Propofol induces a metabolic switch to glycolysis and cell death in a mitochondrial electron transport chain-dependent manner

PLoS One. 2018 Feb 15;13(2):e0192796. doi: 10.1371/journal.pone.0192796. eCollection 2018.

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.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Anesthetics, Intravenous / toxicity*
  • Animals
  • Caspases / metabolism
  • Cell Death / drug effects*
  • Cell Death / physiology
  • Cell Line, Tumor
  • Dose-Response Relationship, Drug
  • Electron Transport / drug effects*
  • Electron Transport / physiology
  • Glycolysis / drug effects*
  • Glycolysis / physiology
  • HeLa Cells
  • Humans
  • Hypoglycemic Agents / pharmacology
  • Membrane Potential, Mitochondrial / drug effects
  • Membrane Potential, Mitochondrial / physiology
  • Metformin / pharmacology
  • Mice
  • Mitochondria / drug effects*
  • Mitochondria / metabolism
  • Muscle Cells / drug effects
  • Muscle Cells / metabolism
  • Neurons / drug effects
  • Neurons / metabolism
  • Oxygen Consumption / drug effects
  • Oxygen Consumption / physiology
  • Propofol / toxicity*
  • Reactive Oxygen Species / metabolism
  • Time Factors

Substances

  • Anesthetics, Intravenous
  • Hypoglycemic Agents
  • Reactive Oxygen Species
  • Metformin
  • Caspases
  • Propofol

Grants and funding

This work was supported by the Japan Society for the Promotion of Science KAKENHI, Grants #26670693 and #24592336 to K.H., #25462457 to K.N., and #15K15577 to T.A., and by a research grant from Katano Kai to K.H. and A.O. This work was supported by the research grant B from Kansai Medical University to A.O., the KMU consortium grant from Kansai Medical University to K.H. and the research grant from Katano Kai to A.O. and K.H. This work was supported by the Japanese Society of Anesthesiologists (JSA) Pitch Contest 2017 to C.S. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.