CC BY 4.0 · J Neuroanaesth Crit Care 2023; 10(03): 155-166
DOI: 10.1055/s-0043-1778075
Narrative Review

Implications for Anesthesia and Beyond

1   Department of Anesthesiology and Intensive Care, Nizam's Institute of Medical Sciences (NIMS), Telangana, Hyderabad, India
,
1   Department of Anesthesiology and Intensive Care, Nizam's Institute of Medical Sciences (NIMS), Telangana, Hyderabad, India
,
Anupama Baradaa
2   Osmania Medical College, Hyderabad, Telangana, India
› Author Affiliations

Abstract

Gamma-aminobutyric acid (GABA), a nonpeptide amino acid transmitter, is a major component of modern neuropharmacology and one of the most crucial target sites for general anesthetics and therapeutic drugs. GABA type A receptors (GABAARs) are the most abundant inhibitory neurotransmitter receptors in the central nervous system. They are part of the rapid-acting, ligand-gated ion channel (LGIC) receptor category, a pentameric Cys-loop superfamily member that mediates inhibitory neurotransmission in the mature brain. GABAARs mainly consist of two α subunits, two β subunits, and one additional subunit from either γ or δ arranged around a central chloride (Cl-) selective channel. Multiple GABAAR subunit subtypes and splice variants have been identified. Each variant of GABAAR exhibits distinct biophysical and pharmacologic properties. Several compounds allosterically modulate the GABAAR positively or negatively. The widely used positive GABAAR modulators include benzodiazepines (anxiolytic and anticonvulsant), general anesthetics (volatile agents like isoflurane, and intravenous agents like barbiturates, etomidate, and propofol), long-chain alcohols, some anticonvulsants, and neuroactive steroids. The binding sites for each drug are distinctly different. The anesthetic drugs enhance receptor-mediated synaptic transmission and thus interrupt the thalamocortical transmission, which controls the sleep–wake patterns. Abnormality in the GABAAR function has been implicated in several neurological conditions, such as sleep disorders, seizures, depression, cognitive function, neurological recovery after injury, and neuroplasticity. Understanding the GABAAR lays the foundation for the development of highly specific drugs in the treatment of neurological disorders and general anesthesia.



Publication History

Article published online:
27 March 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

 
  • References

  • 1 Markwardt SJ, Dieni CV, Wadiche JI, Overstreet-Wadiche L. Ivy/neurogliaform interneurons coordinate activity in the neurogenic niche. Nat Neurosci 2011; 14 (11) 1407-1409
  • 2 Malcangio M, Bowery NG. GABA and its receptors in the spinal cord. Trends Pharmacol Sci 1996; 17 (12) 457-462
  • 3 Miller PS, Smart TG. Binding, activation and modulation of Cys-loop receptors. Trends Pharmacol Sci 2010; 31 (04) 161-174
  • 4 Beleboni RO, Carolino RO, Pizzo AB. et al. Pharmacological and biochemical aspects of GABAergic neurotransmission: pathological and neuropsychobiological relationships. Cell Mol Neurobiol 2004; 24 (06) 707-728
  • 5 Evenseth LSM, Gabrielsen M, Sylte I. The GABAB receptor-structure, ligand binding and drug development. Molecules 2020; 25 (13) 25
  • 6 Fuxe K, Dahlström AB, Jonsson G. et al. The discovery of central monoamine neurons gave volume transmission to the wired brain. Prog Neurobiol 2010; 90 (02) 82-100
  • 7 Olsen RW, Sieghart W. GABA A receptors: subtypes provide diversity of function and pharmacology. Neuropharmacology 2009; 56 (01) 141-148
  • 8 Ghit A, Assal D, Al-Shami AS, Hussein DEE. GABAA receptors: structure, function, pharmacology, and related disorders. J Genet Eng Biotechnol 2021; 19 (01) 123
  • 9 Zhu S, Noviello CM, Teng J, Walsh Jr RM, Kim JJ, Hibbs RE. Structure of a human synaptic GABAA receptor. Nature 2018; 559 (7712) 67-72
  • 10 Mody I, Pearce RA. Diversity of inhibitory neurotransmission through GABA(A) receptors. Trends Neurosci 2004; 27 (09) 569-575
  • 11 Laverty D, Desai R, Uchański T. et al. Cryo-EM structure of the human α1β3γ2 GABAA receptor in a lipid bilayer. Nature 2019; 565 (7740) 516-520
  • 12 Hwang JH, Yaksh TL. The effect of spinal GABA receptor agonists on tactile allodynia in a surgically-induced neuropathic pain model in the rat. Pain 1997; 70 (01) 15-22
  • 13 Munro G, Lopez-Garcia JA, Rivera-Arconada I. et al. Comparison of the novel subtype-selective GABAA receptor-positive allosteric modulator NS11394 [3′-[5-(1-hydroxy-1-methyl-ethyl)-benzoimidazol-1-yl]-biphenyl-2-carbonitrile] with diazepam, zolpidem, bretazenil, and gaboxadol in rat models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 2008; 327 (03) 969-981
  • 14 Tong Y. Seizures caused by pyridoxine (vitamin B6) deficiency in adults: a case report and literature review. Intractable Rare Dis Res 2014; 3 (02) 52-56
  • 15 Raymond LA. Striatal synaptic dysfunction and altered calcium regulation in Huntington disease. Biochem Biophys Res Commun 2017; 483 (04) 1051-1062
  • 16 Nusser Z, Sieghart W, Somogyi P. Segregation of different GABAA receptors to synaptic and extrasynaptic membranes of cerebellar granule cells. J Neurosci 1998; 18 (05) 1693-1703
  • 17 Farrant M, Nusser Z. Variations on an inhibitory theme: phasic and tonic activation of GABA(A) receptors. Nat Rev Neurosci 2005; 6 (03) 215-229
  • 18 Nakamura Y, Darnieder LM, Deeb TZ, Moss SJ. Regulation of GABAARs by phosphorylation. Adv Pharmacol 2015; 72: 97-146
  • 19 Gielen M, Corringer PJ. The dual-gate model for pentameric ligand-gated ion channels activation and desensitization. J Physiol 2018; 596 (10) 1873-1902
  • 20 Papke D, Gonzalez-Gutierrez G, Grosman C. Desensitization of neurotransmitter-gated ion channels during high-frequency stimulation: a comparative study of Cys-loop, AMPA and purinergic receptors. J Physiol 2011; 589 (Pt 7): 1571-1585
  • 21 Jones MV, Westbrook GL. Desensitized states prolong GABAA channel responses to brief agonist pulses. Neuron 1995; 15 (01) 181-191
  • 22 Bright DP, Renzi M, Bartram J. et al. Profound desensitization by ambient GABA limits activation of δ-containing GABAA receptors during spillover. J Neurosci 2011; 31 (02) 753-763
  • 23 Field M, Dorovykh V, Thomas P, Smart TG. Physiological role for GABAA receptor desensitization in the induction of long-term potentiation at inhibitory synapses. Nat Commun 2021; 12 (01) 2112
  • 24 Forman SA, Miller KW. Anesthetic sites and allosteric mechanisms of action on Cys-loop ligand-gated ion channels. Can J Anaesth 2011; 58 (02) 191-205
  • 25 Olsen RW. GABAA receptor: positive and negative allosteric modulators. Neuropharmacology 2018; 136 (Pt A): 10-22
  • 26 Katayama S, Irifune M, Kikuchi N. et al. Increased gamma-aminobutyric acid levels in mouse brain induce loss of righting reflex, but not immobility, in response to noxious stimulation. Anesth Analg 2007; 104 (06) 1422-1429 table of contents
  • 27 Li GD, Chiara DC, Sawyer GW, Husain SS, Olsen RW, Cohen JB. Identification of a GABAA receptor anesthetic binding site at subunit interfaces by photolabeling with an etomidate analog. J Neurosci 2006; 26 (45) 11599-11605
  • 28 Forman SA, Chiara DC, Miller KW. Anesthetics target interfacial transmembrane sites in nicotinic acetylcholine receptors. Neuropharmacology 2015; 96 (Pt B): 169-177
  • 29 Liu K, Jounaidi Y, Forman SA, Feng HJ. Etomidate uniquely modulates the desensitization of recombinant α1β3δ GABA(A) receptors. Neuroscience 2015; 300: 307-313
  • 30 Enna SJ, McCarson KE. The Role of GABA in the Mediation and Perception of Pain. Adv Pharmacol 2006; 54: 1-27
  • 31 Kleingoor C, Wieland HA, Korpi ER, Seeburg PH, Kettenmann H. Current potentiation by diazepam but not GABA sensitivity is determined by a single histidine residue. Neuroreport 1993; 4 (02) 187-190
  • 32 Brohan J, Goudra BG. The role of GABA receptor agonists in anesthesia and sedation. CNS Drugs 2017; 31 (10) 845-856
  • 33 Krasowski MD, O'Shea SM, Rick CE. et al. α subunit isoform influences GABA(A) receptor modulation by propofol. Neuropharmacology 1997; 36 (07) 941-949
  • 34 Drafts BC, Fisher JL. Identification of structures within GABAA receptor alpha subunits that regulate the agonist action of pentobarbital. J Pharmacol Exp Ther 2006; 318 (03) 1094-1101
  • 35 Akk G, Bracamontes J, Steinbach JH. Activation of GABA(A) receptors containing the alpha4 subunit by GABA and pentobarbital. J Physiol 2004; 556 (Pt 2): 387-399
  • 36 Zhang Y, Wang C, Zhang Y, Zhang L, Yu T. GABAA receptor in the thalamic specific relay system contributes to the propofol-induced somatosensory cortical suppression in rat. PLoS One 2013; 8 (12) e82377
  • 37 Jia F, Yue M, Chandra D, Homanics GE, Goldstein PA, Harrison NL. Isoflurane is a potent modulator of extrasynaptic GABA(A) receptors in the thalamus. J Pharmacol Exp Ther 2008; 324 (03) 1127-1135
  • 38 Garcia PS, Kolesky SE, Jenkins A. General anesthetic actions on GABA(A) receptors. Curr Neuropharmacol 2010; 8 (01) 2-9
  • 39 Fu B, Wang Y, Yang H, Yu T. Effects of etomidate on GABAergic and glutamatergic transmission in rat thalamocortical slices. Neurochem Res 2016; 41 (12) 3181-3191
  • 40 Koyanagi Y, Oi Y, Yamamoto K, Koshikawa N, Kobayashi M. Fast-spiking cell to pyramidal cell connections are the most sensitive to propofol-induced facilitation of GABAergic currents in rat insular cortex. Anesthesiology 2014; 121 (01) 68-78
  • 41 Steinbach JH, Akk G. Modulation of GABA(A) receptor channel gating by pentobarbital. J Physiol 2001; 537 (Pt 3): 715-733
  • 42 Savechenkov PY, Chiara DC, Desai R. et al. Synthesis and pharmacological evaluation of neurosteroid photoaffinity ligands. Eur J Med Chem 2017; 136: 334-347
  • 43 Weir CJ, Mitchell SJ, Lambert JJ. Role of GABAA receptor subtypes in the behavioural effects of intravenous general anaesthetics. Br J Anaesth 2017; 119 (Suppl. 01) i167-i175
  • 44 Lam DW, Reynolds JN. Modulatory and direct effects of propofol on recombinant GABAA receptors expressed in xenopus oocytes: influence of alpha- and gamma2-subunits. Brain Res 1998; 784 (1-2): 179-187
  • 45 Sanna E, Mascia MP, Klein RL, Whiting PJ, Biggio G, Harris RA. Actions of the general anesthetic propofol on recombinant human GABAA receptors: influence of receptor subunits. J Pharmacol Exp Ther 1995; 274 (01) 353-360
  • 46 Hevers W, Hadley SH, Lüddens H, Amin J. Ketamine, but not phencyclidine, selectively modulates cerebellar GABA(A) receptors containing alpha6 and delta subunits. J Neurosci 2008; 28 (20) 5383-5393
  • 47 Chaudhuri KR, Martinez-Martin P. Quantitation of non-motor symptoms in Parkinson's disease. Eur J Neurol 2008; 15 (Suppl. 02) 2-7
  • 48 Chaudhuri KR. The dopaminergic basis of sleep dysfunction and non motor symptoms of Parkinson's disease: evidence from functional imaging. Exp Neurol 2009; 216 (02) 247-248
  • 49 Macdonald RL, Kang JQ, Gallagher MJ. Mutations in GABAA receptor subunits associated with genetic epilepsies. J Physiol 2010; 588 (Pt 11): 1861-1869
  • 50 Errington AC, Cope DW, Crunelli V. Augmentation of tonic GABA(A) inhibition in absence epilepsy: therapeutic value of inverse agonists at extrasynaptic GABA(A) receptors. Adv Pharmacol Sci 2011; 2011: 790590
  • 51 Mody I. Aspects of the homeostaic plasticity of GABAA receptor-mediated inhibition. J Physiol 2005; 562 (Pt 1): 37-46
  • 52 Martin LJ, Bonin RP, Orser BA. The physiological properties and therapeutic potential of alpha5-GABAA receptors. Biochem Soc Trans 2009; 37 (Pt 6): 1334-1337
  • 53 Zurek AA, Yu J, Wang DS. et al. Sustained increase in α5GABAA receptor function impairs memory after anesthesia. J Clin Invest 2014; 124 (12) 5437-5441
  • 54 Clarkson AN, Huang BS, Macisaac SE, Mody I, Carmichael ST. Reducing excessive GABA-mediated tonic inhibition promotes functional recovery after stroke. Nature 2010; 468 (7321) 305-309
  • 55 Levy LM, Ziemann U, Chen R, Cohen LG. Rapid modulation of GABA in sensorimotor cortex induced by acute deafferentation. Ann Neurol 2002; 52 (06) 755-761
  • 56 Mitchell EA, Herd MB, Gunn BG, Lambert JJ, Belelli D. Neurosteroid modulation of GABAA receptors: molecular determinants and significance in health and disease. Neurochem Int 2008; 52 (4-5): 588-595
  • 57 Chisari M, Eisenman LN, Covey DF, Mennerick S, Zorumski CF. The sticky issue of neurosteroids and GABA(A) receptors. Trends Neurosci 2010; 33 (07) 299-306
  • 58 Li P, Bracamontes JR, Manion BD. et al. The neurosteroid 5β-pregnan-3α-ol-20-one enhances actions of etomidate as a positive allosteric modulator of α1β2γ2L GABAA receptors. Br J Pharmacol 2014; 171 (23) 5446-5457
  • 59 Cotten JF, Forman SA, Laha JK. et al. Carboetomidate: a pyrrole analog of etomidate designed not to suppress adrenocortical function. Anesthesiology 2010; 112 (03) 637-644
  • 60 Husain SS, Stewart D, Desai R. et al. p-Trifluoromethyldiazirinyl-etomidate: a potent photoreactive general anesthetic derivative of etomidate that is selective for ligand-gated cationic ion channels. J Med Chem 2010; 53 (17) 6432-6444