A true enigma of modern medicine has persisted for over 150 years. How can volatile anesthetics, as a class of drugs, simultaneously cause a patient to reversibly lose consciousness, feel no pain, and have no memory of surgery? The mechanism by which volatile anesthetics (VAs) produce reversible loss of consciousness and render an organism insensate to a surgical incision remains an unsolved mystery. Our laboratory has exploited a very simple animal model, the nematode C. elegans, to investigate the molecular mechanisms of volatile anesthetic action. Previously, we demonstrated that mitochondrial complex I, an entry point of the electron transport chain, specifically controls the sensitivity of C. elegans to volatile anesthetics. We also found that complex I defects control anesthetic sensitivity in humans. We now have an exciting opportunity to extend those findings into mice using the knockout animal, Ndufs4. The Ndufs4 KO mice lack a subunit of complex I, which results in complex I dysfunction. We anesthetized young (PN23-27) Ndusf4 and wild-type (WT) mice with isoflurane or halothane. For either VA, the KO mice became unresponsive to a tail pinch at a dose 2.8-fold lower than for WT controls. These KO mice display the greatest change in VA sensitivity ever described in a mammal. We also measured the EC50s for loss of righting reflex (LORR) of the KO mice for anesthetics whose targets are well characterized. Surprisingly, the animals were actually resistant to the effects of ketamine. The effects of the complex I KO are specific in terms of anesthetic and not simply the result of generalized CNS depression. Complex I dysfunction causes hypersensitivity to volatile anesthetics across phylogeny. However, the question remains, how do complex I defects affect VA sensitivity. We hypothesize that complex I depression (i.e. energy depletion) leads to defective synaptic function in specific cell types, making them more susceptible to neuronal silencing by VAs. We will knock out Ndufs4 in GABAergic, cholinergic or glutamatergic neurons, in neuronal support cells known as astrocytes, as well as in specific regions of the brain. We will also perform electrophysiologic measurements of WT and mutant mice with and without volatile anesthetics. Our initial results indicate that the VA hypersensitivity in Ndufs4 mice is mediated through glutamatergic neurons.
Our aims are to characterize which neurons and regions of the brain are important for the anesthetic phenotype of these animals, as well as to discover how basic neuronal physiology is altered in the KO animals. Our overarching goal is to understand how the VAs functions. We have linked mitochondrial function to behavior in VAs in worms, mice, and man, a finding the field must consider. Our proposed studies will tease out which cell types mediate the anesthetic response, and will give important insights into the basic mechanisms of action of VAs.

Public Health Relevance

We have discovered a link between mitochondrial complex I and volatile anesthetic sensitivity that holds true for multiple species. We will determine which nervous system cell types and regions are important for determining anesthetic sensitivity, and test the electrophysiologic effects of this defect on neuronal function. Our studies are intended to uncover basic mechanisms by which volatile anesthetics exert their effects and eliminate their adverse side effects.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM105696-04
Application #
9225232
Study Section
Special Emphasis Panel (ZRG1-SBIB-V (02)S)
Program Officer
Cole, Alison E
Project Start
2014-05-01
Project End
2018-02-28
Budget Start
2017-03-01
Budget End
2018-02-28
Support Year
4
Fiscal Year
2017
Total Cost
$489,945
Indirect Cost
$238,691
Name
Seattle Children's Hospital
Department
Type
Independent Hospitals
DUNS #
048682157
City
Seattle
State
WA
Country
United States
Zip Code
98101
Carspecken, Charles William; Chanprasert, Sirisak; Kalume, Franck et al. (2018) Anesthetics Have Different Effects on the Electrocorticographic Spectra of Wild-type and Mitochondrial Mutant Mice. Anesthesiology 129:744-755
Ramadasan-Nair, Renjini; Hui, Jessica; Zimin, Pavel I et al. (2017) Regional knockdown of NDUFS4 implicates a thalamocortical circuit mediating anesthetic sensitivity. PLoS One 12:e0188087
Bennett, Christopher F; Kwon, Jane J; Chen, Christine et al. (2017) Transaldolase inhibition impairs mitochondrial respiration and induces a starvation-like longevity response in Caenorhabditis elegans. PLoS Genet 13:e1006695
van der Bliek, Alexander M; Sedensky, Margaret M; Morgan, Phil G (2017) Cell Biology of the Mitochondrion. Genetics 207:843-871
Zimin, Pavel I; Woods, Christian B; Quintana, Albert et al. (2016) Glutamatergic Neurotransmission Links Sensitivity to Volatile Anesthetics with Mitochondrial Function. Curr Biol 26:2194-201