardioprotection to ROS-mediated cardioprotection. CONCLUSIONS These divergent effects of routinely used drugs in clinical anesthesia may influence the translatability of cardioprotective therapies such as Intralipid postconditioning.
Collapse
Affiliation(s)
| | | | - Manoj Gandhi
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | | | - Michael Zaugg
- From the Department of Anesthesiology and Pain Medicine.,Department of Pharmacology, University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Berkowitz BA, Romero R, Podolsky RH, Lins-Childers KM, Shen Y, Rosales T, Wadghiri YZ, Hoang DM, Arenas-Hernandez M, Garcia-Flores V, Schwenkel G, Panaitescu B, Gomez-Lopez N. QUEST MRI assessment of fetal brain oxidative stress in utero. Neuroimage 2019; 200:601-606. [PMID: 31158477 DOI: 10.1016/j.neuroimage.2019.05.069] [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] [Received: 03/09/2019] [Revised: 05/15/2019] [Accepted: 05/27/2019] [Indexed: 01/29/2023] Open
Abstract
PURPOSE To achieve sufficient precision of R1 (=1/T1) maps of the fetal brain in utero to perform QUEnch-assiSTed (QUEST) MRI in which a significant anti-oxidant-induced reduction in R1 indicates oxidative stress. METHODS C57BL/6 mouse fetuses in utero were gently and non-surgically isolated and secured using a homemade 3D printed clip. Using a commercial receive-only surface coil, brain maps of R1, an index sensitive to excessive and continuous free radical production, were collected using either a conventional Cartesian or a non-Cartesian (periodically rotated overlapping parallel lines with enhanced reconstruction) progressive saturation sequence. Data were normalized to the shortest TR time to remove bias. To assess oxidative stress, brain R1 maps were acquired on the lipopolysaccharide (LPS) model of preterm birth ± rosiglitazone (ROSI, which has anti-oxidant properties); phosphate buffered saline (PBS) controls ± ROSI were similarly studied. RESULTS Sufficient quality R1 maps were generated by a combination of the 3D printed clip, surface coil detection, non-Cartesian sequence, and normalization scheme ensuring minimal fetal movement, good detection sensitivity, reduced motion artifacts, and minimal baseline variations, respectively. In the LPS group, the combined caudate-putamen and thalamus region R1 was reduced (p < 0.05) with ROSI treatment consistent with brain oxidative stress; no evidence for oxidative stress was found in the pons region. In the PBS control group, brain R1's did not change with ROSI treatment. CONCLUSION The sensitivity and reproducibility of the combined approaches described herein enabled first-time demonstration of regional oxidative stress measurements of the fetal brain in utero using QUEST MRI.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA.
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan, 48109, USA; Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, Michigan, 48824, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, 48201, USA; Detroit Medical Center, Detroit, Michigan, 48201, USA
| | - Robert H Podolsky
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, 48073, USA
| | | | - Yimin Shen
- Department of Radiology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Tilman Rosales
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Youssef Zaim Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, New York, 10016, USA
| | - D Minh Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Bernard and Irene Schwartz Center for Biomedical Imaging, NYU School of Medicine and NYU Langone Health, New York, New York, 10016, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - George Schwenkel
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, 20847, and Detroit, Michigan, 48201, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA.
| |
Collapse
|
19
|
Identification of Candidate Genes and Pathways in Dexmedetomidine-Induced Cardioprotection in the Rat Heart by Bioinformatics Analysis. Int J Mol Sci 2019; 20:ijms20071614. [PMID: 30939728 PMCID: PMC6480577 DOI: 10.3390/ijms20071614] [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: 02/09/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022] Open
Abstract
Dexmedetomidine (DEX), a highly selective alpha2 adrenergic receptor agonist, directly protects hearts against ischemia/reperfusion (I/R) injury. However, the detailed mechanism has not been fully elucidated. We studied differentially expressed mRNAs and miRNAs after DEX administration in rat hearts by comprehensive analysis. Additionally, bioinformatics analysis was applied to explore candidate genes and pathways that might play important roles in DEX-induced cardioprotection. The results of microarray analysis showed that 165 mRNAs and 6 miRNAs were differentially expressed after DEX administration. Through bioinformatics analysis using differentially expressed mRNAs, gene ontology (GO) terms including MAP kinase tyrosine/serine/threonine phosphatase activity and pathways including the p53 pathway were significantly enriched in the down-regulated mRNAs. Dusp1 and Atm were associated with the GO term of MAP kinase tyrosine/serine/threonine phosphatase activity and the p53 pathway, respectively. On the other hand, no significant pathway was found in the target mRNAs of deregulated miRNAs. The results indicated some possible key genes and pathways that seem to be of significance in DEX-induced cardioprotection, although miRNAs seem to be unlikely to contribute to cardioprotection induced by DEX.
Collapse
|
20
|
Liu HJ, Liu B. Inhibition of MicroRNA-23 Contributes to the Isoflurane-Mediated Cardioprotection Against Oxidative Stress. Cardiovasc Toxicol 2019; 18:450-458. [PMID: 29627934 DOI: 10.1007/s12012-018-9455-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Isoflurane is one of the most frequently used volatile anesthetics in clinical practice for inhalational anesthesia. It is widely studied that isoflurane mediates cardioprotection during multiple pathological processes. However, the precise mechanisms have not been fully elucidated. Neonatal cardiomyocytes were isolated and cultured, followed by treatments with isoflurane at 0, 50, 100 or 200 µM. Rat cardiomyoblast cell line, H9c2, was treated with H2O2. Expression of miR-23 was measured by qRT-PCR. The cell survival rate of H9c2 in response to H2O2 treatments was evaluated by MTT assay. The ROS and GSH/GSSG levels were measured using Superoxide Detection Kit and GSH/GSSG Ratio Detection Assay Kit. In this study, we report an isoflurane-miR-23-antioxidant axis in cardiomyocyte. We observed that miR-23 was suppressed by isoflurane treatments at 50, 100 or 200 µM. Moreover, cardiomyocyte with isoflurane exposure was insensitive to H2O2 treatment in vitro. Inhibition of miR-23 protected cardiomyocyte against oxidative stress induced by H2O2 treatments at 30, 60, 90 or 120 µM. In addition, overexpression of miR-23 induced ROS generation over twofolds and rendered cardiomyocyte sensitive to H2O2 treatments. We demonstrate that miR-23 inhibited intracellular GSH, an antioxidant against oxidative stress. Our results reveal that with isoflurane exposure, overexpression of miR-23 rendered cardiomyocyte sensitive to H2O2 treatments at 20, 30, 40, 50 µM. Pretreatments with GSH in miR-23 overexpressing cells rescued the cell death under oxidative stress. In summary, our results illustrate that the isoflurane-mediated protection of cardiomyocytes under oxidative stress is through inhibition of miR-23. This study provides an aspect for the miRNAs-modulated cardiomyocyte sensitivity to oxidative stress, contributing to the development of therapeutic agents.
Collapse
Affiliation(s)
- Hai-Jian Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Anesthesiology, Zhoupu Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Bin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
21
|
Kundumani-Sridharan V, Subramani J, Raghavan S, Maiti GP, Owens C, Walker T, Wasnick J, Idell S, Das KC. Short-duration hyperoxia causes genotoxicity in mouse lungs: protection by volatile anesthetic isoflurane. Am J Physiol Lung Cell Mol Physiol 2019; 316:L903-L917. [PMID: 30810065 DOI: 10.1152/ajplung.00142.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High concentrations of oxygen (hyperoxia) are routinely used during anesthesia, and supplemental oxygen is also administered in connection with several other clinical conditions. Although prolonged hyperoxia is known to cause acute lung injury (ALI), whether short-duration hyperoxia causes lung toxicity remains unknown. We exposed mice to room air (RA or 21% O2) or 60% oxygen alone or in combination with 2% isoflurane for 2 h and determined the expression of oxidative stress marker genes, DNA damage and DNA repair genes, and expression of cell cycle regulatory proteins using quantitative PCR and Western analyses. Furthermore, we determined cellular apoptosis using TUNEL assay and assessed the DNA damage product 8-hydroxy-2'-deoxyguanosine (8-Oxo-dG) in the urine of 60% hyperoxia-exposed mice. Our study demonstrates that short-duration hyperoxia causes mitochondrial and nuclear DNA damage and that isoflurane abrogates this DNA damage and decreases apoptosis when used in conjunction with hyperoxia. In contrast, isoflurane mixed with RA caused significant 8-Oxo-dG accumulations in the mitochondria and nucleus. We further show that whereas NADPH oxidase is a major source of superoxide anion generated by isoflurane in normoxia, isoflurane inhibits superoxide generation in hyperoxia. Additionally, isoflurane also protected the mouse lungs against ALI (95% O2 for 36-h exposure). Our study established that short-duration hyperoxia causes genotoxicity in the lungs, which is abrogated when hyperoxia is used in conjunction with isoflurane, but isoflurane alone causes genotoxicity in the lung when delivered with ambient air.
Collapse
Affiliation(s)
| | - Jaganathan Subramani
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Somasundaram Raghavan
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Guru P Maiti
- Oklahoma Medical Research Foundation , Oklahoma City, Oklahoma
| | - Cade Owens
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Trevor Walker
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - John Wasnick
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| |
Collapse
|
22
|
Steinhauser J, Wespi P, Kwiatkowski G, Kozerke S. Assessing the influence of isoflurane anesthesia on cardiac metabolism using hyperpolarized [1- 13 C]pyruvate. NMR IN BIOMEDICINE 2018; 31. [PMID: 29206326 DOI: 10.1002/nbm.3856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 05/07/2023]
Abstract
Isoflurane is a frequently used anesthetic in small-animal dissolution dynamic nuclear polarization-magnetic resonance imaging (DNP-MRI) studies. Although the literature suggests interactions with mitochondrial metabolism, the influence of the compound on cardiac metabolism has not been assessed systematically to date. In the present study, the impact of low versus high isoflurane concentration was examined in a crossover experiment in healthy rats. The results revealed that cardiac metabolism is modulated by isoflurane concentration, showing increased [1-13 C]lactate and reduced [13 C]bicarbonate production during high isoflurane relative to low isoflurane dose [average differences: +16% [1-13 C]lactate/total myocardial carbon, -22% [13 C]bicarbonate/total myocardial carbon; +51% [1-13 C]lactate/[13 C]bicarbonate]. These findings emphasize that reproducible anesthesia is important when studying cardiac metabolism. As the depth of anesthesia is difficult to control in an experimental animal setting, careful study design is required to exclude confounding factors.
Collapse
Affiliation(s)
- Jonas Steinhauser
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Patrick Wespi
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Grzegorz Kwiatkowski
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Sebastian Kozerke
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Bastian C, Politano S, Day J, McCray A, Brunet S, Baltan S. Mitochondrial dynamics and preconditioning in white matter. CONDITIONING MEDICINE 2018; 1:64-72. [PMID: 30135960 PMCID: PMC6101249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Mechanisms of ischemic preconditioning have been extensively studied in gray matter. However, an ischemic episode affects both the gray matter (GM) and white matter (WM) portions of the brain. Inhibition of mitochondrial fission is one of the mechanisms of preconditioning neuronal cell bodies against ischemia. Although axons are anatomical extensions of neuronal cell bodies, injury mechanisms differ between GM and WM. Indeed, axonal dysfunction is responsible for much of the disability associated with clinical deficits observed after stroke; however, the signaling process underlying preconditioning remains unexplored in axons. Using mouse optic nerve, which is a pure isolated WM tract, we show that mitochondria in myelinated axons undergo rapid and profuse fission during oxygen glucose deprivation (OGD) that is mediated by translocation of cytoplasmic Dynamin Related Protein-1 (Drp-1) to mitochondria. OGD-induced mitochondrial fission correlates with reduced mitochondrial motility and loss of axon function. Mitochondrial fragmentation and loss of motility become permanent during the recovery period. Inhibiting mitochondrial fission by administering mitochondrial division inhibitor-1 (Mdivi-1) during OGD preserves mitochondrial shape and motility and promotes axon function recovery. In contrast, preconditioning WM by applying Mdivi-1 only before OGD fails to conserve mitochondrial shape or motility and fails to benefit axon function. Our findings suggest that inhibition of mitochondrial fission during ischemia promotes axon function recovery, but is not sufficient to precondition WM against ischemia. These results raise caution in that approaches to preconditioning neuronal cell bodies may not successfully translate into functional improvement following ischemia.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Stephen Politano
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Jerica Day
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Andrew McCray
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Sylvain Brunet
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Selva Baltan
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| |
Collapse
|
24
|
Nitrite administration improves sepsis-induced myocardial and mitochondrial dysfunction by modulating stress signal responses. J Anesth 2017; 31:885-894. [PMID: 29063286 DOI: 10.1007/s00540-017-2417-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE A specific therapeutic strategy in sepsis-induced myocardial dysfunction remains to be determined. Nitrite may have cardioprotective effects against sepsis-induced myocardial dysfunction. This study investigated the cardioprotective effects of nitrite on myocardial function, mitochondrial bioenergetics, and its underlying molecular mechanisms in severe septic rats. METHODS Sepsis was induced in male Wistar rats by cecal ligation and puncture (CLP). After CLP, we administered normal saline (NS group) or nitrite (nitrite group) subcutaneously. We administered nitrite at different doses (0.1-10 mg/kg) to ascertain the most effective dose and examined cardiac function in an isolated heart experiment 8 h after CLP. We investigated mitochondrial bioenergetics and molecular mechanisms underlying the administration of nitrite in vitro. RESULTS In isolated heart experiments, the left ventricular developed pressure (96 ± 5 mmHg) at a moderate nitrite dose (1.0 mg/kg) was significantly higher than that in the NS group (75 ± 4 mmHg, P < 0.05). Mitochondrial oxidative phosphorylation in the nitrite group was significantly higher than that in the NS group (P < 0.01). Immunoblotting revealed that nitrite significantly increased the phosphorylation of Akt (P < 0.05) and reduced the nuclear translocation of NF-κB (P < 0.05) compared with the NS group. Nitrite was also shown to improve the rate of survival in severe septic rats (P < 0.001). CONCLUSIONS Our results showed that a moderate nitrite dose improved septic myocardial dysfunction at organ, cellular, and molecular levels via modulation of stress signal responses, which resulted in an improvement in survival.
Collapse
|
25
|
Brozović G, Oršolić N, Rozgaj R, Knežević F, Knežević AH, Maričić M, Krsnik D, Benković V. Sevoflurane and isoflurane genotoxicity in kidney cells of mice. Arh Hig Rada Toksikol 2017; 68:228-235. [DOI: 10.1515/aiht-2017-68-2941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 08/01/2017] [Indexed: 11/15/2022] Open
Abstract
Abstract
The aim of this study was to evaluate the DNA damage and repair in kidney cells of Swiss albino mice after repeated exposure to sevoflurane and isoflurane and compare their detrimental effects. We used the alkaline comet assay to establish the genetic damage and measured three parameters: tail length, tail moment, and tail intensity of comets. These parameters were measured immediately after exposure to the above mentioned inhalation anaesthetics, two hours, six hours, and 24 hours later and were compared with the control group. Mean values of all three parameters were significantly higher in experimental groups compared to the control group. DNA damage in kidney cells of mice exposed to sevoflurane increased continuously before it reached its peak 24 hours after exposure. Isoflurane induced the highest DNA damage two hours after exposure. Levels of DNA damage recorded 24 h after cessation of exposure to both tested compounds suggest that sevoflurane was slightly more genotoxic than isoflurane to kidney cells of mice. According to these results, the currently used volatile anaesthetics sevoflurane and isoflurane are able to damage DNA in kidney cells of mice. Such findings suggest a possibility for similar outcomes in humans and that fact must be taken into account in everyday clinical practice.
Collapse
Affiliation(s)
- Gordana Brozović
- Faculty of Medicine, University of Osijek, Osijek , Croația
- Department of Anaesthesiology, Reanimatology and ICU, University Hospital for Tumours, Sestre milosrdnice University Hospital Centre, Zagreb , Croatia
| | - Nada Oršolić
- Division of Animal Physiology, Department of Biology, Faculty of Science, University of Zagreb , Croatia
| | - Ružica Rozgaj
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb , Croatia
| | - Fabijan Knežević
- Department of Pathology, Sveti Duh University Hospital, Zagreb , Croatia
| | - Anica Horvat Knežević
- Division of Animal Physiology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb , Croatia
| | - Martina Maričić
- Department of Anaesthesiology, Reanimatology and ICU, University Hospital for Tumours, Sestre milosrdnice University Hospital Centre, Zagreb , Croatia
| | - Dajana Krsnik
- Division of Animal Physiology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb , Croatia
| | - Vesna Benković
- Division of Animal Physiology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb , Croatia
| |
Collapse
|
26
|
Ma L, Zhu J, Gao Q, Rebecchi MJ, Wang Q, Liu L. Restoring Pharmacologic Preconditioning in the Aging Heart: Role of Mitophagy/Autophagy. J Gerontol A Biol Sci Med Sci 2017; 72:489-498. [PMID: 27565512 DOI: 10.1093/gerona/glw168] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/27/2016] [Indexed: 12/16/2022] Open
Abstract
We previously reported that pretreatment with the potent antioxidant TEMPOL improves mitochondrial function and restores preconditioning in the aging heart. Because mitophagy is implicated in cardiac preconditioning and declines with age, this study was designed to investigate how age influences mitophagy in response to preconditioning and whether TEMPOL pretreatment improves it. Old (22-24 months) rats were pretreated with or without 4-week TEMPOL and compared with young (4-6 months) untreated rats. Cardioprotection induced by isoflurane (ISO) in vivo and in isolated cardiomyocytes in vitro was assessed following ischemia/reperfusion and simulated hypoxia/reoxygenation, respectively. Mitophagy was determined by comparing the levels/subcellular locations of key mitophagic markers using Western blotting and immunofluorescence techniques. ISO preconditioned the young but not old heart in vivo and in vitro. Aging impaired ISO-induced mitochondrial accumulation of PINK1 and Parkin, as well as mitochondrial ubiquitination, and baseline and ISO-induced autophagic flux assessed by LC3 puncta, membrane associated LC3-II and p62. Pretreatment with TEMPOL improved these processes and restored ISO preconditioning. Inhibition of autophagy abolished ISO-induced protection in cardiomyocytes from young and TEMPOL pretreated old rats. Thus, antioxidant pretreatment significantly improves mitophagic response to ISO in old myocardium, which may contribute to restoration of cardioprotection in senescent animals.
Collapse
Affiliation(s)
- Li Ma
- Department of Anesthesiology, School of Medicine, Stony Brook University, New York
| | - Jiang Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Qun Gao
- Department of Anesthesiology, School of Medicine, Stony Brook University, New York
| | - Mario J Rebecchi
- Department of Anesthesiology, School of Medicine, Stony Brook University, New York
| | - Qiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Lixin Liu
- Department of Anesthesiology, School of Medicine, Stony Brook University, New York
| |
Collapse
|
27
|
Harisseh R, Chiari P, Villedieu C, Sueur P, Abrial M, Fellahi JL, Ovize M, Gharib A. Cyclophilin D Modulates the Cardiac Mitochondrial Target of Isoflurane, Sevoflurane, and Desflurane. J Cardiovasc Pharmacol 2017; 69:326-334. [PMID: 28328748 DOI: 10.1097/fjc.0000000000000479] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Volatile anesthetics are known to limit myocardial ischemia-reperfusion injuries. Mitochondria were shown to be major contributors to cardioprotection. Cyclophilin D (CypD) is one of the main regulators of mitochondria-induced cell death. We compared the effect of isoflurane, sevoflurane, and desflurane in the presence or absence of CypD, to clarify its role in the mechanism of cardioprotection induced by these anesthetics. METHODS Oxidative phosphorylation, mitochondrial membrane potential, and H2O2 production were measured in isolated mitochondria from wild-type (WT) or CypD knockout mice in basal conditions and after hypoxia-reoxygenation in the presence or absence of volatile anesthetics. RESULTS All volatile anesthetics inhibited mitochondrial state 3 of complex I, decreased membrane potential, and increased adenosine diphosphate consumption duration in both WT and CypD knockout mice. However, they differently modified H2O2 production after stimulation by succinate: CypD ablation reduced H2O2 production, isoflurane decreased H2O2 level in WT but not in CypD knockout mice, sevoflurane affected both lines whereas desflurane increased H2O2 production in CypD knockout and had no effect on WT mice. CONCLUSIONS This study showed different effects of isoflurane, sevoflurane, and desflurane on mitochondrial functions and highlighted the implication of CypD in the regulation of adenosine diphosphate consumption and complex I-induced radical oxygen species production.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Anesthetics, Inhalation/pharmacology
- Animals
- Peptidyl-Prolyl Isomerase F
- Cyclophilins/deficiency
- Cyclophilins/genetics
- Cyclophilins/metabolism
- Cytoprotection
- Desflurane
- Energy Metabolism/drug effects
- Genotype
- Hydrogen Peroxide/metabolism
- Isoflurane/analogs & derivatives
- Isoflurane/pharmacology
- Male
- Membrane Potential, Mitochondrial/drug effects
- Methyl Ethers/pharmacology
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidative Phosphorylation/drug effects
- Phenotype
- Protective Agents/pharmacology
- Sevoflurane
- Time Factors
Collapse
Affiliation(s)
- Rania Harisseh
- *INSERM UMR 1060, CarMeN Laboratory, Univ Lyon1, IHU OPERA, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France; †Service d'Anesthésie Réanimation, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France; and ‡Service d'Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Comparative analysis of resuscitation using human serum albumin and crystalloids or 130/0.4 hydroxyethyl starch and crystalloids on skeletal muscle metabolic profile during experimental haemorrhagic shock in swine. Eur J Anaesthesiol 2017; 34:89-97. [DOI: 10.1097/eja.0000000000000537] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
The genetics of isoflurane-induced developmental neurotoxicity. Neurotoxicol Teratol 2016; 60:40-49. [PMID: 27989695 DOI: 10.1016/j.ntt.2016.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/07/2016] [Accepted: 10/27/2016] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Neurotoxicity induced by early developmental exposure to volatile anesthetics is a characteristic of organisms across a wide range of species, extending from the nematode C. elegans to mammals. Prevention of anesthetic-induced neurotoxicity (AIN) will rely upon an understanding of its underlying mechanisms. However, no forward genetic screens have been undertaken to identify the critical pathways affected in AIN. By characterizing such pathways, we may identify mechanisms to eliminate isoflurane induced AIN in mammals. METHODS Chemotaxis in adult C. elegans after larval exposure to isoflurane was used to measure AIN. We initially compared changes in chemotaxis indices between classical mutants known to affect nervous system development adding mutants in response to data. Activation of specific genes was visualized using fluorescent markers. Animals were then treated with rapamycin or preconditioned with isoflurane to test effects on AIN. RESULTS Forty-four mutations, as well as pharmacologic manipulations, identified two pathways, highly conserved from invertebrates to humans, that regulate AIN in C. elegans. Activation of one stress-protective pathway (DAF-2 dependent) eliminates AIN, while activation of a second stress-responsive pathway (endoplasmic reticulum (ER) associated stress) causes AIN. Pharmacologic inhibition of the mechanistic Target of Rapamycin (mTOR) blocks ER-stress and AIN. Preconditioning with isoflurane prior to larval exposure also inhibited AIN. DISCUSSION Our data are best explained by a model in which isoflurane acutely inhibits mitochondrial function causing activation of responses that ultimately lead to ER-stress. The neurotoxic effect of isoflurane can be completely prevented by manipulations at multiple points in the pathways that control this response. Endogenous signaling pathways can be recruited to protect organisms from the neurotoxic effects of isoflurane.
Collapse
|
30
|
Zhang H, Sun XR, Wang J, Zhang ZZ, Zhao HT, Li HH, Ji MH, Li KY, Yang JJ. Reactive Oxygen Species-mediated Loss of Phenotype of Parvalbumin Interneurons Contributes to Long-term Cognitive Impairments After Repeated Neonatal Ketamine Exposures. Neurotox Res 2016; 30:593-605. [DOI: 10.1007/s12640-016-9653-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 01/29/2023]
|
31
|
Wu J, Zhang M, Li H, Sun X, Hao S, Ji M, Yang J, Li K. BDNF pathway is involved in the protective effects of SS-31 on isoflurane-induced cognitive deficits in aging mice. Behav Brain Res 2016; 305:115-21. [DOI: 10.1016/j.bbr.2016.02.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/24/2016] [Accepted: 02/28/2016] [Indexed: 01/21/2023]
|
32
|
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]
|
33
|
Teixeira G, Chiari P, Fauconnier J, Abrial M, Couture-Lepetit E, Harisseh R, Pillot B, Lacampagne A, Tourneur Y, Gharib A, Ovize M. Involvement of Cyclophilin D and Calcium in Isoflurane-induced Preconditioning. Anesthesiology 2015; 123:1374-84. [PMID: 26460965 DOI: 10.1097/aln.0000000000000876] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The mitochondrial permeability transition pore (PTP) has been established as an important mediator of ischemia-reperfusion-induced cell death. The matrix protein cyclophilin D (CypD) is the best known regulator of PTP opening. Therefore, the authors hypothesized that isoflurane, by inhibiting the respiratory chain complex I, another regulator of PTP, might reinforce the myocardial protection afforded by CypD inhibition. METHODS Adult mouse or isolated cardiomyocytes from wild-type or CypD knockout (CypD-KO) mice were subjected to ischemia or hypoxia followed by reperfusion or reoxygenation. Infarct size was assessed in vivo. Mitochondrial membrane potential and PTP opening were assessed using tetramethylrhodamine methyl ester perchlorate and calcein-cobalt fluorescence, respectively. Fluo-4 AM and rhod-2 AM staining allowed the measurement, by confocal microscopy, of Ca transient and Ca transfer from sarcoplasmic reticulum (SR) to mitochondria after caffeine stimulation. RESULTS Both inhibition of CypD and isoflurane significantly reduced infarct size (-50 and -37%, respectively) and delayed PTP opening (+63% each). Their combination had no additive effect (n = 6/group). CypD-KO mice displayed endogenous protection against ischemia-reperfusion. Isoflurane depolarized the mitochondrial membrane (-28%, n = 5), decreased oxidative phosphorylation (-59%, n = 5), and blunted the caffeine-induced Ca transfer from SR to mitochondria (-22%, n = 7) in the cardiomyocytes of wild-type mice. Importantly, this transfer was spontaneously decreased in the cardiomyocytes of CypD-KO mice (-25%, n = 4 to 5). CONCLUSIONS The results suggest that the partial inhibitory effect of isoflurane on respiratory complex I is insufficient to afford a synergy to CypD-induced protection. Isoflurane attenuates the Ca transfer from SR to mitochondria, which is also the prominent role of CypD, and finally prevents PTP opening.
Collapse
Affiliation(s)
- Geoffrey Teixeira
- From INSERM UMR-1060, CarMeN Laboratory, Université Lyon-1, Faculté de Médecine Rockefeller, Lyon, France (G.T., P.C., M.A., E.C.-L., R.H., B.P., Y.T., A.G., M.O.); Service d'Anesthésie Réanimation, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France (P.C.); INSERM UMR-1046, Université Montpellier 1, Université Montpellier 2, Centre Hospitalier Universitaire de Montpellier, Montpellier, France (J.F., A.L.); and Service d'Explorations Fonctionnelles Cardiovasculaires and CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France (M.O.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Braz MG, Braz LG, Freire CMM, Lucio LMC, Braz JRC, Tang G, Salvadori DMF, Yeum KJ. Isoflurane and Propofol Contribute to Increasing the Antioxidant Status of Patients During Minor Elective Surgery: A Randomized Clinical Study. Medicine (Baltimore) 2015; 94:e1266. [PMID: 26252290 PMCID: PMC4616612 DOI: 10.1097/md.0000000000001266] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/04/2015] [Accepted: 07/09/2015] [Indexed: 02/07/2023] Open
Abstract
Isoflurane is a volatile halogenated anesthetic used especially for anesthesia maintenance whereas propofol is a venous anesthetic utilized for anesthesia induction and maintenance, and reportedly an antioxidant. However, there are still controversies related to isoflurane-induced oxidative stress and it remains unanswered whether the antioxidant effects occur in patients under propofol anesthesia.Taking into account the importance of better understanding the role of anesthetics on oxidative stress in anesthetized patients, the present study was designed to evaluate general anesthesia maintained with isoflurane or propofol on antioxidant status in patients who underwent minimally invasive surgeries.We conducted a prospective randomized trial in 30 adult patients without comorbidities who underwent elective minor surgery (septoplasty) lasting at least 2 h admitted to a Brazilian tertiary hospital.The patients were randomly allocated into 2 groups, according to anesthesia maintenance (isoflurane, n = 15 or propofol, n = 15). Peripheral blood samples were drawn before anesthesia (baseline) and 2-h after anesthesia induction.The primary outcomes were to investigate the effect of either isoflurane or propofol anesthesia on aqueous plasma oxidizability and total antioxidant performance (TAP) by fluorometry as well as several individual antioxidants by high-performance liquid chromatography. As secondary outcome, oxidized genetic damage (7,8-dihydro-8-oxoguanine, known as 8-oxo-Gua) was investigated by the comet assay.Both anesthesia techniques (isoflurane or propofol) for a 2-h period resulted in a significant decrease of plasma α-tocopherol, but not other antioxidants including uric acid, carotenoids, and retinol (P > 0.05). Propofol, in contrast to isoflurane anesthesia, significantly increased (P < 0.001) anti-inflammatory/antioxidant plasma γ-tocopherol concentration in patients. Both anesthesia types significantly enhanced hydrophilic antioxidant capacity and TAP, with no significant difference between them, and 8-oxo-Gua remained unchanged during anesthesia in both groups. In addition, both anesthetics showed antioxidant capacity in vitro.This study shows that anesthesia maintained with either propofol or isoflurane increase both hydrophilic and total antioxidant capacity in plasma, but only propofol anesthesia increases plasma γ-tocopherol concentration. Additionally, both types of anesthetics do not lead to oxidative DNA damage in patients without comorbidities undergoing minimally invasive surgery.
Collapse
Affiliation(s)
- Mariana G Braz
- From the Faculdade de Medicina de Botucatu, UNESP-Univ Estadual Paulista, Botucatu, SP, Brazil (MGB, LGB, CMMF, LMCL, JRCB, DMFS); Jean Mayer USDA-Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA (MGB, GT, K-JY); and College of Biomedical and Health Sciences, Konkuk University, Chungcheongbuk-do, South Korea (K-JY)
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
BACKGROUND Anesthetic cardioprotection reduces myocardial infarct size after ischemia-reperfusion injury. Currently, the role of microRNA in this process remains unknown. MicroRNAs are short, noncoding nucleotide sequences that negatively regulate gene expression through degradation or suppression of messenger RNA. In this study, the authors uncovered the functional role of microRNA-21 (miR-21) up-regulation after anesthetic exposure. METHODS MicroRNA and messenger RNA expression changes were analyzed by quantitative real-time polymerase chain reaction in cardiomyocytes after exposure to isoflurane. Lactate dehydrogenase release assay and propidium iodide staining were conducted after inhibition of miR-21. miR-21 target expression was analyzed by Western blot. The functional role of miR-21 was confirmed in vivo in both wild-type and miR-21 knockout mice. RESULTS Isoflurane induces an acute up-regulation of miR-21 in both in vivo and in vitro rat models (n = 6, 247.8 ± 27.5% and 258.5 ± 9.0%), which mediates protection to cardiomyocytes through down-regulation of programmed cell death protein 4 messenger RNA (n = 3, 82.0 ± 4.9% of control group). This protective effect was confirmed by knockdown of miR-21 and programmed cell death protein 4 in vitro. In addition, the protective effect of isoflurane was abolished in miR-21 knockout mice in vivo, with no significant decrease in infarct size compared with nonexposed controls (n = 8, 62.3 ± 4.6% and 56.2 ± 3.2%). CONCLUSIONS The authors demonstrate for the first time that isoflurane mediates protection of cardiomyocytes against oxidative stress via an miR-21/programmed cell death protein 4 pathway. These results reveal a novel mechanism by which the damage done by ischemia/reperfusion injury may be decreased.
Collapse
|
36
|
Lotz C, Kehl F. Volatile Anesthetic-Induced Cardiac Protection: Molecular Mechanisms, Clinical Aspects, and Interactions With Nonvolatile Agents. J Cardiothorac Vasc Anesth 2015; 29:749-60. [DOI: 10.1053/j.jvca.2014.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Indexed: 02/07/2023]
|
37
|
Gonzalez S, Fernando R, Berthelot J, Perrin-Tricaud C, Sarzi E, Chrast R, Lenaers G, Tricaud N. In vivo time-lapse imaging of mitochondria in healthy and diseased peripheral myelin sheath. Mitochondrion 2015; 23:32-41. [PMID: 26031781 DOI: 10.1016/j.mito.2015.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 10/23/2022]
Abstract
The myelin sheath that covers a large amount of neurons is critical for their homeostasis, and myelinating glia mitochondria have recently been shown to be essential for neuron survival. However morphological and physiological properties of these organelles remain elusive. Here we report a method to analyze mitochondrial dynamics and morphology in myelinating Schwann cells of living mice using viral transduction and time-lapse multiphoton microscopy. We describe the distribution, shape, size and dynamics of mitochondria in live cells. We also report mitochondrial alterations in Opa1(delTTAG) mutant mice cells at presymptomatic stages, suggesting that mitochondrial defects in myelin contribute to OPA1 related neuropathy and represent a biomarker for the disease.
Collapse
Affiliation(s)
- Sergio Gonzalez
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France
| | - Ruani Fernando
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France
| | - Jade Berthelot
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France
| | - Claire Perrin-Tricaud
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France
| | - Emmanuelle Sarzi
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France
| | - Roman Chrast
- Karolinska Institutet, Department of Clinical Neuroscience, Department of Neuroscience, Stockhom 171 77, Sweden
| | - Guy Lenaers
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France; Mitochondrial Medicine Research Centre, Pôle de Recherche et d'Enseignement en Médecine Mitochondriale, Université d'Angers, Angers 49933, France
| | - Nicolas Tricaud
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier 34091, France.
| |
Collapse
|
38
|
Sevoflurane Induces DNA Damage Whereas Isoflurane Leads to Higher Antioxidative Status in Anesthetized Rats. BIOMED RESEARCH INTERNATIONAL 2015; 2015:264971. [PMID: 26101770 PMCID: PMC4458518 DOI: 10.1155/2015/264971] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/06/2015] [Accepted: 05/10/2015] [Indexed: 11/17/2022]
Abstract
Taking into account that there are controversial antioxidative effects of inhalational anesthetics isoflurane and sevoflurane and absence of comparison of genotoxicity of both anesthetics in animal model, the aim of this study was to compare DNA damage and antioxidant status in Wistar rats exposed to a single time to isoflurane or sevoflurane. The alkaline single-cell gel electrophoresis assay (comet assay) was performed in order to evaluate DNA damage in whole blood cells of control animals (unexposed; n = 6) and those exposed to 2% isoflurane (n = 6) or 4% sevoflurane (n = 6) for 120 min. Plasma antioxidant status was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. There was no statistically significant difference between isoflurane and sevoflurane groups regarding hemodynamic and temperature variables (P > 0.05). Sevoflurane significantly increased DNA damage compared to unexposed animals (P = 0.02). In addition, Wistar rats anesthetized with isoflurane showed higher antioxidative status (MTT) than control group (P = 0.019). There were no significant differences in DNA damage or antioxidant status between isoflurane and sevoflurane groups (P > 0.05). In conclusion, our findings suggest that, in contrast to sevoflurane exposure, isoflurane increases systemic antioxidative status, protecting cells from DNA damage in rats.
Collapse
|
39
|
Kunst G, Klein AA. Peri-operative anaesthetic myocardial preconditioning and protection - cellular mechanisms and clinical relevance in cardiac anaesthesia. Anaesthesia 2015; 70:467-82. [PMID: 25764404 PMCID: PMC4402000 DOI: 10.1111/anae.12975] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2014] [Indexed: 12/11/2022]
Abstract
Preconditioning has been shown to reduce myocardial damage caused by ischaemia–reperfusion injury peri-operatively. Volatile anaesthetic agents have the potential to provide myocardial protection by anaesthetic preconditioning and, in addition, they also mediate renal and cerebral protection. A number of proof-of-concept trials have confirmed that the experimental evidence can be translated into clinical practice with regard to postoperative markers of myocardial injury; however, this effect has not been ubiquitous. The clinical trials published to date have also been too small to investigate clinical outcome and mortality. Data from recent meta-analyses in cardiac anaesthesia are also not conclusive regarding intra-operative volatile anaesthesia. These inconclusive clinical results have led to great variability currently in the type of anaesthetic agent used during cardiac surgery. This review summarises experimentally proposed mechanisms of anaesthetic preconditioning, and assesses randomised controlled clinical trials in cardiac anaesthesia that have been aimed at translating experimental results into the clinical setting.
Collapse
Affiliation(s)
- G Kunst
- Department of Anaesthetics, King's College Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|
40
|
Sosunov SA, Ameer X, Niatsetskaya ZV, Utkina-Sosunova I, Ratner VI, Ten VS. Isoflurane anesthesia initiated at the onset of reperfusion attenuates oxidative and hypoxic-ischemic brain injury. PLoS One 2015; 10:e0120456. [PMID: 25799166 PMCID: PMC4370491 DOI: 10.1371/journal.pone.0120456] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/22/2015] [Indexed: 01/16/2023] Open
Abstract
This study demonstrates that in mice subjected to hypoxia-ischemia (HI) brain injury isoflurane anesthesia initiated upon reperfusion limits a release of mitochondrial oxidative radicals by inhibiting a recovery of complex-I dependent mitochondrial respiration. This significantly attenuates an oxidative stress and reduces the extent of HI brain injury. Neonatal mice were subjected to HI, and at the initiation of reperfusion were exposed to isoflurane with or without mechanical ventilation. At the end of HI and isoflurane exposure cerebral mitochondrial respiration, H2O2 emission rates were measured followed by an assessment of cerebral oxidative damage and infarct volumes. At 8 weeks after HI navigational memory and brain atrophy were assessed. In vitro, direct effect of isoflurane on mitochondrial H2O2 emission was compared to that of complex-I inhibitor, rotenone. Compared to controls, 15 minutes of isoflurane anesthesia inhibited recovery of the compex I-dependent mitochondrial respiration and decreased H2O2 production in mitochondria supported with succinate. This was associated with reduced oxidative brain injury, superior navigational memory and decreased cerebral atrophy compared to the vehicle-treated HI-mice. Extended isoflurane anesthesia was associated with sluggish recovery of cerebral blood flow (CBF) and the neuroprotection was lost. However, when isoflurane anesthesia was supported with mechanical ventilation the CBF recovery improved, the event associated with further reduction of infarct volume compared to HI-mice exposed to isoflurane without respiratory support. Thus, in neonatal mice brief isoflurane anesthesia initiated at the onset of reperfusion limits mitochondrial release of oxidative radicals and attenuates an oxidative stress. This novel mechanism contributes to neuroprotective action of isoflurane. The use of mechanical ventilation during isoflurane anesthesia counterbalances negative effect of isoflurane anesthesia on recovery of cerebral circulation which potentiates protection against reperfusion injury.
Collapse
Affiliation(s)
- Sergey A. Sosunov
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
| | - Xavier Ameer
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
| | - Zoya V. Niatsetskaya
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
| | - Irina Utkina-Sosunova
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
| | - Veniamin I. Ratner
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
| | - Vadim S. Ten
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
41
|
Kikuchi C, Dosenovic S, Bienengraeber M. Anaesthetics as cardioprotectants: translatability and mechanism. Br J Pharmacol 2015; 172:2051-61. [PMID: 25322898 DOI: 10.1111/bph.12981] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/06/2014] [Accepted: 10/11/2014] [Indexed: 12/22/2022] Open
Abstract
The pharmacological conditioning of the heart with anaesthetics, such as volatile anaesthetics or opioids, is a phenomenon whereby a transient exposure to an anaesthetic agent protects the heart from the harmful consequences of myocardial ischaemia and reperfusion injury. The cellular and molecular mechanisms of anaesthetic conditioning appear largely to mimic those of ischaemic pre- and post-conditioning. Progress has been made on the understanding of the underlying mechanisms although the order of events and the specific targets of anaesthetics that trigger protection are not always clear. In the laboratory, the protection afforded by certain anaesthetics against cardiac ischaemia and reperfusion injury is powerful and reproducible but this has not necessarily translated into similarly robust clinical benefits. Indeed, clinical studies and meta-analyses delivered variable results when comparing in the laboratory setting protective and non-protective anaesthetics. Reasons for this include underlying conditions such as age, obesity and diabetes. Animal models for disease or ageing, human cardiomyocytes derived from stem cells of patients and further clinical studies are employed to better understand the underlying causes that prevent a more robust protection in patients.
Collapse
Affiliation(s)
- C Kikuchi
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan
| | | | | |
Collapse
|
42
|
Kikuchi C, Dosenovic S, Bienengraeber M. Anaesthetics as cardioprotectants: translatability and mechanism. Br J Pharmacol 2015. [PMID: 25322898 DOI: 10.1111/bph.2015.172.issue-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The pharmacological conditioning of the heart with anaesthetics, such as volatile anaesthetics or opioids, is a phenomenon whereby a transient exposure to an anaesthetic agent protects the heart from the harmful consequences of myocardial ischaemia and reperfusion injury. The cellular and molecular mechanisms of anaesthetic conditioning appear largely to mimic those of ischaemic pre- and post-conditioning. Progress has been made on the understanding of the underlying mechanisms although the order of events and the specific targets of anaesthetics that trigger protection are not always clear. In the laboratory, the protection afforded by certain anaesthetics against cardiac ischaemia and reperfusion injury is powerful and reproducible but this has not necessarily translated into similarly robust clinical benefits. Indeed, clinical studies and meta-analyses delivered variable results when comparing in the laboratory setting protective and non-protective anaesthetics. Reasons for this include underlying conditions such as age, obesity and diabetes. Animal models for disease or ageing, human cardiomyocytes derived from stem cells of patients and further clinical studies are employed to better understand the underlying causes that prevent a more robust protection in patients.
Collapse
Affiliation(s)
- C Kikuchi
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan
| | | | | |
Collapse
|
43
|
Cardioprotective efficacy depends critically on pharmacological dose, duration of ischaemia, health status of animals and choice of anaesthetic regimen: a case study with folic acid. J Transl Med 2014; 12:325. [PMID: 25432364 PMCID: PMC4265322 DOI: 10.1186/s12967-014-0325-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/11/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Acute, high-dose folic acid (FA) administration has recently been shown to possess unprecedented effective cardioprotection against ischaemia/reperfusion (I/R) injury. Here we explore the translation potential of FA as treatment modality for cardiac I/R. METHODS Dependency of FA protection on dose, ischaemia duration, and eNOS was examined in an isolated mouse heart I/R model, whereas dependency on animal health status and anaesthesia was examined in an in vivo rat model of regional cardiac I/R. RESULTS 50 μM FA provided maximal reduction (by 95%) of I/R-induced cell death following 25 min ischaemia in isolated wild-type hearts, with protection associated with increased coupled eNOS protein. No protection was observed with 35 min I or in eNOS(-/-) hearts. Acute intravenous administration of FA during a 25 min ischaemic period reduced infarct size by 45% in in vivo pentobarbital-anaesthetised young, healthy rats. FA did not reduce infarct size in aged or pre-diabetic rats, although it did preserve hemodynamics in the pre-diabetic rats. Finally, using a clinically-relevant anaesthetic regimen of fentanyl-propofol anaesthesia, FA treatment was ineffective in young, aged and pre-diabetic animals. CONCLUSIONS The protective potential of an initially promising cardioprotective treatment of high dose FA against cardiac I/R infarction, is critically dependent on experimental conditions with relevance to the clinical condition. Our data indicates the necessity of expanded pre-clinical testing of cardioprotective interventions before embarking on clinical testing, in order to prevent too many "lost-in-translation" drugs and unnecessary clinical studies.
Collapse
|
44
|
Alleman RJ, Katunga LA, Nelson MAM, Brown DA, Anderson EJ. The "Goldilocks Zone" from a redox perspective-Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front Physiol 2014; 5:358. [PMID: 25278906 PMCID: PMC4166897 DOI: 10.3389/fphys.2014.00358] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 01/17/2023] Open
Abstract
Consequences of oxidative stress may be beneficial or detrimental in physiological systems. An organ system's position on the “hormetic curve” is governed by the source and temporality of reactive oxygen species (ROS) production, proximity of ROS to moieties most susceptible to damage, and the capacity of the endogenous cellular ROS scavenging mechanisms. Most importantly, the resilience of the tissue (the capacity to recover from damage) is a decisive factor, and this is reflected in the disparate response to ROS in cardiac and skeletal muscle. In myocytes, a high oxidative capacity invariably results in a significant ROS burden which in homeostasis, is rapidly neutralized by the robust antioxidant network. The up-regulation of key pathways in the antioxidant network is a central component of the hormetic response to ROS. Despite such adaptations, persistent oxidative stress over an extended time-frame (e.g., months to years) inevitably leads to cumulative damages, maladaptation and ultimately the pathogenesis of chronic diseases. Indeed, persistent oxidative stress in heart and skeletal muscle has been repeatedly demonstrated to have causal roles in the etiology of heart disease and insulin resistance, respectively. Deciphering the mechanisms that underlie the divergence between adaptive and maladaptive responses to oxidative stress remains an active area of research for basic scientists and clinicians alike, as this would undoubtedly lead to novel therapeutic approaches. Here, we provide an overview of major types of ROS in striated muscle and the divergent adaptations that occur in response to them. Emphasis is placed on highlighting newly uncovered areas of research on this topic, with particular focus on the mitochondria, and the diverging roles that ROS play in muscle health (e.g., exercise or preconditioning) and disease (e.g., cardiomyopathy, ischemia, metabolic syndrome).
Collapse
Affiliation(s)
- Rick J Alleman
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Lalage A Katunga
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Margaret A M Nelson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Ethan J Anderson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
45
|
Agarwal B, Stowe DF, Dash RK, Bosnjak ZJ, Camara AKS. Mitochondrial targets for volatile anesthetics against cardiac ischemia-reperfusion injury. Front Physiol 2014; 5:341. [PMID: 25278902 PMCID: PMC4165278 DOI: 10.3389/fphys.2014.00341] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022] Open
Abstract
Mitochondria are critical modulators of cell function and are increasingly recognized as proximal sensors and effectors that ultimately determine the balance between cell survival and cell death. Volatile anesthetics (VA) are long known for their cardioprotective effects, as demonstrated by improved mitochondrial and cellular functions, and by reduced necrotic and apoptotic cell death during cardiac ischemia and reperfusion (IR) injury. The molecular mechanisms by which VA impart cardioprotection are still poorly understood. Because of the emerging role of mitochondria as therapeutic targets in diseases, including ischemic heart disease, it is important to know if VA-induced cytoprotective mechanisms are mediated at the mitochondrial level. In recent years, considerable evidence points to direct effects of VA on mitochondrial channel/transporter protein functions and electron transport chain (ETC) complexes as potential targets in mediating cardioprotection. This review furnishes an integrated overview of targets that VA impart on mitochondrial channels/transporters and ETC proteins that could provide a basis for cation regulation and homeostasis, mitochondrial bioenergetics, and reactive oxygen species (ROS) emission in redox signaling for cardiac cell protection during IR injury.
Collapse
Affiliation(s)
- Bhawana Agarwal
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - David F. Stowe
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
- Zablocki VA Medical CenterMilwaukee, WI, USA
- Department of Biomedical Engineering, Marquette UniversityMilwaukee, WI, USA
| | - Ranjan K. Dash
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Biomedical Engineering, Marquette UniversityMilwaukee, WI, USA
- Biotechnology and Bioengineering Center, Medical College of WisconsinMilwaukee, WI, USA
| | - Zeljko J. Bosnjak
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
| | - Amadou K. S. Camara
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
| |
Collapse
|
46
|
Yamakage M. Can age-related mitochondrial dysfunction affect volatile anesthetic potency? J Anesth 2014; 28:805-6. [PMID: 25150578 DOI: 10.1007/s00540-014-1907-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/11/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Michiaki Yamakage
- Department of Anesthesiology, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan,
| |
Collapse
|
47
|
Lou PH, Lucchinetti E, Zhang L, Affolter A, Gandhi M, Hersberger M, Warren BE, Lemieux H, Sobhi HF, Clanachan AS, Zaugg M. Loss of Intralipid®- but not sevoflurane-mediated cardioprotection in early type-2 diabetic hearts of fructose-fed rats: importance of ROS signaling. PLoS One 2014; 9:e104971. [PMID: 25127027 PMCID: PMC4134246 DOI: 10.1371/journal.pone.0104971] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/15/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Insulin resistance and early type-2 diabetes are highly prevalent. However, it is unknown whether Intralipid® and sevoflurane protect the early diabetic heart against ischemia-reperfusion injury. METHODS Early type-2 diabetic hearts from Sprague-Dawley rats fed for 6 weeks with fructose were exposed to 15 min of ischemia and 30 min of reperfusion. Intralipid® (1%) was administered at the onset of reperfusion. Peri-ischemic sevoflurane (2 vol.-%) served as alternative protection strategy. Recovery of left ventricular function was recorded and the activation of Akt and ERK 1/2 was monitored. Mitochondrial function was assessed by high-resolution respirometry and mitochondrial ROS production was measured by Amplex Red and aconitase activity assays. Acylcarnitine tissue content was measured and concentration-response curves of complex IV inhibition by palmitoylcarnitine were obtained. RESULTS Intralipid® did not exert protection in early diabetic hearts, while sevoflurane improved functional recovery. Sevoflurane protection was abolished by concomitant administration of the ROS scavenger N-2-mercaptopropionyl glycine. Sevoflurane, but not Intralipid® produced protective ROS during reperfusion, which activated Akt. Intralipid® failed to inhibit respiratory complex IV, while sevoflurane inhibited complex I. Early diabetic hearts exhibited reduced carnitine-palmitoyl-transferase-1 activity, but palmitoylcarnitine could not rescue protection and enhance postischemic functional recovery. Cardiac mitochondria from early diabetic rats exhibited an increased content of subunit IV-2 of respiratory complex IV and of uncoupling protein-3. CONCLUSIONS Early type-2 diabetic hearts lose complex IV-mediated protection by Intralipid® potentially due to a switch in complex IV subunit expression and increased mitochondrial uncoupling, but are amenable to complex I-mediated sevoflurane protection.
Collapse
Affiliation(s)
- Phing-How Lou
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Eliana Lucchinetti
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Liyan Zhang
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Andreas Affolter
- Department of Clinical Chemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Manoj Gandhi
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Martin Hersberger
- Department of Clinical Chemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Blair E. Warren
- Campus Saint-Jean, University of Alberta, Edmonton, Alberta, Canada
| | - Hélène Lemieux
- Campus Saint-Jean, University of Alberta, Edmonton, Alberta, Canada
| | - Hany F. Sobhi
- Coppin Center for Organic Synthesis, Coppin State University, Baltimore, Maryland, United States of America
| | | | - Michael Zaugg
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
48
|
Abstract
BACKGROUND Diabetes alters mitochondrial bioenergetics and consequently disrupts cardioprotective signaling. The authors investigated whether mitochondrial DNA (mtDNA) modulates anesthetic preconditioning (APC) and cardiac susceptibility to ischemia-reperfusion injury by using two strains of rats, both sharing nuclear genome of type 2 diabetes mellitus (T2DN) rats and having distinct mitochondrial genomes of Wistar and fawn-hooded hypertensive (FHH) rat strains (T2DN(mtWistar) and T2DN(mtFHH), respectively). METHODS Myocardial infarct size was measured in Wistar, T2DN(mtWistar), and T2DN(mtFHH) rats with or without APC (1.4% isoflurane) in the presence or absence of antioxidant N-acetylcysteine. Flavoprotein fluorescence intensity, a marker of mitochondrial redox state, 5-(and-6)-chloromethyl-2',7'-dichlorofluorescein fluorescence intensity, a marker of reactive oxygen species generation, and mitochondrial permeability transition pore opening were assessed in isolated rat ventricular cardiomyocytes with or without isoflurane (0.5 mmol/l). RESULTS Myocardial infarct size was decreased by APC in Wistar and T2DN(mtWistar) rats (to 42 ± 6%, n = 8; and 44 ± 7%, n = 8; of risk area, respectively) compared with their respective controls (60 ± 3%, n = 6; and 59 ± 9%, n = 7), but not in T2DN(mtFHH) rats (60 ± 2%, n = 8). N-acetylcysteine applied during isoflurane treatment restored APC in T2DN(mtFHH) (39 ± 6%, n = 7; and 38 ± 5%, n = 7; 150 and 75 mg/kg N-acetylcysteine, respectively), but abolished protection in control rats (54 ± 8%, n = 6). Similar to the data on infarct size, APC delayed mitochondrial permeability transition pore opening in T2DN(mtWistar) but not in T2DN(mtFHH) cardiomyocytes. Isoflurane increased flavoprotein and 5-(and-6)-chloromethyl-2',7'-dichlorofluorescein fluorescence intensity in all rat strains, with the greatest effect in T2DN(mtFHH) cardiomyocytes. CONCLUSION Differences in the mitochondrial genome modulate isoflurane-induced generation of reactive oxygen species which translates into differential susceptibility to APC and ischemia-reperfusion injury in diabetic rats.
Collapse
|
49
|
Time-Dependent Effects of Anesthetic Isoflurane on Reactive Oxygen Species Levels in HEK-293 Cells. Brain Sci 2014; 4:311-20. [PMID: 24961763 PMCID: PMC4101479 DOI: 10.3390/brainsci4020311] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 11/17/2022] Open
Abstract
The inhalation anesthetic isoflurane has been reported to induce caspase activation and apoptosis, which may lead to learning and memory impairment. However, the underlying mechanisms of these effects are largely unknown. Isoflurane has been shown to induce elevation of cytosol calcium levels, accumulation of reactive oxygen species (ROS), opening of the mitochondrial permeability transition pore, reduction in mitochondria membrane potential, and release of cytochrome c. The time course of these effects, however, remains to be determined. Therefore, we performed a pilot study to determine the effects of treatment with isoflurane for various times on ROS levels in HEK-293 cells. The cells were treated with 2% isoflurane plus 21% O2 and 5% CO2 for 15, 30, 60, or 90 min. We then used fluorescence imaging and microplate fluorometer to detect ROS levels. We show that 2% isoflurane for 60 or 90 min, but not 15 or 30 min, induced ROS accumulation in the cells. These data illustrated that isoflurane could cause time-dependent effects on ROS levels. These findings have established a system to further determine the time course effects of isoflurane on cellular and mitochondria function. Ultimately, the studies would elucidate, at least partially, the underlying mechanisms of isoflurane-induced cellular toxicity.
Collapse
|
50
|
Isoflurane anesthetic hypersensitivity and progressive respiratory depression in a mouse model with isolated mitochondrial complex I deficiency. J Anesth 2014; 28:807-14. [PMID: 24522811 DOI: 10.1007/s00540-014-1791-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 01/10/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Children with mitochondrial disorders are frequently anesthetized for a wide range of operations. These disorders may interfere with the response to surgery and anesthesia. We examined anesthetic sensitivity to and respiratory effects of isoflurane in the Ndufs4 knockout (KO) mouse model. These mice exhibit an isolated mitochondrial complex I (CI) deficiency of the respiratory chain, and they also display clinical signs and symptoms resembling those of patients with mitochondrial CI disease. METHODS We investigated seven Ndufs4(-/-) knockout (KO), five Ndufs4(+/-) heterozygous (HZ) and five Ndufs4(+/+) wild type (WT) mice between 22 and 25 days and again between 31 and 34 days post-natal. Animals were placed inside an airtight box, breathing spontaneously while isoflurane was administered in increasing concentrations. Minimum alveolar concentration (MAC) was determined with the bracketing study design, using the response to electrical stimulation to the hind paw. RESULTS MAC for isoflurane was significantly lower in KO mice than in HZ and WT mice: 0.81% ± 0.01 vs 1.55 ± 0.05% and 1.55 ± 0.13%, respectively, at 22-25 days, and 0.65 ± 0.05%, 1.65 ± 0.08% and 1.68 ± 0.08% at 31-34 days. The KO mice showed severe respiratory depression at lower isoflurane concentrations than the WT and HZ mice. CONCLUSION We observed an increased isoflurane anesthetic sensitivity and severe respiratory depression in the KO mice. The respiratory depression during anesthesia was strongly progressive with age. Since the pathophysiological consequences from complex I deficiency are mainly reflected in the central nervous system and our mouse model involves progressive encephalopathy, further investigation of isoflurane effects on brain mitochondrial function is warranted.
Collapse
|