1
|
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
|
2
|
Ebrahimi M, Dabbagh A, Madadi F. Propofol-induced hippocampal Neurotoxicity: A mitochondrial perspective. Brain Res 2024; 1831:148841. [PMID: 38428475 DOI: 10.1016/j.brainres.2024.148841] [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: 01/19/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Propofol is a frequently used anesthetic. It can induce neurodegeneration and inhibit neurogenesis in the hippocampus. This effect may be temporary. It can, however, become permanent in vulnerable populations, such as the elderly, who are more susceptible to Alzheimer's disease, and neonates and children, whose brains are still developing and require neurogenesis. Current clinical practice strategies have failed to provide an effective solution to this problem. In addition, the molecular mechanism of this toxicity is not fully understood. Recent advances in molecular research have revealed that apoptosis, in close association with mitochondria, is a crucial mechanism through which propofol contributes to hippocampal toxicity. Preventing the toxicity of propofol on the hippocampus has shown promise in in-vivo, in-vitro, and to a lesser extent human studies. This study seeks to provide a comprehensive literature review of the effects of propofol toxicity on the hippocampus via mitochondria and to suggest translational suggestions based on these molecular results.
Collapse
Affiliation(s)
- Moein Ebrahimi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Dabbagh
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firoozeh Madadi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Kilicaslan B, Akinci SB, Saricaoglu F, Yılbas SO, Ozkaya BA. Effects of coenzyme Q10 in a propofol infusion syndrome model of rabbits. ASIAN BIOMED 2023; 17:173-184. [PMID: 37860674 PMCID: PMC10584382 DOI: 10.2478/abm-2023-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Background Coenzyme Q (CoQ) might be the main site of interaction with propofol on the mitochondrial respiratory chain in the propofol infusion syndrome (PRIS) because of the structural similarity between coenzyme Q10 (CoQ10) and propofol. Aim To investigate the effects of CoQ10 on survival and organ injury in a PRIS model in rabbits. Methods Sixteen male New Zealand white rabbits were divided into 4 groups: (1) propofol infusion group, (2) propofol infusion and CoQ10, 100 mg/kg was administered intravenously, (3) sevoflurane inhalation was administered, and (4) sevoflurane inhalation and CoQ10, 100 mg/kg intravenously, was administered. Arterial blood gas and biochemical analyses were repeated every 2 h and every 12 h, respectively. Animals that were alive on the 24th hour after anesthesia induction were euthanized. The organ damages were investigated under light and transmission electron microscopy (TEM). Results The propofol infusion group had the highest troponin T levels when compared with the other three groups at the 12th hour. The propofol + CoQ10 group had lower troponin T levels when compared with the propofol and sevoflurane groups (P < 0.05). Administration of CoQ10 decreased total liver injury scores and total organ injury scores both in the propofol and sevoflurane groups. The propofol and sevoflurane organ toxicities were attenuated with CoQ10 in liver, gallbladder, urinary bladder, and spleen. Conclusion The addition of CoQ10 to propofol and sevoflurane anesthesia prevented the propofol-associated increase in troponin T levels at the 12th hour of infusion and decreased anesthetic-induced total liver and organ injury scores.
Collapse
Affiliation(s)
- Banu Kilicaslan
- Department of Anesthesiology and Intensive Care, Hacettepe University, Ankara06230, Turkey
| | - Seda B Akinci
- Department of Anesthesiology and Intensive Care, Hacettepe University, Ankara06230, Turkey
| | - Fatma Saricaoglu
- Department of Anesthesiology and Intensive Care, Hacettepe University, Ankara06230, Turkey
| | - Savas O Yılbas
- Department of Anesthesiology and Intensive Care, Hacettepe University, Ankara06230, Turkey
| | - Burcu A Ozkaya
- Department of Anesthesiology and Intensive Care, Hacettepe University, Ankara06230, Turkey
| |
Collapse
|
4
|
Zhang S, Deng Y, Gao Y. Malignant hyperthermia-like syndrome in acute chlorfenapyr poisoning- a case report. Heliyon 2022; 8:e10051. [PMID: 35992001 PMCID: PMC9382259 DOI: 10.1016/j.heliyon.2022.e10051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/22/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Background Chlorfenapyr is a pesticide that interferes with mitochondrial oxidative phosphorylation, resulting in the disruption of ATP production and cellular death. We present a fatal case of chlorfenapyr poisoning presented with malignant hyperthermia- like syndrome after intubation. Case presentation A 49-year-old male presented with fatigue and diaphoresis four days after ingesting emamectin chlorfenapyr. IV hydration was given, and two sessions of hemoperfusion were performed. He was intubated for airway protection on Day 3 because of drowsiness. Immediately after intubation, he developed tachycardia and hyperthermia (temperature 41 °C), followed by cardiac arrest. During resuscitation, we noted he had severe diaphoresis and generalized muscle rigidity. Peri-arrest ABG showed abrupt onset of severe type 2 respiratory failure, lactate acidosis, and hyperkalemia. The clinical manifestation and ABG result lead to the possible diagnosis of malignant hyperthermia. The resuscitation was unsuccessful, and the patient eventually passed away. Propofol might be the culprit drug in this case as it is known to affect mitochondrial metabolism via uncoupling oxidative phosphorylation. Conclusion We suggest monitoring for signs and symptoms of malignant hyperthermia in chlorfenapyr poisoning, especially after intubation. Propofol should be avoided or used with caution during induction for intubation. Further research on the possible antidote and usage of early RRT in ED is needed.
Collapse
|
5
|
Barajas MB, Brunner SD, Wang A, Griffiths KK, Levy RJ. Propofol toxicity in the developing mouse heart mitochondria. Pediatr Res 2022; 92:1341-1349. [PMID: 35173299 PMCID: PMC9378757 DOI: 10.1038/s41390-022-01985-1] [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: 07/27/2021] [Revised: 12/16/2021] [Accepted: 01/30/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Propofol infusion syndrome (PRIS) is a potentially lethal consequence of long-term propofol administration. Children are vulnerable and cardiac involvement is often prominent and associated with mortality. We aimed to determine the mechanism of propofol toxicity in newborn mice, hypothesizing that propofol would induce discrete defects within immature cardiac mitochondria. METHODS Newborn murine cardiac mitochondria were exposed to propofol or intralipid in vitro. Non-exposed mitochondria served as controls. Mitochondrial respiration and membrane potential (ΔΨ) were measured and respiratory chain complex kinetics were determined. RESULTS Propofol and intralipid exerted biological activity in isolated mitochondria. Although intralipid effects were a potential confounder, we found that propofol induced a dose-dependent increase in proton leak and caused a defect in substrate oxidation at coenzyme Q (CoQ). These impairments prevented propofol-exposed cardiomyocyte mitochondria from generating an adequate ΔΨ. The addition of the quinone analog, CoQ0, blocked propofol-induced leak and increased Complex II+III activity. CONCLUSIONS Propofol uncoupled immature cardiomyocyte mitochondria by inducing excessive CoQ-sensitive leak and interfered with electron transport at CoQ. The findings provide new insight into the mechanisms of propofol toxicity in the developing heart and may help explain why children are vulnerable to developing PRIS. IMPACT Propofol uncouples immature cardiomyocyte mitochondria by inducing excessive coenzyme Q (CoQ)-sensitive proton leak. Propofol also interferes with electron transport at the level of CoQ. These defects provide new insight into propofol toxicity in the developing heart.
Collapse
Affiliation(s)
- Matthew B. Barajas
- grid.239585.00000 0001 2285 2675Department of Anesthesiology, Columbia University Medical Center, New York, NY USA
| | - Sarah D. Brunner
- grid.239585.00000 0001 2285 2675Department of Pediatrics, Division of Pediatric Critical Care Medicine, Columbia University Medical Center, New York, NY USA
| | - Aili Wang
- grid.239585.00000 0001 2285 2675Department of Anesthesiology, Columbia University Medical Center, New York, NY USA
| | - Keren K. Griffiths
- grid.239585.00000 0001 2285 2675Department of Anesthesiology, Columbia University Medical Center, New York, NY USA
| | - Richard J. Levy
- grid.239585.00000 0001 2285 2675Department of Anesthesiology, Columbia University Medical Center, New York, NY USA
| |
Collapse
|
6
|
Host bioenergetic parameters reveal cytotoxicity of anti-tuberculosis drugs undetected using conventional viability assays. Antimicrob Agents Chemother 2021; 65:e0093221. [PMID: 34339269 PMCID: PMC8448146 DOI: 10.1128/aac.00932-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
High attrition rates in tuberculosis (TB) drug development have been largely attributed to safety, which is likely due to the use of endpoint assays measuring cell viability to detect drug cytotoxicity. In drug development for cancer, metabolic, and neurological disorders and for antibiotics, cytotoxicity is increasingly being assessed using extracellular flux (XF) analysis, which measures cellular bioenergetic metabolism in real time. Here, we adopt the XF platform to investigate the cytotoxicity of drugs currently used in TB treatment on the bioenergetic metabolism of HepG2 cells, THP-1 macrophages, and human monocyte-derived macrophages (hMDMs). We found that the XF analysis reveals earlier drug-induced effects on the cells’ bioenergetic metabolism prior to cell death, measured by conventional viability assays. Furthermore, each cell type has a distinct response to drug treatment, suggesting that more than one cell type should be considered to examine cytotoxicity in TB drug development. Interestingly, chemically unrelated drugs with different modes of action on Mycobacterium tuberculosis have similar effects on the bioenergetic parameters of the cells, thus discouraging the prediction of potential cytotoxicity based on chemical structure and mode of action of new chemical entities. The clustering of the drug-induced effects on the hMDM bioenergetic parameters are reflected in the clustering of the effects of the drugs on cytokine production in hMDMs, demonstrating concurrence between the effects of the drugs on the metabolism and functioning of the macrophages. These findings can be used as a benchmark to establish XF analysis as a new tool to assay cytotoxicity in TB drug development.
Collapse
|
7
|
Elucidating the interaction of propofol as an intravenous anesthetic drug with blood components: IgG and peripheral blood mononuclear cell as targets. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.102965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
8
|
Ren Z, Hu R. Downregulation of long noncoding RNA SNHG6 rescued propofol-induced cytotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc Diagn Ther 2020; 10:811-819. [PMID: 32968636 DOI: 10.21037/cdt-20-443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Propofol (PPF) overdose is a rare but lethal condition, which may lead to severe cardiac failure. In this study, we established an in vitro PPF-induced cardiac cytotoxicity model, and investigate the functional role of long non-coding RNA (lncRNA) small nucleolar RNA host gene 6 (SNHG6). Methods Human induced pluripotent stem cell-derived cardiomyocytes (HiPSC-CMs) were exposed to PPF in vitro. PPF-induced cytotoxic effects were measured. PPF-induced SNHG6 expression change in HiPSC-CMs were monitored by qRT-PCR. SNHG6 was downregulated in HiPSC-CMs to examine its role in PPF-induced cardiac cytotoxicity. The expression of competing endogenous RNA (ceRNA) candidate of SNHG6, human microRNA-186-5p (hsa-miR-186-5p) was also investigated in PPF-exposed HiPSC-CMs. Functions of hsa-miR-186-5p were further investigated in PPF-exposed and SNHG6-downregulated HiPSC-CMs. Results PPF induced significant cytotoxicity, as well as SNHG6 upregulation in HiPSC-CMs. SNHG6 downregulation had rescuing effects on PPF-induced cardiac cytotoxicity. Dual-luciferase activity assay confirmed that hsa-miR-186-5p was the ceRNA candidate of SNHG6. QRT-PCR showed hsa-miR-186-5p expression was reversely correlated with SNHG6 in PPF-exposed HiPSC-CMs. Suppressing hsa-miR-186-5p reduced the rescuing effects of SNHG6-downregulation on PPF-induced cardiac cytotoxicity. Conclusions SNHG6/hsa-miR-186-5p can modulate PPF-induced cardiac cytotoxicity in HiPSC-CMs, and thus may be a future drug target to prevent PPF infusion syndrome.
Collapse
Affiliation(s)
- Zhongguo Ren
- Department of Anesthesiology, The People's Hospital of China Three Gorges University, Yichang, China
| | - Rong Hu
- Department of Geriatrics, The People's Hospital of China Three Gorges University, Yichang, China
| |
Collapse
|
9
|
He L, Wang X, Zheng S. Inhibition of the electron transport chain in propofol induced neurotoxicity in zebrafish embryos. Neurotoxicol Teratol 2020; 78:106856. [PMID: 31923456 DOI: 10.1016/j.ntt.2020.106856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 01/20/2023]
Abstract
Fetal and neonatal exposure to propofol can lead to neuronal death and long-term neurobehavioral deficiencies in both rodents and nonhuman primates. Zebrafish embryo, which is fertilized ex-utero, has provided us a new model species to study the effects of general anesthetics on developing brain. Inhibited electron transport chain leads to mitochondrial dysfunction and insufficient energy production. The aim of this study was to dissect the role of electron transport chain in propofol-induced neurotoxicity. 6 h post fertilization (hpf) zebrafish embryos were exposed to control or 1, 2 or 4 μg/ml propofol until 48hpf. Acridine orange staining was used to assess cell apoptosis in the brain of zebrafish embryos. The activity of mitochondrial electron transport chain complex was assessed using colorimetric method. Expression of key subunit of cytochrome c oxidase was assessed by western blot and transcription level of cox4i1 was assessed by quantitative real time-PCR. The mitochondrial membrane potential and ATP content were assessed. Exposure to 1, 2 and 4 μg/ml propofol induced significant increases in cell apoptosis in the brain of zebrafish embryos in a dose-dependent manner and led to significant decreases in electron transport chain complex IV activity from (0.161 ± 0.023)μmol/mg/min in blank control-treated group to (0.096 ± 0.015)μmol/mg/min, (0.083 ± 0.013)μmol/mg/min and (0.045 ± 0.014)μmol/mg/min respectively, accompanied by decreased expression of key regulatory subunit of cytochrome c oxidase-subunit IV and decreased transcription level of cox4i1. Propofol exposure also decreased the mitochondrial membrane potential and ATP content. Our findings demonstrate that inhibition of the electron transport chain is involved in the mechanisms by which propofol induces neurotoxicity in the developing brain.
Collapse
Affiliation(s)
- Lin He
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Xuan Wang
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Shan Zheng
- Department of Surgery, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Wang HW, Zhang Y, Tan PP, Jia LS, Chen Y, Zhou BH. Mitochondrial respiratory chain dysfunction mediated by ROS is a primary point of fluoride-induced damage in Hepa1-6 cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 255:113359. [PMID: 31614248 DOI: 10.1016/j.envpol.2019.113359] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/12/2019] [Accepted: 10/06/2019] [Indexed: 05/20/2023]
Abstract
To evaluate the mechanism of fluoride (F) mitochondrial toxicity, we cultured Hepa1-6 cells with different F concentrations (0, 1 and 2 mmoL/L) and determined cell pathological morphology, mitochondrial respiratory chain damage and cell cycle change. Results showed that the activities and mRNA expression levels of antioxidant enzymes considerably decreased, whereas the contents of reactive oxygen species (ROS), malondialdehyde (MDA) and nitric oxide (NO) markedly increased. Breakage of mitochondrial cristae and substantial vacuolated mitochondria were observed by transmission electron microscopy. These results indicate the F-induced oxidative damage in Hepa1-6 cells. The enzyme activities of mitochondrial complexes I, II, III and IV were disordered in Hepa1-6 cells treated by excessive F, thereby indicating a remarkable down-regulation. Further research showed that complex subunits also demonstrated the development of disorder, in which the protein expressions levels of NDUFV2 and SDHA were substantially down-regulated, whereas those of CYC1 and COX Ⅳ were markedly up-regulated. Reductions in ATP and mitochondrial membrane potential were detected with the dysfunction of the mitochondrial respiratory chain. The G2/M phase arrest of the cell cycle in Hepa1-6 cells was measured via flow cytometry, and the up-regulated protein expressions of Cyt c, caspase 9, caspase 3 and substantial apoptotic cells were determined. In summary, this study demonstrated that ROS-mediated mitochondrial respiratory chain dysfunction causes F-induced Hepa1-6 cell damage.
Collapse
Affiliation(s)
- Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Yan Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Pan-Pan Tan
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Liu-Shu Jia
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Yu Chen
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China
| | - Bian-Hua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China.
| |
Collapse
|
11
|
Qin J, Li Y, Wang K. Propofol induces impairment of mitochondrial biogenesis through inhibiting the expression of peroxisome proliferator-activated receptor-γ coactivator-1α. J Cell Biochem 2019; 120:18288-18297. [PMID: 31190345 DOI: 10.1002/jcb.29138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 11/07/2022]
Abstract
Propofol is a commonly used general anesthetic in patient care. Recent studies have shown that propofol has neurological side effects especially in young children, which raises a concern regarding the safety of its use. We explored the effects of the molecular mechanism of propofol on neuronal mitochondrial function in SH-SY5Y cells. Our results demonstrate that clinically relevant doses of propofol reduce the expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) in a dose- and time-dependent manner. At a concentration of 2%, propofol suppresses the mitochondrial regulator nuclear respiratory factor 1 and mitochondrial transcription factor A and impairs neuronal mitochondrial biogenesis. These impairments involve reduction of mitochondrial mass and reduction of the ratio of mitochondrial to nuclear DNA as well as reduction of cytochrome C oxidase activity. Propofol treatment reduces intracellular adenosine triphosphate (ATP) production, the mitochondrial respiratory rate, and increases mitochondrial reactive oxygen species production, implying that it disturbs neuronal mitochondrial function. Overexpression of PGC-1α rescued propofol-induced reduced mitochondrial mass, ATP production, and respiratory rate, indicating that PGC-1α is the mediator of the effect of propofol on mitochondrial function. Finally, we demonstrate that propofol suppresses PGC-1α by inhibiting cAMP-response element binding protein (CREB) activation and promoting PKA RI expression, and the addition of cyclic adenosine monophosphate rescues propofol-mediated reduced PGC-1α. In conclusion, PGC-1α is the central mediator of propofol-induced impairment of mitochondrial biogenesis and neuronal mitochondrial dysfunction. Our study demonstrates the molecular mechanism behind propofol-induced neurotoxicity and provides valuable information regarding its side effects in clinical practice.
Collapse
Affiliation(s)
- Jing Qin
- Department of Anesthesiology, Linyi Central Hospital, Linyi, Shandong, China
| | - Yizhen Li
- Department of Anesthesiology, Weifang People's Hospital, Weifang, Shandong, China
| | - Kai Wang
- Department of Breast Surgery, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
12
|
Tamoxifen citrate/Coenzyme Q10 as smart nanocarriers Bitherapy for Breast Cancer: Cytotoxicity, genotoxicity, and antioxidant activity. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Nutraceuticals Inspiring the Current Therapy for Lifestyle Diseases. Adv Pharmacol Sci 2019; 2019:6908716. [PMID: 30755770 PMCID: PMC6348880 DOI: 10.1155/2019/6908716] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/16/2018] [Indexed: 12/17/2022] Open
Abstract
Nutraceuticals are the pharmaceutically blended products that possess both nutritional as well as the medicinal value. Such a product is designed to improve the physical health, fight against day-to-day challenges such as stress, increase longevity, etc. Nowadays, emphasis is given to those herbs which are used as food and medicine due to its greater acceptance. Due to dynamic action, the popularity of nutraceuticals among people as well as healthcare providers has been increased over medicines and health supplements. This review documents herbs with a wide variety of therapeutic values such as immunity booster, antidiabetic, anticancer, antimicrobial, and gastroprotective. These herbs could be better options to formulate as nutraceuticals. Several nutraceuticals are described based on their availability as food, chemical nature, and mechanism of action.
Collapse
|
14
|
Berndt N, Rösner J, Haq RU, Kann O, Kovács R, Holzhütter HG, Spies C, Liotta A. Possible neurotoxicity of the anesthetic propofol: evidence for the inhibition of complex II of the respiratory chain in area CA3 of rat hippocampal slices. Arch Toxicol 2018; 92:3191-3205. [PMID: 30143847 PMCID: PMC6132669 DOI: 10.1007/s00204-018-2295-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
Propofol is the most frequently used intravenous anesthetic for induction and maintenance of anesthesia. Propofol acts first and formost as a GABAA-agonist, but effects on other neuronal receptors and voltage-gated ion channels have been described. Besides its direct effect on neurotransmission, propofol-dependent impairment of mitochondrial function in neurons has been suggested to be responsible for neurotoxicity and postoperative brain dysfunction. To clarify the potential neurotoxic effect in more detail, we investigated the effects of propofol on neuronal energy metabolism of hippocampal slices of the stratum pyramidale of area CA3 at different activity states. We combined oxygen-measurements, electrophysiology and flavin adenine dinucleotide (FAD)-imaging with computational modeling to uncover molecular targets in mitochondrial energy metabolism that are directly inhibited by propofol. We found that high concentrations of propofol (100 µM) significantly decrease population spikes, paired pulse ratio, the cerebral metabolic rate of oxygen consumption (CMRO2), frequency and power of gamma oscillations and increase FAD-oxidation. Model-based simulation of mitochondrial FAD redox state at inhibition of different respiratory chain (RC) complexes and the pyruvate-dehydrogenase show that the alterations in FAD-autofluorescence during propofol administration can be explained with a strong direct inhibition of the complex II (cxII) of the RC. While this inhibition may not affect ATP availability under normal conditions, it may have an impact at high energy demand. Our data support the notion that propofol may lead to neurotoxicity and neuronal dysfunction by directly affecting the energy metabolism in neurons.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.,Institute for Computational and Imaging Science in Cardiovascular Medicine Charité, Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Jörg Rösner
- Neuroscience Research Center, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Rizwan Ul Haq
- Neuroscience Research Center, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, 69120, Heidelberg, Germany
| | - Richard Kovács
- Institute for Neurophysiology, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | | | - Claudia Spies
- Department of Anesthesiology and Intensive Care, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.,Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Agustin Liotta
- Neuroscience Research Center, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany. .,Department of Anesthesiology and Intensive Care, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany. .,Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
15
|
Diquigiovanni C, Bergamini C, Evangelisti C, Isidori F, Vettori A, Tiso N, Argenton F, Costanzini A, Iommarini L, Anbunathan H, Pagotto U, Repaci A, Babbi G, Casadio R, Lenaz G, Rhoden KJ, Porcelli AM, Fato R, Bowcock A, Seri M, Romeo G, Bonora E. Mutant MYO1F alters the mitochondrial network and induces tumor proliferation in thyroid cancer. Int J Cancer 2018; 143:1706-1719. [PMID: 29672841 DOI: 10.1002/ijc.31548] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 03/21/2018] [Accepted: 04/11/2018] [Indexed: 12/26/2022]
Abstract
Familial aggregation is a significant risk factor for the development of thyroid cancer and familial non-medullary thyroid cancer (FNMTC) accounts for 5-7% of all NMTC. Whole exome sequencing analysis in the family affected by FNMTC with oncocytic features where our group previously identified a predisposing locus on chromosome 19p13.2, revealed a novel heterozygous mutation (c.400G > A, NM_012335; p.Gly134Ser) in exon 5 of MYO1F, mapping to the linkage locus. In the thyroid FRTL-5 cell model stably expressing the mutant MYO1F p.Gly134Ser protein, we observed an altered mitochondrial network, with increased mitochondrial mass and a significant increase in both intracellular and extracellular reactive oxygen species, compared to cells expressing the wild-type (wt) protein or carrying the empty vector. The mutation conferred a significant advantage in colony formation, invasion and anchorage-independent growth. These data were corroborated by in vivo studies in zebrafish, since we demonstrated that the mutant MYO1F p.Gly134Ser, when overexpressed, can induce proliferation in whole vertebrate embryos, compared to the wt one. MYO1F screening in additional 192 FNMTC families identified another variant in exon 7, which leads to exon skipping, and is predicted to alter the ATP-binding domain in MYO1F. Our study identified for the first time a role for MYO1F in NMTC.
Collapse
Affiliation(s)
- Chiara Diquigiovanni
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Cecilia Evangelisti
- Department of Biomedical and Neuromotor Sciences, DIBINEM, University of Bologna, Bologna, Italy
| | - Federica Isidori
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Andrea Vettori
- Department of Biology, University of Padova, Padova, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | | | - Anna Costanzini
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Hima Anbunathan
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Uberto Pagotto
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Andrea Repaci
- Endocrinology Unit, St. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Giorgio Lenaz
- Department of Biomedical and Neuromotor Sciences, DIBINEM, University of Bologna, Bologna, Italy
| | - Kerry J Rhoden
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Romana Fato
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Bologna, Italy
| | - Anne Bowcock
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Marco Seri
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Giovanni Romeo
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Elena Bonora
- Department of Medical and Surgical Sciences, DIMEC, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Propofol Reversed Hypoxia-Induced Docetaxel Resistance in Prostate Cancer Cells by Preventing Epithelial-Mesenchymal Transition by Inhibiting Hypoxia-Inducible Factor 1 α. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4174232. [PMID: 29568752 PMCID: PMC5820676 DOI: 10.1155/2018/4174232] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/10/2017] [Accepted: 11/23/2017] [Indexed: 12/27/2022]
Abstract
Prostate cancer is the second most frequently diagnosed cancer worldwide. Hypoxia-induced epithelial–mesenchymal transition (EMT), driven by hypoxia-inducible factor 1α (HIF-1α), is involved in cancer progression and metastasis. The present study was designed to explore the role of propofol in hypoxia-induced resistance of prostate cancer cells to docetaxel. We used the Cell Counting Kit-8 and 5-ethynyl-2′-deoxyuridine incorporation assay to measure cell viability and cell proliferation, respectively, in prostate cancer cell lines. Then, we detected HIF-1α, E-cadherin, and vimentin expression using western blotting. Propofol reversed the hypoxia-induced docetaxel resistance in the prostate cancer cell lines. Propofol not only decreased hypoxia-induced HIF-1α expression, but also reversed hypoxia-induced EMT by suppressing HIF-1α. Furthermore, small interfering RNA–mediated silencing of HIF-1α reversed the hypoxia-induced docetaxel resistance, although there was little change in docetaxel sensitivity between the hypoxia group and propofol group. The induction of hypoxia did not affect E-cadherin and vimentin expression, and under the siRNA knockdown conditions, the effects of propofol were obviated. These data support a role for propofol in regulating EMT in prostate cancer cells. Taken together, our findings demonstrate that propofol plays an important role in hypoxia-induced docetaxel sensitivity and EMT in prostate cancer cells and that it is a potential drug for overcoming drug resistance in prostate cancer cells via HIF-1α suppression.
Collapse
|
17
|
Cytotoxicity of propofol in human induced pluripotent stem cell-derived cardiomyocytes. J Anesth 2017; 32:120-131. [PMID: 29288336 PMCID: PMC5797219 DOI: 10.1007/s00540-017-2441-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/16/2017] [Indexed: 12/04/2022]
Abstract
Purpose Propofol infusion syndrome (PRIS) is a lethal condition caused by propofol overdose. Previous studies suggest that pathophysiological mechanisms underlying PRIS involve mitochondrial dysfunction; however, these mechanisms have not been fully elucidated. This study aimed to establish an experimental model of propofol-induced cytotoxicity using cultured human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to determine the mechanisms behind propofol-induced mitochondrial dysfunction, and to evaluate the protective effects of coenzyme Q10 (CoQ10). Methods Human iPSC-derived cardiomyocytes were exposed to propofol (0, 2, 10, or 50 µg/ml) with or without 5 µM CoQ10. Mitochondrial function was assessed by measuring intracellular ATP, lactate concentrations in culture media, NAD+/NADH ratio, and the mitochondrial membrane potential. Propofol-induced cytotoxicity was evaluated by analysis of cell viability. Expression levels of genes associated with mitochondrial energy metabolism were determined by PCR. Intracellular morphological changes were analyzed by confocal microscopy. Results Treatment with 50 µg/ml propofol for 48 h reduced cell viability. High concentrations of propofol (≥ 10 µg/ml) induced mitochondrial dysfunction accompanied by downregulation of gene expression of PGC-1alpha and its downstream targets (NDUFS8 and SDHB, which are involved in the respiratory chain reaction; and CPT1B, which regulates beta-oxidation). Cardiomyocytes co-treated with 5 µM CoQ10 exhibited resistance to propofol-induced toxicity through recovery of gene expression. Conclusions Propofol-induced cytotoxicity in human iPSC-derived cardiomyocytes may be associated with mitochondrial dysfunction via downregulation of PGC-1alpha-regulated genes associated with mitochondrial energy metabolism. Co-treatment with CoQ10 protected cardiomyocytes from propofol-induced cytotoxicity.
Collapse
|
18
|
Acute action of rotenone on excitability of catecholaminergic neurons in rostral ventrolateral medulla. Brain Res Bull 2017; 134:151-161. [DOI: 10.1016/j.brainresbull.2017.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/09/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022]
|
19
|
Cata JP, Bhavsar S, Hagan KB, Arunkumar R, Grasu R, Dang A, Carlson R, Arnold B, Popat K, Rao G, Potylchansky Y, Lipski I, Ratty S, Nguyen AT, McHugh T, Feng L, Rahlfs TF. Intraoperative serum lactate is not a predictor of survival after glioblastoma surgery. J Clin Neurosci 2017; 43:224-228. [PMID: 28601568 DOI: 10.1016/j.jocn.2017.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/21/2017] [Accepted: 05/21/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Cancer cells can produce lactate in high concentrations. Two previous studies examined the clinical relevance of serum lactate as a biomarker in patients with brain tumors. Patients with high-grade tumors have higher serum concentrations of lactate than those with low-grade tumors. We hypothesized that serum lactic could be used of biomarker to predictor of survival in patients with glioblastoma (GB). METHODS This was a retrospective study. Demographic, lactate concentrations and imaging data from 275 adult patients with primary GB was included in the analysis. The progression free survival (PFS) and overall survival (OS) rates were compared in patients who had above and below the median concentrations of lactate. We also investigated the correlation between lactate concentrations and tumor volume. Multivariate analyses were conducted to test the association lactate, tumor volume and demographic variables with PFS and OS. RESULTS The median serum concentration of lactate was 2.3mmol/L. A weak correlation was found between lactate concentrations and tumor volume. Kaplan-Meier curves demonstrated similar survival in patients with higher or lower than 2.3mmol/L of lactate. The multivariate analysis indicated that the intraoperative levels of lactate were not independently associated with changes in survival. On another hand, a preoperative T1 volume was an independent predictor PFS (HR 95%CI: 1.41, 1.02-1.82, p=0.006) and OS (HR 95%CI: 1.47, 1.11-1.96, p=0.006). CONCLUSION This retrospective study suggests that the serum concentrations of lactate cannot be used as a biomarker to predict survival after GB surgery. To date, there are no clinically available serum biomarkers to determine prognosis in patients with high-grade gliomas. These tumors may produce high levels of lactic acid. We hypothesized that serum lactic could be used of biomarker to predictor of survival in patients with glioblastoma (GB). In this study, we collected perioperative and survival data from 275 adult patients with primary high-grade gliomas to determine whether intraoperative serum acid lactic concentrations can serve as a marker of prognosis. The median serum concentration of lactate was 2.3mmol/L. Our analysis indicated the intraoperative levels of lactate were not independently associated with changes in survival. This retrospective study suggests that the serum concentrations of lactate cannot be used as a biomarker to predict survival after GB surgery.
Collapse
Affiliation(s)
- J P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.
| | - S Bhavsar
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - K B Hagan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Arunkumar
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Grasu
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - A Dang
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Carlson
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - B Arnold
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - K Popat
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Y Potylchansky
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - I Lipski
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Sally Ratty
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - A T Nguyen
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas McHugh
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - L Feng
- Department of Biostatistics, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - T F Rahlfs
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
Hsieh VC, Krane EJ, Morgan PG. Mitochondrial Disease and Anesthesia. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409817707770] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Vincent C. Hsieh
- Department of Anesthesiology and Perioperative Medicine, University of Washington, Seattle, WA, USA
| | - Elliot J. Krane
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Philip G. Morgan
- Department of Anesthesiology and Perioperative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|