1
|
Lansky S, Wang Z, Clarke OB, Chipot C, Scheuring S. Structural dynamics and permeability of the TRPV3 pentamer. Nat Commun 2025; 16:4520. [PMID: 40374654 PMCID: PMC12081643 DOI: 10.1038/s41467-025-59798-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 05/03/2025] [Indexed: 05/17/2025] Open
Abstract
TRPV3 belongs to the large superfamily of tetrameric transient receptor potential (TRP) ion channels. Recently, using high-speed atomic force microscopy (HS-AFM), we discovered a rare and transient pentameric state for TRPV3 that is in equilibrium with the tetrameric state, and, using cryo-EM, we solved a low-resolution structure of the TRPV3 pentamer, in which, however, many residues were unresolved. Here, we present a higher resolution and more complete structure of the pentamer, revealing a domain-swapped architecture, a collapsed vanilloid binding site, and a large pore. Molecular dynamics simulations and potential of mean force calculations of the pentamer establish high protein dynamics and permeability to large cations. Subunit interface analysis, together with thermal denaturation experiments, led us to propose a molecular mechanism of the tetramer-to-pentamer transition, backed experimentally by HS-AFM observations. Collectively, our data demonstrate that the TRPV3 pentamer is in a hyper-activated state with unique, highly permissive permeation properties.
Collapse
Affiliation(s)
- Shifra Lansky
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Zhaokun Wang
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Oliver B Clarke
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Christophe Chipot
- Laboratoire International Associé CNRS and University of Illinois at Urbana-Champaign, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Simon Scheuring
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Philip AB, Brohan J, Goudra B. The Role of GABA Receptors in Anesthesia and Sedation: An Updated Review. CNS Drugs 2025; 39:39-54. [PMID: 39465449 PMCID: PMC11695389 DOI: 10.1007/s40263-024-01128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/29/2024]
Abstract
GABA (γ-aminobutyric acid) receptors are constituents of many inhibitory synapses within the central nervous system. They are formed by 5 subunits out of 19 various subunits: α1-6, β1-3, γ1-3, δ, ε, θ, π, and ρ1-3. Two main subtypes of GABA receptors have been identified, namely GABAA and GABAB. The GABAA receptor (GABAAR) is formed by a variety of combinations of five subunits, although both α and β subunits must be included to produce a GABA-gated ion channel. Other subunits are γ, δ, ε, π, and ϴ. GABAAR has many isoforms, that dictate, among other properties, their differing affinities and conductance. Drugs acting on GABAAR form the cornerstone of anesthesia and sedation practice. Some such GABAAR agonists used in anesthesia practice are propofol, etomidate, methohexital, thiopental, isoflurane, sevoflurane, and desflurane. Ketamine, nitrous oxide, and xenon are not GABAR agonists and instead inhibit glutamate receptors-mainly NMDA receptors. Inspite of its many drawbacks such as pain in injection, quick and uncontrolled conversion from sedation to general anesthesia and dose-related cardiovascular depression, propofol remains the most popular GABAR agonist employed by anesthesia providers. In addition, being formulated in a lipid emulsion, contamination and bacterial growth is possible. Literature is rife with newer propofol formulations, aiming to address many of these drawbacks, and with some degree of success. A nonemulsion propofol formulation has been developed with cyclodextrins, which form inclusion complexes with drugs having lipophilic properties while maintaining aqueous solubility. Inhalational anesthetics are also GABA agonists. The binding sites are primarily located within α+/β- and β+/α- subunit interfaces, with residues in the α+/γ- interface. Isoflurane and sevoflurane might have slightly different binding sites providing unexpected degree of selectivity. Methoxyflurane has made a comeback in Europe for rapid provision of analgesia in the emergency departments. Penthrox (Galen, UK) is the special device designed for its administration. With better understanding of pharmacology of GABAAR agonists, newer sedative agents have been developed, which utilize "soft pharmacology," a term pertaining to agents that are rapidly metabolized into inactive metabolites after producing desired therapeutic effect(s). These newer "soft" GABAAR agonists have many properties of ideal sedative agents, as they can offer well-controlled, titratable activity and ultrashort action. Remimazolam, a modified midazolam and methoxycarbonyl-etomidate (MOC-etomidate), an ultrashort-acting etomidate analog are two such examples. Cyclopropyl methoxycarbonyl metomidate is another second-generation soft etomidate analog that has a greater potency and longer half-life than MOC-etomidate. Additionally, it might not cause adrenal axis suppression. Carboetomidate is another soft analog of etomidate with low affinity for 11β-hydroxylase and is, therefore, unlikely to have clinically significant adrenocortical suppressant effects. Alphaxalone, a GABAAR agonist, is recently formulated in combination with 7-sulfobutylether-β-cyclodextrin (SBECD), which has a low hypersensitivity profile.
Collapse
Affiliation(s)
| | | | - Basavana Goudra
- Department of Anesthesiology, Jefferson Surgical Center Endoscopy, Sidney Kimmel Medical College, Jefferson Health, 111 S 11th Street, #7132, Philadelphia, PA, 19107, USA.
| |
Collapse
|
3
|
Kumamoto E. Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na + and TRP Channels. Biomolecules 2024; 14:1619. [PMID: 39766326 PMCID: PMC11727300 DOI: 10.3390/biom14121619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Nociceptive information is transmitted by action potentials (APs) through primary afferent neurons from the periphery to the central nervous system. Voltage-gated Na+ channels are involved in this AP production, while transient receptor potential (TRP) channels, which are non-selective cation channels, are involved in receiving and transmitting nociceptive stimuli in the peripheral and central terminals of the primary afferent neurons. Peripheral terminal TRP vanilloid-1 (TRPV1), ankylin-1 (TRPA1) and melastatin-8 (TRPM8) activation produces APs, while central terminal TRP activation enhances the spontaneous release of L-glutamate from the terminal to spinal cord and brain stem lamina II neurons that play a pivotal role in modulating nociceptive transmission. There is much evidence demonstrating that chemical compounds involved in Na+ channel (or nerve AP conduction) inhibition modify TRP channel functions. Among these compounds are local anesthetics, anti-epileptics, α2-adrenoceptor agonists, antidepressants (all of which are used as analgesic adjuvants), general anesthetics, opioids, non-steroidal anti-inflammatory drugs and plant-derived compounds, many of which are involved in antinociception. This review mentions the modulation of Na+ channels and TRP channels including TRPV1, TRPA1 and TRPM8, both of which modulations are produced by pain-related compounds.
Collapse
Affiliation(s)
- Eiichi Kumamoto
- Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
4
|
Fang JY, Yamamoto T, Yamamoto S. Propofol-Induced Fasciculations in a Patient With Obstructive Sleep Apnea: A Case Report. Cureus 2024; 16:e75559. [PMID: 39803114 PMCID: PMC11723773 DOI: 10.7759/cureus.75559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
We report a case of a 39-year-old male patient who developed propofol-induced fasciculations during the induction of general anesthesia. The patient had a history of moderate obstructive sleep apnea and was intolerant to continuous positive airway pressure therapy. He subsequently underwent the insertion of a hypoglossal nerve stimulator as a viable surgical intervention. The patient had a drug-induced sleep endoscopy that showed a 100% obstruction at the velum and the oropharynx, mainly in the anteroposterior and lateral directions. The patient experienced a smooth induction and emergence from general anesthesia, except for a brief episode of myoclonus-like movement in the bilateral upper extremities after propofol administration. The patient recovered well and reported an improvement in his sleep quality and daytime symptoms.
Collapse
Affiliation(s)
- Jaden Y Fang
- Anesthesiology, University of Texas Medical Branch, Galveston, USA
| | - Tomohiro Yamamoto
- Department of Medicine, Gunma University School of Medicine, Maebashi, JPN
| | - Satoshi Yamamoto
- Anesthesiology, University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
5
|
Li A, Li N, Zhu L, Xu Z, Wang Y, Li J, Zhang G. The efficacy and safety of ciprofol versus propofol in patients undergoing painless hysteroscopy: a randomized, double-blind, controlled trial. BMC Anesthesiol 2024; 24:411. [PMID: 39533194 PMCID: PMC11555848 DOI: 10.1186/s12871-024-02787-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Studies have reported that ciprofol has the advantage of reducing injection pain compared to propofol during gastroscopy, colonoscopy, and fiberoptic bronchoscopy. The effect of ciprofol on the injection pain in painless hysteroscopy needs to further explore. METHODS A double-blind randomized controlled trial (RCT) was designed, and patients were recruited from the First Central Hospital of Baoding from March 2024 to June 2024. The eligible participants were allocated into ciprofol group (ciprofol combined with alfentanil) and propofol group (propofol combined with alfentanil) at 1:1 ratio. The primary outcome was injection pain. The secondary outcomes included sedation success rate, anesthesia success rate, adverse events, patient satisfaction, and comparison of vital signs before and after administration. RESULTS A total of 217 participants were included for analysis, with 109 participants in the ciprofol group and 108 participants in the propofol group. The injection pain rate of ciprofol group (18.35%) was significantly lower than the propofol group (40.74%). Both the ciprofol group and propofol group had 100% of the sedation success rate. The anesthesia success rate between the two groups was comparable (P > 0.05). The rate of adverse events was lower (27.52% vs. 45.37%) and patient satisfaction was higher (9.84 ± 0.45 vs. 9.65 ± 0.85) in the ciprofol group than the propofol group. In addition, values of systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP) in propofol group were significantly lower than those in ciprofol group at the time of cervical dilation and consciousness recovery. CONCLUSIONS Ciprofol exhibits comparable efficacy to that of propofol, and is associated with less injection pain rate, fewer adverse events, higher patient satisfaction, and more stable hemodynamics when used for general anesthesia during the painless hysteroscopy. CLINICAL TRIAL NUMBER NCT06413862.
Collapse
Affiliation(s)
- Aijun Li
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| | - Ning Li
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| | - Lei Zhu
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China.
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Baoding, P.R. China.
| | - Zige Xu
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| | - Yifan Wang
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| | - Junjie Li
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| | - Gerong Zhang
- Department of Anesthesiology, the First Central Hospital of Baoding, No.443 Wusi East Road, Lianchi District, Baoding, 071000, P.R. China
| |
Collapse
|
6
|
Sahoo TK, Trivedi S, Pedhadiva M, Gupta S, Trivedi G. Efficacy of 6% Hydroxyethyl Starch and 2% Lidocaine in Reducing the Pain of Propofol Injection: A randomised, placebo-controlled trial. Sultan Qaboos Univ Med J 2024; 24:556-561. [PMID: 39634794 PMCID: PMC11614019 DOI: 10.18295/squmj.11.2024.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/25/2024] [Accepted: 08/15/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives Propofol administration is associated with pain, mediated by the activation of vascular endothelium. Hydroxyethyl starch (HES) inhibits endothelial membrane activation by various nociceptive substances. Thus, this study hypothesised that pre-administration of HES can reduce pain on propofol administration. This study aimed to compare the proportion of patients with no pain on propofol administration and to compare the severity of pain and any change in pre- and post-induction haemodynamic variables. Methods This prospective, randomised, placebo-controlled clinical trial was conducted at Chirayu Medical College & Hospital, Bhopal, India, between August 2023 and December 2023 and included patients undergoing elective surgery under general anaesthesia. Patients were randomly assigned to 3 groups to receive either 100 mL of 6% HES followed by propofol (Group HES), 100 mL normal saline (NS) followed by propofol premixed with 2% lidocaine (Group L) or 100 mL NS followed by propofol induction (Group P). Results A total of 339 patients were included. The proportion of patients with no pain on propofol injection was significantly higher in the HES group (n = 75) than in the lignocaine (n = 33) and placebo (n = 13) groups (P <0.0001 each). The median pain scores were 0 (interquartile range [IQR]: 0-1), 1 (IQR: 0-1) and 2 (IQR: 2-3) in the HES, lignocaine and placebo groups, respectively. The proportion of patients with moderate (n = 44) and severe (n = 48) pain scores was significantly higher in the placebo group than in the HES and lignocaine groups (P <0.0001 each). Conclusion The proportion of patients experiencing pain on propofol injection was found to be significantly less with the pre-administration of 100 mL 6% HES compared to the pre-administration of lidocaine.
Collapse
Affiliation(s)
- Tapan K. Sahoo
- Department of Anaesthesia, Chirayu Medical College & Hospital, Bhopal, India
| | - Saurabh Trivedi
- Department of Trauma & Emergency Medicine, All India Institute of Medical Sciences, Bhopal, India
| | - Monika Pedhadiva
- Department of Anaesthesia, Chirayu Medical College & Hospital, Bhopal, India
| | - Seema Gupta
- Department of Anaesthesia, Chirayu Medical College & Hospital, Bhopal, India
| | - Gaurav Trivedi
- Department of Radiation Oncology, All India Institute of Medical Sciences, Raebareli, India
| |
Collapse
|
7
|
Yu H, Gao D, Yang Y, Liu L, Zhao X, Na R. The Interaction Mechanism Between C14-Polyacetylene Compounds and the Rat TRPA1 Receptor: An In Silico Study. Int J Mol Sci 2024; 25:11290. [PMID: 39457072 PMCID: PMC11508972 DOI: 10.3390/ijms252011290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Polyacetylene (PA) compounds, as natural products, exhibit remarkable properties and distinctive chemical activities. Three structurally similar C14-PA compounds-Echinophorin D, Echinophorin B, and Echinophorin A-extracted from plants demonstrate varying biological activities on the Transient Receptor Potential Channel A1 (TRPA1) protein, which belongs to the TRP (Transient Receptor Potential) family. In the current study, we investigated the binding modes of these three PA compounds with TRPA1 using molecular dynamics (MD), molecular docking, binding free energy calculations, and quantum mechanics/molecular mechanics (QM/MM) methods. Initially, a putative binding site (site-II) in TRPA1 was identified for these compounds; Echinophorin B was found to stabilize the upward A-loop of TRPA1, which is critical for its activation. Furthermore, the binding affinity calculations of PA compounds through molecular fragment decomposition indicate that the arrangement of two triple bonds and one double bond in C14-PA compounds is vital for regulating TRPA1 bioactivity. Additionally, the lipophilic and electronic properties of the three molecules were analyzed in relation to binding affinity, establishing a correlation between TRPA1 activity and these molecular properties.
Collapse
Affiliation(s)
- Hui Yu
- College of Science, Beihua University, Jilin 132013, China;
| | - Denghui Gao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China;
| | - Ying Yang
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (Y.Y.); (R.N.)
| | - Lu Liu
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130000, China;
| | - Xi Zhao
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130000, China;
| | - Risong Na
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (Y.Y.); (R.N.)
| |
Collapse
|
8
|
Sohns K, Kostenko A, Behrendt M, Schmelz M, Rukwied R, Carr R. Depolarization of mouse DRG neurons by GABA does not translate into acute pain or hyperalgesia in healthy human volunteers. PLoS One 2024; 19:e0307668. [PMID: 39186592 PMCID: PMC11346724 DOI: 10.1371/journal.pone.0307668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/09/2024] [Indexed: 08/28/2024] Open
Abstract
The majority of somatosensory DRG neurons express GABAA receptors (GABAAR) and depolarise in response to its activation based on the high intracellular chloride concentration maintained by the Na-K-Cl cotransporter type 1 (NKCC1). The translation of this response to peripheral nerve terminals in people is so far unclear. We show here that GABA (EC50 = 16.67μM) acting via GABAAR produces an influx of extracellular calcium in approximately 20% (336/1720) of isolated mouse DRG neurons. In contrast, upon injection into forearm skin of healthy volunteers GABA (1mM, 100μl) did not induce any overt sensations nor a specific flare response and did not sensitize C-nociceptors to slow depolarizing electrical sinusoidal stimuli. Block of the inward chloride transporter NKCC1 by furosemide (1mg/100μl) did not reduce electrically evoked pain ratings nor did repetitive GABA stimulation in combination with an inhibited NKCC1 driven chloride replenishment by furosemide. Finally, we generated a sustained period of C-fiber firing by iontophoretically delivering codeine or histamine to induce tonic itch. Neither the intensity nor the duration of histamine or codeine itch was affected by prior injection of furosemide. We conclude that although GABA can evoke calcium transients in a proportion of isolated mouse DRG neurons, it does not induce or modify pain or itch ratings in healthy human skin even when chloride gradients are altered by inhibition of the sodium coupled NKCC1 transporter.
Collapse
Affiliation(s)
- Kyra Sohns
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Anna Kostenko
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Marc Behrendt
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Martin Schmelz
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Roman Rukwied
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Richard Carr
- Experimental Pain Research, MCTN, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
9
|
Kasuya K, Takahashi K, Hashimoto M, Ohta T. Nociceptive transient receptor potential ankyrin 1 (TRPA1) in sensory neurons are targets of the antifungal drug econazole. BMC Pharmacol Toxicol 2024; 25:53. [PMID: 39169383 PMCID: PMC11337588 DOI: 10.1186/s40360-024-00779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Econazole is a widely used imidazole derivative antifungal for treating skin infections. The molecular targets for its frequent adverse effects of skin irritation symptoms, such as pruritus, burning sensation, and pain, have not been clarified. Transient receptor potential (TRP) channels, non-selective cation channels, are mainly expressed in peripheral sensory neurons and serve as sensors for various irritants. METHODS We investigated the effect of econazole on TRP channel activation by measuring intracellular calcium concentration ([Ca2+]i) through fluorescent ratio imaging in mouse dorsal root ganglion (DRG) neurons isolated from wild-type, TRPA1(-/-) and TRPV1(-/-) mice, as well as in heterologously TRP channel-expressed cells. A cheek injection model was employed to assess econazole-induced itch and pain in vivo. RESULTS Econazole evoked an increase in [Ca2+]i, which was abolished by the removal of extracellular Ca2+ in mouse DRG neurons. The [Ca2+]i responses to econazole were suppressed by a TRPA1 blocker but not by a TRPV1 blocker. Attenuation of the econazole-induced [Ca2+]i responses was observed in the TRPA1(-/-) mouse DRG neurons but was not significant in the TRPV1(-/-) neurons. Econazole increased the [Ca2+]i in HEK293 cells expressing TRPA1 (TRPA1-HEK) but not in those expressing TRPV1, although at higher concentrations, it induced Ca2+ mobilization from intracellular stores in untransfected naïve HEK293 cells. Miconazole, which is a structural analog of econazole, also increased the [Ca2+]i in mouse DRG neurons and TRPA1-HEK, and its nonspecific action was larger than econazole. Fluconazole, a triazole drug failed to activate TRPA1 and TRPV1 in mouse DRG neurons and TRPA1-HEK. Econazole induced itch and pain in wild-type mice, with reduced responses in TRPA1(-/-) mice. CONCLUSIONS These findings suggested that the imidazole derivatives econazole and miconazole may induce skin irritation by activating nociceptive TRPA1 in the sensory neurons. Suppression of TRPA1 activation may mitigate the adverse effects of econazole.
Collapse
Affiliation(s)
- Kaoru Kasuya
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Kenji Takahashi
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Miho Hashimoto
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Toshio Ohta
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan.
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan.
| |
Collapse
|
10
|
Ahlström S, Reiterä P, Jokela R, Olkkola KT, Kaunisto MA, Kalso E. Influence of Clinical and Genetic Factors on Propofol Dose Requirements: A Genome-wide Association Study. Anesthesiology 2024; 141:300-312. [PMID: 38700459 DOI: 10.1097/aln.0000000000005036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
BACKGROUND Propofol is a widely used intravenous hypnotic. Dosing is based mostly on weight, with great interindividual variation in consumption. Suggested factors affecting propofol requirements include age, sex, ethnicity, anxiety, alcohol consumption, smoking, and concomitant valproate use. Genetic factors have not been widely explored. METHODS This study considered 1,000 women undergoing breast cancer surgery under propofol and remifentanil anesthesia. Depth of anesthesia was monitored with State Entropy (GE Healthcare, Finland). Propofol requirements during surgery were recorded. DNA from blood was genotyped with a genome-wide array. A multivariable linear regression model was used to assess the relevance of clinical variables and select those to be used as covariates in a genome-wide association study. Imputed genotype data were used to explore selected loci further. In silico functional annotation was used to explore possible consequences of the discovered genetic variants. Additionally, previously reported genetic associations from candidate gene studies were tested. RESULTS Body mass index, smoking status, alcohol use, remifentanil dose (ln[mg · kg-1 · min-1]), and average State Entropy during surgery remained statistically significant in the multivariable model. Two loci reached genome-wide significance (P < 5 × 10-8). The most significant associations were for single-nucleotide polymorphisms rs997989 (30 kb from ROBO3), likely affecting expression of another nearby gene, FEZ1, and rs9518419, close to NALCN (sodium leak channel); rs10512538 near KCNJ2 encoding the Kir2.1 potassium channel showed suggestive association (P = 4.7 × 10-7). None of these single-nucleotide polymorphisms are coding variants but possibly affect the regulation of nearby genes. None of the single-nucleotide polymorphisms previously reported as affecting propofol pharmacokinetics or pharmacodynamics showed association in the data. CONCLUSIONS In this first genome-wide association study exploring propofol requirements, This study discovered novel genetic associations suggesting new biologically relevant pathways for propofol and general anesthesia. The roles of the gene products of ROBO3/FEZ1, NALCN, and KCNJ2 in propofol anesthesia warrant further studies. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Sirkku Ahlström
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Paula Reiterä
- Department of Public Health, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Ritva Jokela
- HUS Shared Group Services, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Klaus T Olkkola
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland; INDIVIDRUG Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Mari A Kaunisto
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Eija Kalso
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| |
Collapse
|
11
|
Medrado AS, Santiago NAS, Moraes ER, Kushmerick C, Naves LA. Long-term application of adrenergic agonists modulates nociceptive ion channels. Neurosci Lett 2024; 822:137628. [PMID: 38191088 DOI: 10.1016/j.neulet.2024.137628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024]
Abstract
Dorsal root ganglia (DRG) neurons transduce and convey somatosensory information from the periphery to the central nervous system. Adrenergic mediators are known to modulate nociceptive inputs in DRG neurons, acting as up- or down-regulators of neuronal excitability. They are also important in the development of sympathetic neuropathy. ATP-activated P2X channels and capsaicin-activated TRPV1 channels are directly involved in the transduction of nociceptive stimuli. In this work, we show that long-term (up to 3 days) in vitro stimulation of DRG neurons with selective α1-adrenergic agonist increased slow but not fast ATP-activated currents, with no effect on capsaicin currents. Selective agonists for α2, β1 and β3-adrenergic receptors decreased capsaicin activated currents and had no effect on ATP currents. Capsaicin currents were associated with increased neuronal excitability, while none of the adrenergic modulators produced change in rheobase. These results demonstrate that chronic adrenergic activation modulates two nociceptive transducer molecules, increasing or decreasing channel current depending on the adrenergic receptor subtype. These observations aid our understanding of nociceptive or antinociceptive effects of adrenergic agonists.
Collapse
Affiliation(s)
- Aline S Medrado
- Department of Physiology and Biophysics, Institute of Biological Sciences-ICB, Federal University of Minas Gerais-UFMG., Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, Zip-code: 31270-901, Brazil
| | - Naiara A S Santiago
- Department of Physiology and Biophysics, Institute of Biological Sciences-ICB, Federal University of Minas Gerais-UFMG., Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, Zip-code: 31270-901, Brazil
| | - Eder R Moraes
- Department of Physiology and Biophysics, Institute of Biological Sciences-ICB, Federal University of Minas Gerais-UFMG., Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, Zip-code: 31270-901, Brazil.
| | - Christopher Kushmerick
- Department of Physiology and Biophysics, Institute of Biological Sciences-ICB, Federal University of Minas Gerais-UFMG., Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, Zip-code: 31270-901, Brazil
| | - Lígia A Naves
- Department of Physiology and Biophysics, Institute of Biological Sciences-ICB, Federal University of Minas Gerais-UFMG., Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, Zip-code: 31270-901, Brazil
| |
Collapse
|
12
|
Yoshida A, Nishibata M, Maruyama T, Sunami S, Isono K, Kawamata T. Activation of Transient Receptor Potential Vanilloid 1 Is Involved in Both Pain and Tumor Growth in a Mouse Model of Cancer Pain. Neuroscience 2024; 538:80-92. [PMID: 38157977 DOI: 10.1016/j.neuroscience.2023.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/03/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Activation of calcitonin gene-related peptide (CGRP)-positive sensory neurons in the tumor microenvironment has been shown to be involved in tumor growth. However, how CGRP-positive sensory neurons are activated requires elucidation. In this study, we focused on transient receptor potential vanilloid 1 (TRPV1) and examined the contribution of TRPV1 to tumor growth and cancer pain in a mouse cancer model in which Lewis lung carcinoma was subcutaneously inoculated in the left plantar region. Tumor inoculation gradually increased the volumes of the hind paws of wild type (WT) mice over time, but those of both αCGRP knockout mice and TRPV1 knockout mice were significantly smaller than those of WT mice after tumor inoculation. Both TRPV1 and CGRP are therefore suggested to be involved in tumor growth. In an immunohistochemical study, the percentage of phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB)-positive profiles in CGRP-positive dorsal root ganglion (DRG) neurons in WT mice was significantly increased after tumor inoculation. The percentage of p-CREB-positive profiles in CGRP-positive DRG neurons in TRPV1 knockout mice was also increased after tumor inoculation, but was significantly lower than that in WT mice, indicating the contribution of TRPV1 to activation of CGRP-positive DRG neurons. Cancer pain in TRPV1 knockout mice was significantly lower than that in WT mice. In conclusion, TRPV1 is involved in both tumor growth and cancer pain, potentially leading to a novel strategy for the treatment of cancer pain and cancer development. Cancer pain is also suggested to facilitate tumor growth.
Collapse
Affiliation(s)
- Akari Yoshida
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan.
| | - Masayuki Nishibata
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Tomoyuki Maruyama
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Shogo Sunami
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Kyoichi Isono
- Laboratory Animal Center, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Tomoyuki Kawamata
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| |
Collapse
|
13
|
Noguchi S, Kajimoto T, Kumamoto T, Shingai M, Narasaki S, Urabe T, Imamura S, Harada K, Hide I, Tanaka S, Yanase Y, Nakamura SI, Tsutsumi YM, Sakai N. Features and mechanisms of propofol-induced protein kinase C (PKC) translocation and activation in living cells. Front Pharmacol 2023; 14:1284586. [PMID: 38026993 PMCID: PMC10662334 DOI: 10.3389/fphar.2023.1284586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background and purpose: In this study, we aimed to elucidate the action mechanisms of propofol, particularly those underlying propofol-induced protein kinase C (PKC) translocation. Experimental approach: Various PKCs fused with green fluorescent protein (PKC-GFP) or other GFP-fused proteins were expressed in HeLa cells, and their propofol-induced dynamics were observed using confocal laser scanning microscopy. Propofol-induced PKC activation in cells was estimated using the C kinase activity receptor (CKAR), an indicator of intracellular PKC activation. We also examined PKC translocation using isomers and derivatives of propofol to identify the crucial structural motifs involved in this process. Key results: Propofol persistently translocated PKCα conventional PKCs and PKCδ from novel PKCs (nPKCs) to the plasma membrane (PM). Propofol translocated PKCδ and PKCη of nPKCs to the Golgi apparatus and endoplasmic reticulum, respectively. Propofol also induced the nuclear translocation of PKCζ of atypical PKCs or proteins other than PKCs, such that the protein concentration inside and outside the nucleus became uniform. CKAR analysis revealed that propofol activated PKC in the PM and Golgi apparatus. Moreover, tests using isomers and derivatives of propofol predicted that the structural motifs important for the induction of PKC and nuclear translocation are different. Conclusion and implications: Propofol induced the subtype-specific intracellular translocation of PKCs and activated PKCs. Additionally, propofol induced the nuclear translocation of PKCs and other proteins, probably by altering the permeability of the nuclear envelope. Interestingly, propofol-induced PKC and nuclear translocation may occur via different mechanisms. Our findings provide insights into the action mechanisms of propofol.
Collapse
Affiliation(s)
- Soma Noguchi
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taketoshi Kajimoto
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuya Kumamoto
- Department of Synthetic Organic Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masashi Shingai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Soshi Narasaki
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Urabe
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Serika Imamura
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuhki Yanase
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shun-Ichi Nakamura
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuo M. Tsutsumi
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
14
|
Wehrmann T, Riphaus A, Eckardt AJ, Klare P, Kopp I, von Delius S, Rosien U, Tonner PH. Updated S3 Guideline "Sedation for Gastrointestinal Endoscopy" of the German Society of Gastroenterology, Digestive and Metabolic Diseases (DGVS) - June 2023 - AWMF-Register-No. 021/014. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:e654-e705. [PMID: 37813354 DOI: 10.1055/a-2165-6388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Affiliation(s)
- Till Wehrmann
- Clinic for Gastroenterology, DKD Helios Clinic Wiesbaden, Wiesbaden, Germany
| | - Andrea Riphaus
- Internal Medicine, St. Elisabethen Hospital Frankfurt Artemed SE, Frankfurt, Germany
| | - Alexander J Eckardt
- Clinic for Gastroenterology, DKD Helios Clinic Wiesbaden, Wiesbaden, Germany
| | - Peter Klare
- Department Internal Medicine - Gastroenterology, Diabetology, and Hematology/Oncology, Hospital Agatharied, Hausham, Germany
| | - Ina Kopp
- Association of the Scientific Medical Societies in Germany e.V. (AWMF), Berlin, Germany
| | - Stefan von Delius
- Medical Clinic II - Internal Medicine - Gastroenterology, Hepatology, Endocrinology, Hematology, and Oncology, RoMed Clinic Rosenheim, Rosenheim, Germany
| | - Ulrich Rosien
- Medical Clinic, Israelite Hospital, Hamburg, Germany
| | - Peter H Tonner
- Anesthesia and Intensive Care, Clinic Leer, Leer, Germany
| |
Collapse
|
15
|
Mager ML, Ciotu CI, Gold-Binder M, Heber S, Fischer MJM. TRPA1-dependent and -independent activation by commonly used preservatives. Front Pharmacol 2023; 14:1248558. [PMID: 37860113 PMCID: PMC10582264 DOI: 10.3389/fphar.2023.1248558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
Background and purpose: Addition of preservatives ensures microbial stability, especially in multidose containers of parenterally administered pharmaceuticals. These compounds can cause side effects, and particularly at the site of application, might elicit or facilitate pain. TRPA1 is a cation channel expressed in peripheral neurons which contributes to pain and inflammation and is sensitive to many irritants. The most commonly used preservatives, in particular with a focus on parenteral formulations, were investigated for their potential to activate TRPA1. Experimental approach: Sixteen preservatives were screened for mediating calcium influx in human TRPA1-transfected HEK293t cells. Untransfected cells served as control, results were further validated in mouse sensory neurons. In addition, proinflammatory mediators serotonin, histamine and prostaglandin E2 were co-administered to probe a potential sensitisation of preservative-induced TRPA1 activation. Key results: Butylparaben, propylparaben, ethylparaben, bronopol, methylparaben, phenylethyl alcohol and phenol induced a TRPA1-dependent calcium influx in transfected HEK293t cells at concentrations used for preservation. Other preservatives increased calcium within the used concentration ranges, but to a similar degree in untransfected controls. Serotonin, histamine, and prostaglandin enhanced TRPA1 activation of phenylethyl alcohol, bronopol, ethylparaben, propylparaben and butylparaben. Conclusion and implications: Systematic screening of common preservatives applied for parenterally administered drugs resulted in identifying several preservatives with substantial TRPA1 channel activation. This activation was enhanced by the addition of proinflammatory meditators. This allows selecting a preservative without TRPA1 activation, particularly in case of pharmaceuticals that could act proinflammatory.
Collapse
Affiliation(s)
| | | | | | | | - Michael J. M. Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Wehrmann T, Riphaus A, Eckardt AJ, Klare P, Kopp I, von Delius S, Rosien U, Tonner PH. Aktualisierte S3-Leitlinie „Sedierung in der gastrointestinalen Endoskopie“ der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS). ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:1246-1301. [PMID: 37678315 DOI: 10.1055/a-2124-5333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Affiliation(s)
- Till Wehrmann
- Klinik für Gastroenterologie, DKD Helios Klinik Wiesbaden, Wiesbaden, Deutschland
| | - Andrea Riphaus
- Innere Medizin, St. Elisabethen Krankenhaus Frankfurt Artemed SE, Frankfurt, Deutschland
| | - Alexander J Eckardt
- Klinik für Gastroenterologie, DKD Helios Klinik Wiesbaden, Wiesbaden, Deutschland
| | - Peter Klare
- Abteilung Innere Medizin - Gastroenterologie, Diabetologie und Hämato-/Onkologie, Krankenhaus Agatharied, Hausham, Deutschland
| | - Ina Kopp
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin, Deutschland
| | - Stefan von Delius
- Medizinische Klinik II - Innere Medizin - Gastroenterologie, Hepatologie, Endokrinologie, Hämatologie und Onkologie, RoMed Klinikum Rosenheim, Rosenheim, Deutschland
| | - Ulrich Rosien
- Medizinische Klinik, Israelitisches Krankenhaus, Hamburg, Deutschland
| | - Peter H Tonner
- Anästhesie- und Intensivmedizin, Klinikum Leer, Leer, Deutschland
| |
Collapse
|
17
|
Kumar D, Dubey PK, Singh K. Evaluation of Peripheral Versus Central Route of Ondansetron as Pretreatment to Prevent Pain on the Injection of Propofol: A Randomized Controlled Study. Turk J Anaesthesiol Reanim 2023; 51:249-254. [PMID: 37455522 DOI: 10.4274/tjar.2023.221112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Objective We evaluated whether systemic ondansetron was also useful in the attenuation of propofol injection pain similar to ondansetron pretreatment. Methods Eighty patients were enrolled. Patients in group S received ondansetron 4 mg in saline in the right hand followed 30 min later by 5 mL saline in the left hand along with venous occlusion. Group L patients received 4 mL of saline in the right hand followed by 5 mL 4 mg ondansetron in the left hand after 30 min. Two minutes later the occlusion was released. Patients received one-fourth of the calculated total dose of propofol, and their level of pain was graded on a scale of 0 to 3, with 0 denoting no discomfort. Mean blood pressure and heart rates were also recorded. Continuous variables were checked for normality using Shapiro-Wilks test. Normal continuous variables were expressed as mean standard deviation and non-normal continuous variables were expressed as median interquartile range. T-test for the difference in the mean and paired test were used for normally distributed continuous variable whereas Mann-Whitney U test-Wilcoxon test and sign test were used for non-normally distributed variables. Repeated measure analysis of variance was used for a variable measured over different periods of time to control for the baseline effect on subsequent measures. Results Our results demonstrated that both systemic administration 30 min before and local venous pretreatment with ondansetron were equally beneficial in reducing pain during propofol injection. Conclusion A systemic administration of ondansetron may play a role in the attenuation of propofol injection pain.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Anaesthesiology and Critical Care Medicine, Indira Gandhi Institute of Medical Sciences, Patna, India
| | - Prakash K Dubey
- Department of Anaesthesiology and Critical Care Medicine, Indira Gandhi Institute of Medical Sciences, Patna, India
| | - Kunal Singh
- Department of Anaesthesiology and Critical Care Medicine, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
18
|
Modification of the TRP Channel TRPA1 as a Relevant Factor in Migraine-Related Intracranial Hypersensitivity. Int J Mol Sci 2023; 24:ijms24065375. [PMID: 36982450 PMCID: PMC10049246 DOI: 10.3390/ijms24065375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Recently, the transient receptor potential ankyrin 1 (TRPA1) has gained more attention in migraine-related research. The involvement of the TRPA1 receptor in migraine headaches is proposed by the fact that TRPA1 may be a target of some migraine-triggering factors. Although it is doubtful that activation of TRPA1 alone is sufficient to induce pain, behavioral studies have demonstrated that TRPA1 is involved in injury- and inflammation-induced hypersensitivity. Here, we review the functional relevance of TRPA1 in headaches and its therapeutic potential, mainly focusing on its role in the development of hypersensitivity, referring to its altered expression in pathological conditions, and its functional interaction with other TRP channels.
Collapse
|
19
|
Isolation of human TRPA1 channel from transfected HEK293 cells and identification of alkylation sites after sulfur mustard exposure. Arch Toxicol 2023; 97:429-439. [PMID: 36371551 PMCID: PMC9859856 DOI: 10.1007/s00204-022-03411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022]
Abstract
Transient receptor potential (TRP) channels are important in the sensing of pain and other stimuli. They may be triggered by electrophilic agonists after covalent modification of certain cysteine residues. Sulfur mustard (SM) is a banned chemical warfare agent and its reactivity is also based on an electrophilic intermediate. The activation of human TRP ankyrin 1 (hTRPA1) channels by SM has already been documented, however, the mechanism of action is not known in detail. The aim of this work was to purify hTRPA1 channel from overexpressing HEK293 cells for identification of SM-induced alkylation sites. To confirm hTRPA1 isolation, Western blot analysis was performed showing a characteristic double band at 125 kDa. Immunomagnetic separation was carried out using either an anti-His-tag or an anti-hTRPA1 antibody to isolate hTRPA1 from lysates of transfected HEK293 cells. The identity of the channel was confirmed by micro liquid chromatography-electrospray ionization high-resolution tandem-mass spectrometry. Following SM exposure, hTRPA1 channel modifications were found at Cys462 and Cys665, as well as at Asp339 and Glu341 described herein for the first time. Since Cys665 is a well-known target of hTRPA1 agonists and is involved in hTRPA1 activation, SM-induced modifications of cysteine, as well as aspartic acid and glutamic acid residues may play a role in hTRPA1 activation. Considering hTRPA1 as a target of other SM-related chemical warfare agents, analogous adducts may be predicted and identified applying the analytical approach described herein.
Collapse
|
20
|
de Melo IB, Oliveira-Paula GH, Ferezin LP, Ferreira GC, Pinheiro LC, Tanus-Santos JE, Garcia LV, Lacchini R, Paula-Garcia WN. TRPA1 Polymorphisms Modify the Hypotensive Responses to Propofol with No Change in Nitrite or Nitrate Levels. Curr Issues Mol Biol 2022; 44:6333-6345. [PMID: 36547093 PMCID: PMC9777046 DOI: 10.3390/cimb44120432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Anesthesia with propofol is frequently associated with hypotension. The TRPA1 gene contributes to the vasodilator effect of propofol. Hypotension is crucial for anesthesiologists because it is deleterious in the perioperative period. We tested whether the TRPA1 gene polymorphisms or haplotypes interfere with the hypotensive responses to propofol. PCR-determined genotypes and haplotype frequencies were estimated. Nitrite, nitrates, and NOx levels were measured. Propofol induced a more expressive lowering of the blood pressure (BP) without changing nitrite or nitrate levels in patients carrying CG+GG genotypes for the rs16937976 TRPA1 polymorphism and AG+AA genotypes for the rs13218757 TRPA1 polymorphism. The CGA haplotype presented the most remarkable drop in BP. Heart rate values were not impacted. The present exploratory analysis suggests that TRPA1 genotypes and haplotypes influence the hypotensive responses to propofol. The mechanisms involved are probably other than those related to NO bioavailability. With better genetic knowledge, planning anesthesia with fewer side effects may be possible.
Collapse
Affiliation(s)
- Isabela Borges de Melo
- Department of Orthopedics and Anesthesiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Gustavo H. Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Letícia Perticarrara Ferezin
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Graziele C. Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Lucas C. Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Luis V. Garcia
- Department of Orthopedics and Anesthesiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
| | - Waynice N. Paula-Garcia
- Department of Orthopedics and Anesthesiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14048900, SP, Brazil
- Correspondence: ; Tel.: +55-16-3602-2814
| |
Collapse
|
21
|
Torres KV, Pantke S, Rudolf D, Eberhardt MM, Leffler A. The coumarin osthole is a non-electrophilic agonist of TRPA1. Neurosci Lett 2022; 789:136878. [PMID: 36115537 DOI: 10.1016/j.neulet.2022.136878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/30/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022]
Abstract
The naturally occurring coumarin osthole has antipruritic properties, and recent reports suggest that this effect is due an inhibition or desensitization of the cation channels TRPV1 and TRPV3. Osthole was also suggested to activate TRPA1, an effect that should rather be pruritic than antipruritic. Here we characterized the effects of osthole on TRPA1 by means of ratiometric calcium imaging and patch clamp electrophysiology. In HEK 293 expressing human (h) TRPA1, osthole induced a concentration-dependent increase in intracellular calcium that was inhibited by the TRPA1-inhibitor A967079. In mouse dorsal root ganglion (DRG) cells, osthole induced a strong calcium-influx that was partly mediated by TRPA1. Osthole evoked fully reversible membrane currents in whole-cell as well as cell-free inside-out recordings on hTRPA1. Osthole failed to activate the mutant hTRPA1-S873V/T874L, a previously described binding site for the non-electrophilic TRPA1-agonists menthol and carvacrol. The combined application of osthole and carvacrol diminished channel activation, suggesting a competitive binding. Finally, osthole failed to activate TRPM8 and TRPV4 but induced a modest activation of hTRPV1 expressed in HEK 293 cells. We conclude that osthole is a potent non-electrophilic agonist of TRPA1. The relevance of this property for the antipruritic effects needs to be further explored.
Collapse
Affiliation(s)
- Karen V Torres
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Sebastian Pantke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Daniel Rudolf
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Mirjam M Eberhardt
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover 30625, Germany
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
22
|
Ahmed F, Dubey PK, . A. The Valsalva Maneuver Is Not as Effective as Lidocaine for the Attenuation of Pain on Injection of Propofol: A Randomized Controlled Study. Cureus 2022; 14:e25678. [PMID: 35816661 PMCID: PMC9257113 DOI: 10.7759/cureus.25678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background and objective Lidocaine pretreatment is considered the gold standard for attenuating pain on injection of propofol. Valsalva maneuver (VM) causes baroreceptor reflex arc-induced antinociception by increasing the intrathoracic pressure. We aimed to evaluate the efficacy of VM in alleviating the pain on injection of propofol in this randomized comparative study. Methods A total of 90 patients were recruited for this randomized study. They were classified into two groups. Patients in group D received 5-mL 4% lidocaine in saline intravenously while the venous drainage was occluded. Then they were asked to press a rubber ball as hard as they could. Patients in group V received 5-ml saline pretreatment. They were then asked to perform VM by blowing into rubber tubing connected to a manometer and raising and holding the pressure up to 40 mmHg. The verbal response and behavioral signs were recorded with a score corresponding to no, mild, moderate, or severe pain. A t-test was performed to compare the mean of variables between the two groups. The Kolmogorov-Smirnov test was used for testing the equality of the distribution function of pain scores between the groups. Repeated measures analysis of variance (ANOVA) was performed to test the heart rate and mean arterial pressure (MAP) at different points of observation. Results The incidence of pain and pain scores were significantly higher among the patients in the VM group as compared to those in the lidocaine with distraction group. Conclusions VM performed immediately before the injection failed to attenuate the pain produced by propofol as compared to lidocaine pretreatment along with distraction.
Collapse
|
23
|
Mahajan N, Khare P, Kondepudi KK, Bishnoi M. TRPA1: Pharmacology, natural activators and role in obesity prevention. Eur J Pharmacol 2021; 912:174553. [PMID: 34627805 DOI: 10.1016/j.ejphar.2021.174553] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/26/2022]
Abstract
Transient receptor potential ankyrin 1 (TRPA1) channel is a calcium permeable, non-selective cation channel, expressed in the sensory neurons and non-neuronal cells of different tissues. Initially studied for its role in pain and inflammation, TRPA1 has now functionally involved in multiple other physiological functions. TRPA1 channel has been extensively studied for modulation by pungent compounds present in the spices and herbs. In the last decade, the role of TRPA1 agonism in body weight reduction, secretion of hunger and satiety hormones, insulin secretion and thermogenesis, has unveiled the potential of the TRPA1 channel to be used as a preventive target to tackle obesity and associated comorbidities including insulin resistance in type 2 diabetes. In this review, we summarized the recent findings of TRPA1 based dietary/non-dietary modulation for its role in obesity prevention and therapeutics.
Collapse
Affiliation(s)
- Neha Mahajan
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India; Regional Centre for Biotechnology, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Pragyanshu Khare
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India
| | - Kanthi Kiran Kondepudi
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India
| | - Mahendra Bishnoi
- Centre of Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-food Biotechnology Institute (NABI), Knowledge City-Sector-81, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
24
|
Mini-review: The nociceptive sensory functions of the polymodal receptor Transient Receptor Potential Ankyrin Type 1 (TRPA1). Neurosci Lett 2021; 764:136286. [PMID: 34624396 DOI: 10.1016/j.neulet.2021.136286] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 01/23/2023]
Abstract
Over the last 17 years since its cloning in 2003, the receptor-channel TRPA1 has received increasing attention due to its polymodal features and prominent role in pain signaling in a variety of human disease states. While evidence has been accumulating for non-neuronal TRPA1 expression, it is the presence of this channel in nociceptive nerve endings which has taken centre stage, due to its potential clinical ramifications. As a consequence, we shall focus in this review on the sensory functions of TRPA1 related to its expression in the peripheral nervous system. While substantial research has been focused on the putative role of TRPA1 in detecting irritant compounds, noxious cold and mechanical stimuli, the current overall picture is, to some extent, still cloudy. The chemosensory function of the channel is well demonstrated, as well as its involvement in the detection of oxidative and nitrosative stress; however, the other sensory features of TRPA1 have not been fully elucidated yet. The current state of the experimental evidence for these physiological roles of TRPA1 in mammals, and particularly in humans, will be discussed in this review.
Collapse
|
25
|
Rhythmic Change of Cortical Hemodynamic Signals Associated with Ongoing Nociception in Awake and Anesthetized Individuals: An Exploratory Functional Near Infrared Spectroscopy Study. Anesthesiology 2021; 135:877-892. [PMID: 34610092 DOI: 10.1097/aln.0000000000003986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Patients undergoing surgical procedures are vulnerable to repetitive evoked or ongoing nociceptive barrage. Using functional near infrared spectroscopy, the authors aimed to evaluate the cortical hemodynamic signal power changes during ongoing nociception in healthy awake volunteers and in surgical patients under general anesthesia. The authors hypothesized that ongoing nociception to heat or surgical trauma would induce reductions in the power of cortical low-frequency hemodynamic oscillations in a similar manner as previously reported using functional magnetic resonance imaging for ongoing pain. METHODS Cortical hemodynamic signals during noxious stimuli from the fontopolar cortex were evaluated in two groups: group 1, a healthy/conscious group (n = 15, all males) where ongoing noxious and innocuous heat stimulus was induced by a contact thermode to the dorsum of left hand; and group 2, a patient/unconscious group (n = 13, 3 males) receiving general anesthesia undergoing knee surgery. The fractional power of low-frequency hemodynamic signals was compared across stimulation conditions in the healthy awake group, and between patients who received standard anesthesia and those who received standard anesthesia with additional regional nerve block. RESULTS A reduction of the total fractional power in both groups-specifically, a decrease in the slow-5 frequency band (0.01 to 0.027 Hz) of oxygenated hemoglobin concentration changes over the frontopolar cortex-was observed during ongoing noxious stimuli in the healthy awake group (paired t test, P = 0.017; effect size, 0.70), and during invasive procedures in the surgery group (paired t test, P = 0.003; effect size, 2.16). The reduction was partially reversed in patients who received a regional nerve block that likely diminished afferent nociceptive activity (two-sample t test, P = 0.002; effect size, 2.34). CONCLUSIONS These results suggest common power changes in slow-wave cortical hemodynamic oscillations during ongoing nociceptive processing in conscious and unconscious states. The observed signal may potentially promote future development of a surrogate signal to assess ongoing nociception under general anesthesia. EDITOR’S PERSPECTIVE
Collapse
|
26
|
Startek JB, Milici A, Naert R, Segal A, Alpizar YA, Voets T, Talavera K. The Agonist Action of Alkylphenols on TRPA1 Relates to Their Effects on Membrane Lipid Order: Implications for TRPA1-Mediated Chemosensation. Int J Mol Sci 2021; 22:ijms22073368. [PMID: 33806007 PMCID: PMC8037438 DOI: 10.3390/ijms22073368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
The Transient Receptor Potential Ankyrin 1 cation channel (TRPA1) is a broadly-tuned chemosensor expressed in nociceptive neurons. Multiple TRPA1 agonists are chemically unrelated non-electrophilic compounds, for which the mechanisms of channel activation remain unknown. Here, we assess the hypothesis that such chemicals activate TRPA1 by inducing mechanical perturbations in the plasma membrane. We characterized the activation of mouse TRPA1 by non-electrophilic alkylphenols (APs) of different carbon chain lengths in the para position of the aromatic ring. Having discarded oxidative stress and the action of electrophilic mediators as activation mechanisms, we determined whether APs induce mechanical perturbations in the plasma membrane using dyes whose fluorescence properties change upon alteration of the lipid environment. APs activated TRPA1, with potency increasing with their lipophilicity. APs increased the generalized polarization of Laurdan fluorescence and the anisotropy of the fluorescence of 1,6-diphenyl-1,3,5-hexatriene (DPH), also according to their lipophilicity. Thus, the potency of APs for TRPA1 activation is an increasing function of their ability to induce lipid order and membrane rigidity. These results support the hypothesis that TRPA1 senses non-electrophilic compounds by detecting the mechanical alterations they produce in the plasma membrane. This may explain how structurally unrelated non-reactive compounds induce TRPA1 activation and support the role of TRPA1 as an unspecific sensor of potentially noxious compounds.
Collapse
Affiliation(s)
- Justyna B. Startek
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Alina Milici
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Robbe Naert
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Yeranddy A. Alpizar
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; (J.B.S.); (A.M.); (R.N.); (A.S.); (Y.A.A.); (T.V.)
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-330469
| |
Collapse
|
27
|
Maglie R, Souza Monteiro de Araujo D, Antiga E, Geppetti P, Nassini R, De Logu F. The Role of TRPA1 in Skin Physiology and Pathology. Int J Mol Sci 2021; 22:3065. [PMID: 33802836 PMCID: PMC8002674 DOI: 10.3390/ijms22063065] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential ankyrin 1 (TRPA1), a member of the TRP superfamily of channels, acts as 'polymodal cellular sensor' on primary sensory neurons where it mediates the peripheral and central processing of pain, itch, and thermal sensation. However, the TRPA1 expression extends far beyond the sensory nerves. In recent years, much attention has been paid to its expression and function in non-neuronal cell types including skin cells, such as keratinocytes, melanocytes, mast cells, dendritic cells, and endothelial cells. TRPA1 seems critically involved in a series of physiological skin functions, including formation and maintenance of physico-chemical skin barriers, skin cells, and tissue growth and differentiation. TRPA1 appears to be implicated in mechanistic processes in various immunological inflammatory diseases and cancers of the skin, such as atopic and allergic contact dermatitis, psoriasis, bullous pemphigoid, cutaneous T-cell lymphoma, and melanoma. Here, we report recent findings on the implication of TRPA1 in skin physiology and pathophysiology. The potential use of TRPA1 antagonists in the treatment of inflammatory and immunological skin disorders will be also addressed.
Collapse
Affiliation(s)
- Roberto Maglie
- Department of Health Sciences, Section of Dermatology, University of Florence, 50139 Florence, Italy; (R.M.); (E.A.)
| | - Daniel Souza Monteiro de Araujo
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Emiliano Antiga
- Department of Health Sciences, Section of Dermatology, University of Florence, 50139 Florence, Italy; (R.M.); (E.A.)
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (P.G.); (F.D.L.)
| |
Collapse
|
28
|
Souza Monteiro de Araujo D, Nassini R, Geppetti P, De Logu F. TRPA1 as a therapeutic target for nociceptive pain. Expert Opin Ther Targets 2020; 24:997-1008. [PMID: 32838583 PMCID: PMC7610834 DOI: 10.1080/14728222.2020.1815191] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction Chronic pain affects approximatively 30–50% of the population globally. Pathologies such as migraine, diabetic neuropathy, nerve injury and treatment with chemotherapeutic agents, can induce chronic pain. Members of the transient receptor potential (TRP) channels, including the TRP ankyrin 1 (TRPA1), have a major role in pain. Areas covered We focus on TRPA1 as a therapeutic target for pain relief. The structure, localization, and activation of the channel and its implication in different pathways to signal pain are described. This paper underlines the role of pharmacological interventions on TRPA1 to reduce pain in numerous pain conditions. We conducted a literature search in PubMed up to and including July 2020. Expert opinion Our understanding of the molecular mechanisms underlying the sensitization of central and peripheral nociceptive pathways is limited. Preclinical evidence indicates that, in murine models of pain diseases, numerous mechanisms converge on the pathway that encompasses oxidative stress and Schwann cell TRPA1 to sustain chronic pain. Programs to identify and develop treatments to attenuate TRPA1-mediated chronic pain have emerged from this knowledge. Antagonists explored as a novel class of analgesics have a new and promising target in the TRPA1 expressed by peripheral glial cells.
Collapse
Affiliation(s)
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence , Florence, Italy
| |
Collapse
|
29
|
Ton HT, Phan TX, Ahern GP. Inhibition of Ligand-Gated TRPA1 by General Anesthetics. Mol Pharmacol 2020; 98:185-191. [PMID: 32580996 PMCID: PMC7406985 DOI: 10.1124/mol.119.118851] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 05/27/2020] [Indexed: 11/22/2022] Open
Abstract
Several general anesthetics (GAs) produce pain or irritation upon administration, and this occurs predominantly through the activation of the nociceptive ion channel, transient receptor potential ankyrin type 1 (TRPA1). However, the effects of GAs on agonist-mediated TRPA1 activity are unclear. Here we show that a diverse range of noxious and non-noxious volatile anesthetics, at clinically relevant concentrations, inhibit ligand-activated TRPA1 currents. These effects are species-specific; GAs blocks rodent TRPA1 without affecting the Drosophila ortholog. Furthermore, propofol inhibits rodent but not human TRPA1. Analysis of chimeric TRPA1 proteins and mutagenesis combined reveals two amino acid residues located in the S5 domain, Ser876 and Thr877, that are critical for the inhibitory effects of isoflurane and propofol. Introduction of these residues into Drosophila TRPA1 confers anesthetic inhibition. Furthermore, several residues lining the presumptive binding pocket for noxious GAs are not required for the inhibitory effects of GAs. We conclude that anesthetics inhibit TRPA1 by interacting at a site distinct from the activation site. The inhibitory effects of GAs at TRPA1 may contribute to the diverse pharmacological action of these drugs. SIGNIFICANCE STATEMENT: We show that both noxious and non-noxious general anesthetics inhibit agonist-evoked transient receptor potential ankyrin type 1 (TRPA1) activity and identify critical amino acid residues located in the pore domain. Inhibition of TRPA1 may affect pain and vascular signaling during anesthesia.
Collapse
Affiliation(s)
- Hoai T Ton
- Department of Pharmacology and Physiology, Georgetown University, Washington DC (H.T.T., T.X.P., G.P.A.) and Department of Biology, Vinh University, Vinh city, Nghe An, Vietnam (H.T.T., T.X.P.)
| | - Thieu X Phan
- Department of Pharmacology and Physiology, Georgetown University, Washington DC (H.T.T., T.X.P., G.P.A.) and Department of Biology, Vinh University, Vinh city, Nghe An, Vietnam (H.T.T., T.X.P.)
| | - Gerard P Ahern
- Department of Pharmacology and Physiology, Georgetown University, Washington DC (H.T.T., T.X.P., G.P.A.) and Department of Biology, Vinh University, Vinh city, Nghe An, Vietnam (H.T.T., T.X.P.)
| |
Collapse
|
30
|
Kant R, Dubey PK, Ranjan A. Palonosetron Pretreatment is not as Effective as Lignocaine for Attenuation of Pain on Injection of Propofol. Turk J Anaesthesiol Reanim 2020; 48:196-201. [PMID: 32551446 PMCID: PMC7279865 DOI: 10.5152/tjar.2019.79477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/09/2019] [Indexed: 11/24/2022] Open
Abstract
Objective 5-HT3 receptor antagonists are known to possess local anesthetic properties and are commonly used for the alleviation of pain following propofol injection. Palonosetron, a newer molecule, has shown contradictory results for this property. The aim of this study was to compare the effect of palonosetron pretreatment in alleviating propofol injection pain with that of lignocaine. Their comparative effect on various hemodynamic parameters was also evaluated. Methods A total of 100 adult patients were randomly assigned to one of two groups: group L received lignocaine 40 mg in 5 mL of 0.9% saline pretreatment solution and group P received 0.075 mg palonosetron in 5 mL 0.9% of saline pretreatment solution. After 2 minutes, the tourniquet was released and one-fourth of the total calculated dose of propofol was administered, after which the pain assessment was made. The Students t-test was used for comparing the difference of mean between the two groups after testing for equality of variance using F-statistics. Categorical variables were expressed as a percentage, and the Chi-square test was performed to assess the independence of attributes. Repeated-measure analysis of variance was used to compare the change in heart rate and mean arterial pressure over three time points between the two groups. Results The proportion of pain reported by the subjects in the lignocaine group was significantly lower as compared to the subjects in the palonosetron group (p=0.001). No significant difference of mean heart rate and mean arterial pressure was observed between the two groups following these interventions. Conclusion The efficacy of palonosetron in alleviating the pain on injection of propofol was significantly less than that of lignocaine.
Collapse
Affiliation(s)
- Ravi Kant
- Indira Gandhi Institute of Medical Sciences, Patna, India
| | | | - Alok Ranjan
- All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
31
|
Chung CL, Lin YS, Chan NJ, Chen YY, Hsu CC. Hypersensitivity of Airway Reflexes Induced by Hydrogen Sulfide: Role of TRPA1 Receptors. Int J Mol Sci 2020; 21:ijms21113929. [PMID: 32486252 PMCID: PMC7312894 DOI: 10.3390/ijms21113929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/14/2023] Open
Abstract
The activation of capsaicin-sensitive lung vagal (CSLV) afferents can elicit airway reflexes. Hypersensitivity of these afferents is known to contribute to the airway hypersensitivity during airway inflammation. Hydrogen sulfide (H2S) has been suggested as a potential therapeutic agent for airway hypersensitivity diseases, such as asthma, because of its relaxing effect on airway smooth muscle and anti-inflammatory effect. However, it is still unknown whether H2S affects airway reflexes. Our previous study demonstrated that exogenous application of H2S sensitized CSLV afferents and enhanced Ca2+ transients in CSLV neurons. The present study aimed to determine whether the H2S-induced sensitization leads to functional changes in airway reflexes and elevates the electrical excitability of the CSLV neurons. Our results showed that, first and foremost, in anesthetized, spontaneously breathing rats, the inhalation of aerosolized sodium hydrosulfide (NaHS, a donor of H2S; 5 mg/mL, 3 min) caused an enhancement in apneic response evoked by several stimulants of the CSLV afferents. This enhancement effect was found 5 min after NaHS inhalation and returned to control 30 min later. However, NaHS no longer enhanced the apneic response after perineural capsaicin treatment on both cervical vagi that blocked the conduction of CSLV fibers. Furthermore, the enhancing effect of NaHS on apneic response was totally abolished by pretreatment with intravenous HC-030031 (a TRPA1 antagonist; 8 mg/kg), whereas the potentiating effect was not affected by the pretreatment with the vehicle of HC-030031. We also found that intracerebroventricular infusion pretreated with HC-030031 failed to alter the potentiating effect of NaHS on the apneic response. Besides, the cough reflex elicited by capsaicin aerosol was enhanced by inhalation of NaHS in conscious guinea pigs. Nevertheless, this effect was entirely eliminated by pretreatment with HC-030031, not by its vehicle. Last but not least, voltage-clamp electrophysiological analysis of isolated rat CSLV neurons showed a similar pattern of potentiating effects of NaHS on capsaicin-induced inward current, and the involvement of TRPA1 receptors was also distinctly shown. In conclusion, these results suggest that H2S non-specifically enhances the airway reflex responses, at least in part, through action on the TRPA1 receptors expressed on the CSLV afferents. Therefore, H2S should be used with caution when applying for therapeutic purposes in airway hypersensitivity diseases.
Collapse
Affiliation(s)
- Chi-Li Chung
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - You Shuei Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (N.-J.C.); (Y.-Y.C.)
| | - Nai-Ju Chan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (N.-J.C.); (Y.-Y.C.)
| | - Yueh-Yin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (N.-J.C.); (Y.-Y.C.)
| | - Chun-Chun Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (N.-J.C.); (Y.-Y.C.)
- Correspondence:
| |
Collapse
|
32
|
Petersen EN, Pavel MA, Wang H, Hansen SB. Disruption of palmitate-mediated localization; a shared pathway of force and anesthetic activation of TREK-1 channels. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183091. [PMID: 31672538 PMCID: PMC6907892 DOI: 10.1016/j.bbamem.2019.183091] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/15/2019] [Accepted: 09/17/2019] [Indexed: 12/22/2022]
Abstract
TWIK related K+ channel (TREK-1) is a mechano- and anesthetic sensitive channel that when activated attenuates pain and causes anesthesia. Recently the enzyme phospholipase D2 (PLD2) was shown to bind to the channel and generate a local high concentration of phosphatidic acid (PA), an anionic signaling lipid that gates TREK-1. In a biological membrane, the cell harnesses lipid heterogeneity (lipid compartments) to control gating of TREK-1 using palmitate-mediated localization of PLD2. Here we discuss the ability of mechanical force and anesthetics to disrupt palmitate-mediated localization of PLD2 giving rise to TREK-1's mechano- and anesthetic-sensitive properties. The likely consequences of this indirect lipid-based mechanism of activation are discussed in terms of a putative model for excitatory and inhibitory mechano-effectors and anesthetic sensitive ion channels in a biological context. Lastly, we discuss the ability of locally generated PA to reach mM concentrations near TREK-1 and the biophysics of localized signaling. Palmitate-mediated localization of PLD2 emerges as a central control mechanism of TREK-1 responding to mechanical force and anesthetic action. This article is part of a Special Issue entitled: Molecular biophysics of membranes and membrane proteins.
Collapse
Affiliation(s)
- E Nicholas Petersen
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Mahmud Arif Pavel
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Hao Wang
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Scott B Hansen
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
33
|
Kusunoki M, Hayashi M, Shoji T, Uba T, Tanaka H, Sumi C, Matsuo Y, Hirota K. Propofol inhibits stromatoxin-1-sensitive voltage-dependent K + channels in pancreatic β-cells and enhances insulin secretion. PeerJ 2019; 7:e8157. [PMID: 31824770 PMCID: PMC6894434 DOI: 10.7717/peerj.8157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Background Proper glycemic control is an important goal of critical care medicine, including perioperative patient care that can influence patients’ prognosis. Insulin secretion from pancreatic β-cells is generally assumed to play a critical role in glycemic control in response to an elevated blood glucose concentration. Many animal and human studies have demonstrated that perioperative drugs, including volatile anesthetics, have an impact on glucose-stimulated insulin secretion (GSIS). However, the effects of the intravenous anesthetic propofol on glucose metabolism and insulin sensitivity are largely unknown at present. Methods The effect of propofol on insulin secretion under low glucose or high glucose was examined in mouse MIN6 cells, rat INS-1 cells, and mouse pancreatic β-cells/islets. Cellular oxygen or energy metabolism was measured by Extracellular Flux Analyzer. Expression of glucose transporter 2 (GLUT2), potassium channels, and insulin mRNA was assessed by qRT-PCR. Protein expression of voltage-dependent potassium channels (Kv2) was also assessed by immunoblot. Propofol’s effects on potassium channels including stromatoxin-1-sensitive Kv channels and cellular oxygen and energy metabolisms were also examined. Results We showed that propofol, at clinically relevant doses, facilitates insulin secretion under low glucose conditions and GSIS in MIN6, INS-1 cells, and pancreatic β-cells/islets. Propofol did not affect intracellular ATP or ADP concentrations and cellular oxygen or energy metabolism. The mRNA expression of GLUT2 and channels including the voltage-dependent calcium channels Cav1.2, Kir6.2, and SUR1 subunit of KATP, and Kv2 were not affected by glucose or propofol. Finally, we demonstrated that propofol specifically blocks Kv currents in β-cells, resulting in insulin secretion in the presence of glucose. Conclusions Our data support the hypothesis that glucose induces membrane depolarization at the distal site, leading to KATP channel closure, and that the closure of Kv channels by propofol depolarization in β-cells enhances Ca2+ entry, leading to insulin secretion. Because its activity is dependent on GSIS, propofol and its derivatives are potential compounds that enhance and initiate β-cell electrical activity.
Collapse
Affiliation(s)
- Munenori Kusunoki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Mikio Hayashi
- Department of Cell Physiology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Tomohiro Shoji
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takeo Uba
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Hiromasa Tanaka
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Chisato Sumi
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
34
|
Talavera K, Startek JB, Alvarez-Collazo J, Boonen B, Alpizar YA, Sanchez A, Naert R, Nilius B. Mammalian Transient Receptor Potential TRPA1 Channels: From Structure to Disease. Physiol Rev 2019; 100:725-803. [PMID: 31670612 DOI: 10.1152/physrev.00005.2019] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The transient receptor potential ankyrin (TRPA) channels are Ca2+-permeable nonselective cation channels remarkably conserved through the animal kingdom. Mammals have only one member, TRPA1, which is widely expressed in sensory neurons and in non-neuronal cells (such as epithelial cells and hair cells). TRPA1 owes its name to the presence of 14 ankyrin repeats located in the NH2 terminus of the channel, an unusual structural feature that may be relevant to its interactions with intracellular components. TRPA1 is primarily involved in the detection of an extremely wide variety of exogenous stimuli that may produce cellular damage. This includes a plethora of electrophilic compounds that interact with nucleophilic amino acid residues in the channel and many other chemically unrelated compounds whose only common feature seems to be their ability to partition in the plasma membrane. TRPA1 has been reported to be activated by cold, heat, and mechanical stimuli, and its function is modulated by multiple factors, including Ca2+, trace metals, pH, and reactive oxygen, nitrogen, and carbonyl species. TRPA1 is involved in acute and chronic pain as well as inflammation, plays key roles in the pathophysiology of nearly all organ systems, and is an attractive target for the treatment of related diseases. Here we review the current knowledge about the mammalian TRPA1 channel, linking its unique structure, widely tuned sensory properties, and complex regulation to its roles in multiple pathophysiological conditions.
Collapse
Affiliation(s)
- Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Justyna B Startek
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Julio Alvarez-Collazo
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Brett Boonen
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Yeranddy A Alpizar
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Alicia Sanchez
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Robbe Naert
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
35
|
Logashina YA, Korolkova YV, Kozlov SA, Andreev YA. TRPA1 Channel as a Regulator of Neurogenic Inflammation and Pain: Structure, Function, Role in Pathophysiology, and Therapeutic Potential of Ligands. BIOCHEMISTRY (MOSCOW) 2019; 84:101-118. [PMID: 31216970 DOI: 10.1134/s0006297919020020] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
TRPA1 is a cation channel located on the plasma membrane of many types of human and animal cells, including skin sensory neurons and epithelial cells of the intestine, lungs, urinary bladder, etc. TRPA1 is the major chemosensor that also responds to thermal and mechanical stimuli. Substances that activate TRPA1, e.g., allyl isothiocyanates (pungent components of mustard, horseradish, and wasabi), cinnamaldehyde from cinnamon, organosulfur compounds from garlic and onion, tear gas, acrolein and crotonaldehyde from cigarette smoke, etc., cause burning, mechanical and thermal hypersensitivity, cough, eye irritation, sneezing, mucus secretion, and neurogenic inflammation. An increased activity of TRPA1 leads to the emergence of chronic pruritus and allergic dermatitis and is associated with episodic pain syndrome, a hereditary disease characterized by episodes of debilitating pain triggered by stress. TRPA1 is now considered as one of the targets for developing new anti-inflammatory and analgesic drugs. This review summarizes information on the structure, function, and physiological role of this channel, as well as describes known TRPA1 ligands and their significance as therapeutic agents in the treatment of inflammation-associated pain.
Collapse
Affiliation(s)
- Yu A Logashina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia
| | - Yu V Korolkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - S A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Ya A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia. .,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia
| |
Collapse
|
36
|
Takayama Y, Derouiche S, Maruyama K, Tominaga M. Emerging Perspectives on Pain Management by Modulation of TRP Channels and ANO1. Int J Mol Sci 2019; 20:E3411. [PMID: 31336748 PMCID: PMC6678529 DOI: 10.3390/ijms20143411] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022] Open
Abstract
Receptor-type ion channels are critical for detection of noxious stimuli in primary sensory neurons. Transient receptor potential (TRP) channels mediate pain sensations and promote a variety of neuronal signals that elicit secondary neural functions (such as calcitonin gene-related peptide [CGRP] secretion), which are important for physiological functions throughout the body. In this review, we focus on the involvement of TRP channels in sensing acute pain, inflammatory pain, headache, migraine, pain due to fungal infections, and osteo-inflammation. Furthermore, action potentials mediated via interactions between TRP channels and the chloride channel, anoctamin 1 (ANO1), can also generate strong pain sensations in primary sensory neurons. Thus, we also discuss mechanisms that enhance neuronal excitation and are dependent on ANO1, and consider modulation of pain sensation from the perspective of both cation and anion dynamics.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Sandra Derouiche
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Makoto Tominaga
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
37
|
Liu Y, Yang H, Sun C, Wang Z, Liu Z. Protective effects of TRPV1 inhibition against sevoflurane-induced cell death. Neurosci Lett 2019; 707:134270. [PMID: 31102705 DOI: 10.1016/j.neulet.2019.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/11/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022]
Abstract
Sevoflurane is a widely used inhalational anesthetic that can induce developmental neurotoxicity, leading to cognitive dysfunction. In this study, we assessed the role of transient receptor potential vanilloid 1 (TRPV1) in mediating sevoflurane activation and whether the TRPV1 antagonist could prevent anesthesia-induced cell death. Here, we demonstrated that the expression of TRPV1 was increased after sevoflurane treatment, and pretreatment with TRPV1 antagonist SB366791 could attenuate the effect of sevoflurane on TRPV1 expression. Moreover, the inhibition of TRPV1 could prevent sevoflurane-induced cell death. The findings of this study provide novel insights into the treatment of general anesthesia-induced developmental neurotoxicity and even cognitive impairment.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Han Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Chengsan Sun
- Department of Psychology, PO Box 400400, University of Virginia, Charlottesville, VA, 22904-4400, United States
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Zhiheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
38
|
Conklin DJ, Guo Y, Nystoriak MA, Jagatheesan G, Obal D, Kilfoil PJ, Hoetker JD, Guo L, Bolli R, Bhatnagar A. TRPA1 channel contributes to myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2019; 316:H889-H899. [PMID: 30735434 PMCID: PMC6483018 DOI: 10.1152/ajpheart.00106.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 01/18/2019] [Accepted: 01/31/2019] [Indexed: 11/22/2022]
Abstract
Myocardial ischemia-reperfusion (I/R) results in the generation of free radicals, accumulation of lipid peroxidation-derived unsaturated aldehydes, variable angina (pain), and infarction. The transient receptor potential ankyrin 1 (TRPA1) mediates pain signaling and is activated by unsaturated aldehydes, including acrolein and 4-hydroxynonenal. The contribution of TRPA1 (a Ca2+-permeable channel) to I/R-induced myocardial injury is unknown. We tested the hypothesis that cardiac TRPA1 confers myocyte sensitivity to aldehyde accumulation and promotes I/R injury. Although basal cardiovascular function in TRPA1-null mice was similar to that in wild-type (WT) mice, infarct size was significantly smaller in TRPA1-null mice than in WT mice (34.1 ± 9.3 vs. 14.3 ± 9.9% of the risk region, n = 8 and 7, respectively, P < 0.05), despite a similar I/R-induced area at risk (40.3 ±8.4% and 42.2 ± 11.3% for WT and TRPA1-null mice, respectively) after myocardial I/R (30 min of ischemia followed by 24 h of reperfusion) in situ. Positive TRPA1 immunofluorescence was present in murine and human hearts and was colocalized with connexin43 at intercalated disks in isolated murine cardiomyocytes. Cardiomyocyte TRPA1 was confirmed by quantitative RT-PCR, DNA sequencing, Western blot analysis, and electrophysiology. A role of TRPA1 in cardiomyocyte toxicity was demonstrated in isolated cardiomyocytes exposed to acrolein, an I/R-associated toxin that induces Ca2+ accumulation and hypercontraction, effects significantly blunted by HC-030031, a TRPA1 antagonist. Protection induced by HC-030031 was quantitatively equivalent to that induced by SN-6, a Na+/Ca2+ exchange inhibitor, further supporting a role of Ca2+ overload in acrolein-induced cardiomyocyte toxicity. These data indicate that cardiac TRPA1 activation likely contributes to I/R injury and, thus, that TRPA1 may be a novel therapeutic target for decreasing myocardial I/R injury. NEW & NOTEWORTHY Transient receptor potential ankyrin 1 (TRPA1) activation mediates increased blood flow, edema, and pain reception, yet its role in myocardial ischemia-reperfusion (I/R) injury is unknown. Genetic ablation of TRPA1 significantly decreased myocardial infarction after I/R in mice. Functional TRPA1 in cardiomyocytes was enriched in intercalated disks and contributed to acrolein-induced Ca2+ overload and hypercontraction. These data indicate that I/R activation of TRPA1 worsens myocardial infarction; TRPA1 may be a potential target to mitigate I/R injury.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Yiru Guo
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Matthew A Nystoriak
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Ganapathy Jagatheesan
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Detlef Obal
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Anesthesiology and Perioperative Medicine, University of Louisville , Louisville, Kentucky
| | - Peter J Kilfoil
- Smidt Heart Institute, Cedars-Sinai Hospital , Los Angeles, California
| | - Joseph David Hoetker
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Luping Guo
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Roberto Bolli
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| |
Collapse
|
39
|
Kita T, Uchida K, Kato K, Suzuki Y, Tominaga M, Yamazaki J. FK506 (tacrolimus) causes pain sensation through the activation of transient receptor potential ankyrin 1 (TRPA1) channels. J Physiol Sci 2019; 69:305-316. [PMID: 30478741 PMCID: PMC10717736 DOI: 10.1007/s12576-018-0647-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
FK506 (tacrolimus) is an immunosuppressant widely used as an ointment in the treatment of atopic dermatitis. However, local application of FK506 can evoke burning sensations in atopic dermatitis patients, and its mechanisms are unknown. In this study, we found that FK506 activates transient receptor potential ankyrin 1 (TRPA1) channels. In Ca2+-imaging experiments, increases in intracellular Ca2+ concentrations ([Ca2+]i) by FK506 were observed in HEK293T cells expressing hTRPA1 or hTRPM8. FK506-induced currents were observed in HEK293T cells expressing hTRPA1 or mTRPA1, but less or not at all in cells expressing hTRPV1 or hTRPM8 using a patch-clamp technique. FK506 also evoked single-channel opening of hTRPA1 in an inside-out configuration. FK506-induced [Ca2+]i increases were also observed in TRPA1-expressing mouse primary sensory neurons. Furthermore, injection of FK506 evoked licking or biting behaviors and these behaviors were almost abolished in TRPA1 knockout mice. These results indicate that FK506 might cause pain sensations through TRPA1 activation.
Collapse
Affiliation(s)
- Tomo Kita
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Kunitoshi Uchida
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan.
| | - Kenichi Kato
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Yoshiro Suzuki
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
| | - Jun Yamazaki
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| |
Collapse
|
40
|
Finley J. Cellular stress and AMPK links metformin and diverse compounds with accelerated emergence from anesthesia and potential recovery from disorders of consciousness. Med Hypotheses 2019; 124:42-52. [PMID: 30798915 DOI: 10.1016/j.mehy.2019.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/19/2019] [Indexed: 01/23/2023]
Abstract
The neural correlates of consciousness and the mechanisms by which general anesthesia (GA) modulate such correlates to induce loss of consciousness (LOC) has been described as one of the biggest mysteries of modern medicine. Several cellular targets and neural circuits have been identified that play a critical role in LOC induced by GA, including the GABAA receptor and ascending arousal nuclei located in the basal forebrain, hypothalamus, and brain stem. General anesthetics (GAs) including propofol and inhalational agents induce LOC in part by potentiating chloride influx through the GABAA receptor, leading to neural inhibition and LOC. Interestingly, nearly all GAs used clinically may also induce paradoxical excitation, a phenomenon in which GAs promote neuronal excitation at low doses before inducing unconsciousness. Additionally, emergence from GA, a passive process that occurs after anesthetic removal, is associated with lower anesthetic concentrations in the brain compared to doses associated with induction of GA. AMPK, an evolutionarily conserved kinase activated by cellular stress (e.g. increases in calcium [Ca2+] and/or reactive oxygen species [ROS], etc.) increases lifespan and healthspan in several model organisms. AMPK is located throughout the mammalian brain, including in neurons of the thalamus, hypothalamus, and striatum as well as in pyramidal neurons in the hippocampus and cortex. Increases in ROS and Ca2+ play critical roles in neuronal excitation and glutamate, the primary excitatory neurotransmitter in the human brain, activates AMPK in cortical neurons. Nearly every neurotransmitter released from ascending arousal circuits that promote wakefulness, arousal, and consciousness activates AMPK, including acetylcholine, histamine, orexin-A, dopamine, and norepinephrine. Several GAs that are commonly used to induce LOC in human patients also activate AMPK (e.g. propofol, sevoflurane, isoflurane, dexmedetomidine, ketamine, midazolam). Various compounds that accelerate emergence from anesthesia, thus mitigating problematic effects associated with delayed emergence such as delirium, also activate AMPK (e.g. nicotine, caffeine, forskolin, carbachol). GAs and neurotransmitters also act as preconditioning agents and the GABAA receptor inhibitor bicuculline, which reverses propofol anesthesia, also activates AMPK in cortical neurons. We propose the novel hypothesis that cellular stress-induced AMPK activation links wakefulness, arousal, and consciousness with paradoxical excitation and accelerated emergence from anesthesia. Because AMPK activators including metformin and nicotine promote proliferation and differentiation of neural stem cells located in the subventricular zone and the dentate gyrus, AMPK activation may also enhance brain repair and promote potential recovery from disorders of consciousness (i.e. minimally conscious state, vegetative state, coma).
Collapse
|
41
|
Wang XL, Cui LW, Liu Z, Gao YM, Wang S, Li H, Liu HX, Yu LJ. Effects of TRPA1 activation and inhibition on TRPA1 and CGRP expression in dorsal root ganglion neurons. Neural Regen Res 2019; 14:140-148. [PMID: 30531088 PMCID: PMC6262987 DOI: 10.4103/1673-5374.243719] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a key player in pain and neurogenic inflammation, and is localized in nociceptive primary sensory dorsal root ganglion (DRG) neurons. TRPA1 plays a major role in the transmission of nociceptive sensory signals. The generation of neurogenic inflammation appears to involve TRPA1-evoked release of calcitonin gene-related peptide (CGRP). However, it remains unknown whether TRPA1 or CGRP expression is affected by TRPA1 activation. Thus, in this study, we examined TRPA1 and CGRP expression in DRG neurons in vitro after treatment with the TRPA1 activator formaldehyde or the TRPA1 blocker menthol. In addition, we examined the role of extracellular signal-regulated protein kinase 1/2 (ERK1/2) in this process. DRG neurons in culture were exposed to formaldehyde, menthol, the ERK1/2 inhibitor PD98059 + formaldehyde, or PD98059 + menthol. After treatment, real-time polymerase chain reaction, western blot assay and double immunofluorescence labeling were performed to evaluate TRPA1 and CGRP expression in DRG neurons. Formaldehyde elevated mRNA and protein levels of TRPA1 and CGRP, as well as the proportion of TRPA1- and CGRP-positive neurons. In contrast, menthol reduced TRPA1 and CGRP expression. Furthermore, the effects of formaldehyde, but not menthol, on CGRP expression were blocked by pretreatment with PD98059. PD98059 pretreatment did not affect TRPA1 expression in the presence of formaldehyde or menthol.
Collapse
Affiliation(s)
- Xiao-Lei Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Li-Wei Cui
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zhen Liu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yue-Ming Gao
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Sheng Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hao Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hu-Xiang Liu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Ling-Jia Yu
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
42
|
Inhibition by general anesthetic propofol of compound action potentials in the frog sciatic nerve and its chemical structure. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:359-369. [PMID: 30519707 DOI: 10.1007/s00210-018-01596-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
Although the intravenous general anesthetic propofol (2,6-diisopropylphenol) has an ability to inhibit nerve conduction, this has not been fully examined. Various agents inhibit compound action potentials (CAPs) in a manner dependent on their chemical structures. To determine propofol's chemical structure that is important in nerve conduction inhibition, we examined the effects of propofol and its related compounds on fast-conducting CAPs recorded from the frog sciatic nerve by using the air-gap method. Propofol concentration-dependently reduced the peak amplitude of the CAP with a half-maximal inhibitory concentration (IC50) value of 0.14 mM. A similar inhibition was produced by other phenols, 4-sec-butylphenol and 4-amylphenol (IC50 values: 0.33 and 0.20 mM, respectively). IC50 values for these and more phenols (4-isopropylphenol, 4-tert-butylphenol, and 4-ter-amylphenol; data published previously) were correlated with the logarithm of their octanol-water partition coefficients. A phenol having ketone group (raspberry ketone) and alcohols (3-phenyl-1-propanol and 2-phenylethylalcohol) inhibited CAPs less effectively than the above-mentioned phenols. The local anesthetic (LA) benzocaine reduced CAP peak amplitudes with an IC50 of 0.80 mM, a value larger than that of propofol. When compared with other LAs, propofol activity was close to those of ropivacaine, levobupivacaine, and pramoxine, while benzocaine activity was similar to those of cocaine and lidocaine. It is concluded that propofol inhibits nerve conduction, possibly owing to isopropyl and hydroxyl groups bound to the benzene ring of propofol and to its lipophilicity; propofol's efficacy is comparable to those of some LAs. These results could serve to develop propofol-related agents exhibiting analgesia when applied topically.
Collapse
|
43
|
Abstract
Propofol is an intravenous hypnotic drug that is used for induction and maintenance of sedation and general anaesthesia. It exerts its effects through potentiation of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) at the GABAA receptor, and has gained widespread use due to its favourable drug effect profile. The main adverse effects are disturbances in cardiopulmonary physiology. Due to its narrow therapeutic margin, propofol should only be administered by practitioners trained and experienced in providing general anaesthesia. Many pharmacokinetic (PK) and pharmacodynamic (PD) models for propofol exist. Some are used to inform drug dosing guidelines, and some are also implemented in so-called target-controlled infusion devices, to calculate the infusion rates required for user-defined target plasma or effect-site concentrations. Most of the models were designed for use in a specific and well-defined patient category. However, models applicable in a more general population have recently been developed and published. The most recent example is the general purpose propofol model developed by Eleveld and colleagues. Retrospective predictive performance evaluations show that this model performs as well as, or even better than, PK models developed for specific populations, such as adults, children or the obese; however, prospective evaluation of the model is still required. Propofol undergoes extensive PK and PD interactions with both other hypnotic drugs and opioids. PD interactions are the most clinically significant, and, with other hypnotics, tend to be additive, whereas interactions with opioids tend to be highly synergistic. Response surface modelling provides a tool to gain understanding and explore these complex interactions. Visual displays illustrating the effect of these interactions in real time can aid clinicians in optimal drug dosing while minimizing adverse effects. In this review, we provide an overview of the PK and PD of propofol in order to refresh readers' knowledge of its clinical applications, while discussing the main avenues of research where significant recent advances have been made.
Collapse
Affiliation(s)
- Marko M. Sahinovic
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- University Medical Center Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Michel M. R. F. Struys
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Anaesthesia and Peri-Operative Medicine, Ghent University, Ghent, Belgium
| | - Anthony R. Absalom
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
44
|
Abstract
The transient receptor potential ankyrin 1 (TRPA1) ion channel is expressed in pain-sensing neurons and other tissues and has become a major target in the development of novel pharmaceuticals. A remarkable feature of the channel is its long list of activators, many of which we are exposed to in daily life. Many of these agonists induce pain and inflammation, making TRPA1 a major target for anti-inflammatory and analgesic therapies. Studies in human patients and in experimental animals have confirmed an important role for TRPA1 in a number of pain conditions. Over the recent years, much progress has been made in elucidating the molecular structure of TRPA1 and in discovering binding sites and modulatory sites of the channel. Because the list of published mutations and important molecular sites is steadily growing and because it has become difficult to see the forest for the trees, this review aims at summarizing the current knowledge about TRPA1, with a special focus on the molecular structure and the known binding or gating sites of the channel.
Collapse
Affiliation(s)
- Jannis E Meents
- Institute of Physiology, University Hospital RWTH Aachen , Aachen , Germany
| | - Cosmin I Ciotu
- Center for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
45
|
Niedermirtl F, Eberhardt M, Namer B, Leffler A, Nau C, Reeh PW, Kistner K. Etomidate and propylene glycol activate nociceptive TRP ion channels. Mol Pain 2018; 14:1744806918811699. [PMID: 30345869 PMCID: PMC6856977 DOI: 10.1177/1744806918811699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
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.
Collapse
Affiliation(s)
- Florian Niedermirtl
- 1 Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany
| | - Mirjam Eberhardt
- 2 Department for Anaesthesia and Critical Care Medicine, Hannover Medical School, Hannover, Germany
| | - Barbara Namer
- 1 Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany
| | - Andreas Leffler
- 2 Department for Anaesthesia and Critical Care Medicine, Hannover Medical School, Hannover, Germany
| | - Carla Nau
- 3 Department of Anesthesiology and Intensive Care, University of Lübeck, Lübeck, Germany
| | - Peter W Reeh
- 1 Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany
| | - Katrin Kistner
- 1 Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany
| |
Collapse
|
46
|
Photosensitization of TRPA1 and TRPV1 by 7-dehydrocholesterol: implications for the Smith-Lemli-Opitz syndrome. Pain 2018; 158:2475-2486. [PMID: 28891864 DOI: 10.1097/j.pain.0000000000001056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Loss-of-function mutations in the enzyme 7-dehydrocholesterol reductase are responsible for the Smith-Lemli-Opitz syndrome, in which 7-dehydrocholesterol (7-DHC) levels are markedly increased in the plasma and tissues of patients. This increase in 7-DHC is probably associated with the painful and itchy photosensitivity reported by the majority of patients with Smith-Lemli-Opitz syndrome. To identify the molecular targets involved in the activation and photosensitization of primary afferents by 7-DHC, we focused on TRPA1 and TRPV1, two ion channels expressed in nociceptive nerve endings and previously shown to respond to ultraviolet and visible light under pathophysiological circumstances. Recombinant human TRPA1 is activated and photosensitized in the presence of 7-DHC. Prolonged preexposure to 7-DHC causes more pronounced photosensitization, and while TRPV1 contributes less to the acute effect, it too becomes highly photosensitive upon preincubation with 7-DHC for 1 to 15 hours. Dorsal root ganglion neurons in primary culture display acute sensitivity to 7-DHC in the dark and also light-evoked responses in the presence of 7-DHC, which are exclusively dependent on TRPA1 and TRPV1. Similarly, prolonged exposure of mouse dorsal root ganglion neurons to 7-DHC renders these cells photosensitive in a largely TRPA1- and TRPV1-dependent manner. Single-fiber recordings in mouse skin-nerve preparations demonstrate violet light-evoked activation and a sensitization to 7-DHC exposure. Vice versa, 7-DHC pretreatment of the isolated trachea leads to a TRPA1- and TRPV1-dependent increase of the light-induced calcitonin gene-related peptide release. Taken together, our results implicate TRPA1 and TRPV1 channels as potential pharmacological targets to address the 7-DHC-induced hypersensitivity to light in patients.
Collapse
|
47
|
Abstract
The precise mechanism by which propofol enhances GABAergic transmission remains unclear, but much progress has been made regarding the underlying structural and dynamic mechanisms. Furthermore, it is now clear that propofol has additional molecular targets, many of which are functionally influenced at concentrations achieved clinically. Focusing primarily on molecular targets, this brief review attempts to summarize some of this recent progress while pointing out knowledge gaps and controversies. It is not intended to be comprehensive but rather to stimulate further thought, discussion, and study on the mechanisms by which propofol produces its pleiotropic effects.
Collapse
Affiliation(s)
- Pei Tang
- Department of Anesthesiology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Roderic Eckenhoff
- Department of Anesthesiology & Critical Care, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104, USA
| |
Collapse
|
48
|
Abstract
Crotalphine is a structural analogue to a novel analgesic peptide that was first identified in the crude venom from the South American rattlesnake Crotalus durissus terrificus. Although crotalphine's analgesic effect is well established, its direct mechanism of action remains unresolved. The aim of the present study was to investigate the effect of crotalphine on ion channels in peripheral pain pathways. We found that picomolar concentrations of crotalphine selectively activate heterologously expressed and native TRPA1 ion channels. TRPA1 activation by crotalphine required intact N-terminal cysteine residues and was followed by strong and long-lasting desensitization of the channel. Homologous desensitization of recombinant TRPA1 and heterologous desensitization in cultured dorsal root ganglia neurons was observed. Likewise, crotalphine acted on peptidergic TRPA1-expressing nerve endings ex vivo as demonstrated by suppression of calcitonin gene-related peptide release from the trachea and in vivo by inhibition of chemically induced and inflammatory hypersensitivity in mice. The crotalphine-mediated desensitizing effect was abolished by the TRPA1 blocker HC030031 and absent in TRPA1-deficient mice. Taken together, these results suggest that crotalphine is the first peptide to mediate antinociception selectively and at subnanomolar concentrations by targeting TRPA1 ion channels.
Collapse
|
49
|
Abstract
GABA (γ-aminobutyric acid) receptors, of which there are two types, are involved in inhibitory synapses within the central nervous system. The GABAA receptor (GABAAR) has a central role in modern anesthesia and sedation practice, which is evident from the high proportion of agents that target the GABAAR. Many GABAAR agonists are used in anesthesia practice and sedation, including propofol, etomidate, methohexital, thiopental, isoflurane, sevoflurane, and desflurane. There are advantages and disadvantages to each GABAAR agonist currently in clinical use. With increasing knowledge regarding the pharmacology of GABAAR agonists, however, newer sedative agents have been developed which employ 'soft pharmacology', a term used to describe the pharmacology of agents whereby their chemical configuration allows rapid metabolism into inactive metabolites after the desired therapeutic effect(s) has occurred. These newer 'soft' GABAAR agonists may well approach ideal sedative agents, as they can offer well-controlled, titratable activity and ultrashort action. This review provides an overview of the role that GABAAR agonists currently play in sedation and anesthesia, in addition to discussing the future role of novel GABAAR agonists in anesthesia and sedation.
Collapse
|
50
|
Woll KA, Skinner KA, Gianti E, Bhanu NV, Garcia BA, Carnevale V, Eckenhoff RG, Gaudet R. Sites Contributing to TRPA1 Activation by the Anesthetic Propofol Identified by Photoaffinity Labeling. Biophys J 2017; 113:2168-2172. [PMID: 28935134 DOI: 10.1016/j.bpj.2017.08.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/07/2017] [Accepted: 08/24/2017] [Indexed: 11/16/2022] Open
Abstract
In addition to inducing anesthesia, propofol activates a key component of the pain pathway, the transient receptor potential ankyrin 1 ion channel (TRPA1). Recent mutagenesis studies suggested a potential activation site within the transmembrane domain, near the A-967079 cavity. However, mutagenesis cannot distinguish between protein-based and ligand-based mechanisms, nor can this site explain the complex modulation by propofol. Thus more direct approaches are required to reveal potentially druggable binding sites. Here we apply photoaffinity labeling using a propofol derivative, meta-azipropofol, for direct identification of binding sites in mouse TRPA1. We confirm that meta-azipropofol activates TRPA1 like the parent anesthetic, and identify two photolabeled residues (V954 and E969) in the S6 helix. In combination with docking to closed and open state models of TRPA1, photoaffinity labeling suggested that the A-967079 cavity is a positive modulatory site for propofol. Further, the photoaffinity labeling of E969 indicated pore block as a likely mechanism for propofol inhibition at high concentrations. The direct identification of drug-binding sites clarifies the molecular mechanisms of important TRPA1 agonists, and will facilitate drug design efforts to modulate TRPA1.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth A Skinner
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| | - Eleonora Gianti
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Natarajan V Bhanu
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin A Garcia
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Rachelle Gaudet
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts.
| |
Collapse
|