Etomidate and propylene glycol activate nociceptive TRP ion channels

Florian Niedermirtl, Mirjam Eberhardt, Barbara Namer, Andreas Leffler, Carla Nau, Peter W Reeh, Katrin Kistner, Florian Niedermirtl, Mirjam Eberhardt, Barbara Namer, Andreas Leffler, Carla Nau, Peter W Reeh, Katrin Kistner

Abstract

Background: Etomidate is a preferred drug for the induction of general anesthesia in cardiovascular risk patients. As with propofol and other perioperatively used anesthetics, the application of aqueous etomidate formulations causes an intensive burning pain upon injection. Such algogenic properties of etomidate have been attributed to the solubilizer propylene glycol which represents 35% of the solution administered clinically. The aim of this study was to investigate the underlying molecular mechanisms which lead to injection pain of aqueous etomidate formulations.

Results: Activation of the nociceptive transient receptor potential (TRP) ion channels TRPA1 and TRPV1 was studied in a transfected HEK293t cell line by whole-cell voltage clamp recordings of induced inward ion currents. Calcium influx in sensory neurons of wild-type and trp knockout mice was ratiometrically measured by Fura2-AM staining. Stimulated calcitonin gene-related peptide release from mouse sciatic nerves was detected by enzyme immunoassay. Painfulness of different etomidate formulations was tested in a translational human pain model. Etomidate as well as propylene glycol proved to be effective agonists of TRPA1 and TRPV1 ion channels at clinically relevant concentrations. Etomidate consistently activated TRPA1, but there was also evidence for a contribution of TRPV1 in dependence of drug concentration ranges and species specificities. Distinct N-terminal cysteine and lysine residues seemed to mediate gating of TRPA1, although the electrophile scavenger N-acetyl-L-cysteine did not prevent its activation by etomidate. Propylene glycol-induced activation of TRPA1 and TRPV1 appeared independent of the concomitant high osmolarity. Intradermal injections of etomidate as well as propylene glycol evoked severe burning pain in the human pain model that was absent with emulsification of etomidate.

Conclusions: Data in our study provided evidence that pain upon injection of clinical aqueous etomidate formulations is not an unspecific effect of hyperosmolarity but rather due to a specific action mediated by activated nociceptive TRPA1 and TRPV1 ion channels in sensory neurons.

Keywords: Etomidate; TRPA1; TRPV1; anesthetic; human pain model; injection pain; nociception; propylene glycol; sensory neuron; transient receptor potential ion channel.

Figures

Figure 1.
Figure 1.
Etomidate activates and desensitizes mTRPA1. (a) Representative current traces of etomidate-evoked inward currents in HEK293t cells transiently expressing mTRPA1. To prevent desensitization only one concentration was tested on each cell. Cells were held at −60 mV, etomidate was applied until the current had reached a steady state. (b) Representative current traces evoked by three consecutive applications of 1000 µM etomidate, each applied at intervals of 2 min. (c) Concentration–response curve for etomidate-evoked inward currents in mTRPA1-expressing HEK293t cells; 6 to 10 cells were tested at each concentration. Data were fitted to the Hill equation. (d) Representative etomidate-evoked inward current blocked by the TRPA1 antagonist AP-18. 300 µM etomidate was applied until the current had reached a steady state, followed by a combined application of etomidate and 50 µM AP-18. (e) Normalized current amplitudes ± SEM measured in (d). Currents were normalized to the peak inward current during the steady state. TRP: transient receptor potential.
Figure 2.
Figure 2.
Etomidate activates rTRPV1. (a) Representative current traces evoked by 0.01–2500 µM etomidate in rTRPV1-expressing HEK293t cells. To prevent desensitization, only one concentration was tested on each cell. Cells were held at −60 mV, etomidate was applied for at least 20 s. Note the changed scales at 500 and 2500 µM etomidate. (b) Concentration–response curve for etomidate-evoked inward currents in rTRPV1-expressing HEK293t cells; between 6 and 29 cells were tested at each concentration. Note the bell-shaped curve progression suggesting channel block at etomidate concentrations > 1 µM. (c) Representative capsaicin-evoked inward current of a rTRPV1-expressing HEK293t cell blocked by 100 µM etomidate. Capsaicin (100 nM) was applied until the current had reached a steady state followed by a combined application of capsaicin and 100 µM etomidate. (d) Etomidate did not activate rTRPV2-, rTRPV3-, rTRPV4- or rTRPM8-expressing HEK293t cells. Cells were held at −60 mV, etomidate was applied for at least 20 s. Note the leak current-stabilizing effect of etomidate on rTRPM8. Channel expression was verified by a subsequent application of the TRPV2 and TRPV3 agonist 2-APB, the TRPV4 agonist 4αPDD and the TRPM8 agonist menthol (not shown); at least four cells were tested for each TRP channel. TRP: transient receptor potential.
Figure 3.
Figure 3.
Activation of hTRPA1 by etomidate involves N-terminal cysteine and lysine residues. (a) Representative current traces of etomidate-evoked inward currents in HEK293t cells transiently expressing hTRPA1. To prevent desensitization, only one concentration was tested on each cell. Cells were held at −60 mV, etomidate was applied until the current had reached a steady state. (b) Concentration–response curve for etomidate-evoked inward currents in hTRPA1- and hTRPA1-C621S/C641S/C665S (3C)-expressing HEK293t cells; 6 to 10 cells were tested at each concentration. Data were fitted to the Hill equation. Note the plateau followed by a second slope at concentrations over 100 µM in the curve of hTRPA1. (c) Representative current traces of the hTRPA1 mutants C621S/C641S/C665S (3C) and C621S/C641S/C665S/K710R (3CK) evoked by 1000 µM etomidate. Cells were held at −60 mV, etomidate was applied for at least 25 s. In 3CK-expressing cells, channel expression was verified by a subsequent and effective application of the TRPA1 agonist 2-APB (not shown), while the functionality was tested using a single application of 2-APB 1000 µM per cell. TRP: transient receptor potential; WT: wild type; 2-APB: 2-amino-phenyl borane.
Figure 4.
Figure 4.
Etomidate induces an increase in [Ca2+]i in sensory neurons. (a) Etomidate 300 µM lead to an increase in [Ca2+]i in capsaicin- and AITC-sensitive DRG neurons from C57BL/6 mice during blockade of GABAA-receptors by 100 µM PTX. Etomidate and GABA (30 µM) were applied for 30 s, AITC (100 µM) for 20 s, and capsaicin (0.3 µM) for 10 s. (b) Etomidate 300 µM induced an increase in [Ca2+]i in DRG neurons from C57BL/6 mice. PTX 100 µM reduced etomidate-induced Ca2+ responses. Co-application of PTX and the TRPA1-antagonist AP-18 (15 µM) completely inhibited etomidate-induced Ca2+ increase. Etomidate was applied for 30 s, application of PTX either alone or in combination with AP-18 was started 30 s before and ended 120 s after etomidate. (c) Etomidate 300 µM did not lead to an increase in [Ca2+]i in DRG neurons from Trpa1−/−-mice during blockade of GABAA receptors by 100 µM PTX. Although neurons did not respond to 100 µM AITC applied as a control, 0.3 µM capsaicin evoked an immediate increase in [Ca2+]i in a subpopulation of neurons. Etomidate was applied for 30 s, AITC for 20 s, and capsaicin for 10 s. (d) Etomidate (100 µM)-evoked Ca2+ increases in DRG neurons from C57BL/6 mice correlated better to responses evoked by 30 µM GABA than to responses evoked by 100 µM AITC. Note the inverse correlation at an etomidate concentration of 600 µM. Panels provide the product–momentum correlation coefficient r. (e) Concentration–response curves for etomidate-evoked [Ca2+]i increases in DRG neurons derived from C57BL/6 mice (with and without 100 µM PTX) and Trpa1−/− mice. Etomidate concentrations from 10 to 2000 µM were tested, the curves were fitted to the Hill equation. TRP: transient receptor potential; PTX: picrotoxin.
Figure 5.
Figure 5.
Propylene glycol (PG) activates hTRPA1 and rTRPV1. (a) Representative current traces of PG-evoked inward currents in non-transfected (MOCK) HEK293t cells or cells transiently expressing hTRPA1. To prevent desensitization, only one concentration was tested on each cell. Cells were held at −60 mV, PG was applied until the current had reached a steady state. (b) Concentration–response curve for PG-induced inward currents in hTRPA1-expressing HEK293t cells; 5 to 13 cells were tested at each concentration. Data were fitted to the Hill equation. (c) Representative PG-evoked inward current in an hTRPA1-expressing HEK293t cell blocked by the TRPA1 antagonist HC-030031. PG 10% was applied until the current had reached a steady state followed by a combined application of PG and 50 µM HC-030031. (d) Representative inward current of 10% PG in HEK293t cells expressing rTRPV1. Cells were held at −60 mV, PG was applied until the current had reached a steady state. (e) Representative PG-evoked inward current in an rTRPV1-expressing HEK293t cell blocked by the selective TRPV1 antagonist BCTC. PG 10% was applied until the current had reached a steady state followed by a combined application of PG and 10 µM BCTC. (f) Representative current in response to 1.4 M glucose (osmolarity corresponds approximately to the osmolarity of 10% PG). Note the loss of seal integrity during application of glucose. TRP: transient receptor potential.
Figure 6.
Figure 6.
Etomidate and PG induce release of CGRP. (a) 10-fold diluted clinical formulations of Etomidat-® Lipuro and Hypnomidate® (both 0.8 mM etomidate) did not induce relevant CGRP release from isolated sciatic nerves of C57BL/6 mice. CGRP release was also not caused by 0.8 mM etomidate in SIF with dimethyl sulfoxide as solubilizer. A 10-fold higher etomidate concentration (8 mM) evoked significant release of CGRP as did a 35% PG solution in SIF. (b) Etomidat-induced CGRP release was significantly reduced in sciatic nerves from Trpa1−/−-, Trpv1−/−-, and Trpv1/Trpa1=/=-knockout mice. TRP: transient receptor potential; PG: propylene glycol; CGRP: calcitonin gene-related peptide.
Figure 7.
Figure 7.
Etomidate formulations induce pain and axon reflex vasodilatation in human volunteers. (a) 100 µl Etomidat-® Lipuro (8.2 mM etomidate) or Lipofundin® injected intradermally in volar forearms of volunteers did not induce pain. 100 µl Hypnomidate® (8.2 mM etomidate) or 35% PG in Ringer’s solution caused high values on the numerical rating scale (NRS) in all volunteers indicating severe pain. An 8.2 mM etomidate formulation with methanol as solubilizer caused highest values on the NRS; methanol alone in Ringer`s solution produces significantly lower values on the NRS. (b) Measurements of regional skin perfusion by LDI 2 min after injection. Intradermal injections of 100 µl of Etomidat-® Lipuro, Lipofundin®, Hypnomidate® or 35% PG in Ringer’s solution caused local vasodilatation. Hypnomidate® and 35% PG in Ringer’s solution produced larger flare responses than lipid emulsion-based formulations after 2 min. Left-hand panels display baseline skin perfusion before injection. (c) Quantitative analyses of the flare sizes as area in cm2 developing over time based on the data from (b), showing wider and extended flare reactions upon Hypnomidate® and PG than after Etomidat-® Lipuro. NRS: numerical rating scale.

References

    1. Bovill JG. Intravenous anesthesia for the patient with left ventricular dysfunction. Semin.Cardiothorac.Vasc.Anesth 2006; 10: 43–48.
    1. Desai PM, Kane D, Sarkar MS. Cardioversion: What to choose? Etomidate or propofol. Ann Card Anaesth 2015; 18: 306–311.
    1. Ayhan Y, Akbulut BB, Karahan S, Gecmez G, Öz G, Gurel SC, Basar K. Etomidate is associated with longer seizure duration, lower stimulus intensity, and lower number of failed trials in electroconvulsive therapy compared with thiopental. J Ect 2015; 31: 26–30.
    1. Wagner RL, White PF, Kan PB, Rosenthal MH, Feldman D. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med. 1984; 310: 1415–1421.
    1. Zolle IM, Berger ML, Hammerschmidt F, Hahner S, Schirbel A, Peric-Simov B. New selective inhibitors of steroid 11beta-hydroxylation in the adrenal cortex. Synthesis and structure-activity relationship of potent etomidate analogues. J Med Chem 2008; 51: 2244–2253.
    1. Absalom A, Pledger D, Kong A. Adrenocortical function in critically ill patients 24 h after a single dose of etomidate. Anaesthesia 1999; 54: 861–867.
    1. Malerba G, Romano-Girard F, Cravoisy A, Dousset B, Nace L, Levy B, Bollaert PE. Risk factors of relative adrenocortical deficiency in intensive care patients needing mechanical ventilation. Intensive Care Med 2005; 31: 388–392.
    1. Cuthbertson BH, Sprung CL, Annane D, Chevret S, Garfield M, Goodman S, Laterre PF, Vincent JL, Freivogel K, Reinhart K, Singer M, Payen D, Weiss YG. The effects of etomidate on adrenal responsiveness and mortality in patients with septic shock. Intensive Care Med 2009; 35: 1868–1876.
    1. Fellows IW, Bastow MD, Byrne AJ, Allison SP. Adrenocortical suppression in multiply injured patients: a complication of etomidate treatment. Br Med J (Clin Res Ed.) 1983; 287: 18357.
    1. Bruder EA, Ball IM, Ridi S, Pickett W, Hohl C. Single induction dose of etomidate versus other induction agents for endotracheal intubation in critically ill patients. Cochrane Database Syst Rev 2015; 1: CD010225.
    1. Hendry JG, Miller BM, Lees NW. Etomidate in a new solvent. A clinical evaluation. Anaesthesia 1977; 32: 996–999.
    1. Fragen RJ, Caldwell N, Brunner EA. Clinical use of etomidate for anesthesia induction: a preliminary report. Anesth.Analg 1976; 55: 730–733.
    1. Doenicke AW, Roizen MF, Hoernecke R, Lorenz W, Ostwald P. Solvent for etomidate may cause pain and adverse effects. Br J Anaesth 1999; 83: 464–466.
    1. Doenicke A, Roizen MF, Nebauer AE, Kugler A, Hoernecke R, Beger-Hintzen H. A comparison of two formulations for etomidate, 2-hydroxypropyl-beta-cyclodextrin (HPCD) and propylene glycol. Anesth Analg 1994; 79: 933–939.
    1. Doenicke A, Nebauer AE, Hoernecke R, Mayer M, Roizen MF. Osmolalities of propylene glycol-containing drug formulations for parenteral use. Should propylene glycol be used as a solvent? Anesth Analg 1992; 75: 431–435.
    1. Leffler A, Fischer MJ, Rehner D, Kienel S, Kistner K, Sauer SK, Gavva NR, Reeh PW, Nau C. The vanilloid receptor TRPV1 is activated and sensitized by local anesthetics in rodent sensory neurons. J Clin Invest 2008; 118: 763–776.
    1. Matta JA, Cornett PM, Miyares RL, Abe K, Sahibzada N, Ahern GP. General anesthetics activate a nociceptive ion channel to enhance pain and inflammation. Proc Natl Acad Sci USA 2008; 105: 8784–8789.
    1. Cornett PM, Matta JA, Ahern GP. General anesthetics sensitize the capsaicin receptor transient receptor potential V1. Mol Pharmacol 2008; 74: 1261–1268.
    1. Leffler A, Lattrell A, Kronewald S, Niedermirtl F, Nau C. Activation of TRPA1 by membrane permeable local anesthetics. Mol Pain 2011; 7: 62.
    1. Jordt SE, Bautista DM, Chuang HH, McKemy DD, Zygmunt PM, Hogestatt ED, Meng ID, Julius D. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 2004; 427: 260–265.
    1. Bautista DM, Movahed P, Hinman A, Axelsson HE, Sterner O, Hogestatt ED, Julius D, Jordt SE, Zygmunt PM. Pungent products from garlic activate the sensory ion channel TRPA1. Proc Natl Acad Sci USA 2005; 102: 12248–12252.
    1. Bautista DM, Jordt SE, Nikai T, Tsuruda PR, Read AJ, Poblete J, Yamoah EN, Basbaum AI, Julius D. TRPA1 mediates the inflammatory actions of environmental irritants and proalgesic agents. Cell 2006; 124: 1269–1282.
    1. Kichko TI, Niedermirtl F, Leffler A, Reeh PW. Irritant volatile anesthetics induce neurogenic inflammation through TRPA1 and TRPV1 channels in the isolated mouse trachea. Anesth Analg 2015; 120: 467–471.
    1. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997; 389: 816–824.
    1. Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 2000; 288: 306–313.
    1. Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983; 16: 109–110.
    1. Davis JB, Gray J, Gunthorpe MJ, Hatcher JP, Davey PT, Overend P, Harries MH, Latcham J, Clapham C, Atkinson K, Hughes SA, Rance K, Grau E, Harper AJ, Pugh PL, Rogers DC, Bingham S, Randall A, Sheardown SA. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 2000; 405: 183–187.
    1. Kwan KY, Allchorne AJ, Vollrath MA, Christensen AP, Zhang DS, Woolf CJ, Corey DP. TRPA1 contributes to cold, mechanical, and chemical nociception but is not essential for hair-cell transduction. Neuron 2006; 50: 277–289.
    1. Mohapatra DP, Nau C. Desensitization of capsaicin-activated currents in the vanilloid receptor TRPV1 is decreased by the cyclic AMP-dependent protein kinase pathway. J Biol Chem 2003; 278: 50080–50090.
    1. Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell 2009; 139: 267–284.
    1. Xiao B, Dubin AE, Bursulaya B, Viswanath V, Jegla TJ, Patapoutian A. Identification of transmembrane domain 5 as a critical molecular determinant of menthol sensitivity in mammalian TRPA1 channels. J.Neurosci 2008; 28: 9640–9651.
    1. Macpherson LJ, Dubin AE, Evans MJ, Marr F, Schultz PG, Cravatt BF, Patapoutian A. Noxious compounds activate TRPA1 ion channels through covalent modification of cysteines. Nature 2007; 445: 541–545.
    1. Hinman A, Chuang HH, Bautista DM, Julius D. TRP channel activation by reversible covalent modification. Proc Natl Acad Sci USA 2006; 103: 19564–19568.
    1. Fischer MJ, Leffler A, Niedermirtl F, Kistner K, Eberhardt M, Reeh PW, Nau C. The general anesthetic propofol excites nociceptors by activating TRPV1 and TRPA1 rather than GABAA receptors. J Biol Chem 2010; 285: 34781–34792.
    1. Carr RW, Sittl R, Fleckenstein J, Grafe P. GABA increases electrical excitability in a subset of human unmyelinated peripheral axons. PLoS One 2010; 5: e8780.
    1. Numazaki M, Tominaga T, Toyooka H, Tominaga M. Direct phosphorylation of capsaicin receptor VR1 by protein kinase Cepsilon and identification of two target serine residues. J Biol Chem 2002; 277: 13375–13378.
    1. Lintin DJ. Painful intravenous injections of diazepam. Anaesthesia 1983; 38: 179–180.
    1. Ciura S, Liedtke W, Bourque CW. Hypertonicity sensing in organum vasculosum lamina terminalis neurons: a mechanical process involving TRPV1 but not TRPV4. J Neurosci 2011; 31: 14669–14676.
    1. Zhang XF, Chen J, Faltynek CR, Moreland RB, Neelands TR. Transient receptor potential A1 mediates an osmotically activated ion channel. Eur J Neurosci 2008; 27: 605–611.
    1. Nishihara E, Hiyama TY, Noda M. Osmosensitivity of transient receptor potential vanilloid 1 is synergistically enhanced by distinct activating stimuli such as temperature and protons. PLoS One 2011; 6: e22246.
    1. Fischer MJ, Reeh PW, Sauer SK. Proton-induced calcitonin gene-related peptide release from rat sciatic nerve axons, in vitro, involving TRPV1. Eur J Neurosci 2003; 18: 803–810.
    1. Mogil JS, Miermeister F, Seifert F, Strasburg K, Zimmermann K, Reinold H, Austin JS, Bernardini N, Chesler EJ, Hofmann HA, Hordo C, Messlinger K, Nemmani KV, Rankin AL, Ritchie J, Siegling A, Smith SB, Sotocinal S, Vater A, Lehto SG, Klussmann S, Quirion R, Michaelis M, Devor M, Reeh PW. Variable sensitivity to noxious heat is mediated by differential expression of the CGRP gene. Proc Natl Acad Sci USA 2005; 102: 12938–12943.
    1. Planells-Cases R, Garcia-Sanz N, Morenilla-Palao C, Ferrer-Montiel A. Functional aspects and mechanisms of TRPV1 involvement in neurogenic inflammation that leads to thermal hyperalgesia. Pflugers Arch. 2005; 451: 151–159.
    1. Bandell M, Macpherson LJ, Patapoutian A. From chills to chilis: mechanisms for thermosensation and chemesthesis via thermoTRPs. Curr Opin Neurobiol 2007; 17: 490–497.
    1. Nilius B, Appendino G, Owsianik G. The transient receptor potential channel TRPA1: from gene to pathophysiology. Pflugers Arch 2012; 464: 425–458.
    1. de la Roche J, Eberhardt MJ, Klinger AB, Stanslowsky N, Wegner F, Koppert W, Reeh PW, Lampert A, Fischer MJ, Leffler A. The molecular basis for species-specific activation of human TRPA1 protein by protons involves poorly conserved residues within transmembrane domains 5 and 6. J Biol Chem 2013; 288: 20280–20292.
    1. Eberhardt MJ, Filipovic MR, Leffler A, de la Roche J, Kistner K, Fischer MJ, Fleming T, Zimmermann K, Ivanovic-Burmazovic I, Nawroth PP, Bierhaus A, Reeh PW, Sauer SK. Methylglyoxal activates nociceptors through transient receptor potential channel A1 (TRPA1): a possible mechanism of metabolic neuropathies. J Biol Chem 2012; 287: 28291–28306.
    1. Kim S, Kang C, Shin CY, Hwang SW, Yang YD, Shim WS, Park MY, Kim E, Kim M, Kim BM, Cho H, Shin Y, Oh U. TRPV1 recapitulates native capsaicin receptor in sensory neurons in association with Fas-associated factor 1. J.Neurosci. 2006; 26: 2403–2412.
    1. (accessed 31 October 2018).
    1. Werley MS, McDonald P, Lilly P, Kirkpatrick D, Wallery J, Byron P, Venitz J. Non-clinical safety and pharmacokinetic evaluations of propylene glycol aerosol in Sprague-Dawley rats and Beagle dogs. Toxicology 2011; 287: 76–90.
    1. Clarke RS. Adverse effects of intravenously administered drugs used in anaesthetic practice. Drugs 1981; 22: 26–41.
    1. Klement W, Arndt JO. Pain on injection of propofol: effects of concentration and diluent. Br.J.Anaesth 1991; 67: 281–284.
    1. Soltesz S, Silomon M, Graf G, Mencke T, Boulaadass S, Molter GP. Effect of a 0.5% dilution of propofol on pain on injection during induction of anesthesia in children. Anesthesiology 2007; 106: 80–84.
    1. Zurborg S, Yurgionas B, Jira JA, Caspani O, Heppenstall PA. Direct activation of the ion channel TRPA1 by Ca2+. Nat Neurosci 2007; 10: 277–279.
    1. Spahn V, Stein C, Zöllner C. Modulation of transient receptor vanilloid 1 activity by transient receptor potential ankyrin 1. Mol Pharmacol 2014; 85: 335–344.
    1. Brewster MS, Gaudet R. How the TRPA1 receptor transmits painful stimuli: Inner workings revealed by electron cryomicroscopy. Bioessays 2015; 37: 1184–1192.

Source: PubMed

3
Se inscrever