Next Article in Journal
Rational Design of an Epidermal Growth Factor Receptor Vaccine: Immunogenicity and Antitumor Research
Previous Article in Journal
Anti-Inflammatory Activity of the Major Triterpenic Acids of Chios Mastic Gum and Their Semi-Synthetic Analogues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na+ and TRP Channels

Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
Biomolecules 2024, 14(12), 1619; https://doi.org/10.3390/biom14121619
Submission received: 27 November 2024 / Revised: 11 December 2024 / Accepted: 12 December 2024 / Published: 18 December 2024

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.

1. Introduction

Noxious stimuli delivered to the periphery are conveyed to the central nervous system (CNS) by action potentials (APs) that travel along primary afferent neuron fibers (see [1] for review). Primary afferent neurons express transient receptor potential (TRP) channels, i.e., non-selective cation channels, that are involved in transmitting sensory information, such as nociception and temperature (see [2] for review). Among these channels are TRPV (vanilloid; 1–6) involved in pain and hot, TRPA (ankyrin; 1) involved in pain and nociceptive cold (<17 °C), TRPM (melastatin; 1–8) involved in pain and mild cold, and TRPC (canonical; 1–7) involved in pain (see [3,4,5] for review). In fact, the cell bodies of primary afferent [dorsal root ganglion (DRG) and trigeminal ganglion (TG)] neurons express mRNA and protein for TRPV1, TRPA1 and TRPM8 [6,7,8,9,10]. The other types of TRP channel, such as TRPM2, TRPM3, TRPC5 and TRPC6, are also located in primary afferent neurons [11,12,13,14].
TRP channels are involved not only in pain and temperature but also in mechano-sensation (TRPC3 and 6 [15]), itch (TRPA1, TRPV1, 3 and 4 [16]) and a variety of sensory allergic reactions (TRPA1 [17]). TRP channels synthesized in the cell bodies are conveyed by axonal transport to their peripheral and central terminals and are thus involved in transmitting sensory information. Peripheral terminal TRP activation is involved in the production of AP, while central terminal TRP activation results in enhanced spontaneous release of L-glutamate from the terminal on spinal cord and brain stem lamina II neurons (see [18,19,20] for TRPV1; see [21,22,23,24] for TRPA1; see [20,23,24,25,26,27] for TRPM8; see [28] for TRPC3/6; for review, see [3,5,29,30]). The enhanced spontaneous L-glutamate release is caused by an increase in intracellular Ca2+ concentration, which is due to activation of voltage-gated Ca2+ channels via membrane depolarization occurring as a result of opening of TRP, or by Ca2+ passing through the TRP itself [3]. The lamina II neurons play a pivotal role in regulating nociceptive transmission from the periphery [31,32,33]. Consistent with this idea, glutamatergic excitatory transmission in lamina II neurons is suppressed by various endogenous analgesics, including neuropeptides (for example, see [34,35,36]).
Regarding nociceptive transmission, in primary afferent neurons, which transmit information as APs involving Na+ channel activation, TRPs at their peripheral terminals receive nociceptive stimuli, while TRPs at their central terminals modify the transmission of nociceptive information. The involvement of TRP in nociceptive transmission is supported by (1) upregulated TRPV1 expression in the spinal lamina II following peripheral inflammation [37], (2) reversal by intrathecally-applied TRPA1 antagonist of mechanical and cold hyperalgesia (an increased sensitivity to pain) caused by the intra-plantar injection of complete Freund’s adjuvant [38], (3) chronic neuropathic pain (chronic pain lasting more than three months after nerve injury or dysfunction; see [39] for review) alleviation by TRPM8 activation in primary afferent neurons [40], (4) reduced heat-induced thermal hyperalgesia following inflammation in TRPM3-deficeint mouse models [12], (5) reduced nociceptive (thermal and mechanical) thresholds following inflammation in TRPC5-knockout mouse models [41], (6) the observation that knockdown of TRPM2 in the DRG and spinal cord alleviated neuropathic pain produced by sciatic nerve chronic constriction injury [42], and (7) analgesia produced by a TRPC6 inhibitor in rat neuropathic pain models induced by spared nerve injury [14].
The TRP channels are targets of various compounds derived from plants (see [17,43,44] for review). For instance, TRPV1, TRPA1 and TRPM8 are activated by capsaicin (8-methyl-N-vanillyl-6-nonenamide, the major pungent ingredient in hot peppers [45], having a vanillyl group), allyl isothiocyanate (AITC, the pungent principal in mustard oil or wasabi [46]) and menthol (2-isopropyl-5-methylcyclohexanol, a secondary alcohol in peppermint or other mint oils [47]), respectively (see [4] for review).
Although many of the properties of the TRP channels are examined in the cell body of primary afferent neuron, there seems to be a difference in activation by plant-derived chemicals between primary afferent cell body and central terminal TRP, as indicated by previous studies (see [29] for review). Such a difference may be due to interactions between TRP, such as TRPA1-TRPV1 complex [48], the presence of TRP splice variants [49,50], or TRP modulation by intracellular second messengers [51,52,53]. Many plant-derived compounds, which increased spontaneous L-glutamate release through central terminal TRP activation in lamina II neurons, inhibited nerve AP conduction and Na+ channels involved in AP production (see [54] for review). As with plant-derived compounds, both TRP and Na+ channels are modulated by local anesthetics, general anesthetics, anti-epileptics, opioids, adrenoceptor agonists, antidepressants and non-steroidal anti-inflammatory drugs (NSAIDs), all of which are involved in antinociception. In this review, how these drugs and the other pain-related drugs affect the primary afferent neuron, its central terminal, cloned TRP and Na+ channels (nerve conduction) is shown, and then a modulatory site on TRP is suggested to be similar in chemical structure to that of Na+ channels.

2. Local Anesthetics

Local anesthetics are used not only for AP conduction blocking (see [55,56] for review) in surgical anesthesia but also for chronic pain alleviation as analgesic adjuvant (see [57,58] for review). Leffler et al. [59,60] have reported that a typical local anesthetic lidocaine, which blocks Na+ channels (with a half-maximal inhibitory concentration (IC50) value of 0.204 mM in Xenopus laevis sciatic nerve fibers [61]), activates TRPV1 and TRPA1 (half-maximal effective concentration (EC50): 3.4 and 5.7 mM, respectively) expressed in rat DRG neurons. Consistent with this observation, lidocaine at concentrations higher than 2 mM activated primary afferent central terminal TRPA1, resulting in spontaneous L-glutamate release enhancement in rat spinal lamina II neurons (spontaneous excitatory postsynaptic current (sEPSC) frequency increase: 132% at 5 mM [62]). The primary afferent central terminal TRPA1 in the rat spinal lamina II was also activated by another local anesthetic tetracaine (307% sEPSC frequency increase at 5 mM [63]), which blocked Na+ channels in Xenopus laevis sciatic nerve fibers (IC50 = 0.0007 mM [61]). Lidocaine and tetracaine inhibited Na+ channel-mediated frog sciatic nerve compound action potentials (CAPs; IC50 for lidocaine and tetracaine: 0.74 mM [64] and 0.014 mM [65], respectively). Bupivacaine, which inhibited Na+ channels (at 0.3 mM in rat brain [66]; IC50 = 0.027 mM in Xenopus laevis sciatic nerve fibers [61]) and frog sciatic nerve CAPs (76% amplitude reduction at 0.5 mM [67]), activated TRPA1 in rat DRG neurons (EC50 = 0.41 mM [68]). CAP inhibition similar to that in the frog sciatic nerve has been also reported in mammalian nerve fibers. For example, lidocaine suppressed rat sciatic nerve CAPs with an IC50 value of 0.28 mM [69]. Tetracaine and bupivacaine depressed rabbit fast-conducting A fiber CAPs (IC50: 0.009 and 0.048 mM, respectively [70]).
A cationic lidocaine derivative QX-314 (N-ethyl-lidocaine), which inhibits Na+ channels when intracellularly applied, is known to have an ability to go through TRPV1 [71,72]. This indicates that QX-314 suppresses Na+ channels in primary afferent neurons expressing TRPV1. QX-314 exhibited a biphasic effect on human TRPV1 expressed in Xenopus laevis oocytes, activating at high concentrations (such as 30–60 mM) and inhibiting at low concentration (IC50 = 8 μM for capsaicin response [73]), showing that QX-314 has an effect on TRPV1. Moreover, QX-314 is shown to activate not only human TRPV1, but also TRPA1 expressed in human embryonic kidney (HEK) 293 cells [74]. The results of these local anesthetic actions are summarized in Table 1.

3. General Anesthetics

General anesthetics are thought to produce not only the loss of consciousness but also analgesia by acting on ligand-gated ion channels (see [75,76] for review), for example, in a pain inhibitory circuit of the amygdala [77]. A general anesthetic propofol (2,6-diisopropylphenol) activated rat TRPV1 [78] and TRPA1 (at 3–300 μM [79]) expressed in HEK293 cells, and also TRPV1 and TRPA1 located in rodent DRG neurons (at 10–30 μM [80,81]), while inhibiting Na+ channels (IC50 = 20 μM in the human brain [82]; at >60 μM in Xenopus laevis myelinated nerve fibers [83]; IC50 = 46 μM in rat cerebro-cortical synaptosomes [84]; IC50 = 10 μM for rat brain IIa Na+ channels expressed in Chinese hamster ovary (CHO) cells [85]; see [86] for review). Ton et al. [87] have reported that propofol has a modulatory action on rodent but not human TRPA1, indicating a species difference. Frog sciatic nerve CAPs were inhibited by propofol (IC50 = 0.14 mM); this inhibition required a specific chemical structure of propofol [88].
Table 1. Analgesic, analgesic adjuvants and other drugs exhibiting both TRP and Na+ channel (or CAP) modulation.
Table 1. Analgesic, analgesic adjuvants and other drugs exhibiting both TRP and Na+ channel (or CAP) modulation.
Chemical CompoundsCentral Terminal TRP ModulationPrimary Afferent Neuron and Cloned TRP ModulationVoltage-Gated Na+ ChannelCAPReferences
Local anesthetics
LidocaineTRPA1 ↑TRPA1 ↑
TRPV1 ↑
[59,60,61,62,64,69]
TetracaineTRPA1 ↑n.d.[61,63,65,70]
Bupivacainen.d.TRPA1 ↑[61,66,67,68,70]
QX-314n.d.TRPV1 (↑, ↓)
TRPA1 ↑

(intracellularly-applied)
n.d.[73,74]
General anesthetics
Propofoln.d.TRPV1 ↑
TRPA1 ↑
[78,79,80,81,82,83,84,85,87,88]
Isofluranen.d.TRPA1 (↑, ↓)n.d.[79,87,89]
Antiepileptics
Phenytoinn.d.TRPA1 ↑[67,90,91,92]
Carbamazepinen.d.TRPV1 ↓[67,93,94]
Opioids
Morphinen.d.TRPV1 ↑
TRPA1 ↑
[95,96,97,98,99]
Tramadoln.d.TRPV1 (↑, →)
TRPA1 ↓
[64,97,100,101,102,103]
O-Desmethyl tramadoln.d.TRPA1 ↓n.d.[64,103]
α2-Adrenoceptor agonist
Dexmedetomidinen.d.TRPV1 ↓
TRPM2 ↓
[65,104,105,106,107,108,109]
Antidepressants
Duloxetinen.d.TRPV1 ↓
TRPM2 ↓
TRPC5 ↓
[110,111,112,113,114]
Maprotilinen.d.TRPM3 ↓[113,115,116]
Amitriptylinen.d.TRPV1 ↓[113,116,117,118,119,120,121]
Fluoxetinen.d.TRPV1 ↓[113,116,122,123]
Desipraminen.d.TRPV1 ↓[113,116,123,124]
NSAIDs
Diclofenacn.d.TRPA1 ↑
TRPM3 ↓
[115,125,126,127,128]
Etodolacn.d.TRPA1 ↑n.d.[126,129]
Tolfenamic acidn.d.TRPM3 ↓
TRPC6 ↓
[126,130,131]
Meclofenamic acidn.d.TRPM3 ↓
TRPC6 ↓
[126,130,131]
Mefenamic acidn.d.TRPM3 ↓
TRPC6 ↓
[126,130,131]
Flufenamic acidn.d.TRPM3 ↓
TRPC6 ↓
[126,130,131,132]
Ibuprofenn.d.TRPA1 ↑
(at 10 mM)
No effect at 1 mMn.d.[126,133]
Salicylaten.d.TRPM7 ↓n.d.[134,135]
Other drugs
MethylglyoxalTRPV1 ↑
TRPA1 ↑
TRPA1 ↑Nav1.8 ↑n.d.[136,137,138]
Nicotinen.d.TRPV1 ↑n.d.[139]
Pregnenolone sulfaten.d.TRPV1 ↓
TRPM3 ↑
n.d.[115,140,141,142]
Riluzolen.d.TRPC5 ↑n.d.[143,144]
AnandamideTRPV1 ↑TRPV1 ↑n.d.[19,145,146,147]
n.d.: not determined; ↑: activation; ↓: inhibition; →: no change.
Another general anesthetic, isoflurane ((RS)-2-chloro-2-(difluoro-methoxy)-1,1,1-trifluoroethane), is shown to activate rat TRPA1 expressed in HEK293 cells (EC50 = 0.18 mM [79]) while having both rodent TRPA1 inhibition (IC50 = 0.28 mM for AITC response at a holding potential of −100 mV [87]) and Na+ channel inhibition in pyramidal neurons of the mouse prefrontal cortex (36% amplitude reduction at 0.45 mM at a holding potential of −70 mV [89]). The results of these general anesthetic actions are summarized in Table 1.

4. Antiepileptics

Antiepileptics are widely used as analgesic adjuvants to alleviate neuropathic and inflammatory pain (caused by inflammation leading to inflammatory cytokine release; see [148,149] for review). A typical anti-epileptic phenytoin (5,5-diphenylhydantoin) activated human TRPA1 but not TRPV1 and TRPM8 expressed in HEK cells (at 0.1 mM [90]), while inhibiting Na+ channels in rat cortical neurons (IC50 = 51 μM [91]) and N4TG1 mouse neuroblastoma cells (IC50 = 58 μM [92]), and frog sciatic nerve CAPs (16% amplitude reduction at 0.1 mM [67]). Carbamazepine (an iminostilbene derivative; 5H-dibenz[b,f]azepine-5-carboxamide) having an ability to suppress Na+ channels in mouse central neurons [93] and frog sciatic nerve CAPs (IC50 = 0.50 mM [67]) is suggested to inhibit rat TRPV1 from the observation that capsaicin-induced hyperalgesia was attenuated by carbamazepine [94]. TRPV1 is thought to be a potential target of anti-epileptics [150]. The results of these antiepileptic actions are summarized in Table 1.

5. Opioids

Opioids can exert analgesic effects not only in the CNS [see [151,152,153] for review] but also in the peripheral nervous system by activating opioid receptors (see [154,155] for review). TRPV1 and TRPA1 located in mouse DRG neurons were activated by morphine with an EC50 value of 2.6 mM [95]; morphine inhibited Na+ channels (frog Ranvier node [96]; IC50 = ca. 0.3 mM for TTX-sensitive currents in DRG neuroblastoma hybridoma cell line ND7/23 cells [97]) and frog sciatic nerve CAPs (15% amplitude reduction at 5 mM [98]). A similar AP conduction inhibition by morphine has been reported in the rat sciatic nerve; morphine reduced the peak amplitude of AP recorded intracellularly from DRG neurons with an IC50 value of 2.9 mM [99].
A clinically-used and orally-active μ-opioid receptor agonist tramadol ((1RS; 2RS)-2-[(dimethyl-amino) methyl]-1-(3-methoxyphenyl)-cyclohexanol hydrochloride [156]), which inhibited Na+ channels (IC50 = 0.103 mM for TTX-sensitive Nav1.2 [100]; IC50 = ca. 0.2 mM for TTX-sensitive channels in ND7/23 cells [97]) and frog sciatic nerve CAPs (IC50 = 2.3 mM [64]), activated rat TRPV1 expressed in CHO cells (EC50 = 0.08 μM [101]). An inhibitory action of tramadol on CAPs has been reported for the rat sciatic nerve [102]. Since tramadol (5 mg/kg) inhibits the spontaneous release of L-glutamate from nerve terminals by activating opioid receptors in the rat spinal cord in vivo ([157]; see [158,159] for EPSC suppression by μ-opioid receptor activation in the lamina II of tissue slice preparations), it is difficult to determine whether tramadol acts on primary afferent central terminal TRP. Miyano et al. [103] have reported that tramadol and its metabolite O-desmethyl tramadol (M1; at each 10 μM [160]) do not activate human TRPV1 and TRPA1 expressed in HEK293 cells while suppressing human TRPA1 (activated by AITC), but not human TRPV1 (activated by capsaicin), expressed in HEK293 cells. The difference between Marincsák et al. [101]’s and Miyano et al. [103]’s results on TRPV1 activation has been attributed to the distinction in cell line (CHO or HEK293) used to express TRPV1, rather than different species (human or rat [103]). Unlike tramadol, M1 exhibited a small reduction in frog sciatic nerve CAP peak amplitude (9% amplitude reduction at 5 mM [64]). The frog sciatic nerve CAP inhibitions produced by morphine and tramadol may be due to a membrane hyperpolarization occurring as a result of opioid-receptor activation. However, this is unlikely, because the morphine and tramadol activities were resistant to a non-selective opioid-receptor antagonist naloxone [64,98]. The results of these opioid actions are summarized in Table 1.

6. α2-Adrenoceptor Agonist

α2-Adrenoceptor agonists are widely used not only as an analgesic (which inhibits nociceptive transmission by activating α2 adrenoceptors in the CNS; for example, see [161,162]) but also as an analgesic adjuvant to alleviate chronic pain (see [163,164,165] for review). An α2-adrenoceptor agonist dexmedetomidine (DEX; ((+)-(S)-4-[1-(2,3-dimethylphenyl)-ethyl]-1H-imidazole; see [166,167] for review) inhibited Na+ channels in NG108-15 neuronal cells (22% amplitude reduction at 10 μM [104]) and also frog sciatic nerve CAPs (IC50 = 0.40 mM [65]). A similar CAP inhibitory action of DEX has been reported in the rat sciatic nerve [105]. On the other hand, TRPV1 activation by capsaicin in rat DRG neurons was suppressed by DEX (1 pM); this action has been attributed to inhibited adenylate cyclase/cyclic AMP/protein kinase A pathway following adrenoceptor activation [106]. DEX-induced TRPV1 inhibition has been also reported in mouse DRG neurons (30% capsaicin response suppression at 10 μM [107]). DEX inhibited not only TRPV1 (activated by capsaicin) but also TRPM2 (activated by its agonist cumene hydroperoxide) in mouse DRG and hippocampal neurons [108]. On the other hand, primary afferent central terminal TRPV1 in the rat spinal lamina II was unaffected by DEX at 0.3 μM [109]. This may have been due to the fact that this concentration is lower than those that inhibit Na+ channels in NG108-15 cells (see above; [104]) and frog sciatic nerve CAPs (76% amplitude reduction at 0.5 mM [65]). Alternatively, the lack of the inhibitory effect of DEX on central terminal TRPV1 may indicate that endogenous TRPV1 agonists such as anandamide (arachidonoylethanolamide; see [168,169] for review) do not constitutively promote spontaneous L-glutamate release in the lamina II. The results of the α2-adrenoceptor agonist actions are summarized in Table 1.

7. Antidepressants

Various types of antidepressants are used as analgesic adjuvants for treating neuropathic pain (see [170,171,172] for review). An antidepressant duloxetine (serotonin and noradrenaline reuptake inhibitor, SNRI) used for chronic pain alleviation inhibited both TRP (TRPV1 and TRPM2; activated by capsaicin and cumene hydroperoxide, respectively) in rat DRG neurons [110] and Na+ channels (IC50 = 22 μM for tetrodotoxin (TTX)-sensitive Nav1.7 expressed in HEK293 cells [111]; IC50 = 14 μM for TTX-resistant Nav1.5 [112]). Frog sciatic nerve CAPs have been shown to be inhibited by duloxetine (IC50 = 0.23 mM [113]). Moreover, duloxetine has been reported to inhibit TRPC5 that is involved in neuropathic and inflammatory pain (IC50 = 0.30 μM for a TRPC5 agonist riluzole (2-amino-6-[trifluoromethoxy]benzothiazole) response at negative potentials [114]; see [5] for review). On the other hand, another antidepressant, maprotiline (tetracyclic secondary amine), suppressed mouse TRPM3 expressed in HEK293 cells (IC50 = 1.3 μM for its agonist pregnenolone sulfate response [115]) while inhibiting Na+ channels (IC50 = 28 μM for human Nav1.7 expressed in HEK293 cells [116]) and frog sciatic nerve CAPs (IC50 = 0.95 mM [113]). Behavioral studies suggest that TRPV1 may be suppressed by other antidepressants, amitriptyline (tricyclic tertiary amine [117]), fluoxetine (selective serotonin reuptake inhibitor, SSRI [122]) and desipramine (tricyclic secondary amine [124]), all of which inhibited frog sciatic nerve CAPs (IC50 = 0.26, 1.5 and 1.6 mM, respectively, for amitriptyline, fluoxetine and desipramine [113]) and Na+ channels (amitriptyline: IC50 = 85 μM for human Nav1.7 expressed in HEK293 cells [116], IC50 = 20.2 μM in bovine adrenal chromaffin cells [118], the rat striatum [119], rat cortical neurons [120]; fluoxetine: IC50 = 74 μM for human Nav1.7 [116], IC50 = 1.11 μM at −60 mV in rat hippocampal neurons [123]; desipramine: IC50 = 24 μM for human Nav1.7 [116], IC50 = 1.68 μM at −60 mV in rat hippocampal neurons [123]). Sciatic nerve AP conduction suppression by antidepressants has been reported in mammalian nerve fibers. For instance, amitriptyline at 5 and 10 mM produced a complete nerve conduction blockade of rat sciatic nerve [121]. The results of these antidepressant actions are summarized in Table 1.

8. NSAIDs

Analgesia produced by NSAIDs has been attributed to various actions, including suppression of the synthesis of prostaglandins from arachidonic acid by inhibiting cyclooxygenase (see [173,174,175] for review). For example, at least a part of the antinociceptive action of NSAIDs is due to Na+ channel inhibition, leading to nerve AP conduction inhibition (see [176] for review). In support of this idea, an acetic acid-based NSAID diclofenac inhibited Na+ channels in rat DRG neurons (IC50 = 14 and 97 μM for TTX-sensitive and TTX-resistant Na+ currents, respectively [125]) and frog sciatic nerve CAPs (IC50 = 0.94 mM [126]). When examined in TRP expressed in heterologous cells, diclofenac activated TRPA1 (at 0.3 mM for channels expressed in Xenopus laevis oocytes [127]) while inhibiting TRPM3 (IC50 = 18.8 μM for human TRPM3 isoform (TRPM31325) responses activated by a TRPM3 agonist pregnenolone sulfate in HEK293 cells [128]; IC50 = 6.2 μM for mouse TRPM3 responses activated by pregnenolone sulfate in HEK293 cells [115]). Another acetic acid-based NSAID etodolac activated TRPA1 in mouse DRG neurons and also TRPA1 (but not TRPV1, TRPV2 and TRPM8) expressed in HEK293 cells [129] while inhibiting frog sciatic nerve CAPs (15% amplitude reduction at 1 mM [126]). With respect to fenamic acid-based NSAIDs, all of tolfenamic acid, meclofenamic acid, mefenamic acid and flufenamic acid inhibited frog sciatic nerve CAPs (IC50: 0.29, 0.19 and 0.22 mM, respectively, for tolfenamic acid, meclofenamic acid and flufenamic acid; 16% peak amplitude reduction by mefenamic acid at 0.2 mM [126]). Their NSAIDs inhibited TRPM3 expressed in HEK293 cells (pregnenolone sulfate responses: IC50 = 11.1, 13.3, 33.1 and 6.6 μM for tolfenamic acid, meclofenamic acid, flufenamic acid and mefenamic acid, respectively) and TRPC6 expressed in HEK293 cells (TRPC6 agonist hyperforin responses: IC50 = 12.3, 37.5, 17.1 and >300 μM for tolfenamic acid, meclofenamic acid, flufenamic acid and mefenamic acid, respectively [130]). Na+ channels are reported to be suppressed by tolfenamic acid, mefenamic acid and flufenamic acid (at 100 μM for human Nav1.7 and (TTX-resistant) Nav1.8 expressed in CHO cells [131]; IC50 = 0.189 mM for flufenamic acid to inhibit Na+-channel currents in rat hippocampal pyramidal neurons [132]). A propionic acid-based NSAID ibuprofen at 1 mM did not affect frog sciatic nerve CAPs [126], but TRPA1 in rat TG neurons was activated by ibuprofen at 10 mM [133]. Such a difference in activity may be due to a difference in the concentration of ibuprofen used. An aryl propionic acid-based NSAID salicylate is reported to inhibit TRPM7 activated by Mg2+ depletion in Jurkat T lymphocytes (at 3–30 mM [134]) and Na+ channels in rat inferior colliculus neurons (IC50 = 1.43 mM [135]). The results of these NSAID actions are summarized in Table 1.

9. Other Drugs

Other pain-related drugs that do not fall into the categories mentioned above also act on both Na+ and TRP channels. Behavioral studies have demonstrated that a reduced derivative of pyruvic acid, methylglyoxal (MGO; 2-oxopropanal), which is associated with diabetes resulting in neuropathic pain (diabetic peripheral neuropathy), activates both TRPA1 and Nav1.8 in rat primary afferent neurons [136]. Consistent with this observation, rat Nav1.8 expressed in ND7/23 cells was modified by MGO at 0.1 mM [137]. Primary afferent central terminal TRPA1 and TRPV1 in the rat spinal lamina II were activated by MGO (10 mM); this action was due to reactive oxygen species produced by MGO via advanced glycation end-products [138]. Alternatively, it has been reported in rat TG neurons that an agonist for nicotinic acetylcholine-receptors (involved in nociceptive transmission; see [177] for review), nicotine, at 1 mM inhibits Na+ channels while enhancing capsaicin-activating TRPV1 responses, both of which effects are not mediated by nicotinic acetylcholine receptors [139]. Although nicotine (0.1 mM) as well as capsaicin increased the spontaneous release of L-glutamate in the rat spinal lamina II, this nicotine action was suggested to be mediated by the activation of presynaptic nicotinic acetylcholine receptors [178]. The possibility cannot be ruled out from the above-mentioned experimental result [139] that primary afferent central terminal TRPV1 activated by nicotine plays a role in the presynaptic facilitation in the lamina II. Moreover, a neuroactive steroid pregnenolone sulfate, which inhibits capsaicin-induced nociception in behavioral studies [140] while being a robust TRPM3 activator (EC50 = 23 μM at −80 mV for TRPM3 expressed in HEK293 cells [141]; also see [115]), inhibited Nav1.2 expressed in Xenopus laevis oocytes (IC50 = 53 μM [142]). Riluzole, which depresses nociceptive responses in mice [179], inhibited TTX-sensitive and TTX-resistant Na+ channels in rat DRG neurons (with IC50 values of 90 and 143 μM, respectively [143]), and activated TRPC5 expressed in HEK293 cells (EC50 = 9.2 μM; [144]).
Anandamide is shown to activate TRPV1 in rat DRG neurons (when intracellularly applied [145]) and TG neurons (when bath-applied; EC50 = 10 μM [19]), while suppressing Nav1.2, (TTX-sensitive) Nav1.6, Nav1.7, and Nav1.8 (IC50 = 17, 12, 27 and 40 μM, respectively), expressed in Xenopus laevis oocytes [146], and rat TTX-sensitive and TTX-resistant Na+ channels (IC50: 5.4 and 38.4 μM, respectively [147]). Consistent with the fact that anandamide activates not only TRPV1 but also cannabinoid receptors (see [168,180] for review), anandamide inhibited formalin-induced nociception in a fashion sensitive to a CB1 receptor antagonist SR141716A [181]. An inhibition of primary afferent monosynaptically-evoked C-fiber EPSC (involved in slow pain transmission [1]), as produced by capsaicin (1 μM), was not mimicked by anandamide (10 μM) in the rat spinal lamina II, indicating that anandamide does not activate TRPV1 in primary afferent C-fiber central terminals [182]. On the other hand, spontaneous L-glutamate release in rat brain stem lamina II neurons was enhanced by anandamide (by 79% at 30 μM) in a manner sensitive to a TRPV1 antagonist capsazepine [19]. The possibility that primary afferent central terminal TRPV1 is involved in the modulatory effect of anandamide on nociceptive transmission cannot be ruled out. The results of these other drug actions are summarized in Table 1.

10. Plant-Derived Compounds

Many plant-derived compounds are involved in modulating analgesic activity when orally, intraperitoneally or intrathecally administrated, with fewer side effects than synthetic chemicals (see [183,184] for review). Among them, representative TRP agonists, capsaicin, AITC and menthol, activated TRPV1 [185], TRPA1 [186,187] and TRPM8 [188,189], respectively, are located in the cell body of primary afferent neuron and TRP-expressing heterologous cells (see [3,4] for review). For example, capsaicin activated TRPV1 with an EC50 value of 0.68 μM in rat TG neurons [190,191] and of ca. 0.01 μM in human TRPV1-expressing CHO cells [192]. AITC activated TRPA1 with an EC50 value of 22 μM in mouse TRPA1-expressing CHO cells [186], and menthol activated TRPM8 with an EC50 value of 80 μM in rat TG neurons [188]. On the other hand, Na+ channels were inhibited by capsaicin (IC50 = 0.45 μM in capsaicin-sensitive neurons [193]; 85% amplitude reduction of TTX-resistant Na+ currents by 1 μM capsaicin [194]) and also by menthol (IC50 = 0.571 mM for rat type IIA Na+ channels expressed in HEK293 cells [195]).
Similar modulatory actions have been reported for other plant-derived compounds. Anethole (1-methoxy-4-[(E)-prop-1-enyl]benzene; a phenolic derivative contained in plants such as anise [196]) activated TRPA1 in mouse DRG and TG neurons, and also in human TRPA1-expressing HEK293 cells (EC50 = 80.8 μM [197]), while inhibiting Na+ channels in rat DRG neurons (IC50 = 1.85 mM [198]). Resveratrol (trans-3,5,4′-trihydroxystilbene; a natural stilbenoid contained in grape [199]) inhibited both mouse TRPA1 expressed in HEK 293 cells (IC50 = 0.75 μM for AITC responses [200]) and Na+ channels (ca. 25% amplitude reduction at 20 μM for TTX-sensitive Na+ currents in rat cortical neurons [201]). Curcumin ((1E,6E)-1,7-bis (4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione; the major component of turmeric [202]), which has two vanillyl groups, suppressed capsaicin-induced responses in TG neurons and TRPV1-expressing HEK293 cells [203], while inhibiting frog sciatic nerve CAPs (10% amplitude reduction at 0.05 mM [204]; see [205] for actions of curcumin on ion channels).
With respect to primary afferent central terminal TRP in the spinal lamina II, plant-derived compounds having an ability to activate the channels inhibited nerve AP conduction, possibly Na+ channels, in the frog sciatic nerve. For instance, capsaicin increased spontaneous L-glutamate release by TRPV1 activation (31% sEPSC frequency increase at 1 μM [18]), while suppressing frog sciatic nerve CAPs (36% amplitude reduction at 0.1 mM [204]). Similar spontaneous L-glutamate release enhancement was produced in menthol-induced TRPM8 activation (869% frequency increase at 0.5 mM in postnatal 10–11 days rat [20]; 411% frequency increase at 0.1 mM [25]; 295% frequency increase at 0.5 mM [26]; EC50 = 0.263 mM [23]; 409% frequency increase at 0.5 mM [27]) and in AITC-induced TRPA1 activation (202% frequency increase at 0.1 mM [21]; EC50 = 0.226 mM [23]; at 0.2 mM [22]; 192% frequency increase at 0.1 mM [24]). On the other hand, menthol and AITC inhibited frog sciatic nerve CAPs (IC50: 1.1, 0.93 and 1.5 mM for (-)-menthol, (+)-menthol [206] and AITC [207], respectively).
Moreover, spinal lamina II primary afferent central terminal TRPA1 was activated by various plant-derived compounds, cinnamaldehyde ((2E)-3-phenylprop-2-enal, which is contained in cinnamon [46]; 205% sEPSC frequency increase at 0.1 mM [21]; EC50 = 0.038 mM [23]; 192% frequency increase at 0.3 mM [24]), eugenol (2-methoxy-4-(2-propenyl) phenol, which is contained in clove and bay leaves [46]; EC50 = 3.8 mM [208]), zingerone (4-(4-hydroxy-3-methoxyphenyl)-2-butanone, which is contained in ginger [46]; EC50 = 1.3 mM [209]), carvacrol (5-isopropyl-2-methylphenol, which is contained in oregano and thyme essential oils [46]; EC50 = 0.69 mM [210]), (+)-carvone ((+)-2-methyl-5-(1-methylethenyl)-2-cyclohexenone, which is contained in caraway [46]; EC50 = 0.72 mM [211]), thymol (5-methyl-2-isopropylphenol, which is contained in thyme [46]; EC50 = 0.18 mM [212]), 1,8-cineole (1,3,3-trimethyl-2-oxabicyclo[2.2.2]octane, which is contained in eucalyptus and rosemary [213]; EC50 = 3.2 mM [27]) and citral (3,7-dimethyl-2,6-octadienal, which is contained in lemongrass [214]; EC50 = 0.58 mM [215]). All of the compounds reduced frog sciatic nerve CAP amplitudes (cinnamaldehyde: IC50 = 1.2 mM [207]; eugenol and zingerone: IC50 = 0.81 and 8.3 mM, respectively [204]; carvacrol: IC50 = 0.34 mM [206]; (+)-carvone: IC50 = 2.0 mM [206]; thymol: IC50 = 0.34 mM [206]; 1,8-cineole: IC50 = 5.7 mM [206]; citral: IC50 = 0.46 mM [216]).
On the other hand, a spearmint component (-)-carvone and 1,4-cineole (1-methyl-4-(1-methylethyl)-7-oxabicyclo[2.2.1]heptane, which is a minor component in plants containing 1,8-cineole [217]), exhibited both rat spinal lamina II primary afferent central terminal TRPV1, but not TRPA1, activation ((-)-carvone: EC50 = 0.70 mM [211]; 1,4-cineole: EC50 = 0.42 mM [27]) and frog sciatic nerve CAP inhibition (IC50: 1.4 and 7.2 mM, respectively, for (-)-carvone and 1,4-cineole [206]). (±)-Linalool ((±)-3,7-dimethylocta-1,6-diene-3-ol, which is contained in lavender [46]) and its isomer geraniol (trans-3,7-dimethyl-2,6-octadien-1-ol [46]), both of which suppressed frog sciatic nerve CAPs (IC50 = 1.7 and 0.53 mM, respectively, for (±)-linalool and geraniol [216]), activated different types of spinal lamina II primary afferent central terminal TRP. Thus, (±)-linalool activated both TRPV1 and TRPA1 (EC50 = 2.2 mM), and geraniol activated TRPM8 (EC50 = 1.6 mM [218]). An active ingredient of Zanthoxylum piperitum, hydroxy-α-sanshool((2E,6Z,8E,10E)-N-(2-hydroxy-2-methylpropyl)dodeca-2,6,8,10-tetraenamide), also had an ability to activate both TRPV1 and TRPA1 expressed in HEK cells [219], while inhibiting Na+ channels in mouse DRG neurons, Nav1.7 and Nav1.8 [220], and frog sciatic nerve CAPs (ca. 50% amplitude reduction at 0.05 mM [207]). A non-pungent capsaicin-like drug capsiate ((E)-8-methyl-6-nonenoic acid (4-hydroxy-3-methoxyphenyl)methyl ester; isolated from a non-pungent cultivar of CH-19 sweet red pepper; see [221] for review) activated both cloned TRPV1 [222] and TRPA1 (EC50 = 2.76 μM [223]), while inhibiting frog sciatic nerve CAPs (22% amplitude reduction at 0.1 mM [204]). Primary afferent central terminal TRPV1 in the spinal lamina II was activated by piperine (1-peperoylpiperidine; a pungent component of black pepper, isolated from the fruits of Piper [46]; EC50 = 52.3 μM [224]) that inhibited frog sciatic nerve CAPs (23% amplitude reduction at 0.05 mM; [207]) and Na+ channels (IC50 = 10 μM for rat TTX-sensitive Nav1.4 [225]). A naturally occurring chalcone, cardamonin (2′,4′-dihydroxy-6′-methoxychalcone; isolated from several plants including Alpinia conchigera [226]), which exhibited analgesia in chronic constriction injury-induced neuropathic pain mice model [227], inhibited human TRPA1 expressed in HEK293 cells, activated by cinnamaldehyde (IC50 = 0.454 μM [228]), while suppressing frog sciatic nerve CAPs (ca. 50% amplitude reduction at 3.7 mM [229]). It has been preliminarily shown by molecular docking and molecular dynamic simulation that cardamonin acts on Nav1.7 [230].
CAP inhibition similar to that in the frog sciatic nerve has been reported in mammalian nerve fibers. For instance, in the rat sciatic nerve, eugenol suppressed CAPs (almost completely block at 0.6 mM [231]), and 1,8-cineole did so with an IC50 value of 6–8 mM [232]. Citral, carvacrol, citronellol, (-)-carvone and (+)-carvone inhibited rat sciatic nerve CAPs with the IC50 values of 0.23 [233], 0.50 [234], 2.2 [235], 10.7 and 8.7 mM [236], respectively.
Consistent with the plant-derived compound-induced CAP inhibition, Na+ channels were suppressed by cinnamaldehyde (IC50 = 1.5 and 0.8 mM for TTX-sensitive and TTX-resistant Na+ currents, respectively, in mouse TG neurons [237]), eugenol (IC50 = 0.308 and 0.543 mM for TTX-sensitive and TTX-resistant Na+ currents, respectively, in rat DRG neurons [238]), zingerone (IC50 = 23.7 μM in pituitary tumor (GH3) cells [239]), carvacrol (IC50 = 0.37 mM in rat DRG neurons [234]), (-)-carvone (IC50 = 0.75 mM for Nav1.2 [240]), thymol (IC50 = 0.149 mM for rat type IIA Na+ channels expressed in HEK293 cells [195]), 1,8-cineole (partial blockade at 6 mM in rat superior cervical ganglion neurons [241]) and (±)-linalool (51% current amplitude reduction at 6 mM in rat DRG neurons [242]; see [243] for review).
As seen in capsaicin, AITC, menthol, anethole, resveratrol and curcumin activities, primary afferent (DRG and TG) neuron cell body and cloned TRP were also modulated by various plant-derived compounds, including cinnamaldehyde (EC50 = 61 μM for mouse TRPA1 activation in CHO cells [186]), eugenol (TRPV1 activation in HEK293 cells and TG neurons [244]), zingerone (EC50 = 15 mM for TRPV1 activation in rat TG neurons [190]), carvacrol (at 0.3 mM for human and rodent TRPA1 activation in HEK293 cells [245]; EC50 = 0.49 mM for TRPV3 activation in HEK293 cells [246]), (-)-carvone (EC50 = 1.3 mM for TRPV1 activation in HEK293 cells [247]; TRPV3 activation [246]), thymol (EC50 = 0.127 mM for human TRPA1 activation in HEK293 cells [248]; EC50 = 0.0522 mM for rat TRPM8 activation in HEK293 cells [249]; at 1 mM for human TRPV3 activation in HEK293 cells [250]), piperine (EC50 = 0.035 mM for TRPV1 activation in rat TG neurons [190]), citral (EC50 = 0.465, 0.926 and 0.0335 mM for rat TRPV1, TRPV3 and TRPM8 activation in HEK293 cells, respectively, and ca. 0.2 mM for rat TRPA1 activation threshold [251]), 1,8-cineole (at 5 mM for human TRPM8 activation in HEK293 cells; IC50 = 3.4 mM for AITC response inhibition [252]), 1,4-cineole (at 5 mM for human TRPM8 and TRPA1 activation in HEK293 cells [252]), (±)-linalool (EC50 = 6.7 mM for mouse TRPM8 activation in HEK293 cells [253]; EC50 = 0.117 mM for rat TRPA1 activation in HEK293 cells [254]) and geraniol (EC50 = 5.9 mM for mouse TRPM8 activation in HEK293 cells [253]; at 1 mM for each of rat TRPV1, TRPA1 and TRPM8 activation in CHO cells [255]). The results for these plant-derived compound actions are summarized in Table 2.
Traditional Japanese and Chinese medicines are used for a variety of purposes, including pain relief (see [256,257] for review). Many antinociceptive drugs used in such folk medicines, which are derived from plants, suppress sciatic nerve CAPs [258,259], possibly through Na+ channel inhibition. For example, a traditional Japanese medicine daikenchuto, which has an ability to alleviate intestinal obstruction-related abdominal pain [260], inhibited frog sciatic nerve CAPs (IC50 = 1.1 mg/mL [258]). Another traditional Japanese medicine inchinkoto, used for oral mucositis (see [261] for review), suppressed frog sciatic nerve CAPs (IC50 = 5.4 mg/mL [259]). Mitigation by inchinkoto of oral mucositis-related pain has been attributed to eugenol having an ability to modulate TRP (see [259]). Astragaloside, isolated from a traditional Chinese medicinal plant, which alleviates inflammatory pain [262], inhibited frog sciatic nerve CAPs (25% amplitude reduction at 50 mg/mL [263]). Since many plant-derived compounds modulate TRP, it is possible that traditional medicinal drugs act on both Na+ and TRP channels. In support of this idea, daikenchuto is shown to contain TRP agonists, such as hydroxy-α-sanshool (for instance, see [264]). This issue remains to be further examined.

11. Voltage-Gated Na+ Channels and TRP Channels

As mentioned above, the chemical compounds that affect nerve AP conduction and/or Na+ channels also modify TRP activation (see Table 1 and Table 2). Consistent with this result, TRP channels are similar in chemical structure to Na+ channels. TRP channels are composed of homo- or hetero-tetramers of subunits having six membrane-spanning segments [4], while the α subunit of the Na+ channels, which forms an ion conduction pore, consists of four repeated domains, each of which contains six membrane-spanning segments [265]. In either case, their six segments (S1-S6) contain an ion-permeable pathway formed by S5, S6 and the intervening pore loop region. Their detailed chemical structures have been revealed by using single particle analysis with cryo-electron microscopy (see [266,267,268] for TRPV1; [269] for TRPA1; [270,271] for TRPM8; [272] for TRPM2; [273] for TRPM3; [274] for TRPM7; [275] for TRPC3; [276] for TRPC5; [275,277] for TRPC6; see [278] for review of Na+ channels). The compounds given in Table 1 and Table 2 may act on channel proteins themselves or on cell membrane lipids in close proximity to the proteins, and thereby modify ion channel function. Since their channel proteins have intracellular sites that undergo phosphorylation by second messengers to alter channel function [4,265], the compounds may not act directly on those channels. Although the structure–function relationships of Na+ and TRP channels have been studied in detail [279,280], it is beyond the scope of this review to describe at which sites on these channels the various compounds presented in this review act. In the future, it will be necessary to clarify a common site in two types of the TRP and Na+ channels on which each of the compounds acts by using molecular docking methods, which allow detailed understanding of the interaction between ion channels and ligands (for example, see [281] for review).
When considering the role of Na+ and TRP channels in nociceptive transmission, it may be important to be aware of the differences in the efficiency of drug action on these channels. However, because it is unlikely that a drug would act simultaneously on the peripheral and central terminals and nerve fibers of primary afferent neurons, it may be difficult to obtain meaningful concentration-dependence for drug action via comparisons of the efficacy of a drug acting on each. With respect to practical application, considering that the mM order values of many analgesic substances for CAP inhibition are higher than those required to activate TRPV1 or TRPA1 in primary afferent neurons, topically administrated substances are likely to produce analgesia by inhibiting pain conduction rather than by activating TRPV1 or TRPA1 in the peripheral terminals and causing pain.

12. Conclusions

Nociceptive pain is a physiological mechanism useful to protect a person against injury to tissues, while neuropathic pain is due to a long-lasting excessive increase in the excitability of neurons near injured neuronal tissues and thus negatively impacts on the quality of life and well-being [1].Clinically, nociceptive pain is alleviated by opioids and NSAIDs, while neuropathic pain is mitigated by using analgesic adjuvants, such as local anesthetics, antiepileptics, antidepressants and α2-adrenoceptor agonists. General anesthetics used for consciousness loss in surgery also cause analgesia. Many plant-derived compounds produce analgesia, as evidenced by the use of herbal medicines to relieve pain in traditional medicine. Other drugs, such as methylglyoxal, nicotine, pregnenolone sulfate, riluzole and anandamide, also have an antinociceptive action. This review demonstrated that their chemical compounds exhibit both TRP and Na+ channel modulation. This result indicates that a modulatory site on TRP channel protein may be similar in chemical structure to that of Na+ channel protein. It remains to be clarified what structures common to TRP and Na+ channels the drugs act on, and what drugs beside those mentioned here act on both. Primary afferent peripheral terminal TRP activation produces APs, while Na+ channel inhibition results in depressed AP conduction from the periphery to the CNS. Primary afferent central terminal TRP activation in the spinal and brain stem lamina II leads to nociceptive transmission modulation. Clarifying the common site of drug action in TRP and Na+ channels may be useful for development of a new analgesic substance that act on both of the channels near the peripheral and central terminals of primary afferent neuron.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Fields, H.L. Pain; McGraw-Hill: New York, NY, USA, 1987. [Google Scholar]
  2. Ramsey, S.; Delling, M.; Clapham, D.E. An introduction to TRP channels. Annu. Rev. Physiol. 2006, 68, 619–647. [Google Scholar] [CrossRef]
  3. Patapoutian, A.; Tate, S.; Woolf, C.J. Transient receptor potential channels: Targeting pain at the source. Nat. Rev. Drug Discov. 2009, 8, 55–68. [Google Scholar] [CrossRef]
  4. Julius, D. TRP channels and pain. Annu. Rev. Cell Dev. Biol. 2013, 29, 355–384. [Google Scholar] [CrossRef] [PubMed]
  5. Sun, Z.-C.; Ma, S.-B.; Chu, W.-G.; Jia, D.; Luo, C. Canonical transient receptor potential (TRPC) channels in nociception and pathological pain. Neural Plast. 2020, 2020, 3764193. [Google Scholar] [CrossRef]
  6. Guo, A.; Vulchanova, L.; Wang, J.; Li, X.; Elde, R. Immunocytochemical localization of the vanilloid receptor 1 (VR1): Relationship to neuropeptides, the P2X3 purinoceptor and IB4 binding sites. Eur. J. Neurosci. 1999, 11, 946–958. [Google Scholar] [CrossRef] [PubMed]
  7. Valtschanoff, J.G.; Rustioni, A.; Guo, A.; Hwang, S.J. Vanilloid receptor VR1 is both presynaptic and postsynaptic in the superficial laminae of the rat dorsal horn. J. Comp. Neurol. 2001, 436, 225–235. [Google Scholar] [CrossRef]
  8. Hwang, S.J.; Burette, A.; Rustioni, A.; Valtschanoff, J.G. Vanilloid receptor VR1-positive primary afferents are glutamatergic and contact spinal neurons that co-express neurokinin receptor NK1 and glutamate receptors. J. Neurocytol. 2004, 33, 321–329. [Google Scholar] [CrossRef]
  9. Kobayashi, K.; Fukuoka, T.; Obata, K.; Yamanaka, H.; Dai, Y.; Tokunaga, A.; Noguchi, K. Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with Aδ/C-fibers and colocalization with Trk receptors. J. Comp. Neurol. 2005, 493, 596–606. [Google Scholar] [CrossRef] [PubMed]
  10. Kim, Y.S.; Son, J.Y.; Kim, T.H.; Paik, S.K.; Dai, Y.; Noguchi, K.; Ahn, D.K.; Bae, Y.C. Expression of transient receptor potential ankyrin 1 (TRPA1) in the rat trigeminal sensory afferents and spinal dorsal horn. J. Comp. Neurol. 2010, 518, 687–698. [Google Scholar] [CrossRef] [PubMed]
  11. Elg, S.; Marmigere, F.; Mattsson, J.P.; Ernfors, P. Cellular subtype distribution and developmental regulation of TRPC channel members in the mouse dorsal root ganglion. J. Comp. Neurol. 2007, 503, 35–46. [Google Scholar] [CrossRef]
  12. Vriens, J.; Owsianik, G.; Hofmann, T.; Philipp, S.E.; Stab, J.; Chen, X.; Benoit, M.; Xue, F.; Janssens, A.; Kerselaers, S.; et al. TRPM3 is a nociceptor channel involved in the detection of noxious heat. Neuron 2011, 70, 482–494. [Google Scholar] [CrossRef] [PubMed]
  13. Vandewauw, I.; Owsianik, G.; Voets, T. Systematic and quantitative mRNA expression analysis of TRP channel genes at the single trigeminal and dorsal root ganglion level in mouse. BMC Neurosci. 2013, 14, 21. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, J.; Zhao, M.; Jia, P.; Liu, F.-F.; Chen, K.; Meng, F.-Y.; Hong, J.-H.; Zhang, T.; Jin, X.-H.; Shi, J. The analgesic action of larixyl acetate, a potent TRPC6 inhibitor, in rat neuropathic pain model induced by spared nerve injury. J. Neuroinflammation 2020, 17, 118. [Google Scholar] [CrossRef]
  15. Quick, K.; Zhao, J.; Eijkelkamp, N.; Linley, J.E.; Rugiero, F.; Cox, J.J.; Raouf, R.; Gringhuis, M.; Sexton, J.E.; Abramowitz, J.; et al. TRPC3 and TRPC6 are essential for normal mechanotransduction in subsets of sensory neurons and cochlear hair cells. Open Biol. 2012, 2, 120068. [Google Scholar] [CrossRef]
  16. Kittaka, H.; Tominaga, M. The molecular and cellular mechanisms of itch and the involvemen of TRP channels in the peripheral sensory nervous system and skin. Allergol. Int. 2017, 66, 22–30. [Google Scholar] [CrossRef]
  17. Mihara, S.; Shibamoto, T. The role of flavor and fragrance chemicals in TRPA1 (transient receptor potential cation channel, member A1) activity associated with allergies. Allergy Asthma Clin. Immunol. 2015, 11, 11. [Google Scholar] [CrossRef]
  18. Yang, K.; Kumamoto, E.; Furue, H.; Yoshimura, M. Capsaicin facilitates excitatory but not inhibitory synaptic transmission in substantia gelatinosa of the rat spinal cord. Neurosci. Lett. 1998, 255, 135–138. [Google Scholar] [CrossRef]
  19. Jennings, E.A.; Vaughan, C.W.; Roberts, L.A.; Christie, M.J. The actions of anandamide on rat superficial medullary dorsal horn neurons in vitro. J. Physiol. 2003, 548, 121–129. [Google Scholar] [CrossRef]
  20. Baccei, M.L.; Bardoni, R.; Fitzgerald, M. Development of nociceptive synaptic inputs to the neonatal rat dorsal horn: Glutamate release by capsaicin and menthol. J. Physiol. 2003, 549, 231–242. [Google Scholar] [CrossRef] [PubMed]
  21. Kosugi, M.; Nakatsuka, T.; Fujita, T.; Kuroda, Y.; Kumamoto, E. Activation of TRPA1 channel facilitates excitatory synaptic transmission in substantia gelatinosa neurons of the adult rat spinal cord. J. Neurosci. 2007, 27, 4443–4451. [Google Scholar] [CrossRef] [PubMed]
  22. Sun, B.; Bang, S.-I.; Jin, Y.-H. Transient receptor potential A1 increase glutamate release on brain stem neurons. NeuroReport 2009, 20, 1002–1006. [Google Scholar] [CrossRef] [PubMed]
  23. Wrigley, P.J.; Jeong, H.-J.; Vaughan, C.W. Primary afferents with TRPM8 and TRPA1 profiles target distinct subpopulations of rat superficial dorsal horn neurones. Br. J. Pharmacol. 2009, 157, 371–380. [Google Scholar] [CrossRef] [PubMed]
  24. Uta, D.; Furue, H.; Pickering, A.E.; Rashid, M.H.; Mizuguchi-Takase, H.; Katafuchi, T.; Imoto, K.; Yoshimura, M. TRPA1-expressing primary afferents synapse with a morphologically identified subclass of substantia gelatinosa neurons in the adult rat spinal cord. Eur. J. Neurosci. 2010, 31, 1960–1973. [Google Scholar] [CrossRef] [PubMed]
  25. Tsuzuki, K.; Xing, H.; Ling, J.; Gu, J.G. Menthol-induced Ca2+ release from presynaptic Ca2+ stores potentiates sensory synaptic transmission. J. Neurosci. 2004, 24, 762–771. [Google Scholar] [CrossRef] [PubMed]
  26. Suzuki, S.C.; Furue, H.; Koga, K.; Jiang, N.; Nohmi, M.; Shimazaki, Y.; Katoh-Fukui, Y.; Yokoyama, M.; Yoshimura, M.; Takeichi, M. Cadherin-8 is required for the first relay synapses to receive functional inputs from primary sensory afferents for cold sensation. J. Neurosci. 2007, 27, 3466–3476. [Google Scholar] [CrossRef] [PubMed]
  27. Jiang, C.-Y.; Wang, C.; Xu, N.-X.; Fujita, T.; Murata, Y.; Kumamoto, E. 1,8- and 1,4-cineole enhance spontaneous excitatory transmission by activating different types of transient receptor potential channels in the rat spinal substantia gelatinosa. J. Neurochem. 2016, 136, 764–777. [Google Scholar] [CrossRef] [PubMed]
  28. Sun, Z.-C.; Han, W.-J.; Dou, Z.-W.; Lu, N.; Wang, X.; Wang, F.-D.; Ma, S.-B.; Tian, Z.-C.; Xian, H.; Liu, W.-N.; et al. TRPC3/6 channels mediate mechanical pain hypersensitivity via enhancement of nociceptor excitability and of spinal synaptic transmission. Adv. Sci. 2024, 11, e2404342. [Google Scholar] [CrossRef] [PubMed]
  29. Kumamoto, E.; Fujita, T.; Jiang, C.-Y. TRP channels involved in spontaneous L-glutamate release enhancement in the adult rat spinal substantia gelatinosa. Cells 2014, 3, 331–362. [Google Scholar] [CrossRef]
  30. Kumamoto, E.; Fujita, T. Differential activation of TRP channels in the adult rat spinal substantia gelatinosa. Pharmaceuticals 2016, 9, 46. [Google Scholar] [CrossRef]
  31. Willis, W.D., Jr.; Coggeshall, R.E. Sensory Mechanisms of the Spinal Cord, 2nd ed.; Plenum: New York, NY, USA, 1991. [Google Scholar]
  32. Todd, A.J. Neuronal circuitry for pain processing in the dorsal horn. Nat. Rev. Neurosci. 2010, 11, 823–836. [Google Scholar] [CrossRef]
  33. Zeilhofer, H.U.; Wildner, H.; Yévenes, G.E. Fast synaptic inhibition in spinal sensory processing and pain control. Physiol. Rev. 2012, 92, 193–235. [Google Scholar] [CrossRef]
  34. Fürst, S. Transmitters involved in antinociception in the spinal cord. Brain Res. Bull. 1999, 48, 129–141. [Google Scholar] [CrossRef] [PubMed]
  35. Merighi, A. The histology, physiology, neurochemistry and circuitry of the substantia gelatinosa Rolandi (lamina II) in mammalian spinal cord. Prog. Neurobiol. 2018, 169, 91–134. [Google Scholar] [CrossRef]
  36. Kumamoto, E. Cellular mechanisms for antinociception produced by oxytocin and orexins in the rat spinal lamina II—Comparison with those of other endogenous pain modulators. Pharmaceuticals 2019, 12, 136. [Google Scholar] [CrossRef] [PubMed]
  37. Lappin, S.C.; Randall, A.D.; Gunthorpe, M.J.; Morisset, V. TRPV1 antagonist, SB-366791, inhibits glutamatergic synaptic transmission in rat spinal dorsal horn following peripheral inflammation. Eur. J. Pharmacol. 2006, 540, 73–81. [Google Scholar] [CrossRef] [PubMed]
  38. da Costa, D.S.M.; Meotti, F.C.; Andrade, E.L.; Leal, P.C.; Motta, E.M.; Calixto, J.B. The involvement of the transient receptor potential A1 (TRPA1) in the maintenance of mechanical and cold hyperalgesia in persistent inflammation. Pain 2010, 148, 431–437. [Google Scholar] [CrossRef] [PubMed]
  39. Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic pain. Nat. Rev. Dis. Primers 2017, 3, 17002. [Google Scholar] [CrossRef]
  40. Proudfoot, C.J.; Garry, E.M.; Cottrell, D.F.; Rosie, R.; Anderson, H.; Robertson, D.C.; Fleetwood-Walker, S.M.; Mitchell, R. Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. Curr. Biol. 2006, 16, 1591–1605. [Google Scholar] [CrossRef]
  41. Alawi, K.M.; Russell, F.A.; Aubdool, A.A.; Srivastava, S.; Riffo-Vasquez, Y.; Baldissera, L., Jr.; Thakore, P.; Saleque, N.; Fernandes, E.S.; Walsh, D.A.; et al. Transient receptor potential canonical 5 (TRPC5) protects against pain and vascular inflammation in arthritis and joint inflammation. Ann. Rheum. Dis. 2017, 76, 252–260. [Google Scholar] [CrossRef]
  42. Wang, H.; Song, T.; Wang, W.; Zhang, Z. TRPM2 participates the transformation of acute pain to chronic pain during injury-induced neuropathic pain. Synapse 2019, 73, e22117. [Google Scholar] [CrossRef] [PubMed]
  43. Meotti, F.C.; de Andrade, E.L.; Calixto, J.B. TRP modulation by natural compounds. Hadb. Exp. Pharmacol. 2014, 223, 1177–1238. [Google Scholar]
  44. Premkumar, L.S. Transient receptor potential channels as targets for phytochemicals. ACS Chem. Neurosci. 2014, 5, 1117–1130. [Google Scholar] [CrossRef]
  45. Szolcsányi, J. Forty years in capsaicin research for sensory pharmacology and physiology. Neuropeptides 2004, 38, 377–384. [Google Scholar] [CrossRef] [PubMed]
  46. Vriens, J.; Nilius, B.; Vennekens, R. Herbal compounds and toxins modulating TRP channels. Curr. Neuropharmacol. 2008, 6, 79–96. [Google Scholar] [PubMed]
  47. Eccles, R. Menthol and related cooling compounds. J. Pharm. Pharmacol. 1994, 46, 618–630. [Google Scholar] [CrossRef]
  48. Staruschenko, A.; Jeske, N.A.; Akopian, A.N. Contribution of TRPV1-TRPA1 interaction to the single channel properties of the TRPA1 channel. J. Biol. Chem. 2010, 285, 15167–15177. [Google Scholar] [CrossRef] [PubMed]
  49. Tian, W.; Fu, Y.; Wang, D.H.; Cohen, D.M. Regulation of TRPV1 by a novel renally expressed rat TRPV1 splice variant. Am. J. Physiol. Ren. Physiol. 2006, 290, F117–F126. [Google Scholar] [CrossRef] [PubMed]
  50. Zhou, Y.; Suzuki, Y.; Uchida, K.; Tominaga, M. Identification of a splice variant of mouse TRPA1 that regulates TRPA1 activity. Nat. Commun. 2013, 4, 2399. [Google Scholar] [CrossRef]
  51. Dai, Y.; Wang, S.; Tominaga, M.; Yamamoto, S.; Fukuoka, T.; Higashi, T.; Kobayashi, K.; Obata, K.; Yamanaka, H.; Noguchi, K. Sensitization of TRPA1 by PAR2 contributes to the sensation of inflammatory pain. J. Clin. Investig. 2007, 117, 1979–1987. [Google Scholar] [CrossRef]
  52. Schmidt, M.; Dubin, A.E.; Petrus, M.J.; Earley, T.J.; Patapoutian, A. Nociceptive signals induce trafficking of TRPA1 to the plasma membrane. Neuron 2009, 64, 498–509. [Google Scholar] [CrossRef]
  53. Wang, S.; Dai, Y.; Fukuoka, T.; Yamanaka, H.; Kobayashi, K.; Obata, K.; Cui, X.; Tominaga, M.; Noguchi, K. Phospholipase C and protein kinase A mediate bradykinin sensitization of TRPA1: A molecular mechanism of inflammatory pain. Brain 2008, 131, 1241–1251. [Google Scholar] [CrossRef] [PubMed]
  54. Kumamoto, E. Effects of plant-derived compounds on excitatory synaptic transmission and nerve conduction in the nervous system—Involvement in pain modulation. Curr. Top. Phytochem. 2018, 14, 45–70. [Google Scholar]
  55. Hille, B. Ionic Channels of Excitable Membranes; Sinauer Associates Inc.: Sunderland, MA, USA, 1984. [Google Scholar]
  56. Scholz, A. Mechanisms of (local) anaesthetics on voltage-gated sodium and other ion channels. Br. J. Anaesth. 2002, 89, 52–61. [Google Scholar] [CrossRef]
  57. Backonja, M.-M. Local anesthetics as adjuvant analgesics. J. Pain Symptom Manag. 1994, 9, 491–499. [Google Scholar] [CrossRef] [PubMed]
  58. Kalso, E.; Tramèr, M.R.; McQuay, H.J.; Moore, R.A. Systemic local-anaesthetic-type drugs in chronic pain: A systematic review. Eur. J. Pain 1998, 2, 3–14. [Google Scholar] [CrossRef]
  59. Leffler, A.; Fischer, M.J.; Rehner, D.; Kienel, S.; Kistner, K.; Sauer, S.K.; Gavva, N.R.; Reeh, P.W.; Nau, C. The vanilloid receptor TRPV1 is activated and sensitized by local anesthetics in rodent sensory neurons. J. Clin. Investig. 2008, 118, 763–776. [Google Scholar] [CrossRef]
  60. Leffler, A.; Lattrell, A.; Kronewald, S.; Niedermirtl, F.; Nau, C. Activation of TRPA1 by membrane permeable local anesthetics. Mol. Pain 2011, 7, 62. [Google Scholar] [CrossRef]
  61. Bräu, M.E.; Vogel, W.; Hempelmann, G. Fundamental properties of local anesthetics: Half-maximal blocking concentrations for tonic block of Na+ and K+ channels in peripheral nerve. Anesth. Analg. 1998, 87, 885–889. [Google Scholar]
  62. Piao, L.-H.; Fujita, T.; Jiang, C.-Y.; Liu, T.; Yue, H.-Y.; Nakatsuka, T.; Kumamoto, E. TRPA1 activation by lidocaine in nerve terminals results in glutamate release increase. Biochem. Biophys. Res. Commun. 2009, 379, 980–984. [Google Scholar] [CrossRef] [PubMed]
  63. Piao, L.-H.; Fujita, T.; Yu, T.; Kumamoto, E. Presynaptic facilitation by tetracaine of glutamatergic spontaneous excitatory transmission in the rat spinal substantia gelatinosa—Involvement of TRPA1 channels. Brain Res. 2017, 1657, 245–252. [Google Scholar] [CrossRef] [PubMed]
  64. Katsuki, R.; Fujita, T.; Koga, A.; Liu, T.; Nakatsuka, T.; Nakashima, M.; Kumamoto, E. Tramadol, but not its major metabolite (mono-O-demethyl tramadol) depresses compound action potentials in frog sciatic nerves. Br. J. Pharmacol. 2006, 149, 319–327. [Google Scholar] [CrossRef] [PubMed]
  65. Kosugi, T.; Mizuta, K.; Fujita, T.; Nakashima, M.; Kumamoto, E. High concentrations of dexmedetomidine inhibit compound action potentials in frog sciatic nerves without α2 adrenoceptor activation. Br. J. Pharmacol. 2010, 160, 1662–1676. [Google Scholar] [CrossRef]
  66. Lee-Son, S.; Wang, G.K.; Concus, A.; Crill, E.; Strichartz, G. Stereoselective inhibition of neuronal sodium channels by local anesthetics. Evidence for two sites of action? Anesthesiology 1992, 77, 324–335. [Google Scholar] [CrossRef] [PubMed]
  67. Uemura, Y.; Fujita, T.; Ohtsubo, S.; Hirakawa, N.; Sakaguchi, Y.; Kumamoto, E. Effects of various antiepileptics used to alleviate neuropathic pain on compound action potential in frog sciatic nerves: Comparison with those of local anesthetics. BioMed Res. Int. 2014, 2014, 540238. [Google Scholar] [CrossRef]
  68. Brenneis, C.; Kistner, K.; Puopolo, M.; Jo, S.; Roberson, D.P.; Sisignano, M.; Segal, D.; Cobos, E.J.; Wainger, B.J.; Labocha, S.; et al. Bupivacaine-induced cellular entry of QX-314 and its contribution to differential nerve block. Br. J. Pharmacol. 2014, 171, 438–451. [Google Scholar] [CrossRef] [PubMed]
  69. Yilmaz-Rastoder, E.; Gold, M.S.; Hough, K.A.; Gebhart, G.F.; Williams, B.A. Effect of adjuvant drugs on the action of local anesthetics in isolated rat sciatic nerves. Reg. Anesth. Pain Med. 2012, 37, 403–409. [Google Scholar] [CrossRef] [PubMed]
  70. Gissen, A.J.; Covino, B.G.; Gregus, J. Differential sensitivities of mammalian nerve fibers to local anesthetic agents. Anesthesiology 1980, 53, 467–474. [Google Scholar] [CrossRef]
  71. Binshtok, A.M.; Bean, B.P.; Woolf, C.J. Inhibition of nociceptors by TRPV1-mediated entry of impermeant sodium channel blockers. Nature 2007, 449, 607–610. [Google Scholar] [CrossRef]
  72. Puopolo, M.; Binshtok, A.M.; Yao, G.-L.; Oh, S.B.; Woolf, C.J.; Bean, B.P. Permeation and block of TRPV1 channels by the cationic lidocaine derivative QX-314. J. Neurophysiol. 2013, 109, 1704–1712. [Google Scholar] [CrossRef]
  73. Rivera-Acevedo, R.E.; Pless, S.A.; Ahern, C.A.; Schwarz, S.K.W. The quaternary lidocaine derivative, QX-314, exerts biphasic effects on transient receptor potential vanilloid subtype 1 channels in vitro. Anesthesiology 2011, 114, 1425–1434. [Google Scholar] [CrossRef] [PubMed]
  74. Stueber, T.; Eberhardt, M.J.; Hadamitzky, C.; Jangra, A.; Schenk, S.; Dick, F.; Stoetzer, C.; Kistner, K.; Reeh, P.W.; Binshtok, A.M.; et al. Quaternary lidocaine derivative QX-314 activates and permeates human TRPV1 and TRPA1 to produce inhibition of sodium channels and cytotoxicity. Anesthesiology 2016, 124, 1153–1165. [Google Scholar] [CrossRef] [PubMed]
  75. Hemmings, H.C., Jr.; Akabas, M.H.; Goldstein, P.A.; Trudell, J.R.; Orser, B.A.; Harrison, N.L. Emerging molecular mechanisms of general anesthetic action. Trends in Pharmacol. Sci. 2005, 26, 503–510. [Google Scholar] [CrossRef]
  76. Eilers, H.; Yost, S. General anesthetics. In Basic & Clinical Pharmacology, 14th ed.; Katzung, B.G., Ed.; McGraw-Hill, Medical Publishing Division: New York, NY, USA, 2018; Chapter 25; pp. 440–458. [Google Scholar]
  77. Hua, T.; Chen, B.; Lu, D.; Sakurai, K.; Zhao, S.; Han, B.-X.; Kim, J.; Yin, L.; Chen, Y.; Lu, J.; et al. General anesthetics activate a potent central pain-suppression circuit in the amygdala. Nat. Neurosci. 2020, 23, 854–868. [Google Scholar] [CrossRef] [PubMed]
  78. Tsutsumi, S.; Tomioka, A.; Sudo, M.; Nakamura, A.; Shirakura, K.; Takagishi, K.; Kohama, K. Propofol activates vanilloid receptor channels expressed in human embryonic kidney 293 cells. Neurosci. Lett. 2001, 312, 45–49. [Google Scholar] [CrossRef]
  79. Matta, J.A.; Cornett, P.M.; Miyares, R.L.; Abe, K.; Sahibzada, N.; Ahern, G.P. General anesthetics activate a nociceptive ion channel to enhance pain and inflammation. Proc. Natl. Acad. Sci. USA 2008, 105, 8784–8789. [Google Scholar] [CrossRef]
  80. Fischer, M.J.M.; Leffler, A.; Niedermirtl, F.; Kistner, K.; Eberhardt, M.; Reeh, P.W.; Nau, C. The general anesthetic propofol excites nociceptors by activating TRPV1 and TRPA1 rather than GABAA receptors. J. Biol. Chem. 2010, 285, 34781–34792. [Google Scholar] [CrossRef]
  81. Zhang, H.; Wickley, P.J.; Sinha, S.; Bratz, I.N.; Damron, D.S. Propofol restores transient receptor potential vanilloid receptor subtype-1 sensitivity via activation of transient receptor potential ankyrin receptor subtype-1 in sensory neurons. Anesthesiology 2011, 114, 1169–1179. [Google Scholar] [CrossRef]
  82. Frenkel, C.; Urban, B.W. Human brain sodium channels as one of the molecular target sites for the new intravenous anaesthetic propofol (2,6-diisopropylphenol). Eur. J. Pharmacol. 1991, 208, 75–79. [Google Scholar] [CrossRef]
  83. Veintemilla, F.; Elinder, F.; Århem, P. Mechanisms of propofol action on ion currents in the myelinated axon of Xenopus laevis. Eur. J. Pharmacol. 1992, 218, 59–68. [Google Scholar] [CrossRef]
  84. Ratnakumari, L.; Hemmings, H.C., Jr. Effects of propofol on sodium channel-dependent sodium influx and glutamate release in rat cerebrocortical synaptosomes. Anesthesiology 1997, 86, 428–439. [Google Scholar] [CrossRef]
  85. Rehberg, B.; Duch, D.S. Suppression of central nervous system sodium channels by propofol. Anesthesiology 1999, 91, 512–520. [Google Scholar] [CrossRef] [PubMed]
  86. Kobayashi, M.; Oi, Y. Actions of propofol on neurons in the cerebral cortex. J. Nippon Med. Sch. 2017, 84, 165–169. [Google Scholar] [CrossRef]
  87. Ton, H.T.; Phan, T.X.; Ahern, G.P. Inhibition of ligand-gated TRPA1 by general anesthetics. Mol. Pharmacol. 2020, 98, 185–191. [Google Scholar] [CrossRef] [PubMed]
  88. Magori, N.; Fujita, T.; Mizuta, K.; Kumamoto, E. Inhibition by general anesthetic propofol of compound action potentials in the frog sciatic nerve and its chemical structure. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 359–369. [Google Scholar] [CrossRef] [PubMed]
  89. Qiu, J.X.; Yang, Y.X.; Liu, J.; Zhao, W.L.; Li, Q.; Zhu, T.; Liang, P.; Zhou, C. The volatile anesthetic isoflurane differentially inhibits voltage-gated sodium channel currents between pyramidal and parvalbumin neurons in the prefrontal cortex. Front. Neural Circuits 2023, 17, 1185095. [Google Scholar] [CrossRef] [PubMed]
  90. López-González, M.J.; Luis, E.; Fajardo, O.; Meseguer, V.; Gers-Barlag, K.; Niñerola, S.; Viana, F. TRPA1 channels mediate human gingival fibroblast response to phenytoin. J. Dent. Res. 2017, 96, 832–839. [Google Scholar] [CrossRef]
  91. Molnár, P.; Erdö, S.L. Vinpocetine is as potent as phenytoin to block voltage-gated Na+ channels in rat cortical neurons. Eur. J. Pharmacol. 1995, 273, 303–306. [Google Scholar] [CrossRef] [PubMed]
  92. Lang, D.G.; Wang, C.M.; Cooper, B.R. Lamotrigine, phenytoin and carbamazepine interactions on the sodium current present in N4TG1 mouse neuroblastoma cells. J. Pharmacol. Exp. Ther. 1993, 266, 829–835. [Google Scholar] [PubMed]
  93. McLean, M.J.; Macdonald, R.L. Carbamazepine and 10,11-epoxycarbamazepine produce use- and voltage-dependent limitation of rapidly firing action potentials of mouse central neurons in cell culture. J. Pharmacol. Exp. Ther. 1986, 238, 727–738. [Google Scholar]
  94. Bektas, N.; Tekes, F.A.; Eken, H.; Arslan, R. The antihyperalgesic effects of gabapentinoids, carbamazepine and its keto analogue, oxcarbazepine, in capsaicin-induced thermal hyperalgesia. World J. Pharm. Sci. 2019, 7, 1–6. [Google Scholar]
  95. Forster, A.B.; Reeh, P.W.; Messlinger, K.; Fischer, M.J.M. High concentrations of morphine sensitize and activate mouse dorsal root ganglia via TRPV1 and TRPA1 receptors. Mol. Pain 2009, 5, 17. [Google Scholar] [CrossRef]
  96. Hu, S.; Rubly, N. Effects of morphine on ionic currents in frog node of Ranvier. Eur. J. Pharmacol. 1983, 95, 185–192. [Google Scholar] [CrossRef]
  97. Leffler, A.; Frank, G.; Kistner, K.; Niedermirtl, F.; Koppert, W.; Reeh, P.W.; Nau, C. Local anesthetic-like inhibition of voltage-gated Na+ channels by the partial μ-opioid receptor agonist buprenorphine. Anesthesiology 2012, 116, 1335–1346. [Google Scholar] [CrossRef]
  98. Mizuta, K.; Fujita, T.; Nakatsuka, T.; Kumamoto, E. Inhibitory effects of opioids on compound action potentials in frog sciatic nerves and their chemical structures. Life Sci. 2008, 83, 198–207. [Google Scholar] [CrossRef]
  99. Mizuta, K.; Fujita, T.; Kumamoto, E. Inhibition by morphine and its analogs of action potentials in adult rat dorsal root ganglion neurons. J. Neurosci. Res. 2012, 90, 1830–1841. [Google Scholar] [CrossRef]
  100. Haeseler, G.; Foadi, N.; Ahrens, J.; Dengler, R.; Hecker, H.; Leuwer, M. Tramadol, fentanyl and sufentanil but not morphine block voltage-operated sodium channels. Pain 2006, 126, 234–244. [Google Scholar] [CrossRef] [PubMed]
  101. Marincsák, R.; Tóth, B.I.; Czifra, G.; Szabó, T.; Kovács, L.; Bíró, T. The analgesic drug, tramadol, acts as an agonist of the transient receptor potential vanilloid-1. Anesth. Analg. 2008, 106, 1890–1896. [Google Scholar] [CrossRef] [PubMed]
  102. Mert, T.; Gunes, Y.; Guven, M.; Gunay, I.; Gocmen, C. Differential effects of lidocaine and tramadol on modified nerve impulse by 4-aminopyridine in rats. Pharmacology 2003, 69, 68–73. [Google Scholar] [CrossRef] [PubMed]
  103. Miyano, K.; Minami, K.; Yokoyama, T.; Ohbuchi, K.; Yamaguchi, T.; Murakami, S.; Shiraishi, S.; Yamamoto, M.; Matoba, M.; Uezono, Y. Tramadol and its metabolite M1 selectively suppress transient receptor potential ankyrin 1 activity, but not transient receptor potential vanilloid 1 activity. Anesth. Analg. 2015, 120, 790–798. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, B.-S.; Peng, H.; Wu, S.-N. Dexmedetomidine, an α2-adrenergic agonist, inhibits neuronal delayed-rectifier potassium current and sodium current. Br. J. Anaesth. 2009, 103, 244–254. [Google Scholar] [CrossRef]
  105. Taylan, S.B.; Bariskaner, H. Effects of dexmedetomidine and dexketoprofen on the conduction block of rat sciatic nerve. Neural Regen. Res. 2020, 15, 929–935. [Google Scholar] [CrossRef] [PubMed]
  106. Matsushita, Y.; Manabe, M.; Kitamura, N.; Shibuya, I. Adrenergic receptors inhibit TRPV1 activity in the dorsal root ganglion neurons of rats. PLoS ONE 2018, 13, e0191032. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, B.M.; Jang, Y.; Park, G.; Kim, K.; Oh, S.H.; Shin, T.J.; Chung, G. Dexmedetomidine modulates transient receptor potential vanilloid subtype 1. Biochem. Biophys. Res. Commun. 2019, 522, 832–837. [Google Scholar] [CrossRef]
  108. Akpinar, H.; Naziroğlu, M.; Övey, I.S.; Çiğ, B.; Akpinar, O. The neuroprotective action of dexmedetomidine on apoptosis, calcium entry and oxidative stress in cerebral ischemia-induced rats: Contribution of TRPM2 and TRPV1 channels. Sci. Rep. 2016, 6, 37196. [Google Scholar] [CrossRef] [PubMed]
  109. Ishii, H.; Kohno, T.; Yamakura, T.; Ikoma, M.; Baba, H. Action of dexmedetomidine on the substantia gelatinosa neurons of the rat spinal cord. Eur. J. Neurosci. 2008, 27, 3182–3190. [Google Scholar] [CrossRef]
  110. Demirdaş, A.; Nazıroğlu, M.; Övey, İ.S. Duloxetine reduces oxidative stress, apoptosis, and Ca2+ entry through modulation of TRPM2 and TRPV1 channels in the hippocampus and dorsal root ganglion of rats. Mol. Neurobiol. 2017, 54, 4683–4695. [Google Scholar] [CrossRef]
  111. Wang, S.-Y.; Calderon, J.; Wang, G.K. Block of neuronal Na+ channels by antidepressant duloxetine in a state-dependent manner. Anesthesiology 2010, 113, 655–665. [Google Scholar] [CrossRef]
  112. Stoetzer, C.; Papenberg, B.; Doll, T.; Völker, M.; Heineke, J.; Stoetzer, M.; Wegner, F.; Leffler, A. Differential inhibition of cardiac and neuronal Na+ channels by the selective serotonin-norepinephrine reuptake inhibitors duloxetine and venlafaxine. Eur. J. Pharmacol. 2016, 783, 1–10. [Google Scholar] [CrossRef]
  113. Hirao, R.; Fujita, T.; Sakai, A.; Kumamoto, E. Compound action potential inhibition produced by various antidepressants in the frog sciatic nerve. Eur. J. Pharmacol. 2018, 819, 122–128. [Google Scholar] [CrossRef]
  114. Zimova, L.; Ptakova, A.; Mitro, M.; Krusek, J.; Vlachovam, V. Activity dependent inhibition of TRPC1/4/5 channels by duloxetine involves voltage sensor-like domain. Biomed. Pharmacotherap. 2022, 152, 113262. [Google Scholar] [CrossRef]
  115. Krügel, U.; Straub, I.; Beckmann, H.; Schaefer, M. Primidone inhibits TRPM3 and attenuates thermal nociception in vivo. Pain 2017, 158, 856–867. [Google Scholar] [CrossRef] [PubMed]
  116. Dick, I.E.; Brochu, R.M.; Purohit, Y.; Kaczorowski, G.J.; Martin, W.J.; Priest, B.T. Sodium channel blockade may contribute to the analgesic efficacy of antidepressants. J. Pain 2007, 8, 315–324. [Google Scholar] [CrossRef] [PubMed]
  117. Abdelhamid, R.E.; Kovács, K.J.; Nunez, M.G.; Larson, A.A. Depressive behavior in the forced swim test can be induced by TRPV1 receptor activity and is dependent on NMDA receptors. Pharmacol. Res. 2014, 79, 21–27. [Google Scholar] [CrossRef] [PubMed]
  118. Pancrazio, J.J.; Kamatchi, G.L.; Roscoe, A.K.; Lynch, C., 3rd. Inhibition of neuronal Na+ channels by antidepressant drugs. J. Pharmacol. Exp. Ther. 1998, 284, 208–214. [Google Scholar]
  119. Ishii, Y.; Sumi, T. Amitriptyline inhibits striatal efflux of neurotransmitters via blockade of voltage-dependent Na+ channels. Eur. J. Pharmacol. 1992, 221, 377–380. [Google Scholar] [CrossRef]
  120. Yan, L.; Wang, Q.; Fu, Q.; Ye, Q.; Xiao, H.; Wan, Q. Amitriptyline inhibits currents and decreases the mRNA expression of voltage-gated sodium channels in cultured rat cortical neurons. Brain Res. 2010, 1336, 1–9. [Google Scholar] [CrossRef] [PubMed]
  121. Gerner, P.; Mujtaba, M.; Sinnott, C.J.; Wang, G.K. Amitriptyline versus bupivacaine in rat sciatic nerve blockade. Anesthesiology 2001, 94, 661–667. [Google Scholar] [CrossRef]
  122. Manna, S.S.S.; Umathe, S.N. A possible participation of transient receptor potential vanilloid type 1 channels in the antidepressant effect of fluoxetine. Eur. J. Pharmacol. 2012, 685, 81–90. [Google Scholar] [CrossRef] [PubMed]
  123. Lenkey, N.; Karoly, R.; Kiss, J.P.; Szasz, B.K.; Vizi, E.S.; Mike, A. The mechanism of activity-dependent sodium channel inhibition by the antidepressants fluoxetine and desipramine. Mol. Pharmacol. 2006, 70, 2052–2063. [Google Scholar] [CrossRef]
  124. Shaikh, S.; Deshpande, S.; Gohil, P. The role of transient receptor potential vanilloid 1 receptor in desipramine induced analgesic effect in diabetic mice. Int. J. Pharm. Sci. Res. 2012, 3, 4882–4887. [Google Scholar]
  125. Lee, H.M.; Kim, H.I.; Shin, Y.K.; Lee, C.S.; Park, M.; Song, J.H. Diclofenac inhibition of sodium currents in rat dorsal root ganglion neurons. Brain Res. 2003, 992, 120–127. [Google Scholar] [CrossRef] [PubMed]
  126. Suzuki, R.; Fujita, T.; Mizuta, K.; Kumamoto, E. Inhibition by non-steroidal anti-inflammatory drugs of compound action potentials in frog sciatic nerve fibers. Biomed. Pharmacother. 2018, 103, 326–335. [Google Scholar] [CrossRef] [PubMed]
  127. Hu, H.; Tian, J.; Zhu, Y.; Wang, C.; Xiao, R.; Herz, J.M.; Wood, J.D.; Zhu, M.X. Activation of TRPA1 channels by fenamate nonsteroidal anti-inflammatory drugs. Pflügers Arch. 2010, 459, 579–592. [Google Scholar] [CrossRef] [PubMed]
  128. Suzuki, H.; Sasaki, E.; Nakagawa, A.; Muraki, Y.; Hatano, N.; Muraki, K. Diclofenac, a nonsteroidal anti-inflammatory drug, is an antagonist of human TRPM3 isoforms. Pharmacol. Res. Perspect. 2016, 4, e00232. [Google Scholar] [CrossRef] [PubMed]
  129. Inoue, N.; Ito, S.; Nogawa, M.; Tajima, K.; Kyoi, T. Etodolac blocks the allyl isothiocyanate-induced response in mouse sensory neurons by selective TRPA1 activation. Pharmacology 2012, 90, 47–54. [Google Scholar] [CrossRef] [PubMed]
  130. Klose, C.; Straub, I.; Riehle, M.; Ranta, F.; Krautwurst, D.; Ullrich, S.; Meyerhof, W.; Harteneck, C. Fenamates as TRP channel blockers: Mefenamic acid selectively blocks TRPM3. Br. J. Pharmacol. 2011, 162, 1757–1769. [Google Scholar] [CrossRef] [PubMed]
  131. Sun, J.-F.; Xu, Y.-J.; Kong, X.-H.; Su, Y.; Wang, Z.-Y. Fenamates inhibit human sodium channel Nav1.7 and Nav1.8. Neurosci. Lett. 2019, 696, 67–73. [Google Scholar] [CrossRef]
  132. Yau, H.-J.; Baranauskas, G.; Martina, M. Flufenamic acid decreases neuronal excitability through modulation of voltage-gated sodium channel gating. J. Physiol. 2010, 588, 3869–3882. [Google Scholar] [CrossRef] [PubMed]
  133. des Gachons, C.P.; Uchida, K.; Bryant, B.; Shima, A.; Sperry, J.B.; Dankulich-Nagrudny, L.; Tominaga, M.; Smith III, A.B.; Beauchamp, G.K.; Breslin, P.A.S. Unusual pungency from extra-virgin olive oil is attributable to restricted spatial expression of the receptor of oleocanthal. J. Neurosci. 2011, 31, 999–1009. [Google Scholar] [CrossRef]
  134. Chokshi, R.; Bennett, O.; Zhelay, T.; Kozak, J.A. NSAIDs naproxen, ibuprofen, salicylate, and aspirin inhibit TRPM7 channels by cytosolic acidification. Front. Physiol. 2021, 12, 727549. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, Y.; Li, X. Effects of salicylate on voltage-gated sodium channels in rat inferior colliculus neurons. Hear. Res. 2004, 193, 68–74. [Google Scholar] [CrossRef] [PubMed]
  136. Huang, Q.; Chen, Y.; Gong, N.; Wang, Y.-X. Methylglyoxal mediates streptozotocin-induced diabetic neuropathic pain via activation of the peripheral TRPA1 and Nav1.8 channels. Metabolism 2016, 65, 463–474. [Google Scholar] [CrossRef] [PubMed]
  137. Bierhaus, A.; Fleming, T.; Stoyanov, S.; Leffler, A.; Babes, A.; Neacsu, C.; Sauer, S.K.; Eberhardt, M.; Schnölzer, M.; Lasitschka, F.; et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat. Med. 2012, 18, 926–933. [Google Scholar] [CrossRef] [PubMed]
  138. Ueno, T.; Yamanaka, M.; Taniguchi, W.; Nishio, N.; Matsuyama, Y.; Miyake, R.; Kaimochi, Y.; Nakatsuka, T.; Yamada, H. Methylglyoxal activates transient receptor potential A1/V1 via reactive oxygen species in the spinal dorsal horn. Mol. Pain 2024, 20, 17448069241233744. [Google Scholar] [CrossRef]
  139. Liu, L.; Zhu, W.; Zhang, Z.-S.; Yang, T.; Grant, A.; Oxford, G.; Simon, S.A. Nicotine inhibits voltage-dependent sodium channels and sensitizes vanilloid receptors. J Neurophysiol. 2003, 91, 1482–1491. [Google Scholar] [CrossRef]
  140. Chen, S.-C.; Liu, B.-C.; Chen, C.-W.; Wu, F.-S. Intradermal pregnenolone sulfate attenuates capsaicin-induced nociception in rats. Biochem. Biophys. Res. Commun. 2006, 349, 626–633. [Google Scholar] [CrossRef]
  141. Wagner, T.F.J.; Loch, S.; Lambert, S.; Straub, I.; Mannebach, S.; Mathar, I.; Düfer, M.; Lis, A.; Flockerzi, V.; Philipp, S.E.; et al. Transient receptor potential M3 channels are ionotropic steroid receptors in pancreatic β cells. Nat. Cell Biol. 2008, 10, 1421–1430. [Google Scholar] [CrossRef]
  142. Horishita, T.; Ueno, S.; Yanagihara, N.; Sudo, Y.; Uezono, Y.; Okura, D.; Sata, T. Inhibition by pregnenolone sulphate, a metabolite of the neurosteroid pregnenolone, of voltage-gated sodium channels expressed in Xenopus oocytes. J. Pharmacol. Sci. 2012, 120, 54–58. [Google Scholar] [CrossRef] [PubMed]
  143. Song, J.H.; Huang, C.S.; Nagata, K.; Yeh, J.Z.; Narahashi, T. Differential action of riluzole on tetrodotoxin-sensitive and tetrodotoxin-resistant sodium channels. J. Pharmacol. Exp. Ther. 1997, 282, 707–714. [Google Scholar] [PubMed]
  144. Richter, J.M.; Schaefer, M.; Hill, K. Riluzole activates TRPC5 channels independently of PLC activity. Br. J. Pharmacol. 2014, 171, 158–170. [Google Scholar] [CrossRef]
  145. Evans, R.M.; Scott, R.H.; Ross, R.A. Multiple actions of anandamide on neonatal rat cultured sensory neurons. Br. J. Pharmacol. 2004, 141, 1223–1233. [Google Scholar] [CrossRef] [PubMed]
  146. Okura, D.; Horishita, T.; Ueno, S.; Yanagihara, N.; Sudo, Y.; Uezono, Y.; Sata, T. The endocannabinoid anandamide inhibits voltage-gated sodium channels Nav1.2, Nav1.6, Nav1.7, and Nav1.8 in Xenopus oocytes. Anesth. Analg. 2014, 118, 554–562. [Google Scholar] [CrossRef]
  147. Kim, H.I.; Kim, T.H.; Shin, Y.K.; Lee, C.S.; Park, M.; Song, J.-H. Anandamide suppression of Na+ currents in rat dorsal root ganglion neurons. Brain Res. 2005, 1062, 39–47. [Google Scholar] [CrossRef]
  148. Jensen, T.S. Anticonvulsants in neuropathic pain: Rationale and clinical evidence. Eur. J. Pain 2002, 6 (Suppl. A), 61–68. [Google Scholar] [CrossRef]
  149. Tomić, M.; Pecikoza, U.; Micov, A.; Vučković, S.; Stepanović-Petrović, R. Antiepileptic drugs as analgesics/adjuvants in inflammatory pain: Current preclinical evidence. Pharmacol. Ther. 2018, 192, 42–64. [Google Scholar] [CrossRef]
  150. Naziroğlu, M. TRPV1 channel: A potential drug target for treating epilepsy. Curr. Neuropharmacol. 2015, 13, 239–247. [Google Scholar] [CrossRef] [PubMed]
  151. North, R.A. Opioid actions on membrane ion channels. In Handbook of Experimental Pharmacology; Herz, A., Ed.; Springer: Berlin/Heidelberg, Germany, 1993; Volume 104, pp. 773–797. [Google Scholar]
  152. Yaksh, T.L. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiol. Scand. 1997, 41, 94–111. [Google Scholar] [CrossRef] [PubMed]
  153. Schumacher, M.A.; Basbaum, A.I.; Naidu, R.K. Opioid agonists & antagonists. In Basic & Clinical Pharmacology, 14th ed.; Katzung, B.G., Ed.; McGraw-Hill, Medical Publishing Division: New York, NY, USA, 2018; Chapter 31; pp. 553–574. [Google Scholar]
  154. Stein, C.; Schäfer, M.; Machelska, H. Attacking pain at its source: New perspectives on opioids. Nat. Med. 2003, 9, 1003–1008. [Google Scholar] [CrossRef]
  155. Stein, C.; Lang, L.J. Peripheral mechanisms of opioid analgesia. Curr. Opin. Pharmacol. 2009, 9, 3–8. [Google Scholar] [CrossRef]
  156. Klotz, U. Tramadol—The impact of its pharmacokinetic and pharmacodynamic properties on the clinical management of pain. Arzneimittelforschung 2003, 53, 681–687. [Google Scholar] [CrossRef]
  157. Yamasaki, H.; Funai, Y.; Funao, T.; Mori, T.; Nishikawa, K. Effects of tramadol on substantia gelatinosa neurons in the rat spinal cord: An in vivo patch-clamp analysis. PLoS ONE 2015, 10, e0125147. [Google Scholar] [CrossRef] [PubMed]
  158. Grudt, T.J.; Williams, J.T. μ-Opioid agonists inhibit spinal trigeminal substantia gelatinosa neurons in guinea pig and rat. J. Neurosci. 1994, 14, 1646–1654. [Google Scholar] [CrossRef] [PubMed]
  159. Kohno, T.; Kumamoto, E.; Higashi, H.; Shimoji, K.; Yoshimura, M. Actions of opioids on excitatory and inhibitory transmission in substantia gelatinosa of adult rat spinal cord. J. Physiol. 1999, 518, 803–813. [Google Scholar] [CrossRef]
  160. Lintz, W.; Erlacin, S.; Frankus, E.; Uragg, H. Metabolismus von Tramadol bei Mensch und Tier. Arzneimittelforschung 1981, 31, 1932–1943. [Google Scholar] [PubMed]
  161. Pan, Y.-Z.; Li, D.-P.; Pan, H.-L. Inhibition of glutamatergic synaptic input to spinal lamina IIo neurons by presynaptic α2-adrenergic receptors. J. Neurophysiol. 2002, 87, 1938–1947. [Google Scholar] [CrossRef] [PubMed]
  162. Kawasaki, Y.; Kumamoto, E.; Furue, H.; Yoshimura, M. α2 Adrenoceptor-mediated presynaptic inhibition of primary afferent glutamatergic transmission in rat substantia gelatinosa neurons. Anesthesiology 2003, 98, 682–689. [Google Scholar] [CrossRef] [PubMed]
  163. Giovannoni, M.P.; Ghelardini, C.; Vergelli, C.; Dal Piaz, V. α2-Agonists as analgesic agents. Med. Res. Rev. 2009, 29, 339–368. [Google Scholar] [CrossRef]
  164. Kamibayashi, T.; Maze, M. Clinical uses of α2-adrenergic agonists. Anesthesiology 2000, 93, 1345–1349. [Google Scholar] [CrossRef] [PubMed]
  165. Di Cesare Mannelli, L.; Micheli, L.; Crocetti, L.; Giovannoni, M.P.; Vergelli, C.; Ghelardini, C. α2 Adrenoceptor: A target for neuropathic pain treatment. Mini Rev. Med. Chem. 2017, 17, 95–107. [Google Scholar] [CrossRef] [PubMed]
  166. Bhana, N.; Goa, K.L.; McClellan, K.J. Dexmedetomidine. Drugs 2000, 59, 263–268. [Google Scholar] [CrossRef]
  167. Gertler, R.; Brown, H.C.; Mitchell, D.H.; Silvius, E.N. Dexmedetomidine: A novel sedative-analgesic agent. Bayl. Univ. Med. Cent. Proc. 2001, 14, 13–21. [Google Scholar] [CrossRef] [PubMed]
  168. Ross, R.A. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 2003, 140, 790–801. [Google Scholar] [CrossRef] [PubMed]
  169. Spicarova, D.; Palecek, J. Anandamide-mediated modulation of nociceptive transmission at the spinal cord level. Physiol. Res. 2024, 73 (Suppl. 1), S435–S448. [Google Scholar] [CrossRef] [PubMed]
  170. Watson, C.P.N. Antidepressant drugs as adjuvant analgesics. J. Pain Symptom Manag. 1994, 9, 392–405. [Google Scholar] [CrossRef] [PubMed]
  171. Sawynok, J.; Esser, M.J.; Reid, A.R. Antidepressants as analgesics: An overview of central and peripheral mechanisms of action. J. Psychiatry Neurosci. 2001, 26, 21–29. [Google Scholar]
  172. Obata, H. Analgesic mechanisms of antidepressants for neuropathic pain. Int. J. Mol. Sci. 2017, 18, 2483. [Google Scholar] [CrossRef] [PubMed]
  173. Ferreira, S.H. Prostaglandins, aspirin-like drugs and analgesia. Nat. New Biol. 1972, 240, 200–203. [Google Scholar] [CrossRef] [PubMed]
  174. Cashman, J.N. The mechanisms of action of NSAIDs in analgesia. Drugs 1996, 52 (Suppl 5), 13–23. [Google Scholar] [CrossRef] [PubMed]
  175. Grosser, T.; Smyth, E.; FitzGerald, G.A. Anti-inflammatory, antipyretic, and analgesic agents; pharmacotherapy of gout. In Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 12th ed.; Brunton, L.L., Chabner, B.A., Knollmann, B.C., Eds.; McGraw-Hill, Medical Publishing Division: New York, NY, USA, 2011; pp. 959–1004. [Google Scholar]
  176. Gwanyanya, A.; Macianskiene, R.; Mubagwa, K. Insights into the effects of diclofenac and other non-steroidal anti-inflammatory agents on ion channels. J. Pharm. Pharmacol. 2012, 64, 1359–1375. [Google Scholar] [CrossRef] [PubMed]
  177. Hone, A.J.; McIntosh, J.M. Nicotinic acetylcholine receptors in neuropathic and inflammatory pain. FEBS Lett. 2018, 592, 1045–1062. [Google Scholar] [CrossRef]
  178. Takeda, D.; Nakatsuka, T.; Papke, R.; Gu, J.G. Modulation of inhibitory synaptic activity by a non-α4β2, non-α7 subtype of nicotinic receptors in the substantia gelatinosa of adult rat spinal cord. Pain 2003, 101, 13–23. [Google Scholar] [CrossRef]
  179. Irifune, M.; Kikuchi, N.; Saida, T.; Takarada, T.; Shimizu, Y.; Endo, C.; Morita, K.; Dohi, T.; Sato, T.; Kawahara, M. Riluzole, a glutamate release inhibitor, induces loss of righting reflex, antinociception, and immobility in response to noxious stimulation in mice. Anesth. Analg. 2007, 104, 1415–1421. [Google Scholar] [CrossRef] [PubMed]
  180. Felder, C.C.; Glass, M. Cannabinoid receptors and their endogenous agonists. Annu. Rev. Pharmacol. Toxicol. 1998, 38, 179–200. [Google Scholar] [CrossRef] [PubMed]
  181. Calignano, A.; La Rana, G.; Giuffrida, A.; Piomelli, D. Control of pain initiation by endogenous cannabinoids. Nature 1998, 394, 277–281. [Google Scholar] [CrossRef]
  182. Luo, C.; Kumamoto, E.; Furue, H.; Chen, J.; Yoshimura, M. Anandamide inhibits excitatory transmission to rat substantia gelatinosa neurones in a manner different from that of capsaicin. Neurosci. Lett. 2002, 321, 17–20. [Google Scholar] [CrossRef] [PubMed]
  183. de Cássia da Silveira e Sá, R.; Lima, T.C.; da Nóbrega, F.R.; de Brito, A.E.M.; de Sousa, D.P. Analgesic-like activity of essential oil constituents: An update. Int. J. Mol. Sci. 2017, 18, 2392. [Google Scholar] [CrossRef] [PubMed]
  184. Goyal, S.; Goyal, S.; Goins, A.E.; Alles, S.R.A. Plant-derived natural products targeting ion channels for pain. Neurobiol. Pain 2023, 13, 100128. [Google Scholar] [CrossRef] [PubMed]
  185. Caterina, M.J.; Schumacher, M.A.; Tominaga, M.; Rosen, T.A.; Levine, J.D.; Julius, D. The capsaicin receptor: A heat-activated ion channel in the pain pathway. Nature 1997, 389, 816–824. [Google Scholar] [CrossRef]
  186. Bandell, M.; Story, G.M.; Hwang, S.W.; Viswanath, V.; Eid, S.R.; Petrus, M.J.; Earley, T.J.; Patapoutian, A. Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin. Neuron 2004, 41, 849–857. [Google Scholar] [CrossRef] [PubMed]
  187. Jordt, S.E.; Bautista, D.M.; Chuang, H.H.; McKemy, D.D.; Zygmunt, P.M.; Högestätt, E.D.; Meng, I.D.; Julius, D. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 2004, 427, 260–265. [Google Scholar] [CrossRef] [PubMed]
  188. McKemy, D.D.; Neuhausser, W.M.; Julius, D. Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature 2002, 416, 52–58. [Google Scholar] [CrossRef]
  189. Peier, A.M.; Moqrich, A.; Hergarden, A.C.; Reeve, A.J.; Andersson, D.A.; Story, G.M.; Earley, T.J.; Dragoni, I.; McIntyre, P.; Bevan, S.; et al. A TRP channel that senses cold stimuli and menthol. Cell 2002, 108, 705–715. [Google Scholar] [CrossRef] [PubMed]
  190. Liu, L.; Simon, S.A. Similarities and differences in the currents activated by capsaicin, piperine, and zingerone in rat trigeminal ganglion cells. J. Neurophysiol. 1996, 76, 1858–1869. [Google Scholar] [CrossRef]
  191. Liu, L.; Lo, Y.-C.; Chen, I.-J.; Simon, S.A. The responses of rat trigeminal ganglion neurons to capsaicin and two nonpungent vanilloid receptor agonists, olvanil and glyceryl nonamide. J. Neurosci. 1997, 17, 4101–4111. [Google Scholar] [CrossRef] [PubMed]
  192. Legrand, C.; Merlini, J.M.; de Senarclens-Bezençon, C.; Michlig, S. New natural agonists of the transient receptor potential ankyrin 1 (TRPA1) channel. Sci. Rep. 2020, 10, 11238. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, L.; Oortgiesen, M.; Li, L.; Simon, S.A. Capsaicin inhibits activation of voltage-gated sodium currents in capsaicin-sensitive trigeminal ganglion neurons. J. Neurophysiol. 2001, 85, 745–758. [Google Scholar] [CrossRef] [PubMed]
  194. Cao, X.; Cao, X.; Xie, H.; Yang, R.; Lei, G.; Li, F.; Li, A.; Liu, C.; Liu, L. Effects of capsaicin on VGSCs in TRPV1−/− mice. Brain Res. 2007, 1163, 33–43. [Google Scholar] [CrossRef] [PubMed]
  195. Haeseler, G.; Maue, D.; Grosskreutz, J.; Bufler, J.; Nentwig, B.; Piepenbrock, S.; Dengler, R.; Leuwer, M. Voltage-dependent block of neuronal and skeletal muscle sodium channels by thymol and menthol. Eur. J. Anaesthesiol. 2002, 19, 571–579. [Google Scholar] [CrossRef]
  196. Ritter, A.M.V.; Ames, F.Q.; Otani, F.; de Oliveira, R.M.W.; Cuman, R.K.N.; Bersani-Amado, C.A. Effects of anethole in nociception experimental models. Evid.-Based Complement. Alternat. Med. 2014, 2014, 345829. [Google Scholar] [CrossRef]
  197. Memon, T.; Yarishkin, O.; Reilly, C.A.; Križaj, D.; Olivera, B.M.; Teichert, R.W. trans-Anethole of fennel oil is a selective and nonelectrophilic agonist of the TRPA1 ion channel. Mol. Pharmacol. 2019, 95, 433–441. [Google Scholar] [CrossRef]
  198. Moreira-Junior, L.; Leal-Cardoso, J.H.; Cassola, A.C.; Carvalho-de-Souza, J.L. State-dependent blockade of dorsal root ganglion voltage-gated Na+ channels by anethole. Int. J. Mol. Sci. 2024, 25, 1034. [Google Scholar] [CrossRef]
  199. Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of resveratrol: In vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxid. Med. Cell. Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef]
  200. Yu, L.; Wang, S.; Kogure, Y.; Yamamoto, S.; Noguchi, K.; Dai, Y. Modulation of TRP channels by resveratrol and other stilbenoids. Mol. Pain 2013, 9, 3. [Google Scholar] [CrossRef]
  201. Wang, Y.-J.; Chan, M.-H.; Chen, L.; Wu, S.-N.; Chen, H.H. Resveratrol attenuates cortical neuron activity: Roles of large conductance calcium-activated potassium channels and voltage-gated sodium channels. J. Biomed. Sci. 2016, 23, 47. [Google Scholar] [CrossRef] [PubMed]
  202. Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  203. Yeon, K.Y.; Kim, S.A.; Kim, Y.H.; Lee, M.K.; Ahn, D.K.; Kim, H.J.; Kim, J.S.; Jung, S.J.; Oh, S.B. Curcumin produces an antihyperalgesic effect via antagonism of TRPV1. J. Dent. Res. 2010, 89, 170–174. [Google Scholar] [CrossRef] [PubMed]
  204. Tomohiro, D.; Mizuta, K.; Fujita, T.; Nishikubo, Y.; Kumamoto, E. Inhibition by capsaicin and its related vanilloids of compound action potentials in frog sciatic nerves. Life Sci. 2013, 92, 368–378. [Google Scholar] [CrossRef] [PubMed]
  205. Tabeshpour, J.; Banaeeyeh, S.; Eisvand, F.; Sathyapalan, T.; Hashemzaei, M.; Sahebkar, A. Effects of curcumin on ion channels and pumps: A review. IUBMB Life 2019, 71, 812–820. [Google Scholar] [CrossRef]
  206. Kawasaki, H.; Mizuta, K.; Fujita, T.; Kumamoto, E. Inhibition by menthol and its related chemicals of compound action potentials in frog sciatic nerves. Life Sci. 2013, 92, 359–367. [Google Scholar] [CrossRef] [PubMed]
  207. Matsushita, A.; Ohtsubo, S.; Fujita, T.; Kumamoto, E. Inhibition by TRPA1 agonists of compound action potentials in the frog sciatic nerve. Biochem. Biophys. Res. Commun. 2013, 434, 179–184. [Google Scholar] [CrossRef] [PubMed]
  208. Inoue, M.; Fujita, T.; Goto, M.; Kumamoto, E. Presynaptic enhancement by eugenol of spontaneous excitatory transmission in rat spinal substantia gelatinosa neurons is mediated by transient receptor potential A1 channels. Neuroscience 2012, 210, 403–415. [Google Scholar] [CrossRef] [PubMed]
  209. Yue, H.-Y.; Jiang, C.-Y.; Fujita, T.; Kumamoto, E. Zingerone enhances glutamatergic spontaneous excitatory transmission by activating TRPA1 but not TRPV1 channels in the adult rat substantia gelatinosa. J. Neurophysiol. 2013, 110, 658–671. [Google Scholar] [CrossRef]
  210. Luo, Q.-T.; Fujita, T.; Jiang, C.-Y.; Kumamoto, E. Carvacrol presynaptically enhances spontaneous excitatory transmission and produces outward current in adult rat spinal substantia gelatinosa neurons. Brain Res. 2014, 1592, 44–54. [Google Scholar] [CrossRef] [PubMed]
  211. Kang, Q.; Jiang, C.-Y.; Fujita, T.; Kumamoto, E. Spontaneous L-glutamate release enhancement in rat substantia gelatinosa neurons by (-)-carvone and (+)-carvone which activate different types of TRP channel. Biochem. Biophys. Res. Commun. 2015, 459, 498–503. [Google Scholar] [CrossRef]
  212. Xu, Z.-H.; Wang, C.; Fujita, T.; Jiang, C.-Y.; Kumamoto, E. Action of thymol on spontaneous excitatory transmission in adult rat spinal substantia gelatinosa neurons. Neurosci. Lett. 2015, 606, 94–99. [Google Scholar] [CrossRef]
  213. Liapi, C.; Anifandis, G.; Chinou, I.; Kourounakis, A.P.; Theodosopoulos, S.; Galanopoulou, P. Antinociceptive properties of 1,8-cineole and β-pinene, from the essential oil of Eucalyptus camaldulensis leaves, in rodents. Planta Med. 2007, 73, 1247–1254. [Google Scholar] [CrossRef]
  214. Nishijima, C.M.; Ganev, E.G.; Mazzardo-Martins, L.; Martins, D.F.; Rocha, L.R.M.; Santos, A.R.S.; Hiruma-Lima, C.A. Citral: A monoterpene with prophylactic and therapeutic anti-nociceptive effects in experimental models of acute and chronic pain. Eur. J. Pharmacol. 2014, 736, 16–25. [Google Scholar] [CrossRef] [PubMed]
  215. Zhu, L.; Fujita, T.; Jiang, C.-Y.; Kumamoto, E. Enhancement by citral of glutamatergic spontaneous excitatory transmission in adult rat substantia gelatinosa neurons. NeuroReport 2016, 27, 166–171. [Google Scholar] [CrossRef] [PubMed]
  216. Ohtsubo, S.; Fujita, T.; Matsushita, A.; Kumamoto, E. Inhibition of the compound action potentials of frog sciatic nerves by aroma oil compounds having various chemical structures. Pharmacol. Res. Perspect. 2015, 3, e00127. [Google Scholar] [CrossRef] [PubMed]
  217. Gomes, P.B.; Feitosa, M.L.; Gomes Silva, M.I.; Noronha, E.C.; Moura, B.A.; Venâncio, E.T.; Rios, E.R.V.; de Sousa, D.P.; de Vasconcelos, S.M.M.; de França Fonteles, M.M.; et al. Anxiolytic-like effect of the monoterpene 1,4-cineole in mice. Pharmacol. Biochem. Behav. 2010, 96, 287–293. [Google Scholar] [CrossRef]
  218. Wang, C.; Fujita, T.; Yasuda, H.; Kumamoto, E. Spontaneous excitatory transmission enhancement produced by linalool and its isomer geraniol in rat spinal substantia gelatinosa neurons—Involvement of transient receptor potential channels. Phytomedicine Plus 2022, 2, 100155. [Google Scholar] [CrossRef]
  219. Koo, J.Y.; Jang, Y.; Cho, H.; Lee, C.-H.; Jang, K.H.; Chang, Y.H.; Shin, J.; Oh, U. Hydroxy-α-sanshool activates TRPV1 and TRPA1 in sensory neurons. Eur. J. Neurosci. 2007, 26, 1139–1147. [Google Scholar] [CrossRef]
  220. Tsunozaki, M.; Lennertz, R.C.; Katta, S.; Stucky, C.L.; Bautista, D.M. The plant-derived alkylamide, hydroxy-alpha-sanshool, induces analgesia through inhibition of voltage-gated sodium channels. Biophys. J. 2012, 102, 323a. [Google Scholar] [CrossRef]
  221. Luo, X.-J.; Peng, J.; Li, Y.-J. Recent advances in the study on capsaicinoids and capsinoids. Eur. J. Pharmacol. 2011, 650, 1–7. [Google Scholar] [CrossRef]
  222. Iida, T.; Moriyama, T.; Kobata, K.; Morita, A.; Murayama, N.; Hashizume, S.; Fushiki, T.; Yazawa, S.; Watanabe, T.; Tominaga, M. TRPV1 activation and induction of nociceptive response by a non-pungent capsaicin-like compound, capsiate. Neuropharmacology 2003, 44, 958–967. [Google Scholar] [CrossRef] [PubMed]
  223. Shintaku, K.; Uchida, K.; Suzuki, Y.; Zhou, Y.; Fushiki, T.; Watanabe, T.; Yazawa, S.; Tominaga, M. Activation of transient receptor potential A1 by a non-pungent capsaicin-like compound, capsiate. Br. J. Pharmacol. 2012, 165, 1476–1486. [Google Scholar] [CrossRef] [PubMed]
  224. Yang, L.; Fujita, T.; Jiang, C.-Y.; Piao, L.-H.; Yue, H.-Y.; Mizuta, K.; Kumamoto, E. TRPV1 agonist piperine but not olvanil enhances glutamatergic spontaneous excitatory transmission in rat spinal substantia gelatinosa neurons. Biochem. Biophys. Res. Commun. 2011, 410, 841–845. [Google Scholar] [CrossRef]
  225. Mishra, A.; Punia, J.K.; Bladen, C.; Zamponi, G.W.; Goel, R.K. Anticonvulsant mechanisms of piperine, a piperidine alkaloid. Channels 2015, 9, 317–323. [Google Scholar] [CrossRef]
  226. Gonçalves, L.M.; Valente, I.M.; Rodrigues, J.A. An overview on cardamonin. J. Med. Food 2014, 17, 633–640. [Google Scholar] [CrossRef]
  227. Sambasevam, Y.; Farouk, A.A.O.; Mohamad, T.A.S.T.; Sulaiman, M.R.; Bharatham, B.H.; Perimal, E.K. Cardamonin attenuates hyperalgesia and allodynia in a mouse model of chronic constriction injury-induced neuropathic pain: Possible involvement of the opioid system. Eur. J. Pharmacol. 2017, 796, 32–38. [Google Scholar] [CrossRef] [PubMed]
  228. Wang, S.; Zhai, C.; Zhang, Y.; Yu, Y.; Zhang, Y.; Ma, L.; Li, S.; Qiao, Y. Cardamonin, a novel antagonist of hTRPA1 cation channel, reveals therapeutic mechanism of pathological pain. Molecules 2016, 21, 1145. [Google Scholar] [CrossRef] [PubMed]
  229. Sambasevam, Y.; Jiun, W.S.; Ghazali, F.H.; Ramadan, A.I.A.; Akira, A.; Sulaiman, M.R.; Hussain, M.K.; Perimal, E.K. Inhibitory effects of cardamonin on compound action potentials in frog sciatic nerves and the possible involvement of opioidergic pathway. Life Sci. Med. Biomed. 2017, 1, 3. [Google Scholar] [CrossRef]
  230. Khalid, M.H.; Jamali, M.A.; Azmi, L.; Sulaiman, M.R.; Mohamad, T.A.S.T.; Perimal, E.K. Molecular docking and molecular dynamic simulation of cardamonin on voltage-gated sodium channel 1.7. In Proceedings of the 10th Federation of the Asian and Oceanian Physiological Societies Congress, EXCO, Daegu, Republic of Korea, 1–4 November 2023. [Google Scholar]
  231. Moreira-Lobo, D.C.A.; Linhares-Siqueira, E.D.; Cruz, G.M.P.; Cruz, J.S.; Carvalho-de-Souza, J.L.; Lahlou, S.; Coelho-de-Souza, A.N.; Barbosa, R.; Magalhães, P.J.C.; Leal-Cardoso, J.H. Eugenol modifies the excitability of rat sciatic nerve and superior cervical ganglion neurons. Neurosci. Lett. 2010, 472, 220–224. [Google Scholar] [CrossRef] [PubMed]
  232. Lima-Accioly, P.M.; Lavor-Porto, P.R.; Cavalcante, F.S.; Magalhães, P.J.C.; Lahlou, S.; Morais, S.M.; Leal-Cardoso, J.H. Essential oil of croton nepetaefolius and its main constituent, 1,8-cineole, block excitability of rat sciatic nerve in vitro. Clin. Exp. Pharmacol. Physiol. 2006, 33, 1158–1163. [Google Scholar] [CrossRef] [PubMed]
  233. Sousa, D.G.; Sousa, S.D.G.; Silva, R.E.R.; Silva-Alves, K.S.; Ferreira-da-Silva, F.W.; Kerntopf, M.R.; Menezes, I.R.A.; Leal-Cardoso, J.H.; Barbosa, R. Essential oil of Lippia alba and its main constituent citral block the excitability of rat sciatic nerves. Braz. J. Med. Biol. Res. 2015, 48, 697–702. [Google Scholar] [CrossRef] [PubMed]
  234. Joca, H.C.; Cruz-Mendes, Y.; Oliveira-Abreu, K.; Maia-Joca, R.P.M.; Barbosa, R.; Lemos, T.L.; Lacerda Beirão, P.S.; Leal-Cardoso, J.H. Carvacrol decreases neuronal excitability by inhibition of voltage-gated sodium channels. J. Nat. Prod. 2012, 75, 1511–1517. [Google Scholar] [CrossRef] [PubMed]
  235. de Sousa, D.P.; Gonçalves, J.C.R.; Quintans-Júnior, L.; Cruz, J.S.; Araújo, D.A.M.; de Almeida, R.N. Study of anticonvulsant effect of citronellol, a monoterpene alcohol, in rodents. Neurosci. Lett. 2006, 401, 231–235. [Google Scholar] [CrossRef] [PubMed]
  236. Gonçalves, J.C.R.; Alves, A.M.H.; de Araújo, A.E.V.; Cruz, J.S.; Araújo, D.A.M. Distinct effects of carvone analogues on the isolated nerve of rats. Eur. J. Pharmacol. 2010, 645, 108–112. [Google Scholar] [CrossRef] [PubMed]
  237. Boonen, B.; Alpizar, Y.A.; Benoy, V.; van Den Bosch, L.; Voets, T.; Talavera, K. The TRPA1 agonist cinnamaldehyde acts as a local anesthetic inhibiting voltage-gated sodium channels in sensory neurons. Biophys. J. 2014, 106, 326a–327a. [Google Scholar] [CrossRef]
  238. Cho, J.S.; Kim, J.-M.; Song, J.-H. Effects of eugenol on Na+ currents in rat dorsal root ganglion neurons. Brain Res. 2008, 1243, 53–62. [Google Scholar] [CrossRef]
  239. Lai, M.-C.; Wu, S.-N.; Huang, C.-W. Zingerone modulates neuronal voltage-gated Na+ and L-type Ca2+ currents. Int. J. Mol. Sci. 2022, 23, 3123. [Google Scholar] [CrossRef] [PubMed]
  240. Brosnan, R.J.; Ramos, K.; de Araujo Aguiar, A.J.; Cenani, A.; Knych, H.K. Anesthetic pharmacology of the mint extracts L-carvone and methyl salicylate. Pharmacology 2022, 107, 167–178. [Google Scholar] [CrossRef]
  241. Ferreira-da-Silva, F.W.; da Silva-Alves, K.S.; Alves-Fernandes, T.A.; Coelho-de-Souza, A.N.; Leal-Cardoso, J.H. Effects of 1,8-cineole on Na+ currents of dissociated superior cervical ganglia neurons. Neurosci. Lett. 2015, 595, 45–49. [Google Scholar] [CrossRef]
  242. Leal-Cardoso, J.H.; da Silva-Alves, K.S.; Ferreira-da-Silva, F.W.; dos Santos-Nascimento, T.; Joca, H.C.; de Macedo, F.H.P.; de Albuquerque-Neto, P.M.; Magalhães, P.J.C.; Lahlou, S.; Cruz, J.S.; et al. Linalool blocks excitability in peripheral nerves and voltage-dependent Na+ current in dissociated dorsal root ganglia neurons. Eur. J. Pharmacol. 2010, 645, 86–93. [Google Scholar] [CrossRef] [PubMed]
  243. Tsuchiya, H. Anesthetic agents of plant origin: A review of phytochemicals with anesthetic activity. Molecules 2017, 22, 1369. [Google Scholar] [CrossRef] [PubMed]
  244. Yang, B.H.; Piao, Z.G.; Kim, Y.-B.; Lee, C.-H.; Lee, J.K.; Park, K.; Kim, J.S.; Oh, S.B. Activation of vanilloid receptor 1 (VR1) by eugenol. J. Dent. Res. 2003, 82, 781–785. [Google Scholar] [CrossRef]
  245. de la Roche, J.; Eberhardt, M.J.; Klinger, A.B.; Stanslowsky, N.; Wegner, F.; Koppert, W.; Reeh, P.W.; Lampert, A.; Fischer, M.J.M.; Leffler, A. The molecular basis for species-specific activation of human TRPA1 protein by protons involves poorly conserved residues within transmembrane domains 5 and 6. J. Biol. Chem. 2013, 288, 20280–20292. [Google Scholar] [CrossRef]
  246. Vogt-Eisele, A.K.; Weber, K.; Sherkheli, M.A.; Vielhaber, G.; Panten, J.; Gisselmann, G.; Hatt, H. Monoterpenoid agonists of TRPV3. Br. J. Pharmacol. 2007, 151, 530–540. [Google Scholar] [CrossRef] [PubMed]
  247. Gonçalves, J.C.R.; Silveira, A.L.; de Souza, H.D.N.; Nery, A.A.; Prado, V.F.; Prado, M.A.M.; Ulrich, H.; Araújo, D.A.M. The monoterpene (-)-carvone: A novel agonist of TRPV1 channels. Cytom. Part A 2013, 83, 212–219. [Google Scholar] [CrossRef] [PubMed]
  248. Lee, S.P.; Buber, M.T.; Yang, Q.; Cerne, R.; Cortés, R.Y.; Sprous, D.G.; Bryant, R.W. Thymol and related alkyl phenols activate the hTRPA1 channel. Br. J. Pharmacol. 2008, 153, 1739–1749. [Google Scholar] [CrossRef]
  249. Ortar, G.; Morera, L.; Moriello, A.S.; Morera, E.; Nalli, M.; Di Marzo, V.; De Petrocellis, L. Modulation of thermo-transient receptor potential (thermo-TRP) channels by thymol-based compounds. Bioorg. Med. Chem. Lett. 2012, 22, 3535–3539. [Google Scholar] [CrossRef] [PubMed]
  250. Xu, H.; Delling, M.; Jun, J.C.; Clapham, D.E. Oregano, thyme and clove-derived flavors and skin sensitizers activate specific TRP channels. Nat. Neurosci. 2006, 9, 628–635. [Google Scholar] [CrossRef]
  251. Stotz, S.C.; Vriens, J.; Martyn, D.; Clardy, J.; Clapham, D.E. Citral sensing by transient receptor potential channels in dorsal root ganglion neurons. PLoS ONE 2008, 3, e2082. [Google Scholar] [CrossRef]
  252. Takaishi, M.; Fujita, F.; Uchida, K.; Yamamoto, S.; Sawada (Shimizu), M.; Hatai (Uotsu), C.; Shimizu, M.; Tominaga, M. 1,8-Cineole, a TRPM8 agonist, is a novel natural antagonist of human TRPA1. Mol. Pain 2012, 8, 86. [Google Scholar] [CrossRef]
  253. Behrendt, H.-J.; Germann, T.; Gillen, C.; Hatt, H.; Jostock, R. Characterization of the mouse cold-menthol receptor TRPM8 and vanilloid receptor type-1 VR1 using a fluorometric imaging plate reader (FLIPR) assay. Br. J. Pharmacol. 2004, 141, 737–745. [Google Scholar] [CrossRef]
  254. Riera, C.E.; Menozzi-Smarrito, C.; Affolter, M.; Michlig, S.; Munari, C.; Robert, F.; Vogel, H.; Simon, S.A.; le Coutre, J. Compounds from Sichuan and Melegueta peppers activate, covalently and non-covalently, TRPA1 and TRPV1 channels. Br. J. Pharmacol. 2009, 157, 1398–1409. [Google Scholar] [CrossRef] [PubMed]
  255. Lübbert, M.; Kyereme, J.; Schöbel, N.; Beltrán, L.; Wetzel, C.H.; Hatt, H. Transient receptor potential channels encode volatile chemicals sensed by rat trigeminal ganglion neurons. PLoS ONE 2013, 8, e77998. [Google Scholar] [CrossRef]
  256. Hijikata, Y. Analgesic treatment with Kampo prescription. Expert Rev. Neurother. 2006, 6, 795–802. [Google Scholar] [CrossRef]
  257. Mochiki, E.; Yanai, M.; Ohno, T.; Kuwano, H. The effect of traditional Japanese medicine (Kampo) on gastrointestinal function. Surg. Today 2010, 40, 1105–1111. [Google Scholar] [CrossRef] [PubMed]
  258. Matsushita, A.; Fujita, T.; Ohtsubo, S.; Kumamoto, E. Traditional Japanese medicines inhibit compound action potentials in the frog sciatic nerve. J. Ethnopharmacol. 2016, 178, 272–280. [Google Scholar] [CrossRef]
  259. Nishimura, M.; Taniguchi, S.; Tamaoki, S.; Fujita, T. Inhibition of compound action potentials in the frog sciatic nerve by inchinkoto, a traditional Japanese medicine used for oral mucositis. J. Oral Biosci. 2024, 66, 420–429. [Google Scholar] [CrossRef] [PubMed]
  260. Horiuchi, A.; Nakayama, Y.; Tanaka, N. Effect of traditional Japanese medicine, daikenchuto (TJ-100) in patients with chronic constipation. Gastroenterol. Res. 2010, 3, 151–155. [Google Scholar] [CrossRef] [PubMed]
  261. Veilleux, M.-P.; Moriyama, S.; Yoshioka, M.; Hinode, D.; Grenier, D. A Review of evidence for a therapeutic application of traditional Japanese Kampo medicine for oral diseases/disorders. Medicines 2018, 5, 35. [Google Scholar] [CrossRef] [PubMed]
  262. Yang, Q.; Lu, J.-T.; Zhou, A.-W.; Wang, B.; He, G.-W.; Chen, M.-Z. Antinociceptive effect of astragalosides and its mechanism of action. Acta Pharmacol. Sin. 2001, 22, 809–812. [Google Scholar]
  263. Wang, J.; Shi, L.; Wang, C.; Yao, L.-H.; Li, G.; Wang, S. Astragaloside depresses compound action potential in sciatic nerve of frogs involved in L-type Ca2+-channel dependent mechanism. Nat. Prod. Res. 2024, in press. [Google Scholar]
  264. Hiraishi, K.; Kurahara, L.-H.; Sumiyoshi, M.; Hu, Y.-P.; Koga, K.; Onitsuka, M.; Kojima, D.; Yue, L.; Takedatsu, H.; Jian, Y.-W.; et al. Daikenchuto (Da-Jian-Zhong-Tang) ameliorates intestinal fibrosis by activating myofibroblast transient receptor potential ankyrin 1 channel. World J. Gastroenterol. 2018, 24, 4036–4053. [Google Scholar] [CrossRef]
  265. Catterall, W.A. Voltage-gated sodium channels at 60: Structure, function and pathophysiology. J. Physiol. 2012, 590, 2577–2589. [Google Scholar] [CrossRef] [PubMed]
  266. Liao, M.; Cao, E.; Julius, D.; Cheng, Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature 2013, 504, 107–112. [Google Scholar] [CrossRef]
  267. Cao, E.; Liao, M.; Cheng, Y.; Julius, D. TRPV1 structures in distinct conformations reveal mechanisms of activation. Nature 2013, 504, 113–118. [Google Scholar] [CrossRef] [PubMed]
  268. Gao, Y.; Cao, E.; Julius, D.; Cheng, Y. TRPV1 structures in nanodiscs reveal mechanisms of ligand and lipid action. Nature 2016, 534, 347–351. [Google Scholar] [CrossRef]
  269. Paulsen, C.E.; Armache, J.-P.; Gao, Y.; Cheng, Y.; Julius, D. Structure of the TRPA1 ion channel suggests regulatory mechanisms. Nature 2015, 520, 511–517. [Google Scholar] [CrossRef]
  270. Yin, Y.; Wu, M.; Zubcevic, L.; Borschel, W.F.; Lander, G.C.; Lee, S.-Y. Structure of the cold- and menthol-sensing ion channel TRPM8. Science 2018, 359, 237–241. [Google Scholar] [CrossRef] [PubMed]
  271. Yin, Y.; Le, S.C.; Hsu, A.L.; Borgnia, M.J.; Yang, H.; Lee, S.-Y. Structural basis of cooling agent and lipid sensing by the cold-activated TRPM8 channel. Science 2019, 363, aav9334. [Google Scholar] [CrossRef] [PubMed]
  272. Yin, Y.; Wu, M.; Hsu, A.L.; Borschel, W.F.; Borgnia1, M.J.; Lander, G.C.; Lee, S.-Y. Visualizing structural transitions of ligand-dependent gating of the TRPM2 channel. Nat. Commun. 2019, 10, 3740. [Google Scholar] [CrossRef] [PubMed]
  273. Zhao, C.; MacKinnon, R. Structural and functional analyses of a GPCR-inhibited ion channel TRPM3. Neuron 2023, 111, 81–91. [Google Scholar] [CrossRef]
  274. Nadezhdin, K.D.; Correia, L.; Narangoda, C.; Patel, D.S.; Neuberger, A.; Gudermann, T.; Kurnikova, M.G.; Chubanov, V.; Sobolevsky, A.I. Structural mechanisms of TRPM7 activation and inhibition. Nat. Commun. 2023, 14, 2639. [Google Scholar] [CrossRef] [PubMed]
  275. Guo, W.; Tang, Q.; Wei, M.; Kang, Y.; Wu, J.-X.; Chen, L. Structural mechanism of human TRPC3 and TRPC6 channel regulation by their intracellular calcium-binding sites. Neuron 2022, 110, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  276. Won, J.; Kim, J.; Jeong, H.; Kim, J.; Feng, S.; Jeong, B.; Kwak, M.; Ko, J.; Im, W.; So, I.; et al. Molecular architecture of the Gαi-bound TRPC5 ion channel. Nat. Commun. 2023, 14, 2550. [Google Scholar] [CrossRef]
  277. Bai, Y.; Yu, X.; Chen, H.; Horne, D.; White, R.; Wu, X.; Lee, P.; Gu, Y.; Ghimire-Rijal, S.; Lin, D.C.-H.; et al. Structural basis for pharmacological modulation of the TRPC6 channel. eLife 2020, 9, e53311. [Google Scholar] [CrossRef]
  278. Jiang, D.; Zhang, J.; Xia, Z. Structural advances in voltage-gated sodium channels. Front. Pharmacol. 2022, 13, 908867. [Google Scholar] [CrossRef]
  279. Catterall, W.A. Structure and function of voltage-gated sodium channels at atomic resolution. Exp. Physiol. 2014, 99, 35–51. [Google Scholar] [CrossRef] [PubMed]
  280. Owsianik, G.; D’hoedt, D.; Voets, T.; Nilius, B. Structure-function relationship of the TRP channel superfamily. Rev. Physiol. Biochem. Pharmacol. 2006, 156, 61–90. [Google Scholar]
  281. Meng, X.-Y.; Zhang, H.-X.; Mezei, M.; Cui, M. Molecular docking: A powerful approach for structure-based drug discovery. Curr. Comput. Aided Drug Des. 2011, 7, 146–157. [Google Scholar] [CrossRef] [PubMed]
Table 2. Plant-derived compounds exhibiting both TRP and Na+ channel (or CAP) modulation.
Table 2. Plant-derived compounds exhibiting both TRP and Na+ channel (or CAP) modulation.
Plant-Derived CompoundsCentral Terminal TRP ModulationPrimary Afferent Neuron and Cloned TRP ModulationVoltage-Gated Na+ ChannelCAPReferences
CapsaicinTRPV1 ↑TRPV1 ↑[18,20,185,190,191,192,193,194,204]
AITCTRPA1 ↑TRPA1 ↑n.d.[21,22,23,24,186,187,207]
MentholTRPM8 ↑TRPM8 ↑[20,23,25,26,27,188,189,195,206]
Anetholen.d.TRPA1 ↑n.d.[197,198]
Resveratroln.d.TRPA1 ↓n.d.[200,201]
Curcuminn.d.TRPV1 ↓n.d.[203,204]
CinnamaldehydeTRPA1 ↑TRPA1 ↑[21,23,24,186,207,237]
EugenolTRPA1 ↑TRPV1 ↑[204,208,231,238,244]
ZingeroneTRPA1 ↑TRPV1 ↑[190,204,209,239]
CarvacrolTRPA1 ↑TRPA1 ↑
TRPV3 ↑
[206,210,234,245,246]
(+)-CarvoneTRPA1 ↑n.d.n.d.[206,211,236]
ThymolTRPA1 ↑TRPA1 ↑
TRPM8 ↑
TRPV3 ↑
[195,206,212,248,249,250]
1,8-CineoleTRPA1 ↑TRPM8 ↑
TRPA1 ↓
[27,206,232,241,252]
CitralTRPA1 ↑TRPV1 ↑
TRPV3 ↑
TRPM8 ↑
TRPA1 ↑
n.d.[215,216,233,251]
(-)-CarvoneTRPV1 ↑TRPV1 ↑
TRPV3 ↑
[206,211,236,240,246,247]
1,4-CineoleTRPV1 ↑TRPM8 ↑
TRPA1 ↑
n.d.[27,206,252]
(±)-LinaloolTRPV1 ↑
TRPA1 ↑
TRPM8 ↑
TRPA1 ↑
[216,218,242,253,254]
GeraniolTRPM8 ↑TRPM8 ↑
TRPV1 ↑
TRPA1 ↑
n.d.[216,218,253,255]
Hydroxy-α-sanshooln.d.TRPV1 ↑
TRPA1 ↑
[207,219,220]
Capsiaten.d.TRPV1 ↑
TRPA1 ↑
n.d.[204,222,223]
PiperineTRPV1 ↑TRPV1 ↑[190,207,224,225]
Cardamoninn.d.TRPA1 ↓n.d.[228,229,230]
n.d.: not determined; ↑: activation; ↓: inhibition.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kumamoto, E. Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na+ and TRP Channels. Biomolecules 2024, 14, 1619. https://doi.org/10.3390/biom14121619

AMA Style

Kumamoto E. Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na+ and TRP Channels. Biomolecules. 2024; 14(12):1619. https://doi.org/10.3390/biom14121619

Chicago/Turabian Style

Kumamoto, Eiichi. 2024. "Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na+ and TRP Channels" Biomolecules 14, no. 12: 1619. https://doi.org/10.3390/biom14121619

APA Style

Kumamoto, E. (2024). Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na+ and TRP Channels. Biomolecules, 14(12), 1619. https://doi.org/10.3390/biom14121619

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop