1
|
Sharma KK, Chauhan B. Integrated dexmedetomidine-sevoflurane algorithm for anesthetic induction - A viable asset for neurosurgery. Surg Neurol Int 2024; 15:455. [PMID: 39777190 PMCID: PMC11704436 DOI: 10.25259/sni_934_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Affiliation(s)
- Kunal Kumar Sharma
- Department of Anesthesia, Neuroanesthesia Cell, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India
| | - Bharti Chauhan
- Department of Anesthesia and Critical Care, Neuroanesthesia Division Under, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
2
|
Dos Santos BG, Brisnovali NF, Goedeke L. Biochemical basis and therapeutic potential of mitochondrial uncoupling in cardiometabolic syndrome. Biochem J 2024; 481:1831-1854. [PMID: 39630236 DOI: 10.1042/bcj20240005] [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: 08/05/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
Mild uncoupling of oxidative phosphorylation is an intrinsic property of all mitochondria, allowing for adjustments in cellular energy metabolism to maintain metabolic homeostasis. Small molecule uncouplers have been extensively studied for their potential to increase metabolic rate, and recent research has focused on developing safe and effective mitochondrial uncoupling agents for the treatment of obesity and cardiometabolic syndrome (CMS). Here, we provide a brief overview of CMS and cover the recent mechanisms by which chemical uncouplers regulate CMS-associated risk-factors and comorbidities, including dyslipidemia, insulin resistance, steatotic liver disease, type 2 diabetes, and atherosclerosis. Additionally, we review the current landscape of uncoupling agents, focusing on repurposed FDA-approved drugs and compounds in advanced preclinical or early-stage clinical development. Lastly, we discuss recent molecular insights by which chemical uncouplers enhance cellular energy expenditure, highlighting their potential as a new addition to the current CMS drug landscape, and outline several limitations that need to be addressed before these agents can successfully be introduced into clinical practice.
Collapse
Affiliation(s)
- Bernardo Gindri Dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Niki F Brisnovali
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| |
Collapse
|
3
|
Leister N, Wendt S, Hedergott A, Heindl LM, Rokohl AC, Stoll SE, Gordon E, Böttiger BW, Fricke J, Schick VC. Anaesthesia Concepts in Patients with Chronic Progressive External Ophthalmoplegia Undergoing Ophthalmic Surgery-A Retrospective Cohort Analysis. J Clin Med 2024; 13:4710. [PMID: 39200852 PMCID: PMC11355925 DOI: 10.3390/jcm13164710] [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: 07/07/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND Chronic progressive external ophthalmoplegia (CPEO) belongs to the group of mitochondrial encephalomyopathies. Anaesthesia for patients with CPEO may be associated with an increased risk due to known drug effects on mitochondrial metabolism. Therefore, the aim of this analysis was to evaluate anaesthesiological concepts in patients with CPEO requiring ophthalmic surgery. METHODS This is a retrospective, monocentric cohort analysis of eleven patients with CPEO undergoing ophthalmic surgery either with general anaesthesia or local anaesthesia in a German university hospital from January 2012 to February 2022. RESULTS A total of twelve ophthalmic surgery procedures were performed in eleven adult patients with CPEO. Six patients underwent surgery after receiving local anaesthesia (LA cohort). Five patients underwent six surgical procedures under general anaesthesia (GA cohort). In five cases within the GA cohort, propofol and remifentanil were used for the maintenance of anaesthesia. In one case, balanced anaesthesia with desflurane and remifentanil was used. The median duration of general anaesthesia was 37.5 min (range, 25-65 min). Patients stayed in the recovery room for a median of 48.5 min (range, 35-70 min). All patients were discharged on the first postoperative day. No relevant complications occurred in either the LA or GA cohort. CONCLUSION Both local and general anaesthesia are feasible concepts for patients with CPEO undergoing ophthalmic surgery. Propofol, at least with a short duration (less than one hour) of use, appears to be a feasible hypnotic drug in CPEO patients.
Collapse
Affiliation(s)
- Nicolas Leister
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (S.E.S.); (B.W.B.); (V.C.S.)
| | - Stefanie Wendt
- Department of Cardiothoracic Surgery and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany;
| | - Andrea Hedergott
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (A.H.); (L.M.H.); (A.C.R.); (E.G.); (J.F.)
| | - Ludwig M. Heindl
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (A.H.); (L.M.H.); (A.C.R.); (E.G.); (J.F.)
| | - Alexander C. Rokohl
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (A.H.); (L.M.H.); (A.C.R.); (E.G.); (J.F.)
| | - Sandra E. Stoll
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (S.E.S.); (B.W.B.); (V.C.S.)
| | - Erik Gordon
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (A.H.); (L.M.H.); (A.C.R.); (E.G.); (J.F.)
| | - Bernd W. Böttiger
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (S.E.S.); (B.W.B.); (V.C.S.)
| | - Julia Fricke
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (A.H.); (L.M.H.); (A.C.R.); (E.G.); (J.F.)
| | - Volker C. Schick
- Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany; (S.E.S.); (B.W.B.); (V.C.S.)
| |
Collapse
|
4
|
Slingerland-Boot R, Kummerow M, Arbous SM, van Zanten ARH. Association between first-week propofol administration and long-term outcomes of critically ill mechanically ventilated patients: A retrospective cohort study. Clin Nutr 2024; 43:42-51. [PMID: 38000194 DOI: 10.1016/j.clnu.2023.10.029] [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: 06/20/2023] [Revised: 09/21/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND & AIM Propofol is commonly used in ICUs, but its long-term effects have not been thoroughly studied. In vitro studies suggest it may harm mitochondrial function, potentially affecting clinical outcomes. This study aimed to investigate the association between substantial propofol sedation and clinical outcomes in critically ill patients. METHODS We conducted a single-centre cohort study of critically ill, mechanically ventilated (≥7 days) adults to compare patients who received a substantial dose of propofol (cumulative >500 mg) during the first week of ICU admission with those who did not. The primary outcome was the association between substantial propofol administration and 6-month mortality, adjusted for relevant covariates. Subanalyses were performed for administration in the early (day 1-3) and late (day 4-7) acute phases of critical illness due to the metabolic changes in this period. Secondary outcomes included tracheostomy need and duration, length of ICU and hospital stay (LOS), discharge destinations, ICU, hospital, and 3-month mortality. RESULTS A total of 839 patients were enrolled, with 73.7 % receiving substantial propofol administration (substantial propofol dose group). Six-month all-cause mortality was 32.4 %. After adjusting for relevant variables, we found no statistically significant difference in 6-month mortality between both groups. There were also no significant differences in secondary outcomes. CONCLUSION Our study suggests that substantial propofol administration during the first week of ICU stay in the least sick critically ill, mechanically ventilated adult patients is safe, with no significant associations found with 6-month mortality, ICU or hospital LOS, differences in discharge destinations or need for tracheostomy.
Collapse
Affiliation(s)
- Rianne Slingerland-Boot
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, the Netherlands
| | - Maren Kummerow
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands
| | - Sesmu M Arbous
- Department of Intensive Care Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Arthur R H van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, the Netherlands.
| |
Collapse
|
5
|
Holzer M, Poole JE, Lascarrou JB, Fujise K, Nichol G. A Commentary on the Effect of Targeted Temperature Management in Patients Resuscitated from Cardiac Arrest. Ther Hypothermia Temp Manag 2023; 13:102-111. [PMID: 36378270 PMCID: PMC10625468 DOI: 10.1089/ther.2022.0041] [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] [Indexed: 11/16/2022] Open
Abstract
The members of the International Liaison Committee on Resuscitation (ILCOR) Advanced Life Support Task Force have written a comprehensive summary of trials of the effectiveness of induced hypothermia (IH) or targeted temperature management (TTM) in comatose patients after cardiac arrest (CA). However, in-depth analysis of these studies is incomplete, especially since there was no significant difference in primary outcome between hypothermia versus normothermia in the recently reported TTM2 trial. We critically appraise trials of IH/TTM versus normothermia to characterize reasons for the lack of treatment effect, based on a previously published framework for what to consider when the primary outcome fails. We found a strong biologic rationale and external clinical evidence that IH treatment is beneficial. Recent TTM trials mainly included unselected patients with a high rate of bystander cardiopulmonary resuscitation. The treatment was not applied as intended, which led to a large delay in achievement of target temperature. While receiving intensive care, sedative drugs were likely used that might have led to increased neurologic damage as were antiplatelet drugs that could be associated with increased acute stent thrombosis in hypothermic patients. It is reasonable to still use or evaluate IH treatment in patients who are comatose after CA as there are multiple plausible reasons why IH compared to normothermia did not significantly improve neurologic outcome in the TTM trials.
Collapse
Affiliation(s)
- Michael Holzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Jeanne E. Poole
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | | | - Ken Fujise
- Harborview Medical Center, Heart Institute, University of Washington, Seattle, Washington, USA
| | - Graham Nichol
- Departments of Medicine and Emergency Medicine, University of Washington-Harborview Center for Prehospital Emergency Care, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
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
|
7
|
Therapeutic Hypothermia Following Cardiac Arrest After the TTM2 trial - More Questions Raised Than Answered. Curr Probl Cardiol 2023; 48:101046. [PMID: 34780867 DOI: 10.1016/j.cpcardiol.2021.101046] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/05/2021] [Indexed: 02/06/2023]
Abstract
For almost 20 years, therapeutic hypothermia has been a cornerstone of modern post-cardiac arrest care lowering mortality, and improvin neurologic outcome compared to conventional therapy. This was challenged by the first TTM-trial in 2013, which did not show a benefit for hypothermia at 33°C compared to controlled normothermia at 36°C. Now, the TTM2 trial showed no benefit of hypothermia compared to fever prevention alone. While TTM1 and TTM2 suggest that hypothermia might not be helpful, a deep dive into the trials reveals that this conclusion does not hold true. Here, we focus on patient selection, suboptimal application of hypothermia, interaction of standard sedation with hypothermia, high incidence of post-arrest fever, and withdrawal of life support based on per-protocol neurologic prognostication in the TTM2-trial. Of particular interest, contemporary trials and registries using intravascular cooling in TTM-like patients repeatedly reported much lower mortality rates than those described in both TTM1 and TTM2.
Collapse
|
8
|
Krajčová A, Skagen C, Džupa V, Urban T, Rustan AC, Jiroutková K, Bakalář B, Thoresen GH, Duška F. Effect of noradrenaline on propofol-induced mitochondrial dysfunction in human skeletal muscle cells. Intensive Care Med Exp 2022; 10:47. [DOI: 10.1186/s40635-022-00474-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Abstract
Background
Mitochondrial dysfunction is a hallmark of both critical illness and propofol infusion syndrome and its severity seems to be proportional to the doses of noradrenaline, which patients are receiving. We comprehensively studied the effects of noradrenaline on cellular bioenergetics and mitochondrial biology in human skeletal muscle cells with and without propofol-induced mitochondrial dysfunction.
Methods
Human skeletal muscle cells were isolated from vastus lateralis biopsies from patients undergoing elective hip replacement surgery (n = 14) or healthy volunteers (n = 4). After long-term (96 h) exposure to propofol (10 µg/mL), noradrenaline (100 µM), or both, energy metabolism was assessed by extracellular flux analysis and substrate oxidation assays using [14C] palmitic and [14C(U)] lactic acid. Mitochondrial membrane potential, morphology and reactive oxygen species production were analysed by confocal laser scanning microscopy. Mitochondrial mass was assessed both spectrophotometrically and by confocal laser scanning microscopy.
Results
Propofol moderately reduced mitochondrial mass and induced bioenergetic dysfunction, such as a reduction of maximum electron transfer chain capacity, ATP synthesis and profound inhibition of exogenous fatty acid oxidation. Noradrenaline exposure increased mitochondrial network size and turnover in both propofol treated and untreated cells as apparent from increased co-localization with lysosomes. After adjustment to mitochondrial mass, noradrenaline did not affect mitochondrial functional parameters in naïve cells, but it significantly reduced the degree of mitochondrial dysfunction induced by propofol co-exposure. The fatty acid oxidation capacity was restored almost completely by noradrenaline co-exposure, most likely due to restoration of the capacity to transfer long-chain fatty acid to mitochondria. Both propofol and noradrenaline reduced mitochondrial membrane potential and increased reactive oxygen species production, but their effects were not additive.
Conclusions
Noradrenaline prevents rather than aggravates propofol-induced impairment of mitochondrial functions in human skeletal muscle cells. Its effects on bioenergetic dysfunctions of other origins, such as sepsis, remain to be demonstrated.
Collapse
|
9
|
Singh A, Anjankar AP. Propofol-Related Infusion Syndrome: A Clinical Review. Cureus 2022; 14:e30383. [PMID: 36407194 PMCID: PMC9671386 DOI: 10.7759/cureus.30383] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023] Open
Abstract
Propofol-related infusion syndrome (PRIS) is a lethal condition characterized by multiple organ system failures. It can occur due to prolonged administration of propofol (an anesthetic) in mechanically intubated patients. The main presenting features of this condition include cardiovascular dysfunction with particular emphasis on impairment of cardiovascular contractility, metabolic acidosis, lactic acidosis, rhabdomyolysis, hyperkalaemia, lipidaemia, hepatomegaly, acute renal failure, and eventually mortality in most cases. The significant risk factors that predispose one to PRIS are: critical illnesses, increased serum catecholamines, steroid therapy, obesity, young age (significantly below three years), depleted carbohydrate stores in the body, increased serum lipids, and most importantly, heavy or extended dosage of propofol. The primary pathophysiology behind PRIS is the disruption of the mitochondrial respiratory chain that causes inhibition of adenosine triphosphate (ATP) synthesis and cellular hypoxia. Further, excess lipolysis of adipose tissue occurs, especially in critically ill patients where the energy source is lipid breakdown instead of carbohydrates. This process generates excess free fatty acids (FFAs) that cannot undergo adequate beta-oxidation. These FFAs contribute to the clinical pathology of PRIS. It requires prompt management as it is a fatal condition. The clinicians must observe the patient's electrocardiogram (ECG), serum creatine kinase, lipase, amylase, lactate, liver enzymes, and myoglobin levels in urine, under propofol sedation. Doctors should immediately stop propofol infusion upon noticing any abnormality in these parameters. The other essentials of management of various manifestations of PRIS will be discussed in this article, along with a detailed explanation of the condition, its risk factors, diagnosis, pathophysiology, and presenting features. This article aims to make clinicians more aware of the occurrence of this syndrome so that better ways to manage and treat this condition can be formulated in the future.
Collapse
Affiliation(s)
- Aayushi Singh
- Anesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Ashish P Anjankar
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Wardha, IND
| |
Collapse
|
10
|
Liang Y, Huang Y, Shao R, Xiao F, Lin F, Dai H, Pan L. Propofol produces neurotoxicity by inducing mitochondrial apoptosis. Exp Ther Med 2022; 24:630. [PMID: 36160898 PMCID: PMC9468839 DOI: 10.3892/etm.2022.11567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Propofol is a fast and short-acting intravenous anesthetic widely used in clinical anesthesia and intensive care unit sedation. However, its use can cause abnormal effects on the central nervous system. Thus, the purpose of this study was to investigate the mechanism of propofol on primary hippocampal neuron injury. In addition, we aimed to determine whether a correlation exists between propofol and mitochondrial apoptosis-induced neurotoxicity. Hippocampal neurons cultured for 4 days were exposed to different drugs. The treatment groups were divided according to drug exposure into propofol, a rotenone inhibitor, and a coenzyme Q10 agonist groups. The final concentrations of propofol were 1, 10 and 100 µM. The content of ATP and reactive oxygen species (ROS) in the neurons of each group were detected using commercial kits in the culture supernatant after 3 h of drug exposure. Western blotting was used to analyze the expression of apoptosis-related proteins. The JC-1 kit was used to detect the mitochondrial membrane potential. The results revealed that, compared with the non-propofol treatment groups, the expression of apoptosis-related proteins, ATP content, and mitochondrial membrane potential were significantly decreased while the ROS content was markedly increased in the propofol treatment group. In conclusion, propofol treatment promoted damage to hippocampal neuronal mitochondria in a dose-dependent manner. This damage may lead to neuronal apoptosis and neurotoxicity by inducing the inhibition of mitochondrial respiratory chain complex I.
Collapse
Affiliation(s)
- Yubing Liang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yu Huang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rongge Shao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
11
|
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
|
12
|
Shrestha R, Johnson E, Byrne FL. Exploring the therapeutic potential of mitochondrial uncouplers in cancer. Mol Metab 2021; 51:101222. [PMID: 33781939 PMCID: PMC8129951 DOI: 10.1016/j.molmet.2021.101222] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mitochondrial uncouplers are well-known for their ability to treat a myriad of metabolic diseases, including obesity and fatty liver diseases. However, for many years now, mitochondrial uncouplers have also been evaluated in diverse models of cancer in vitro and in vivo. Furthermore, some mitochondrial uncouplers are now in clinical trials for cancer, although none have yet been approved for the treatment of cancer. SCOPE OF REVIEW In this review we summarise published studies in which mitochondrial uncouplers have been investigated as an anti-cancer therapy in preclinical models. In many cases, mitochondrial uncouplers show strong anti-cancer effects both as single agents, and in combination therapies, and some are more toxic to cancer cells than normal cells. Furthermore, the mitochondrial uncoupling mechanism of action in cancer cells has been described in detail, with consistencies and inconsistencies between different structural classes of uncouplers. For example, many mitochondrial uncouplers decrease ATP levels and disrupt key metabolic signalling pathways such as AMPK/mTOR but have different effects on reactive oxygen species (ROS) production. Many of these effects oppose aberrant phenotypes common in cancer cells that ultimately result in cell death. We also highlight several gaps in knowledge that need to be addressed before we have a clear direction and strategy for applying mitochondrial uncouplers as anti-cancer agents. MAJOR CONCLUSIONS There is a large body of evidence supporting the therapeutic use of mitochondrial uncouplers to treat cancer. However, the long-term safety of some uncouplers remains in question and it will be critical to identify which patients and cancer types would benefit most from these agents.
Collapse
Affiliation(s)
- Riya Shrestha
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia
| | - Edward Johnson
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia
| | - Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, 2052, Australia.
| |
Collapse
|
13
|
Dominguini D, Steckert AV, Michels M, Spies MB, Ritter C, Barichello T, Thompson J, Dal-Pizzol F. The effects of anaesthetics and sedatives on brain inflammation. Neurosci Biobehav Rev 2021; 127:504-513. [PMID: 33992694 DOI: 10.1016/j.neubiorev.2021.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/27/2021] [Accepted: 05/09/2021] [Indexed: 12/17/2022]
Abstract
Microglia are involved in many dynamic processes in the central nervous system (CNS) including the development of inflammatory processes and neuromodulation. Several sedative, analgesic or anaesthetic drugs, such as opioids, ∝2-adrenergic agonists, ketamine, benzodiazepines and propofol can cause both neuroprotective and harmful effects on the brain. The purpose of this review is to present the main findings on the use of these drugs and the mechanisms involved in microglial activation. Alpha 2-adrenergic agonists, propofol and benzodiazepines have several pro- or anti-inflammatory effects on microglia. Long-term use of benzodiazepines and propofol causes neuroapoptotic effects and α2-adrenergic agonists may attenuate these effects. Conversely, morphine and fentanyl may have proinflammatory effects, causing behavioural changes in patients and changes in cell viability in vitro. Conversely, chronic administration of morphine induces CCL5 chemokine expression in microglial cells that promotes their survival.
Collapse
Affiliation(s)
- Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Amanda V Steckert
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Mariana B Spies
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cristiane Ritter
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Jonathan Thompson
- Department of Cardiovascular Sciences, Anaesthesia Critical Care and Pain Management Group, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
14
|
Hau V, Bonilla-Velez J, Low D. Anesthesia management for ENT surgery in a child with X-linked pyruvate kinase deficiency. Paediatr Anaesth 2021; 31:499-500. [PMID: 33295086 DOI: 10.1111/pan.14105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 11/29/2022]
Abstract
Pyruvate dehydrogenase complex deficiency (PDCD) is a rare X-linked disorder that affects glucose metabolism. There are several case reports describing the anesthetic management of patients with PDCD. This is the first case report to describe the anesthetic management of a PDCD patient requiring spontaneous ventilation using dexmedetomidine and ketamine.
Collapse
Affiliation(s)
- Vickie Hau
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | | | - Daniel Low
- Division of Anesthesiology, Seattle Children's Hospital, Seattle, WA, USA
| |
Collapse
|
15
|
Atkins R, Constantin-Teodosiu D, Varadhan KK, Constantin D, Lobo DN, Greenhaff PL. Major elective abdominal surgery acutely impairs lower limb muscle pyruvate dehydrogenase complex activity and mitochondrial function. Clin Nutr 2021; 40:1046-1051. [PMID: 32711950 PMCID: PMC7957361 DOI: 10.1016/j.clnu.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND & AIMS This post hoc study aimed to determine whether major elective abdominal surgery had any acute impact on mitochondrial pyruvate dehydrogenase complex (PDC) activity and maximal mitochondrial ATP production rates (MAPR) in a large muscle group (vastus lateralis -VL) distant to the site of surgical trauma. METHODS Fifteen patients undergoing major elective open abdominal surgery were studied. Muscle biopsies were obtained after the induction of anesthesia from the VL immediately before and after surgery for the determination of PDC and maximal MAPR (utilizing a variety of energy substrates). RESULTS Muscle PDC activity was reduced by >50% at the end of surgery compared with pre-surgery (p < 0.05). Muscle MAPR were comprehensively suppressed by surgery for the substrate combinations: glutamate + succinate; glutamate + malate; palmitoylcarnitine + malate; and pyruvate + malate (all p < 0.05), and could not be explained by a lower mitochondrial yield. CONCLUSIONS PDC activity and mitochondrial ATP production capacity were acutely impaired in muscle distant to the site of surgical trauma. In keeping with the limited data available, we surmise these events resulted from the general anesthesia procedures employed and the surgery related trauma. These findings further the understanding of the acute dysregulation of mitochondrial function in muscle distant to the site of major surgical trauma in patients, and point to the combination of general anesthesia and trauma related inflammation as being drivers of muscle metabolic insult that warrants further investigation. CLINICAL TRIAL REGISTRATION Registered at (NCT01134809).
Collapse
Affiliation(s)
- Ryan Atkins
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Dumitru Constantin-Teodosiu
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Krishna K Varadhan
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Despina Constantin
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Dileep N Lobo
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK; Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK; National Institute of Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Paul L Greenhaff
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK; National Institute of Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
16
|
McCann MR, George De la Rosa MV, Rosania GR, Stringer KA. L-Carnitine and Acylcarnitines: Mitochondrial Biomarkers for Precision Medicine. Metabolites 2021; 11:51. [PMID: 33466750 PMCID: PMC7829830 DOI: 10.3390/metabo11010051] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Biomarker discovery and implementation are at the forefront of the precision medicine movement. Modern advances in the field of metabolomics afford the opportunity to readily identify new metabolite biomarkers across a wide array of disciplines. Many of the metabolites are derived from or directly reflective of mitochondrial metabolism. L-carnitine and acylcarnitines are established mitochondrial biomarkers used to screen neonates for a series of genetic disorders affecting fatty acid oxidation, known as the inborn errors of metabolism. However, L-carnitine and acylcarnitines are not routinely measured beyond this screening, despite the growing evidence that shows their clinical utility outside of these disorders. Measurements of the carnitine pool have been used to identify the disease and prognosticate mortality among disorders such as diabetes, sepsis, cancer, and heart failure, as well as identify subjects experiencing adverse drug reactions from various medications like valproic acid, clofazimine, zidovudine, cisplatin, propofol, and cyclosporine. The aim of this review is to collect and interpret the literature evidence supporting the clinical biomarker application of L-carnitine and acylcarnitines. Further study of these metabolites could ultimately provide mechanistic insights that guide therapeutic decisions and elucidate new pharmacologic targets.
Collapse
Affiliation(s)
- Marc R. McCann
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Mery Vet George De la Rosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Kathleen A. Stringer
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
17
|
Urban T, Waldauf P, Krajčová A, Jiroutková K, Halačová M, Džupa V, Janoušek L, Pokorná E, Duška F. Kinetic characteristics of propofol-induced inhibition of electron-transfer chain and fatty acid oxidation in human and rodent skeletal and cardiac muscles. PLoS One 2019; 14:e0217254. [PMID: 31584947 PMCID: PMC6777831 DOI: 10.1371/journal.pone.0217254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/20/2019] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Propofol causes a profound inhibition of fatty acid oxidation and reduces spare electron transfer chain capacity in a range of human and rodent cells and tissues-a feature that might be related to the pathogenesis of Propofol Infusion Syndrome. We aimed to explore the mechanism of propofol-induced alteration of bioenergetic pathways by describing its kinetic characteristics. METHODS We obtained samples of skeletal and cardiac muscle from Wistar rat (n = 3) and human subjects: vastus lateralis from hip surgery patients (n = 11) and myocardium from brain-dead organ donors (n = 10). We assessed mitochondrial functional indices using standard SUIT protocol and high resolution respirometry in fresh tissue homogenates with or without short-term exposure to a range of propofol concentration (2.5-100 μg/ml). After finding concentrations of propofol causing partial inhibition of a particular pathways, we used that concentration to construct kinetic curves by plotting oxygen flux against substrate concentration during its stepwise titration in the presence or absence of propofol. By spectrophotometry we also measured the influence of the same propofol concentrations on the activity of isolated respiratory complexes. RESULTS We found that human muscle and cardiac tissues are more sensitive to propofol-mediated inhibition of bioenergetic pathways than rat's tissue. In human homogenates, palmitoyl carnitine-driven respiration was inhibited at much lower concentrations of propofol than that required for a reduction of electron transfer chain capacity, suggesting FAO inhibition mechanism different from downstream limitation or carnitine-palmitoyl transferase-1 inhibition. Inhibition of Complex I was characterised by more marked reduction of Vmax, in keeping with non-competitive nature of the inhibition and the pattern was similar to the inhibition of Complex II or electron transfer chain capacity. There was neither inhibition of Complex IV nor increased leak through inner mitochondrial membrane with up to 100 μg/ml of propofol. If measured in isolation by spectrophotometry, propofol 10 μg/ml did not affect the activity of any respiratory complexes. CONCLUSION In human skeletal and heart muscle homogenates, propofol in concentrations that are achieved in propofol-anaesthetized patients, causes a direct inhibition of fatty acid oxidation, in addition to inhibiting flux of electrons through inner mitochondrial membrane. The inhibition is more marked in human as compared to rodent tissues.
Collapse
Affiliation(s)
- Tomáš Urban
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Petr Waldauf
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Adéla Krajčová
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Kateřina Jiroutková
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Milada Halačová
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Valér Džupa
- Department of Orthopaedics and Traumatology, Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Libor Janoušek
- Transplantation Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Pokorná
- Department of Organ Recovery and Transplantation Databases, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - František Duška
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| |
Collapse
|
18
|
Murakami Y, Ueki R, Tachikawa T, Hirose M. The Basic Study of the Mechanism of Propofol-Related Infusion Syndrome Using a Murine Skeletal Muscle Injury Model. Anesth Pain Med 2019; 9:e89417. [PMID: 31497518 PMCID: PMC6712282 DOI: 10.5812/aapm.89417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/05/2019] [Accepted: 04/07/2019] [Indexed: 12/12/2022] Open
Abstract
Background The pathophysiological mechanism of propofol-related infusion syndrome (PRIS) is believed to be due to the injury to the mitochondrial electron transport chain and the resultant metabolic disorders that are caused by both propofol agents and the lipid solvent. However, the mechanisms and causative factors of PRIS have not been fully elucidated. Objectives The aim of this study was to evaluate the possibility of a research model using the culture of differentiated C2C12 cells for fundamental research of PRIS. Methods First, differentiated C2C12 cells were cultured accompanied by several concentrations of chemical reagents of 2,6-diisopropylphenol (2,6 DIP) or dimethyl sulfoxide (DMSO) for 60 hours and the cell death rate was examined by trypan blue staining. Second, The cells were incubated with a commercially available propofol reagent or lipid reagent for 48 hours. The supernatant fluid of the cell culture medium was gathered and the numbers of floating cells were measured by cell counter. To investigate the mitochondrial disorder by the propofol preparation, JC-1, an experiment using fluorescent reagent, was performed for the 48 hours with 100 µg/mL propofol incubation. Results The rate of cell death was increased with elevating concentrations both of chemical reagents of 2,6 DIP group and dimethyl sulfoxide group. The rates of cell death were significantly higher in the 2,6 DIP group than DMSO group. The numbers of floating cells were increased with elevating concentrations both commercially available propofol reagent and lipid reagent groups. The decreased red/green fluorescence ratio by JC-1 staining in the propofol 100µg/mL group proved an attenuated mitochondrial membrane potential. Conclusions The dose-dependent cell damage induced by the propofol reagents and a lipid solvent may provide a proposed model as a basic experimental model for further investigations into PRIS.
Collapse
Affiliation(s)
- Yuryo Murakami
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ryusuke Ueki
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
- Corresponding Author: MD, Ph.D. Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo 663-8501, Japan. Tel: 81-798456392, Fax: 81-798456393,
| | - Taihei Tachikawa
- Department of Anesthesiology, Meiwa Hospital, Nishinomiya, Japan
| | - Munetaka Hirose
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
19
|
Lipid emulsion, but not propofol, induces skeletal muscle damage and lipid peroxidation. J Anesth 2019; 33:628-635. [PMID: 31473808 PMCID: PMC6874638 DOI: 10.1007/s00540-019-02676-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/22/2019] [Indexed: 11/06/2022]
Abstract
Purpose Prolonged propofol infusion induces skeletal muscle damage. However, it is well known that the lipid emulsion that is the solvent of propofol causes various types of tissue damage via lipid peroxidation, and that propofol, conversely, has an anti-lipid peroxidative effect. The purpose of this study was to determine whether propofol or the lipid emulsion is the cause of muscle damage following prolonged administration. Methods Rats were divided into four groups: NI group (no intervention), Cath group (venous catheter insertion only), Prop group (1% propofol (Maruishi) intravenous infusion at 10 mg/kg/h), and Lipid group (10% Lipofundin® intravenous infusion at 100 mg/kg/h) (n = 10, each group). 1% Propofol (Maruishi) or Lipofundin was infused at 1 mL/kg/h for 72 h. The solvent of 1% propofol (Maruishi) is a 10% lipid emulsion. Lipofundin consists of 50% long-chain triacylglycerols and 50% medium-chain triacylglycerols, similar to the propofol solvent. Plasma concentrations of creatine kinase and myoglobin, superoxide production level, and 4-hydroxynonenal and malondialdehyde expression in the gastrocnemius muscle were evaluated 72 h after the interventions. Results Plasma concentrations of creatine kinase and myoglobin in the Lipid group were significantly higher than those in the other three groups. The superoxide production level, and 4-hydroxynonenal and malondialdehyde expression in the Lipid group were also significantly higher than in the other three groups. Conclusion Lipofundin induces skeletal muscle damage via lipid peroxidation, and 1% propofol (Maruishi) conversely suppresses the muscle damage via antioxidant effects.
Collapse
|
20
|
Abstract
OBJECTIVES Propofol may adversely affect the function of mitochondria and the clinical features of propofol infusion syndrome suggest that this may be linked to propofol-related bioenergetic failure. We aimed to assess the effect of therapeutic propofol concentrations on energy metabolism in human skeletal muscle cells. DESIGN In vitro study on human skeletal muscle cells. SETTINGS University research laboratories. SUBJECTS Patients undergoing hip surgery and healthy volunteers. INTERVENTIONS Vastus lateralis biopsies were processed to obtain cultured myotubes, which were exposed to a range of 1-10 μg/mL propofol for 96 hours. MEASUREMENTS AND MAIN RESULTS Extracellular flux analysis was used to measure global mitochondrial functional indices, glycolysis, fatty acid oxidation, and the functional capacities of individual complexes of electron transfer chain. In addition, we used [1-C]palmitate to measure fatty acid oxidation and spectrophotometry to assess activities of individual electron transfer chain complexes II-IV. Although cell survival and basal oxygen consumption rate were only affected by 10 μg/mL of propofol, concentrations as low as 1 μg/mL reduced spare electron transfer chain capacity. Uncoupling effects of propofol were mild, and not dependent on concentration. There was no inhibition of any respiratory complexes with low dose propofol, but we found a profound inhibition of fatty acid oxidation. Addition of extra fatty acids into the media counteracted the propofol effects on electron transfer chain, suggesting inhibition of fatty acid oxidation as the causative mechanism of reduced spare electron transfer chain capacity. Whether these metabolic in vitro changes are observable in other organs and at the whole-body level remains to be investigated. CONCLUSIONS Concentrations of propofol seen in plasma of sedated patients in ICU cause a significant inhibition of fatty acid oxidation in human skeletal muscle cells and reduce spare capacity of electron transfer chain in mitochondria.
Collapse
|
21
|
Herminghaus A, Buitenhuis AJ, Schulz J, Vollmer C, Scheeren TWL, Bauer I, Picker O, Truse R. Propofol improves colonic but impairs hepatic mitochondrial function in tissue homogenates from healthy rats. Eur J Pharmacol 2019; 853:364-370. [PMID: 31009637 DOI: 10.1016/j.ejphar.2019.04.031] [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: 02/19/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 11/24/2022]
Abstract
Evidence suggests that propofol infusion syndrome (PRIS) is caused by an altered mitochondrial function. The aim of this study was to examine the effects of propofol and the vehicle MCT on mitochondrial function in hepatic and colonic tissue. Mitochondrial oxygen consumption was determined in colon and liver homogenates after incubation with buffer (control), propofol (50, 75, 100, 500 μM) or the carrier substances DMSO and MCT. State 2 (substrate-dependent) and state 3 (ADP-dependent respiration) were assessed. RCI (respiratory control index) - an indicator for coupling between electron transport chain system (ETS) and oxidative phosphorylation (OXPHOS) and ADP/O ratio - a parameter for efficacy of OXPHOS were calculated. Data were presented as % of control. In hepatic mitochondria, 500 μM propofol reduced RCI formulation-independently (propofol/MCT 500 μM: complex I: 66.3 ± 8.7%*, complex II: 75.5 ± 9.2%*; propofol/DMSO 500 μM: complex I: 29.1 ± 8.8%*, complex II: 49.3 ± 15.5%*). 75 μM Propofol/MCT reduced ADP/O for complex I (73.5 ± 27.3%*). DMSO did not affect hepatic mitochondria whereas MCT reduced RCI for complex II (87.2 ± 9.8%*) and ADP/O for complex I (93.7 ± 31.7%*). In colon 50 μM Propofol/MCT increased RCI for complex I and II (complex I: 127.2 ± 10.7%*, complex II: 136.8 ± 33.9%*) and 100 μM Propofol/MCT for complex I (131.4 ± 18.7%*). 500 μM Propofol/DMSO increased ADP/O for complex I (139.4 ± 41.4%*). DMSO did not affect RCI but increased ADP/O for both complexes (complex I: 119.9 ± 25.8%*, complex II: 110.2 ± 14.2%*). MCT increased RCI for complex I (123.0 ± 31.6%*). In hepatic mitochondria propofol uncoupled ETS from OXPHOS formulation-independently and propofol/MCT reduced efficacy of OXPHOS. In colonic mitochondria, propofol/MCT strengthened the coupling and propofol/DMSO enhanced the efficacy of OXPHOS.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - A Johannes Buitenhuis
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Jan Schulz
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Christian Vollmer
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Thomas W L Scheeren
- Department of Anaesthesiology, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, the Netherlands.
| | - Inge Bauer
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Olaf Picker
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Richard Truse
- Department of Anaesthesiology, University of Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
22
|
Salik I, Kubal K, Barst S. Anesthetic Implications of a Patient With Kniest Dysplasia and Mitochondrial Disease: A Case Report. A A Pract 2019; 12:267-269. [PMID: 30285969 DOI: 10.1213/xaa.0000000000000906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Kniest dysplasia, or metatropic dysplasia type II, is a rare chondrodysplasia caused by abnormal type II collagen. Clinically, it is characterized by dwarfism, deafness, skeletal derangements, and ocular abnormalities. Mitochondrial diseases produce a spectrum of abnormalities in affected individuals and predominantly impact organs of high energy utilization, including the brain, skeletal muscles, kidneys, and liver. We present the case of a 6-year-old boy with both Kniest dysplasia and underlying mitochondrial disease for examination under anesthesia before cataract surgery. Successful anesthetic management of a patient with Kniest dysplasia and a mitochondrial myopathy is discussed.
Collapse
Affiliation(s)
- Irim Salik
- From the Department of Pediatric Anesthesiology at Westchester Medical Center, New York Medical College, Valhalla, New York
| | | | | |
Collapse
|
23
|
Propofol infusion syndrome: a structured literature review and analysis of published case reports. Br J Anaesth 2019; 122:448-459. [PMID: 30857601 DOI: 10.1016/j.bja.2018.12.025] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/26/2018] [Accepted: 12/20/2018] [Indexed: 12/31/2022] Open
Abstract
Propofol infusion syndrome is a rare, potentially fatal condition first described in children in the 1990s and later reported in adults. We provide a narrative review of what is currently known about propofol infusion syndrome, including a structured analysis of all published case reports; child and adult cases were analysed separately as propofol is no longer used for long-term sedation in children. The review contains an update on current knowledge of the pathophysiology of this condition along with recommendations for its diagnosis, prevention, and management. We reviewed 108 publications documenting 168 cases of propofol infusion syndrome. We evaluated clinical features and analysed factors influencing mortality in child and adult cases using separate multivariate analysis models. We used separate multiple linear regression models to analyse relationships between cumulative dose of propofol and the number of features seen and organ systems involved. Lipidaemia, fever, and hepatomegaly occurred more frequently in children than in adults, whilst rhabdomyolysis and hyperkalaemia were more frequent in adults. Mortality from propofol infusion syndrome is independently associated with fever and hepatomegaly in children, and electrocardiogram changes, hypotension, hyperkalaemia, traumatic brain injury, and a mean propofol infusion rate >5 mg kg-1 h-1 in adults. The cumulative dose of propofol was associated with an increased number of clinical features and the number of organ systems involved in adult cases only. Clinicians should consider propofol infusion syndrome in cases of unexplained metabolic acidosis, ECG changes, and rhabdomyolysis. We recommend early consideration of continuous haemofiltration in the management of propofol infusion syndrome.
Collapse
|
24
|
Multidisciplinary Perioperative Care for Children with Neuromuscular Disorders. CHILDREN-BASEL 2018; 5:children5090126. [PMID: 30213079 PMCID: PMC6162674 DOI: 10.3390/children5090126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022]
Abstract
Children with neuromuscular diseases present unique challenges to providing safe and appropriate perioperative care. Given the spectrum of disease etiologies and manifestations, this is a population that often requires specialized multidisciplinary care from pediatricians, geneticists, neurologists, dieticians, and pulmonologists which must also be coordinated with surgeons and anesthesiologists when these patients present for surgery. Several of these diseases also have specific pharmacologic implications for anesthesia, most notably mitochondrial disease and muscular dystrophies, which put them at additional risk during the perioperative period particularly in patients presenting without a formal diagnosis. Techniques and strategies to fully evaluate and optimize these patients preoperatively, manage them safely intraoperatively, and return them to their baseline status postoperative are particularly important in this vulnerable group of patients. Utilizing a review of inherited neuromuscular conditions, generalized perioperative concerns, and specific complications related to anesthesia, this article provides an overview of pertinent considerations and recommends a framework for management of these patients.
Collapse
|
25
|
Sumi C, Okamoto A, Tanaka H, Nishi K, Kusunoki M, Shoji T, Uba T, Matsuo Y, Adachi T, Hayashi JI, Takenaga K, Hirota K. Propofol induces a metabolic switch to glycolysis and cell death in a mitochondrial electron transport chain-dependent manner. PLoS One 2018; 13:e0192796. [PMID: 29447230 PMCID: PMC5813975 DOI: 10.1371/journal.pone.0192796] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/30/2018] [Indexed: 12/14/2022] Open
Abstract
The intravenous anesthetic propofol (2,6-diisopropylphenol) has been used for the induction and maintenance of anesthesia and sedation in critical patient care. However, the rare but severe complication propofol infusion syndrome (PRIS) can occur, especially in patients receiving high doses of propofol for prolonged periods. In vivo and in vitro evidence suggests that the propofol toxicity is related to the impaired mitochondrial function. However, underlying molecular mechanisms remain unknown. Therefore, we investigated effects of propofol on cell metabolism and death using a series of established cell lines of various origins, including neurons, myocytes, and trans-mitochondrial cybrids, with defined mitochondrial DNA deficits. We demonstrated that supraclinical concentrations of propofol in not less than 50 μM disturbed the mitochondrial function and induced a metabolic switch, from oxidative phosphorylation to glycolysis, by targeting mitochondrial complexes I, II and III. This disturbance in mitochondrial electron transport caused the generation of reactive oxygen species, resulting in apoptosis. We also found that a predisposition to mitochondrial dysfunction, caused by a genetic mutation or pharmacological suppression of the electron transport chain by biguanides such as metformin and phenformin, promoted propofol-induced caspase activation and cell death induced by clinical relevant concentrations of propofol in not more than 25 μM. With further experiments with appropriate in vivo model, it is possible that the processes to constitute the molecular basis of PRIS are identified.
Collapse
Affiliation(s)
- Chisato Sumi
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Akihisa Okamoto
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Hiromasa Tanaka
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Kenichiro Nishi
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Munenori Kusunoki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Tomohiro Shoji
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takeo Uba
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takehiko Adachi
- Department of Anesthesiology, Tazuke Kofukai Medical Institute Kitano Hospital, Osaka, Japan
| | | | - Keizo Takenaga
- Department of Life Science, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
- * E-mail:
| |
Collapse
|
26
|
Félix LM, Correia F, Pinto PA, Campos SP, Fernandes T, Videira R, Oliveira M, Peixoto FP, Antunes LM. Propofol affinity to mitochondrial membranes does not alter mitochondrial function. Eur J Pharmacol 2017; 803:48-56. [DOI: 10.1016/j.ejphar.2017.03.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 01/11/2023]
|
27
|
Lim KHH, Halestrap AP, Angelini GD, Suleiman MS. Propofol Is Cardioprotective in a Clinically Relevant Model of Normothermic Blood Cardioplegic Arrest and Cardiopulmonary Bypass. Exp Biol Med (Maywood) 2016; 230:413-20. [PMID: 15956771 DOI: 10.1177/15353702-0323006-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The general anesthetic propofol has been shown to be cardioprotective. However, its benefits when used in cardioplegia during cardiac surgery have not been demonstrated. In this study, we investigated the effects of propofol on metabolic stress, cardiac function, and injury in a clinically relevant model of normothermic cardioplegic arrest and cardiopulmonary bypass. Twenty anesthetized pigs, randomized to propofol treatment ( n = 8) and control ( n =12) groups, were surgically prepared for cardiopulmonary bypass (CPB) and cardioplegic arrest. Doses of warm blood cardioplegia were delivered at 15-min intervals during a 60-min aortic cross-clamped period. Propofol was continuously infused for the duration of CPB and was therefore present in blood cardioplegia. Myocardial biopsies were collected before, at the end of cardioplegic arrest, and 20 mins after the release of the aortic cross-clamp. Hemodynamic parameters were monitored and blood samples collected for cardiac troponin I measurements. Propofol infusion during CPB and before ischemia did not alter cardiac function or myocardial metabolism. Propofol treatment attenuated the changes in myocardial tissue levels of adenine nucleotides, lactate, and amino acids during ischemia and reduced cardiac troponin I release on reperfusion. Propofol treatment reduced measurable hemodynamic dysfunction after cardioplegic arrest when compared to untreated controls. In conclusion, propofol protects the heart from ischemia-reperfusion injury in a clinically relevant experimental model. Propofol may therefore be a useful adjunct to cardioplegic solutions as well as being an appropriate anesthetic for cardiac surgery.
Collapse
Affiliation(s)
- Kelvin H H Lim
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol BS2 8HW, United Kingdom
| | | | | | | |
Collapse
|
28
|
Finsterer J, Frank M. Propofol Is Mitochondrion-Toxic and May Unmask a Mitochondrial Disorder. J Child Neurol 2016; 31:1489-1494. [PMID: 27488955 DOI: 10.1177/0883073816661458] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/05/2016] [Indexed: 12/17/2022]
Abstract
There are indications that preexisting mitochondrial disorders or beta-oxidation defects predispose for propofol infusion syndrome. This review aimed at investigating if propofol infusion syndrome occurs exclusively in patients with mitochondrial disorder and if propofol can unmask a mitochondrial disorder. Propofol infusion syndrome has been reported in genetically confirmed mitochondrial disorder patients. In addition, muscle biopsy of patients with propofol infusion syndrome revealed complex IV or complex II deficiency. In animal studies propofol disrupted the electron flow along the respiratory chain and decreased complex I, complex II, and complex III of the respiratory chain. In addition, propofol disrupted the permeability transition pore and reduced the mitochondrial membrane potential. In conclusion, propofol is mitochondrion-toxic and mitochondrial disorder patients should not receive propofol in high dosages over a prolonged period of time. Short-term application of propofol should be safe even in mitochondrial disorder patients. Not only does propofol infusion syndrome occur in mitochondrial disorder patients, but mitochondrial disorder patients are likely at higher risk to develop propofol infusion syndrome. Patients who develop propofol infusion syndrome should be screened for mitochondrial disorder. Propofol infusion syndrome is preventable if risk factors are thoroughly assessed, and if long-term propofol is avoided in patients at risk for propofol infusion syndrome.
Collapse
Affiliation(s)
| | - Marlies Frank
- First Medical Department, Krankenanstalt Rudolfstiftung, Vienna, Austria
| |
Collapse
|
29
|
Campos S, Félix L, Venâncio C, de Lurdes Pinto M, Peixoto F, de Pinho PG, Antunes L. In vivo study of hepatic oxidative stress and mitochondrial function in rabbits with severe hypotension after propofol prolonged infusion. SPRINGERPLUS 2016; 5:1349. [PMID: 27588242 PMCID: PMC4987748 DOI: 10.1186/s40064-016-2970-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/29/2016] [Indexed: 11/12/2022]
Abstract
In humans, prolonged sedations with propofol or using high doses have been associated with propofol infusion syndrome. The main objective of this study was to evaluate the effects of prolonged high-dose administration of a specific propofol emulsion (Propofol Lipuro) and an improved lipid formulation (SMOFlipid) in liver mitochondrial bioenergetics and oxidative stress of rabbits, comparatively to a saline control. Twenty-one male New Zealand white rabbits were randomly allocated in three groups that were continuously treated for 20 h. Each group of seven animals received separately: NaCl 0.9 % (saline), SMOFlipid (lipid-based emulsion without propofol) and Lipuro 2 % (propofol lipid emulsion). An intravenous propofol bolus of 20 mg kg−1 was given to the propofol Lipuro group to allow blind orotracheal intubation and mechanical ventilation. Anesthesia was maintained using infusion rates of: 20, 30, 40, 50 and 60 mg kg−1 h−1, according to the clinical scale of anesthetic depth and the index of consciousness values. The SMOFlipid and saline groups received the same infusion rate as the propofol Lipuro group, which were infused during 20 consecutive hours. At the end, the animals were euthanized, livers collected and mitochondria isolated by standard differential centrifugation. Mitochondrial respiration, membrane potential, swelling and oxidative stress were evaluated. Data were processed using one-way ANOVA (p < 0.05). The animals revealed a significant decrease in cardiovascular parameters showing bradycardia and severe hypotension. No statistical differences were observed when using pyruvate as substrate, however, when using succinate as respiratory substrate, significant decrease in ADP-stimulated respiration rate was observed for SMOFlipid group (p = 0.002). Lipid peroxides (p < 0.01) and protein carbonyls (p = 0.01) showed a statistically significant difference between propofol Lipuro and the SMOFlipid groups. These results suggest that lipid-based emulsions can be involved in the regulation of different pathways that ultimately lead to a decrease of state 3 mitochondrial respiration rate. The infusion of propofol Lipuro during prolonged periods, in addition to marked hypotension and hypoperfusion, also showed to have higher anti-oxidant activity and lower impairment of the mitochondrial function comparatively to the improved lipid formulation, SMOFlipid, using the rabbit as animal model.
Collapse
Affiliation(s)
- Sónia Campos
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Veterinary Sciences Department, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, Apartado 1013, 5001-801 Vila Real, Portugal ; Institute for Research and Innovation in Health (i3S), Laboratory Animal Science, Institute of Molecular and Cell Biology (IBMC), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal ; UCIBIO@REQUIMTE-Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Luís Félix
- Institute for Research and Innovation in Health (i3S), Laboratory Animal Science, Institute of Molecular and Cell Biology (IBMC), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal ; Life Sciences and Environment School (ECVA), Department of Chemistry, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Carlos Venâncio
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Veterinary Sciences Department, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, Apartado 1013, 5001-801 Vila Real, Portugal
| | - Maria de Lurdes Pinto
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Veterinary Sciences Department, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, Apartado 1013, 5001-801 Vila Real, Portugal
| | - Francisco Peixoto
- Life Sciences and Environment School (ECVA), Department of Chemistry, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Paula Guedes de Pinho
- UCIBIO@REQUIMTE-Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Luís Antunes
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Veterinary Sciences Department, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, Apartado 1013, 5001-801 Vila Real, Portugal ; Institute for Research and Innovation in Health (i3S), Laboratory Animal Science, Institute of Molecular and Cell Biology (IBMC), University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
30
|
Madathil RJ, Hira RS, Stoeckl M, Sterz F, Elrod JB, Nichol G. Ischemia reperfusion injury as a modifiable therapeutic target for cardioprotection or neuroprotection in patients undergoing cardiopulmonary resuscitation. Resuscitation 2016; 105:85-91. [PMID: 27131843 DOI: 10.1016/j.resuscitation.2016.04.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/07/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
AIMS We sought to review cellular changes that occur with reperfusion to try to understand whether ischemia-reperfusion injury (RI) is a potentially modifiable therapeutic target for cardioprotection or neuroprotection in patients undergoing cardiopulmonary resuscitation. DATA SOURCES Articles written in English and published in PubMed. RESULTS Remote ischemic conditioning (RIC) involves brief episodes of non-lethal ischemia and reperfusion applied to an organ or limb distal to the heart and brain. Induction of hypothermia involves cooling an ischemic organ or body. Both have pluripotent effects that reduce the potential harm associated with RI in the heart and brain by reduced opening of the mitochondrial permeability transition pore. Recent trials of RIC and induced hypothermia did not demonstrate these treatments to be effective. Assessment of the effect of these interventions in humans to date may have been modified by use of concurrent medications including propofol. CONCLUSIONS Ongoing research is necessary to assess whether reduction of RI improves patient outcomes.
Collapse
Affiliation(s)
| | - Ravi S Hira
- University of Washington, Seattle, WA, United States
| | | | - Fritz Sterz
- Medical University of Vienna, Vienna, Austria
| | | | - Graham Nichol
- University of Washington, Seattle, WA, United States.
| |
Collapse
|
31
|
Lemoine S, Zhu L, Gress S, Gérard JL, Allouche S, Hanouz JL. Mitochondrial involvement in propofol-induced cardioprotection: An in vitro study in human myocardium. Exp Biol Med (Maywood) 2016; 241:527-38. [PMID: 26748397 DOI: 10.1177/1535370215622586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/17/2015] [Indexed: 01/12/2023] Open
Abstract
Propofol has been shown to exert cardioprotection, but the underlying mechanisms remain incompletely understood. We examined: (1) whether propofol-induced cardioprotection depended on the time and the dose of administration; (2) the role of mitochondrial adenosine triphosphate-sensitive potassium channels, nitric oxide synthase, and mitochondrial respiratory chain activity in propofol-induced cardioprotection. Human right atrial trabeculae were obtained during cardiopulmonary bypass for coronary artery bypass and aortic valve replacement. Isometric force of contraction of human right atrial trabeculae hanged in an oxygenated Tyrode's solution was recorded during 30-min hypoxia and 60-min reoxygenation (Control). Propofol 0.1, 1, and 10 µM was administered: (1) 5 min before hypoxia until the end of the experiment; (2) 5 min followed by 5-min washout before hypoxia; (3) during the reoxygenation period, propofol 10 µM was administered in presence of 5-hydroxydecanoate (antagonist of mitochondrial adenosine triphosphate-sensitive potassium channels), and NG-nitro-L-arginine methyl ester (inhibitor of nitric oxide synthase). In addition, mitochondria were isolated from human right atrial at 15 min of reoxygenation. The effect of propofol on activity of the mitochondrial respiratory chain complexes was evaluated by spectrophotometry. The force of contraction (% of baseline) and the complex activity between the different groups were compared with an analysis of variance and post hoc test. Propofol 10 µM administered during the reoxygenation period significantly improved the recovery of force of contraction at the end of reoxygenation (82 ± 6% of baseline value vs. 49 ± 6% in Control; P < 0.001). The beneficial effects of propofol 10 µM were abolished by co-administration with 5-hydroxydecanoate (53 ± 8%) or NG-nitro-L-arginine methyl ester (57 ± 6%). Propofol 10 µM significantly increased enzymatic activities of the mitochondrial respiratory chain complexes, in reoxygenation period, compared to their respective untreated controls. In conclusion, in human myocardium, propofol-induced cardioprotection was mediated by mitochondrial adenosine triphosphate-sensitive potassium channels opening, nitric oxide synthase activation and stimulation of mitochondrial respiratory chain complexes, in early reoxygenation period.
Collapse
Affiliation(s)
- Sandrine Lemoine
- Department of Anesthesiology and Intensive Care, Centre Hospitalier Universitaire de Caen, Faculty of Medicine, Normandie Université, EA4650, Caen 14033, France
| | - Lan Zhu
- Department of Anesthesiology and Intensive Care, Centre Hospitalier Universitaire de Caen, Faculty of Medicine, Normandie Université, EA4650, Caen 14033, France
| | - Steeve Gress
- Department of Anesthesiology and Intensive Care, Centre Hospitalier Universitaire de Caen, Faculty of Medicine, Normandie Université, EA4650, Caen 14033, France
| | - Jean-Louis Gérard
- Department of Anesthesiology and Intensive Care, Centre Hospitalier Universitaire de Caen, Faculty of Medicine, Normandie Université, EA4650, Caen 14033, France
| | - Stéphane Allouche
- Department of Biochemistry, Centre Hospitalier Universitaire de Caen, Faculty of Medicine, Normandie Université, EA4650, Caen 14033, France
| | | |
Collapse
|
32
|
Krajčová A, Waldauf P, Anděl M, Duška F. Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care 2015; 19:398. [PMID: 26558513 PMCID: PMC4642662 DOI: 10.1186/s13054-015-1112-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/22/2015] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Propofol infusion syndrome (PRIS) is a rare, but potentially lethal adverse effect of a commonly used drug. We aimed to review and correlate experimental and clinical data about this syndrome. METHODS We searched for all case reports published between 1990 and 2014 and for all experimental studies on PRIS pathophysiology. We analysed the relationship between signs of PRIS and the rate and duration of propofol infusion causing PRIS. By multivariate logistic regression we looked at the risk factors for mortality. RESULTS Knowledge about PRIS keeps evolving. Compared to earlier case reports in the literature, recently published cases describe older patients developing PRIS at lower doses of propofol, in whom arrhythmia, hypertriglyceridaemia and fever are less frequently seen, with survival more likely. We found that propofol infusion rate and duration, the presence of traumatic brain injury and fever are factors independently associated with mortality in reported cases of PRIS (area under receiver operator curve = 0.85). Similar patterns of exposure to propofol (in terms of time and concentration) are reported in clinical cases and experimental models of PRIS. Cardiac failure and metabolic acidosis occur early in a dose-dependent manner, while arrhythmia, other electrocardiographic changes and rhabdomyolysis appear more frequently after prolonged propofol infusions, irrespective of dose. CONCLUSION PRIS can develop with propofol infusion <4 mg/kg per hour and its diagnosis may be challenging as some of its typical features (hypertriglyceridaemia, fever, hepatomegaly, heart failure) are often (>95 %) missing and others (arrhythmia, electrocardiographic changes) occur late.
Collapse
Affiliation(s)
- Adéla Krajčová
- Laboratory for Metabolism and Bioenergetics, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
- Centre for Research on Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - Petr Waldauf
- Department of Anaesthesiology and Intensive Care, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - Michal Anděl
- Laboratory for Metabolism and Bioenergetics, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
- Centre for Research on Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - František Duška
- Laboratory for Metabolism and Bioenergetics, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
- Department of Anaesthesiology and Intensive Care, Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
- Adult Intensive Care Unit, Nottingham University Hospitals NHS Trust, Nottingham, UK.
| |
Collapse
|
33
|
Ktena YP, Ramstad T, Baker EH, Sloan JL, Mannes AJ, Manoli I, Venditti CP. Propofol administration in patients with methylmalonic acidemia and intracellular cobalamin metabolism disorders: a review of theoretical concerns and clinical experiences in 28 patients. J Inherit Metab Dis 2015; 38:847-53. [PMID: 25985870 PMCID: PMC5577977 DOI: 10.1007/s10545-015-9816-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/18/2015] [Accepted: 01/22/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Methylmalonic acidemia and intracellular cobalamin metabolism disorders represent a heterogeneous group of inborn errors of metabolism. Most patients will require diagnostic and/or therapeutic procedures frequently requiring sedation or anesthetic management due to neurological and neurocognitive impairments. It has been stated that propofol is contraindicated in this population. We report our experience with propofol administration in a large series of patients. METHODS Twenty eight patients (14 mut, seven cblC, three cblA, three cblB, one cblG) aged 2-35.6 years enrolled in a natural history study (ClinicalTrials.gov identifier: NCT00078078) and required anesthetics for 39 diagnostic or therapeutic procedures. Data were collected on the anesthetic technique, perianesthetic course, and adverse events related to propofol. RESULTS Propofol was used as the sole induction agent in most cases (36/39) and as the primary maintenance agent in all cases. Infusion rates were 100-400 mcg kg(-1) min(-1) (mean = 214). Infusion duration was 60-325 min (mean = 158) and total doses ranged between 270-3610 mg (mean = 1217). Adverse events were recorded in two cases; neither appeared to be related to propofol administration. CONCLUSIONS Propofol is an effective, safe induction and maintenance agent for elective short procedures requiring anesthesia in patients with MMA and cobalamin metabolism disorders. Despite multiple comorbidities and propensity toward instability, those affected can receive anesthesia with an acceptable safety profile, if metabolically and hemodynamically stabilized prior to the event. SYNOPSIS A review of the perianesthetic records of 28 patients with isolated MMA and intracellular cobalamin metabolism disorders suggests that propofol anesthesia can be administered safely to these patients, in the setting of metabolic stability.
Collapse
Affiliation(s)
- Yiouli P Ktena
- Organic Acid Research Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 49 Convent Drive, Building 49, Room 4A18, Bethesda, MD, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Propofol is an intravenous agent used commonly for the induction and maintenance of anesthesia, procedural, and critical care sedation in children. The mechanisms of action on the central nervous system involve interactions at various neurotransmitter receptors, especially the gamma-aminobutyric acid A receptor. Approved for use in the USA by the Food and Drug Administration in 1989, its use for induction of anesthesia in children less than 3 years of age still remains off-label. Despite its wide use in pediatric anesthesia, there is conflicting literature about its safety and serious adverse effects in particular subsets of children. Particularly as children are not "little adults", in this review, we emphasize the maturational aspects of propofol pharmacokinetics. Despite the myriad of propofol pharmacokinetic-pharmacodynamic studies and the ability to use allometrical scaling to smooth out differences due to size and age, there is no optimal model that can be used in target controlled infusion pumps for providing closed loop total intravenous anesthesia in children. As the commercial formulation of propofol is a nutrient-rich emulsion, the risk for bacterial contamination exists despite the Food and Drug Administration mandating addition of antimicrobial preservative, calling for manufacturers' directions to discard open vials after 6 h. While propofol has advantages over inhalation anesthesia such as less postoperative nausea and emergence delirium in children, pain on injection remains a problem even with newer formulations. Propofol is known to depress mitochondrial function by its action as an uncoupling agent in oxidative phosphorylation. This has implications for children with mitochondrial diseases and the occurrence of propofol-related infusion syndrome, a rare but seriously life-threatening complication of propofol. At the time of this review, there is no direct evidence in humans for propofol-induced neurotoxicity to the infant brain; however, current concerns of neuroapoptosis in developing brains induced by propofol persist and continue to be a focus of research.
Collapse
Affiliation(s)
- Vidya Chidambaran
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 2001, Cincinnati, OH, 45229, USA,
| | | | | |
Collapse
|
35
|
Vanlander AV, Okun JG, de Jaeger A, Smet J, De Latter E, De Paepe B, Dacremont G, Wuyts B, Vanheel B, De Paepe P, Jorens PG, Van Regenmortel N, Van Coster R. Possible pathogenic mechanism of propofol infusion syndrome involves coenzyme q. Anesthesiology 2015; 122:343-52. [PMID: 25296107 DOI: 10.1097/aln.0000000000000484] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Propofol is a short-acting intravenous anesthetic agent. In rare conditions, a life-threatening complication known as propofol infusion syndrome can occur. The pathophysiologic mechanism is still unknown. Some studies suggested that propofol acts as uncoupling agent, others suggested that it inhibits complex I or complex IV, or causes increased oxidation of cytochrome c and cytochrome aa3, or inhibits mitochondrial fatty acid metabolism. Although the exact site of interaction is not known, most hypotheses point to the direction of the mitochondria. METHODS Eight rats were ventilated and sedated with propofol up to 20 h. Sequential biopsy specimens were taken from liver and skeletal muscle and used for determination of respiratory chain activities and propofol concentration. Activities were also measured in skeletal muscle from a patient who died of propofol infusion syndrome. RESULTS In rats, authors detected a decrease in complex II+III activity starting at low tissue concentration of propofol (20 to 25 µM), further declining at higher concentrations. Before starting anesthesia, the complex II+III/citrate synthase activity ratio in liver was 0.46 (0.25) and in skeletal muscle 0.23 (0.05) (mean [SD]). After 20 h of anesthesia, the ratios declined to 0.17 (0.03) and 0.12 (0.02), respectively. When measured individually, the activities of complexes II and III remained normal. Skeletal muscle from one patient taken in the acute phase of propofol infusion syndrome also shows a selective decrease in complex II+III activity (z-score: -2.96). CONCLUSION Propofol impedes the electron flow through the respiratory chain and coenzyme Q is the main site of interaction with propofol.
Collapse
Affiliation(s)
- Arnaud Vincent Vanlander
- From the Department of Pediatrics, Division of Pediatric Neurology and Metabolism (A.V.V., J.S., E.D.L., B.D.P., R.V.C.), Department of Critical Care Medicine, Division of Pediatric Intensive Care Medicine (A.d.J.), Department of Clinical Chemistry (B.W.), Department of Emergency Medicine (P.D.P.), Ghent University Hospital, Ghent, Belgium; Department of General Pediatrics, Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany (J.G.O.); Department of Pediatrics, University of Ghent, Ghent, Belgium (G.D.); Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium (B.V.); Department of Critical Care Medicine, Antwerp University Hospital, Antwerp University, Edegem, Belgium (P.G.J., N.V.R.); and Department of Critical Care Medicine, ZNA Antwerp, Belgium (N.V.R.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Suzuki T, Kikuchi H, Ogura M, Homma MK, Oshima Y, Homma Y. Weight loss by Ppc-1, a novel small molecule mitochondrial uncoupler derived from slime mold. PLoS One 2015; 10:e0117088. [PMID: 25668511 PMCID: PMC4323345 DOI: 10.1371/journal.pone.0117088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/19/2014] [Indexed: 01/06/2023] Open
Abstract
Mitochondria play a key role in diverse processes including ATP synthesis and apoptosis. Mitochondrial function can be studied using inhibitors of respiration, and new agents are valuable for discovering novel mechanisms involved in mitochondrial regulation. Here, we screened small molecules derived from slime molds and other microorganisms for their effects on mitochondrial oxygen consumption. We identified Ppc-1 as a novel molecule which stimulates oxygen consumption without adverse effects on ATP production. The kinetic behavior of Ppc-1 suggests its function as a mitochondrial uncoupler. Serial administration of Ppc-1 into mice suppressed weight gain with no abnormal effects on liver or kidney tissues, and no evidence of tumor formation. Serum fatty acid levels were significantly elevated in mice treated with Ppc-1, while body fat content remained low. After a single administration, Ppc-1 distributes into various tissues of individual animals at low levels. Ppc-1 stimulates adipocytes in culture to release fatty acids, which might explain the elevated serum fatty acids in Ppc-1-treated mice. The results suggest that Ppc-1 is a unique mitochondrial regulator which will be a valuable tool for mitochondrial research as well as the development of new drugs to treat obesity.
Collapse
Affiliation(s)
- Toshiyuki Suzuki
- Fukushima Medical University School of Medicine, Fukushima, 960–1295, Japan
| | - Haruhisa Kikuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980–8678, Japan
| | - Masato Ogura
- Fukushima Medical University School of Medicine, Fukushima, 960–1295, Japan
| | - Miwako K. Homma
- Fukushima Medical University School of Medicine, Fukushima, 960–1295, Japan
| | - Yoshiteru Oshima
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980–8678, Japan
| | - Yoshimi Homma
- Fukushima Medical University School of Medicine, Fukushima, 960–1295, Japan
- * E-mail:
| |
Collapse
|
37
|
Parikh S, Goldstein A, Koenig MK, Scaglia F, Enns GM, Saneto R, Anselm I, Cohen BH, Falk MJ, Greene C, Gropman AL, Haas R, Hirano M, Morgan P, Sims K, Tarnopolsky M, Van Hove JLK, Wolfe L, DiMauro S. Diagnosis and management of mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society. Genet Med 2014; 17:689-701. [PMID: 25503498 DOI: 10.1038/gim.2014.177] [Citation(s) in RCA: 344] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/06/2014] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The purpose of this statement is to review the literature regarding mitochondrial disease and to provide recommendations for optimal diagnosis and treatment. This statement is intended for physicians who are engaged in diagnosing and treating these patients. METHODS The Writing Group members were appointed by the Mitochondrial Medicine Society. The panel included members with expertise in several different areas. The panel members utilized a comprehensive review of the literature, surveys, and the Delphi method to reach consensus. We anticipate that this statement will need to be updated as the field continues to evolve. RESULTS Consensus-based recommendations are provided for the diagnosis and treatment of mitochondrial disease. CONCLUSION The Delphi process enabled the formation of consensus-based recommendations. We hope that these recommendations will help standardize the evaluation, diagnosis, and care of patients with suspected or demonstrated mitochondrial disease.
Collapse
Affiliation(s)
- Sumit Parikh
- Department of Neurology, Center for Child Neurology, Cleveland Clinic Children's Hospital, Cleveland, Ohio, USA
| | - Amy Goldstein
- Department of Pediatrics, Division of Child Neurology, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mary Kay Koenig
- Department of Pediatrics, Division of Child and Adolescent Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Gregory M Enns
- Department of Pediatrics, Division of Medical Genetics, Stanford University Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Russell Saneto
- Department of Neurology, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA.,Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Irina Anselm
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Bruce H Cohen
- Department of Pediatrics, NeuroDevelopmental Science Center, Children's Hospital Medical Center of Akron, Akron, Ohio, USA
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Carol Greene
- Department of Pediatrics, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Andrea L Gropman
- Department of Neurology, Children's National Medical Center and the George Washington University of the Health Sciences, Washington, DC, USA
| | - Richard Haas
- Department of Neurosciences and Pediatrics, UCSD Medical Center and Rady Children's Hospital San Diego, La Jolla, California, USA
| | - Michio Hirano
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| | - Phil Morgan
- Department of Anesthesiology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Katherine Sims
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark Tarnopolsky
- Department of Pediatrics and Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Johan L K Van Hove
- Department of Pediatrics, Clinical Genetics and Metabolism, Children's Hospital Colorado, Denver, Colorado, USA
| | - Lynne Wolfe
- National Institutes of Health, Bethesda, Maryland, USA
| | - Salvatore DiMauro
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
38
|
Propofol protects the immature rabbit heart against ischemia and reperfusion injury: impact on functional recovery and histopathological changes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:601250. [PMID: 25243155 PMCID: PMC4163471 DOI: 10.1155/2014/601250] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/20/2014] [Indexed: 11/17/2022]
Abstract
The general anesthetic propofol protects the adult heart against ischemia and reperfusion injury; however, its efficacy has not been investigated in the immature heart. This work, for the first time, investigates the cardioprotective efficacy of propofol at clinically relevant concentrations in the immature heart. Langendorff perfused rabbit hearts (7–12 days old) were exposed to 30 minutes' global normothermic ischemia followed by 40 minutes' reperfusion. Left ventricular developed pressure (LVDP) and coronary flow were monitored throughout. Lactate release into coronary effluent was measured during reperfusion. Microscopic examinations of the myocardium were monitored at the end of reperfusion. Hearts were perfused with different propofol concentrations (1, 2, 4, and 10 μg/mL) or with cyclosporine A, prior to ischemic arrest and for 20 minutes during reperfusion. Propofol at 4 and 10 μg/mL caused a significant depression in LVDP prior to ischemia. Propofol at 2 μg/mL conferred significant and maximal protection with no protection at 10 μg/mL. This protection was associated with improved recovery in coronary flow, reduced lactate release, and preservation of cardiomyocyte ultrastructure. The efficacy of propofol at 2 μg/mL was similar to the effect of cyclosporine A. In conclusion, propofol at a clinically relevant concentration is cardioprotective in the immature heart.
Collapse
|
39
|
Niezgoda J, Morgan PG. Anesthetic considerations in patients with mitochondrial defects. Paediatr Anaesth 2013; 23:785-93. [PMID: 23534340 PMCID: PMC3711963 DOI: 10.1111/pan.12158] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/25/2013] [Indexed: 12/17/2022]
Abstract
Mitochondrial disease, once thought to be a rare clinical entity, is now recognized as an important cause of a wide range of neurologic, cardiac, muscle, and endocrine disorders . The incidence of disorders of the respiratory chain alone is estimated to be about 1 per 4-5000 live births, similar to that of more well-known neurologic diseases . High-energy requiring tissues are uniquely dependent on the energy delivered by mitochondria and therefore have the lowest threshold for displaying symptoms of mitochondrial disease. Thus, mitochondrial dysfunction most commonly affects function of the central nervous system, the heart and the muscular system . Mutations in mitochondrial proteins cause striking clinical features in those tissues types, including encephalopathies, seizures, cerebellar ataxias, cardiomyopathies, myopathies, as well as gastrointestinal and hepatic disease. Our knowledge of the contribution of mitochondria in causing disease or influencing aging is expanding rapidly . As diagnosis and treatment improve for children with mitochondrial diseases, it has become increasingly common for them to undergo surgeries for their long-term care. In addition, often a muscle biopsy or other tests needing anesthesia are required for diagnosis. Mitochondrial disease represents probably hundreds of different defects, both genetic and environmental in origin, and is thus difficult to characterize. The specter of possible delayed complications in patients caused by inhibition of metabolism by anesthetics, by remaining in a biochemically stressed state such as fasting/catabolism, or by prolonged exposure to pain is a constant worry to physicians caring for these patients. Here, we review the considerations when caring for a patient with mitochondrial disease.
Collapse
Affiliation(s)
- Julie Niezgoda
- Department of Pediatric Anesthesiology, Cleveland Clinic, Cleveland, USA
| | - Phil G Morgan
- Department of Anesthesiology and Pain Medicine, University of Washington and Seattle Children’s Hospital, Seattle, USA
| |
Collapse
|
40
|
|
41
|
Modifiers of the oligomycin sensitivity of the mitochondrial F1F0-ATPase. Mitochondrion 2013; 13:312-9. [PMID: 23597783 DOI: 10.1016/j.mito.2013.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 02/02/2023]
|
42
|
Coleman AE, McNeil N, Kovalchuck AL, Wangsa D, Ried T, Wang H. Cellular exposure to muscle relaxants and propofol could lead to genomic instability in vitro. J Biomed Res 2013; 26:117-24. [PMID: 23554740 PMCID: PMC3597328 DOI: 10.1016/s1674-8301(12)60021-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 01/05/2012] [Accepted: 01/28/2012] [Indexed: 11/25/2022] Open
Abstract
Anesthesia is widely used in several medical settings and accepted as safe. However, there is some evidence that anesthetic agents can induce genomic changes leading to neural degeneration or apoptosis. Although chromosomal changes have not been observed in vivo, this is most likely due to DNA repair mechanisms, apoptosis, or cellular senescence. Potential chromosomal alterations after exposure to common anesthetic agents may be relevant in patients with genomic instability syndromes or with aggressive treatment of malignancies. In this study, the P388 murine B cells were cultured in vitro, and spectral karyotyping (SKY) was utilized to uncover genome-wide changes. Clinically relevant doses of cisatracurium and propofol increased structural and numerical chromosomal instability. These results may be relevant in patients with underlying chromosomal instability syndromes or concurrently being exposed to chemotherapeutic agents. Future studies may include utilization of stimulated peripheral blood lymphocytes to further confirm the significance of these results.
Collapse
Affiliation(s)
- Allen Edward Coleman
- Department of Anesthesiology, Wayne State University, Detroit Medical Center, Detroit, Michigan 48201, USA
| | | | | | | | | | | |
Collapse
|
43
|
Rybakova SR, Dubinin MV, Samartsev VN. The features of activation of free oxidation by α,ω-tetradecanedioic acid in liver mitochondria. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2013. [DOI: 10.1134/s1990747812050157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Complex I and ATP synthase mediate membrane depolarization and matrix acidification by isoflurane in mitochondria. Eur J Pharmacol 2012; 690:149-57. [PMID: 22796646 DOI: 10.1016/j.ejphar.2012.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 06/21/2012] [Accepted: 07/02/2012] [Indexed: 11/22/2022]
Abstract
Short application of the volatile anesthetic isoflurane at reperfusion after ischemia exerts strong protection of the heart against injury. Mild depolarization and acidification of the mitochondrial matrix are involved in the protective mechanisms of isoflurane, but the molecular basis for these changes is not clear. In this study, mitochondrial respiration, membrane potential, matrix pH, matrix swelling, ATP synthesis and -hydrolysis, and H(2)O(2) release were assessed in isolated mitochondria. We hypothesized that isoflurane induces mitochondrial depolarization and matrix acidification through direct action on both complex I and ATP synthase. With complex I-linked substrates, isoflurane (0.5mM) inhibited mitochondrial respiration by 28 ± 10%, and slightly, but significantly depolarized membrane potential and decreased matrix pH. With complex II- and complex IV-linked substrates, respiration was not changed, but isoflurane still decreased matrix pH and depolarized mitochondrial membrane potential. Depolarization and matrix acidification were attenuated by inhibition of ATP synthase with oligomycin, but not by inhibition of mitochondrial ATP- and Ca(2+)-sensitive K(+) channels or uncoupling proteins. Isoflurane did not induce matrix swelling and did not affect ATP synthesis and hydrolysis, but decreased H(2)O(2) release in the presence of succinate in an oligomycin- and matrix pH-sensitive manner. Isoflurane modulated H(+) flux through ATP synthase in an oligomycin-sensitive manner. Our results indicate that isoflurane-induced mitochondrial depolarization and acidification occur due to inhibition of the electron transport chain at the site of complex I and increased proton flux through ATP synthase. K(+) channels and uncoupling proteins appear not to be involved in the direct effects of isoflurane on mitochondria.
Collapse
|
45
|
VANLANDER AV, JORENS PG, SMET J, DE PAEPE B, VERBRUGGHE W, VAN DEN EYNDEN GG, MEIRE F, PAUWELS P, VAN DER AA N, SENECA S, LISSENS W, OKUN JG, VAN COSTER R. Inborn oxidative phosphorylation defect as risk factor for propofol infusion syndrome. Acta Anaesthesiol Scand 2012; 56:520-5. [PMID: 22260353 DOI: 10.1111/j.1399-6576.2011.02628.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2011] [Indexed: 12/24/2022]
Abstract
Propofol is an anesthetic agent widely used for induction and maintenance of anesthesia, and sedation in children. Although generally considered as reliable and safe, administration of propofol can occasionally induce a potentially fatal complication known as propofol infusion syndrome (PRIS). Mitochondrial dysfunction has been implicated in the pathogenesis of PRIS. We report on an adult patient with Leber hereditary optic neuropathy (LHON) who developed PRIS. He was a carrier of the m.3460G>A mutation, one of the major three pathogenic point mutations associated with LHON. The propositus was blind and underwent propofol sedation after severe head injury. Five days after start of propofol infusion, the patient died. The activity of complex I of the oxidative phosphorylation (OXPHOS) system was severely deficient in skeletal muscle. Our observation indicates that fulminate PRIS can occur in an adult patient with an inborn OXPHOS defect and corroborates the hypothesis that PRIS is caused by inhibition of the OXPHOS system.
Collapse
Affiliation(s)
- A. V. VANLANDER
- Department of Pediatrics; Division of Pediatric Neurology and Metabolism; Ghent University Hospital; Ghent; Belgium
| | - P. G. JORENS
- Department of Critical Care Medicine; Antwerp University Hospital, Antwerp University; Edegem; Belgium
| | - J. SMET
- Department of Pediatrics; Division of Pediatric Neurology and Metabolism; Ghent University Hospital; Ghent; Belgium
| | - B. DE PAEPE
- Department of Pediatrics; Division of Pediatric Neurology and Metabolism; Ghent University Hospital; Ghent; Belgium
| | - W. VERBRUGGHE
- Department of Critical Care Medicine; Antwerp University Hospital, Antwerp University; Edegem; Belgium
| | | | - F. MEIRE
- Department of Pediatric Ophthalmology; Hôpital Universitaire des Enfants Reine Fabiola; Brussels; Belgium
| | - P. PAUWELS
- Department of Pathology; Antwerp University; Wilrijk; Belgium
| | - N. VAN DER AA
- Department of Medical Genetics; Antwerp University Hospital, Antwerp University; Edegem; Belgium
| | - S. SENECA
- Center for Medical Genetics; UZ Brussel and Reproduction and Genetics (REGE); Vrije Universiteit Brussel; Brussels; Belgium
| | - W. LISSENS
- Center for Medical Genetics; UZ Brussel and Reproduction and Genetics (REGE); Vrije Universiteit Brussel; Brussels; Belgium
| | - J. G. OKUN
- Department of General Pediatrics; Division of Inborn Metabolic Diseases; University Children's Hospital; Heidelberg; Germany
| | - R. VAN COSTER
- Department of Pediatrics; Division of Pediatric Neurology and Metabolism; Ghent University Hospital; Ghent; Belgium
| |
Collapse
|
46
|
Pessayre D, Fromenty B, Berson A, Robin MA, Lettéron P, Moreau R, Mansouri A. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev 2011; 44:34-87. [PMID: 21892896 DOI: 10.3109/03602532.2011.604086] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A frequent mechanism for drug-induced liver injury (DILI) is the formation of reactive metabolites that trigger hepatitis through direct toxicity or immune reactions. Both events cause mitochondrial membrane disruption. Genetic or acquired factors predispose to metabolite-mediated hepatitis by increasing the formation of the reactive metabolite, decreasing its detoxification, or by the presence of critical human leukocyte antigen molecule(s). In other instances, the parent drug itself triggers mitochondrial membrane disruption or inhibits mitochondrial function through different mechanisms. Drugs can sequester coenzyme A or can inhibit mitochondrial β-oxidation enzymes, the transfer of electrons along the respiratory chain, or adenosine triphosphate (ATP) synthase. Drugs can also destroy mitochondrial DNA, inhibit its replication, decrease mitochondrial transcripts, or hamper mitochondrial protein synthesis. Quite often, a single drug has many different effects on mitochondrial function. A severe impairment of oxidative phosphorylation decreases hepatic ATP, leading to cell dysfunction or necrosis; it can also secondarily inhibit ß-oxidation, thus causing steatosis, and can also inhibit pyruvate catabolism, leading to lactic acidosis. A severe impairment of β-oxidation can cause a fatty liver; further, decreased gluconeogenesis and increased utilization of glucose to compensate for the inability to oxidize fatty acids, together with the mitochondrial toxicity of accumulated free fatty acids and lipid peroxidation products, may impair energy production, possibly leading to coma and death. Susceptibility to parent drug-mediated mitochondrial dysfunction can be increased by factors impairing the removal of the toxic parent compound or by the presence of other medical condition(s) impairing mitochondrial function. New drug molecules should be screened for possible mitochondrial effects.
Collapse
Affiliation(s)
- Dominique Pessayre
- INSERM, U, Centre de Recherche Bichat Beaujon CRB, Faculté de Médecine Xavier-Bichat, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Le syndrome de perfusion du propofol. ACTA ACUST UNITED AC 2010; 29:377-86. [PMID: 20399595 DOI: 10.1016/j.annfar.2010.02.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2009] [Accepted: 02/17/2010] [Indexed: 01/08/2023]
|
48
|
Morota S, Månsson R, Hansson MJ, Kasuya K, Shimazu M, Hasegawa E, Yanagi S, Omi A, Uchino H, Elmér E. Evaluation of putative inhibitors of mitochondrial permeability transition for brain disorders--specificity vs. toxicity. Exp Neurol 2009; 218:353-62. [PMID: 19348797 DOI: 10.1016/j.expneurol.2009.03.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 03/23/2009] [Accepted: 03/26/2009] [Indexed: 12/16/2022]
Abstract
Inhibition of mitochondrial permeability transition (mPT) has emerged as a promising approach for neuroprotection and development of well-tolerated mPT inhibitors with favorable blood-brain barrier penetration is highly warranted. In a recent study, 28 clinically available drugs with a common heterocyclic structure were identified as mPT inhibitors e.g. trifluoperazine, promethazine and nortriptyline. In addition, neuroprotection by structurally unrelated drugs e.g. neurosteroids, 4-hydroxy-tamoxifen and trimetazidine has been attributed to direct inhibition of mPT. The regulation of mPT is complex and highly dependent on the prevailing experimental conditions. Several features of mPT, such as swelling, depolarization or NADH oxidation, can also occur independently of the mPT phenomenon. Here, in isolated rodent brain-derived and human liver mitochondria, we re-evaluate drugs promoted as potent mPT inhibitors. We address the definition of an mPT inhibitor and present strategies to reliably detect mPT inhibition in vitro. Surprisingly, none of the 12 compounds tested displayed convincing mPT inhibition or effects comparable to cyclophilin D inhibition by the non-immunosuppressive cyclophilin inhibitor D-MeAla(3)-EtVal(4)-Cyclosporin (Debio 025). Propofol and 2-aminoethoxydiphenyl borate (2-APB) inhibited swelling in de-energized mitochondria but did not increase calcium retention capacity (CRC). Progesterone, trifluoperazine, allopregnanolone and 4-hydroxy-tamoxifen dose-dependently reduced CRC and respiratory control and were thus toxic rather than beneficial to mitochondrial function. Interestingly, topiramate increased CRC at high concentrations likely by a mechanism separate from direct mPT inhibition. We conclude that a clinically relevant mPT inhibitor should have a mitochondrial target and increase mitochondrial calcium retention at concentrations which can be translated to human use.
Collapse
Affiliation(s)
- Saori Morota
- Department of Clinical Sciences, Lund University, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Certain anesthetics exhibit neurotoxicity in the brains of immature but not mature animals. Gamma-aminobutyric acid (GABA), the primary inhibitory neurotransmitter in the adult brain, is excitatory on immature neurons via its action at the GABAA receptor, due to a reversed transmembrane chloride gradient. GABAA receptor activation in immature neurons is sufficient to open L-type voltage-gated calcium channels. As propofol is a GABAA agonist, we hypothesized that it and more specific GABAA modulators would increase intracellular free calcium ([Ca2+]i), resulting in the death of neonatal rat hippocampal neurons. Neuronal [Ca2+]i was monitored using Fura2-AM fluorescence imaging. Cell death was assessed by double staining with propidium iodide and Hoechst 33258 at 1 hour (acute) and 48 hours (delayed) after 5 hours exposure of neurons to propofol or the GABAA receptor agonist, muscimol, in the presence and absence of the GABA receptor antagonist, bicuculline, or the L-type Ca2+ channel blocker, nifedipine. Fluorescent measurements of caspase-3,-7 activities were performed at 1 hour after exposure. Both muscimol and propofol induced a rapid increase in [Ca2+]i in days in vitro (DIV) 4, but not in DIV 8 neurons, that was inhibited by nifedipine and bicuculline. Caspase-3,-7 activities and cell death increased significantly in DIV 4 but not DIV 8 hippocampal neuronal cultures 1 hour after 5 hours exposure to propofol, but not muscimol, and were inhibited by the presence of bicuculline or nifedipine. We conclude that an increase in [Ca2+]i, due to activation of GABAA receptors and opening of L-type calcium channels, is necessary for propofol-induced death of immature rat hippocampal neurons but that additional mechanisms not elicited by GABAA activation alone also contribute to cell death.
Collapse
|
50
|
Devin A, Nogueira V, Avéret N, Leverve X, Rigoulet M. Profound effects of the general anesthetic etomidate on oxidative phosphorylation without effects on their yield. J Bioenerg Biomembr 2007; 38:137-42. [PMID: 17029016 DOI: 10.1007/s10863-006-9013-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We investigated the effects of the general anesthetic Etomidate on oxidative phosphorylation in isolated rat liver mitochondria. The study of each electron transfer site shows that there is an inhibition: mainly at complex I but also, to a lesser extent, at complex III. Moreover, with succinate as substrate, the increase in non-phosphorylating respiration is accompanied by a decrease in DeltaPsi. However, this effect is not due to classical uncoupling of oxidative phosphorylation, since ADP addition at high Etomidate concentrations restores the transmembrane difference of electrical potential. Also, in the same range of Etomidate concentration, the ATP/O ratio is not significantly affected. In conclusion, the main effect of Etomidate is to decrease the oxidative phosphorylation rate without changing yield. The H(+) leak which appears under non-phosphorylating conditions becomes negligible in physiological conditions.
Collapse
Affiliation(s)
- Anne Devin
- Institut de Biochimie et Génétique Cellulaires du CNRS, Université de BordeauxII, 1 rue Camille Saint-Saëns, 33077, Bordeaux cedex, France
| | | | | | | | | |
Collapse
|