Skip to main content
Top
Published in: Journal of Anesthesia 5/2019

01-10-2019 | Anesthetics | Original Article

Propofol inhibits the local activity and connectivity of nuclei in the cortico-reticulo-thalamic loop in rats

Authors: Jing Yang, Wei Wang, Zheng Yong, Weixiu Yuan, Hong Zhang, Weidong Mi

Published in: Journal of Anesthesia | Issue 5/2019

Login to get access

Abstract

Purpose

The present study aimed to investigate the effects of different dosages of propofol, that induced different depths of anesthesia, on the local activity and connectivity of nuclei within the cortico-reticulo-thalamic loops, as well as the release of amino acids in those nuclei.

Methods

The nonlinear dynamics analysis of electroencephalogram, including approximate entropy (ApEn) and cross-ApEn (C-ApEn), was used to analyze the effects of different dosages of propofol on the local activity and connectivity of the important nuclei, including the primary somatosensory cortex (S1), ventroposteromedial thalamic nucleus (VPM), reticular thalamic nucleus (RTN), and oral part of the pontine reticular nucleus (PnO). The levels of glutamate (Glu), γ-aminobutyric acid (GABA), and glycine (Gly) in the S1, VPM, and RTN were detected using cerebral microdialysis.

Results

ApEn was more significantly reduced in the cortex than in the subcortical nuclei from awake to deep anesthesia state induced by propofol, and C-ApEn was also more significantly reduced between cortical nucleus and subcortical nucleus than between subcortical nuclei from awake to deep anesthesia state induced by propofol. Propofol inhibited the levels of Glu in S1 and VPM, but elevated the levels in RTN. Gly level decreased in S1, and GABA level increased in S1 after infusion of propofol.

Conclusions

The cortex rather than the subcortical structures, and the cortex–subcortical connectivity instead of subcortical connectivity might be the more vulnerable targets of propofol during anesthesia.
Literature
1.
go back to reference Bonhomme V, Boveroux P, Brichant JF, Laureys S, Boly M. Neural correlates of consciousness during general anesthesia using functional magnetic resonance imaging. Arch Ital Biol. 2012;150:155–63.PubMed Bonhomme V, Boveroux P, Brichant JF, Laureys S, Boly M. Neural correlates of consciousness during general anesthesia using functional magnetic resonance imaging. Arch Ital Biol. 2012;150:155–63.PubMed
2.
go back to reference Alkire MT, Miller J. General anesthesia and the neural correlates of consciousness. Prog Brain Res. 2005;150:229–44.CrossRefPubMed Alkire MT, Miller J. General anesthesia and the neural correlates of consciousness. Prog Brain Res. 2005;150:229–44.CrossRefPubMed
3.
go back to reference Alkire MT. Loss of effective connectivity during general anesthesia. Int Anesthesiol Clin. 2008;46:55–73.CrossRefPubMed Alkire MT. Loss of effective connectivity during general anesthesia. Int Anesthesiol Clin. 2008;46:55–73.CrossRefPubMed
4.
5.
go back to reference Steriade M. Impact of network activities on neuronal properties in corticothalamic systems. J Neurophysiol. 2001;86:1–39.CrossRefPubMed Steriade M. Impact of network activities on neuronal properties in corticothalamic systems. J Neurophysiol. 2001;86:1–39.CrossRefPubMed
6.
go back to reference White NS, Alkire MT. Impaired thalamocortical connectivity in humans during general anesthetic induced unconsciousness. Neuroimage. 2003;19:402–11.CrossRefPubMed White NS, Alkire MT. Impaired thalamocortical connectivity in humans during general anesthetic induced unconsciousness. Neuroimage. 2003;19:402–11.CrossRefPubMed
7.
go back to reference Boly M, Moran R, Murphy M, Boveroux P, Bruno MA, Noirhomme Q, Ledoux D, Bonhomme V, Brichant JF, Tononi G, Laureys S, Friston K. Connectivity changes underlying spectral EEG changes during propofol-induced loss of consciousness. J Neurosci. 2012;32:7082–90.CrossRefPubMedPubMedCentral Boly M, Moran R, Murphy M, Boveroux P, Bruno MA, Noirhomme Q, Ledoux D, Bonhomme V, Brichant JF, Tononi G, Laureys S, Friston K. Connectivity changes underlying spectral EEG changes during propofol-induced loss of consciousness. J Neurosci. 2012;32:7082–90.CrossRefPubMedPubMedCentral
8.
go back to reference Mhuircheartaigh RN, Rosenorn-Lanng D, Wise R, Jbabdi S, Rogers R, Tracey I. Cortical and subcortical connectivity changes during decreasing levels of consciousness in humans. J Neurosci. 2010;30:9095–102.CrossRefPubMedPubMedCentral Mhuircheartaigh RN, Rosenorn-Lanng D, Wise R, Jbabdi S, Rogers R, Tracey I. Cortical and subcortical connectivity changes during decreasing levels of consciousness in humans. J Neurosci. 2010;30:9095–102.CrossRefPubMedPubMedCentral
9.
go back to reference Silva A, Cardoso-Cruz H, Silva F, Galhardo V, Antunes L. Comparison of anesthetic depth indexes based on thalamocortical local field potentials in rats. Anesthesiology. 2010;112:355–63.CrossRefPubMed Silva A, Cardoso-Cruz H, Silva F, Galhardo V, Antunes L. Comparison of anesthetic depth indexes based on thalamocortical local field potentials in rats. Anesthesiology. 2010;112:355–63.CrossRefPubMed
10.
go back to reference Martuzzi R, Ramani R, Qiu M, Rajeevan N, Constable RT. Functional connectivity and alterations in baseline brain state in humans. Neuroimage. 2010;49:823–34.CrossRefPubMed Martuzzi R, Ramani R, Qiu M, Rajeevan N, Constable RT. Functional connectivity and alterations in baseline brain state in humans. Neuroimage. 2010;49:823–34.CrossRefPubMed
11.
go back to reference Ching S, Cimenser A, Purdon PL, Brown EN, Kopell NJ. Thalamocortical model for a propofol-induced alpha-rhythm associated with loss of consciousness. Proc Natl Acad Sci USA. 2010;107:22665–70.CrossRefPubMedPubMedCentral Ching S, Cimenser A, Purdon PL, Brown EN, Kopell NJ. Thalamocortical model for a propofol-induced alpha-rhythm associated with loss of consciousness. Proc Natl Acad Sci USA. 2010;107:22665–70.CrossRefPubMedPubMedCentral
12.
go back to reference Seo S, Leitch B. Synaptic changes in GABAA receptor expression in the thalamus of the stargazer mouse model of absence epilepsy. Neuroscience. 2015;306:28–38.CrossRefPubMed Seo S, Leitch B. Synaptic changes in GABAA receptor expression in the thalamus of the stargazer mouse model of absence epilepsy. Neuroscience. 2015;306:28–38.CrossRefPubMed
13.
go back to reference Cortes PM, Hernández-Arteaga E, Sotelo-Tapia C, Guevara MA, Medina AC, Hernández-González M. Effects of inactivation of the ventral tegmental area on prefronto-accumbens activity and sexual motivation in male rats. Physiol Behav. 2019;27:112593.CrossRef Cortes PM, Hernández-Arteaga E, Sotelo-Tapia C, Guevara MA, Medina AC, Hernández-González M. Effects of inactivation of the ventral tegmental area on prefronto-accumbens activity and sexual motivation in male rats. Physiol Behav. 2019;27:112593.CrossRef
14.
go back to reference Jelles B, Strijers RL, Hooijer C, Jonker C, Stam CJ, Jonkman EJ. Nonlinear EEG analysis in early Alzheimer’s disease. Acta Neurol Scand. 1999;100:360–8.CrossRefPubMed Jelles B, Strijers RL, Hooijer C, Jonker C, Stam CJ, Jonkman EJ. Nonlinear EEG analysis in early Alzheimer’s disease. Acta Neurol Scand. 1999;100:360–8.CrossRefPubMed
15.
go back to reference Hudetz AG. Effect of volatile anesthetics on interhemispheric EEG crossapproximate entropy in the rat. Brain Res. 2002;954:123–31.CrossRefPubMed Hudetz AG. Effect of volatile anesthetics on interhemispheric EEG crossapproximate entropy in the rat. Brain Res. 2002;954:123–31.CrossRefPubMed
16.
go back to reference Pincus SM, Mulligan T, Iranmanesh A, Gheorghiu S, Godschalk M, Veldhuis JD. Older males secrete luteinizing hormone and testosterone more irregularly, and jointly more asynchronously, than younger males. Proc Natl Acad Sci USA. 1996;93:14100–5.CrossRefPubMedPubMedCentral Pincus SM, Mulligan T, Iranmanesh A, Gheorghiu S, Godschalk M, Veldhuis JD. Older males secrete luteinizing hormone and testosterone more irregularly, and jointly more asynchronously, than younger males. Proc Natl Acad Sci USA. 1996;93:14100–5.CrossRefPubMedPubMedCentral
17.
go back to reference Kotani Y, Shimazawa M, Yoshimura S, Iwama T, Hara H. The experimental and clinical pharmacology of propofol, an anesthetic agent with neuroprotective properties. CNS Neurosci Ther. 2008;14:95–106.CrossRefPubMedPubMedCentral Kotani Y, Shimazawa M, Yoshimura S, Iwama T, Hara H. The experimental and clinical pharmacology of propofol, an anesthetic agent with neuroprotective properties. CNS Neurosci Ther. 2008;14:95–106.CrossRefPubMedPubMedCentral
18.
go back to reference Wu DY, Cai G, Yuan Y, Liu L, Li GQ, Song WQ, Wang MB. Application of nonlinear dynamics analysis in assessing unconsciousness: a preliminary study. Clin Neurophysiol. 2011;122:490–8.CrossRefPubMed Wu DY, Cai G, Yuan Y, Liu L, Li GQ, Song WQ, Wang MB. Application of nonlinear dynamics analysis in assessing unconsciousness: a preliminary study. Clin Neurophysiol. 2011;122:490–8.CrossRefPubMed
20.
go back to reference Velly LJ, Rey MF, Bruder NJ, Gouvitsos FA, Witjas T, Regis JM, Peragut JC, Gouin FM. Differential dynamic of action on cortical and subcortical structures of anesthetic agents during induction of anesthesia. Anesthesiology. 2007;107:202–12.CrossRefPubMed Velly LJ, Rey MF, Bruder NJ, Gouvitsos FA, Witjas T, Regis JM, Peragut JC, Gouin FM. Differential dynamic of action on cortical and subcortical structures of anesthetic agents during induction of anesthesia. Anesthesiology. 2007;107:202–12.CrossRefPubMed
21.
go back to reference Weng L, Xie Q, Zhao L, Zhang R, Ma Q, Wang J, Jiang W, He Y, Chen Y, Li C, Ni X, Xu Q, Yu R, Huang R. Abnormal structural connectivity between the basal ganglia, thalamus, and frontal cortex in patients with disorders of consciousness. Cortex. 2017;90:71–877.CrossRefPubMed Weng L, Xie Q, Zhao L, Zhang R, Ma Q, Wang J, Jiang W, He Y, Chen Y, Li C, Ni X, Xu Q, Yu R, Huang R. Abnormal structural connectivity between the basal ganglia, thalamus, and frontal cortex in patients with disorders of consciousness. Cortex. 2017;90:71–877.CrossRefPubMed
22.
go back to reference Crone JS, Lutkenhoff ES, Bio BJ, Laureys S, Monti MM. Testing proposed neuronal models of effective connectivity within the cortico-basal ganglia-thalamo-cortical loop during loss of consciousness. Cereb Cortex. 2017;27:2727–38.PubMed Crone JS, Lutkenhoff ES, Bio BJ, Laureys S, Monti MM. Testing proposed neuronal models of effective connectivity within the cortico-basal ganglia-thalamo-cortical loop during loss of consciousness. Cereb Cortex. 2017;27:2727–38.PubMed
23.
go back to reference Franks NP. General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nat Rev Neurosci. 2008;9:370–86.CrossRefPubMed Franks NP. General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nat Rev Neurosci. 2008;9:370–86.CrossRefPubMed
24.
go back to reference Vahle-Hinz C, Detsch O, Siemers M, Kochs E. Contributions of GABAergic and glutamatergic mechanisms to isoflurane-induced suppression of thalamic somatosensory information transfer. Exp Brain Res. 2007;176:159–72.CrossRefPubMed Vahle-Hinz C, Detsch O, Siemers M, Kochs E. Contributions of GABAergic and glutamatergic mechanisms to isoflurane-induced suppression of thalamic somatosensory information transfer. Exp Brain Res. 2007;176:159–72.CrossRefPubMed
25.
go back to reference Schlünzen L, Juul N, Hansen KV, Cold GE. Regional cerebral blood flow and glucose metabolism during propofol anaesthesia in healthy subjects studied with positron emission tomography. Acta Anaesthesiol Scand. 2012;56:248–55.CrossRefPubMed Schlünzen L, Juul N, Hansen KV, Cold GE. Regional cerebral blood flow and glucose metabolism during propofol anaesthesia in healthy subjects studied with positron emission tomography. Acta Anaesthesiol Scand. 2012;56:248–55.CrossRefPubMed
27.
go back to reference Song XX, Yu BW. Anesthetic effects of propofol in the healthy human brain: functional imaging evidence. J Anesth. 2015;29:279–88.CrossRefPubMed Song XX, Yu BW. Anesthetic effects of propofol in the healthy human brain: functional imaging evidence. J Anesth. 2015;29:279–88.CrossRefPubMed
28.
go back to reference Lukatch HS, Kiddoo CE, Maciver MB. Anesthetic-induced burst suppression EEG activity requires glutamate-mediated excitatory synaptic transmission. Cereb Cortex. 2005;15:1322–31.CrossRefPubMed Lukatch HS, Kiddoo CE, Maciver MB. Anesthetic-induced burst suppression EEG activity requires glutamate-mediated excitatory synaptic transmission. Cereb Cortex. 2005;15:1322–31.CrossRefPubMed
29.
go back to reference Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62:405–96.CrossRefPubMedPubMedCentral Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62:405–96.CrossRefPubMedPubMedCentral
Metadata
Title
Propofol inhibits the local activity and connectivity of nuclei in the cortico-reticulo-thalamic loop in rats
Authors
Jing Yang
Wei Wang
Zheng Yong
Weixiu Yuan
Hong Zhang
Weidong Mi
Publication date
01-10-2019
Publisher
Springer Japan
Published in
Journal of Anesthesia / Issue 5/2019
Print ISSN: 0913-8668
Electronic ISSN: 1438-8359
DOI
https://doi.org/10.1007/s00540-019-02667-9

Other articles of this Issue 5/2019

Journal of Anesthesia 5/2019 Go to the issue