Association of Polymorphisms in Pharmacogenetic Candidate Genes with Propofol Susceptibility

Qi Zhong, Xiangdong Chen, Yan Zhao, Ru Liu, Shanglong Yao, Qi Zhong, Xiangdong Chen, Yan Zhao, Ru Liu, Shanglong Yao

Abstract

Significant individual susceptibility to intravenous anesthetic propofol exists. The etiology of individual variability in the response to propofol may be influenced by genetic polymorphisms in metabolic and functional pathways. With current pharmacogenetics and modern molecular biology technologies, it is possible to study the influence of genetic polymorphisms on susceptibility to propofol. When inducing general anesthesia with intravenous propofol, high individual susceptibility to propofol was found. Using Sequenom MassARRAY single-nucleotide polymorphism (SNP) genotyping, we identified a mutation (rs6313) in the 5HT2A gene that was correlated to individual susceptibility to propofol effect-site concentration (Cep) and onset time of propofol induction. Carriers of the minor allele (G) of 5HT2A rs6313 required less propofol (20% decrease in Cep) and less time (40% decrease in onset time) to induce anesthesia. Moreover, associations were found between the gamma-aminobutyric acid (GABA) receptor SNP rs2279020 and the SCN9A SNP rs6746030 and the susceptibility of bispectral index (BIS) after propofol-induced anesthesia. In addition, dominant mutations in GABAA1 rs2279020, GABAA2 rs11503014, and CHRM2 rs1824024 were putatively associated with cardiovascular susceptibility to propofol anesthesia. No gene-gene interactions were found through a standardized measure of linkage disequilibrium and a multifactor dimensionality reduction analysis. Our results suggest that genetic polymorphisms related to mechanisms of propofol anesthesia are involved in propofol susceptibility.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Clinical characteristics of patients during anesthesia induction. Anesthesia was induced with propofol via target-controlled-infusion (TCI) at 4 µg/ml. At OAA/S scores of 5, 4, 3, 2, 1, and 0, the effect-site concentration (Cep), bispectral index (BIS), onset time and total propofol were recorded. (A,B,C and D), left, reflect the Cep, BIS, onset time and total propofol recorded at OAA/S scores of 5, 4, 3, 2, 1, and 0. (A,B,C and D), right, reflect the distribution of Cep, BIS, onset time and total propofol at the score of 0, when patients lost consciousness. The different colors in Fig. 1, right side, obviously reflect the wide distribution of Cep, BIS, onset time and total propofol values at the score of 0, when patients lost consciousness. Variance between different groups was analyzed by one-way ANOVA. *P < 0.05 vs. Cep and BIS at an OAA/S score of 5 in (A and B); *P < 0.05 vs. onset time and total propofol at an OAA/S score of 4 in (C and D).
Figure 2
Figure 2
High individual diversity of cardiovascular responses to propofol anesthesia under the condition of unconsciousness in patients. The MAP and HR values of 179 participants at OAA/S scores of 5, 4, 3, 2, 1, and 0 are shown in (A and B), left. (A and B), right, show the distribution of MAP and HR changes under the condition of unconsciousness. The different colors in Fig. 1, right side, obviously reflect the wide distribution of MAP and HR changes at the score of 0, when patients lost consciousness. The variance between different groups was analyzed by one-way ANOVA. *P < 0.05 vs. changes in MAP and HR at an OAA/S score of 4 in (A and B).
Figure 3
Figure 3
Flow chart. Among the 192 participants enrolled in our study, one participant showed hypertension at the induction of anesthesia, and the remaining 13 early terminations were due to other reasons (no continuous monitoring data, no blood provided, or lack of phenotypic data). Based on the SNP results, the patients were divided into two groups. By comparing the clinical characteristics between the two groups, susceptibility to propofol was determined.

References

    1. Iohom G, et al. An investigation of potential genetic determinants of propofol requirements and recovery from anaesthesia. Eur. J. Anaesthesiol. 2007;24:912–919. doi: 10.1017/S0265021507000476.
    1. Ypsilantis P, et al. Attenuation of propofol tolerance conferred by remifentanil co-administration does not reduce propofol toxicity in rabbits under prolonged mechanical ventilation. J. Surg. Res. 2011;168:253–261. doi: 10.1016/j.jss.2009.08.020.
    1. Yang LQ, Li JJ, Chen SQ, Wang YW. Effect of different depths of anesthesia on perioperative stress response in children undergoing adenoidectomy and tonsillectomy. CNS Neurosci. Ther. 2013;19:134–135. doi: 10.1111/cns.12038.
    1. Cremer OL, et al. Long-term propofol infusion and cardiac failure in adult head-injured patients. Lancet. 2001;357:117–118. doi: 10.1016/S0140-6736(00)03547-9.
    1. Kam PC, Cardone D. Propofol infusion syndrome. Anaesthesia. 2007;62:690–701. doi: 10.1111/j.1365-2044.2007.05055.x.
    1. Jung YS, et al. The optimal anesthetic depth for interventional neuroradiology: comparisons between light anesthesia and deep anesthesia. Korean J. Anesthesiol. 2015;68:148–152. doi: 10.4097/kjae.2015.68.2.148.
    1. Iohom G, Fitzgerald D, Cunningham A. Principles of pharmacogenetics—implications for the anaesthetist. Br. J. Anaesth. 2004;93:440–450. doi: 10.1093/bja/aeh200.
    1. Schnider TW, et al. The influence of method of administration and covariates on the pharmacokinetics of propofol in adult volunteers. Anesthesiology. 1998;88:1170–1182. doi: 10.1097/00000542-199805000-00006.
    1. Loryan I, et al. Influence of sex on propofol metabolism, a pilot study: implications for propofol anesthesia. Eur. J. Clin. Pharmacol. 2012;68:397–406. doi: 10.1007/s00228-011-1132-2.
    1. Beer B, et al. Association of polymorphisms in pharmacogenetic candidate genes (OPRD1, GAL, ABCB1, OPRM1) with opioid dependence in European population: a case-control study. PLoS One. 2013;8:e75359. doi: 10.1371/journal.pone.0075359.
    1. Weiser BP, Woll KA, Dailey WP, Eckenhoff RG. Mechanisms revealed through general anesthetic photolabeling. Curr. Anesthesiol. Rep. 2014;4:57–66. doi: 10.1007/s40140-013-0040-7.
    1. Cook-Sather SD, et al. TAOK3, a novel genome-wide association study locus associated with morphine requirement and postoperative pain in a retrospective pediatric day surgery population. Pain. 2014;155:1773–1783. doi: 10.1016/j.pain.2014.05.032.
    1. Thermes V, et al. Medaka simplet (FAM53B) belongs to a family of novel vertebrate genes controlling cell proliferation. Development. 2006;133:1881–1890. doi: 10.1242/dev.02350.
    1. Campos SP, et al. Expression of CYP1A1 and CYP1A2 in the liver and kidney of rabbits after prolonged infusion of propofol. Exp. Toxicol. Pathol. 2016;68:521–531. doi: 10.1016/j.etp.2016.07.006.
    1. Hauer D, et al. Propofol enhances memory formation via an interaction with the endocannabinoid system. Anesthesiology. 2011;114:1380–1388. doi: 10.1097/ALN.0b013e31821c120e.
    1. Moriyama A, et al. Association between genetic polymorphisms of the beta1-adrenergic receptor and sensitivity to pain and fentanyl in patients undergoing painful cosmetic surgery. J. Pharmacol. Sci. 2013;121:48–57. doi: 10.1254/jphs.12159FP.
    1. Martignoni E, et al. Two patients with COMT inhibitor-induced hepatic dysfunction and UGT1A9 genetic polymorphism. Neurology. 2005;65:1820–1822. doi: 10.1212/01.wnl.0000187066.81162.70.
    1. Nakanishi M, et al. The effects of general anesthetics on P2X7 and P2Y receptors in a rat microglial cell line. Anesth. Analg. 2007;104:1136–1144. doi: 10.1213/01.ane.0000260615.12553.4e.
    1. Nagakawa T, Yamazaki M, Hatakeyama N, Stekiel TA. The mechanisms of propofol-mediated hyperpolarization of in situ rat mesenteric vascular smooth muscle. Anesth. Analg. 2003;97:1639–1645. doi: 10.1213/01.ANE.0000087043.61777.1F.
    1. Xu X, Zheng C, An L, Wang R, Zhang T. Effects of dopamine and serotonin systems on modulating neural oscillations in hippocampus-prefrontal cortex pathway in rats. Brain Topogr. 2016;29:539–551. doi: 10.1007/s10548-016-0485-3.
    1. Zhang Y, Yu T, Liu Y, Qian K, Yu BW. Muscarinic M1 receptors regulate propofol modulation of GABAergic transmission in rat ventrolateral preoptic neurons. J. Mol. Neurosci. 2015;55:830–835. doi: 10.1007/s12031-014-0435-z.
    1. Zhou L, et al. The circadian clock gene Csnk1e regulates rapid eye movement sleep amount, and nonrapid eye movement sleep architecture in mice. Sleep. 2014;37:785–793. doi: 10.5665/sleep.3590.
    1. Kinde MN, et al. Common anesthetic-binding site for inhibition of pentameric ligand-gated ion channels. Anesthesiology. 2016;124:664–673. doi: 10.1097/ALN.0000000000001005.
    1. Han L, et al. Propofol-induced inhibition of catecholamine release is reversed by maintaining calcium influx. Anesthesiology. 2016;124:878–884. doi: 10.1097/ALN.0000000000001015.
    1. Moe AAK, et al. Risperidone induces long-lasting changes in the conditioned avoidance response and accumbal gene expression selectively in animals treated as adolescents. Neuropharmacology. 2016;108:264–274. doi: 10.1016/j.neuropharm.2016.04.035.
    1. van Laar M, Volkerts E, Verbaten M. Subchronic effects of the GABA-agonist lorazepam and the 5-HT2A/2C antagonist ritanserin on driving performance, slow wave sleep and daytime sleepiness in healthy volunteers. Psychopharmacology. 2001;154:189–197. doi: 10.1007/s002130000633.
    1. Forman SA, Miller KW. Mapping general anesthetic sites in heteromeric γ-aminobutyric acid type A receptors reveals a potential for targeting receptor subtypes. Anesth. Analg. 2016;123:1263–1273. doi: 10.1213/ANE.0000000000001368.
    1. Trapani G, Altomare C, Liso G, Sanna E, Biggio G. Propofol in anesthesia. Mechanism of action, structure-activity relationships, and drug delivery. Curr. Med. Chem. 2000;7:249–271. doi: 10.2174/0929867003375335.
    1. Corradin O, et al. Modeling disease risk through analysis of physical interactions between genetic variants within chromatin regulatory circuitry. Nat. Genet. 2016;48:1313–1320. doi: 10.1038/ng.3674.
    1. Duan G, et al. A single-nucleotide polymorphism in SCN9A may decrease postoperative pain sensitivity in the general population. Anesthesiology. 2013;118:436–442. doi: 10.1097/ALN.0b013e31827dde74.
    1. Lingamaneni R, Birch ML, Hemmings HC., Jr. Widespread inhibition of sodium channel-dependent glutamate release from isolated nerve terminals by isoflurane and propofol. Anesthesiology. 2001;95:1460–1466. doi: 10.1097/00000542-200112000-00027.
    1. Blanco JH, et al. Chronic cholinergic stimulation promotes changes in cardiovascular autonomic control in spontaneously hypertensive rats. Auton. Neurosci. 2015;193:97–103. doi: 10.1016/j.autneu.2015.09.002.
    1. Hahn LW, Ritchie MD, Moore JH. Multifactor dimensionality reduction software for detecting gene-gene and gene-environment interactions. Bioinformatics. 2003;19:376–382. doi: 10.1093/bioinformatics/btf869.
    1. Hachenberg T. Perioperative management with short-acting intravenous anesthetics. Anaesthesiol. Reanim. 2000;25:144–150.
    1. Court MH, Duan SX, Hesse LM, Venkatakrishnan K, Greenblatt DJ. Cytochrome P-450 2B6 is responsible for interindividual variability of propofol hydroxylation by human liver microsomes. Anesthesiology. 2001;94:110–119. doi: 10.1097/00000542-200101000-00021.
    1. Restoux A, et al. Pilot study of closed-loop anaesthesia for liver transplantation. Br. J. Anaesth. 2016;117:332–340. doi: 10.1093/bja/aew262.
    1. Mourão AL, de Abreu FG, Fiegenbaum M. Impact of the cytochrome P450 2B6 (CYP2B6) gene polymorphism c. 516G> T (rs3745274) on propofol dose variability. Eur. J. Drug Metab. Pharmacokinet. 2015;41:511–515. doi: 10.1007/s13318-015-0289-y.
    1. Mastrogianni O, et al. Association of the CYP2B6 c. 516G> T polymorphism with high blood propofol concentrations in women from northern Greece. Drug Metab. Pharmacokinet. 2014;29:215–218. doi: 10.2133/dmpk.DMPK-13-NT-092.
    1. Lian Q-Q, et al. Impact of CYP2C9 polymorphism found in the Chinese population on the metabolism of propofol in vitro. Biol. Pharm. Bull. 2015;38:531–535. doi: 10.1248/bpb.b14-00671.
    1. Puig MV, Gener T. Serotonin modulation of prefronto-hippocampal rhythms in health and disease. ACS Chem. Neurosci. 2015;6:1017–1025. doi: 10.1021/cn500350e.
    1. Peinado A. Immature neocortical neurons exist as extensive syncitial networks linked by dendrodendritic electrical connections. J. Neurophysiol. 2001;85:620–629.
    1. Moore JT, et al. Direct activation of sleep-promoting VLPO neurons by volatile anesthetics contributes to anesthetic hypnosis. Curr. Biol. 2012;22:2008–2016. doi: 10.1016/j.cub.2012.08.042.
    1. Okamoto K, et al. Activation of central 5HT2A receptors reduces the craniofacial nociception of rats. Neuroscience. 2007;147:1090–1102. doi: 10.1016/j.neuroscience.2007.05.012.
    1. Shyr M-H, et al. Propofol anesthesia increases dopamine and serotonin activities at the somatosensory cortex in rats: a microdialysis study. Anesth. Analg. 1997;84:1344–1348. doi: 10.1213/00000539-199706000-00031.
    1. Brown RE, Basheer R, McKenna JT, Strecker RE, McCarley RW. Control of sleep and wakefulness. Physiol. Rev. 2012;92:1087–1187. doi: 10.1152/physrev.00032.2011.
    1. Enna S, McCarson KE. The role of GABA in the mediation and perception of pain. Adv. Pharmacol. 2005;54:1–27. doi: 10.1016/S1054-3589(06)54001-3.
    1. Brown EN, Lydic R, Schiff ND. General anesthesia, sleep, and coma. N. Engl. J. Med. 2010;363:2638–2650. doi: 10.1056/NEJMra0808281.
    1. Franks NP. General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nat. Rev. Neurosci. 2008;9:370–386. doi: 10.1038/nrn2372.
    1. Minier F, Sigel E. Positioning of the α-subunit isoforms confers a functional signature to γ-aminobutyric acid type A receptors. Proc. Natl. Acad. Sci. USA. 2004;101:7769–7774. doi: 10.1073/pnas.0400220101.
    1. Franks NP. Molecular targets underlying general anaesthesia. Br. J. Pharmacol. 2006;147:S72–S81. doi: 10.1038/sj.bjp.0706441.
    1. Dib-Hajj SD, Yang Y, Black JA, Waxman SG. The NaV1. 7 sodium channel: from molecule to man. Nat. Rev. Neurosci. 2013;14:49–62. doi: 10.1038/nrn3404.
    1. Bennett DL, Woods CG. Painful and painless channelopathies. Lancet Neurol. 2014;13:587–599. doi: 10.1016/S1474-4422(14)70024-9.
    1. Rehberg B, Duch DS. Suppression of central nervous system sodium channels by propofol. Anesthesiology. 1999;91:512–520. doi: 10.1097/00000542-199908000-00026.
    1. Shirasaka T, Yoshimura Y, Qiu D-L, Takasaki M. The effects of propofol on hypothalamic paraventricular nucleus neurons in the rat. Anesth. Analg. 2004;98:1017–1023. doi: 10.1213/01.ANE.0000107960.89818.35.
    1. El Beheiry H, Mak P. Effects of aging and propofol on the cardiovascular component of the autonomic nervous system. J. Clin. Anesth. 2013;25:637–643. doi: 10.1016/j.jclinane.2013.07.004.
    1. Stocker SD, Lang SM, Simmonds SS, Wenner MM, Farquhar WB. Cerebrospinal fluid hypernatremia elevates sympathetic nerve activity and blood pressure via the rostral ventrolateral medulla. Hypertension. 2015;66:1184–1190.
    1. Dewland TA, Androne AS, Lee FA, Lampert RJ, Katz SD. Effect of acetylcholinesterase inhibition with pyridostigmine on cardiac parasympathetic function in sedentary adults and trained athletes. Am. J. Physiol. Heart Circ. Physiol. 2007;293:H86–H92. doi: 10.1152/ajpheart.01339.2006.
    1. Gamou S, Fukuda S, Ogura M, Sakamoto H, Morita S. Microinjection of propofol into the perifornical area induces sedation with decreasing cortical acetylcholine release in rats. Anesth. Analg. 2010;111:395–402. doi: 10.1213/ANE.0b013e3181e24776.
    1. McCormick DA. Neurotransmitter actions in the thalamus and cerebral cortex and their role in neuromodulation of thalamocortical activity. Prog. Neurobiol. 1992;39:337–388. doi: 10.1016/0301-0082(92)90012-4.
    1. Ge DJ, Qi B, Tang G, Li JY. Intraoperative dexmedetomidine promotes postoperative analgesia and recovery in patients after abdominal hysterectomy: a double-blind, randomized clinical trial. Sci. Rep. 2016;6:21514. doi: 10.1038/srep21514.
    1. Chernik DA, et al. Validity and reliability of the Observer’s Assessment of Alertness/Sedation Scale: study with intravenous midazolam. J. Clin. Psychopharmacol. 1990;10:244–251. doi: 10.1097/00004714-199008000-00003.

Source: PubMed

3
Abonnere