Научная статья на тему 'TRANSIENT RECEPTOR POTENTIAL ANKYRIN 1: STRUCTURE, FUNCTION AND LIGANDS'

TRANSIENT RECEPTOR POTENTIAL ANKYRIN 1: STRUCTURE, FUNCTION AND LIGANDS Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
62
16
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
TRPA1 / TRANSIENT RECEPTOR POTENTIAL ANKYRIN / STRUCTURE / ACTIVATION OF TRPA1

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Pyatigorskaya N.V., Filippova O.V., Nikolenko N.S., Kravchenko A.D.

Introduction: Transient receptor potential ankyrin 1 (TRPA1) is a protein expressed in many living organisms. During the study of TRPA1 its unique biological role as a universal and polymodal sensor of various altering agents was found. The aim of this study is to search and generalize information about structural features and molecular determinants, mechanisms of activation, action and modulation of TRPA1 as a universal pain and inflammation sensor, as well as the nature of activators and antagonists of this target and their therapeutic potential. Materials and methods: This article presents an overview of the results of scientific research of TRPA1, its modulators, as well as an overview of their pharmacological potential over the period from the discovery of these channels to the present, with an emphasis on the last decade. Results and discussion: The main collected data on expression, structural features and molecular determinants, mechanisms of activation and action of TRPA1 indicate its role as a universal and labile element ofthe primary response ofthe body to adverse exogenous and endogenous factors. Regardless of the nature of the stimulus, hyperstimulation of TRPA1 channels can lead to such phenomena as pain, inflammation, itching, edema and other manifestations of alteration, and therefore TRPA1 blockade can be used in the treatment of various diseases accompanied by these pathological conditions. Currently, TRPA1 antagonists are being actively searched for and studied, as evidenced by a high patent activity over the past 14 years; however, the molecular mechanisms of action and pharmacological properties of TRPA1 blockers remain understudied. Conclusion: Acquire of new information about TRPA1 will help in the development of its modulators, which can become promising analgesics, anti-inflammatory drugs, bronchodilators, and agents for the treatment of cardiovascular diseases of new generations.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «TRANSIENT RECEPTOR POTENTIAL ANKYRIN 1: STRUCTURE, FUNCTION AND LIGANDS»

# Research Results in Pharmacology

Research Results in Pharmacology 8(3): 19-29 UDC: 577.25 + 615.31 DOI 10.3897/rrpharmacology. 8.90214

3 Review Article

Transient receptor potential Ankyrin 1: structure, function and ligands

Natalia V. Pyatigorskaya1, Olga V. Filippova1, Natalia S. Nikolenko1, Aleksey D. Kravchenko1

1 I.M. Sechenov First Moscow State Medical University under the Ministry of Health of the Russian Federation (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia

Corresponding author: Aleksey D. Kravchenko (aleksej_kravchenko97@mail.ru)

Academic editor: Mikhail Korokin ♦ Received 27 March 2022 ♦ Accepted 11 July 2022 ♦ Published 19 July 2022

Citation: Pyatigorskaya NV, Filippova OV, Nikolenko NS, Kravchenko AD (2022) Transient receptor potential Ankyrin 1: structure, function and ligands. Research Results in Pharmacology 8(3): 19-29. https://doi.org/10.3897/rrpharmacology.8.90214

Abstract

Introduction: Transient receptor potential ankyrin 1 (TRPA1) is a protein expressed in many living organisms. During the study of TRPA1 its unique biological role as a universal and polymodal sensor of various altering agents was found. The aim of this study is to search and generalize information about structural features and molecular determinants, mechanisms of activation, action and modulation of TRPA1 as a universal pain and inflammation sensor, as well as the nature of activators and antagonists of this target and their therapeutic potential.

Materials and methods: This article presents an overview of the results of scientific research of TRPA1, its modulators, as well as an overview of their pharmacological potential over the period from the discovery of these channels to the present, with an emphasis on the last decade.

Results and discussion: The main collected data on expression, structural features and molecular determinants, mechanisms of activation and action of TRPA1 indicate its role as a universal and labile element ofthe primary response ofthe body to adverse exogenous and endogenous factors. Regardless of the nature of the stimulus, hyperstimulation of TRPA1 channels can lead to such phenomena as pain, inflammation, itching, edema and other manifestations of alteration, and therefore TRPA1 blockade can be used in the treatment of various diseases accompanied by these pathological conditions. Currently, TRPA1 antagonists are being actively searched for and studied, as evidenced by a high patent activity over the past 14 years; however, the molecular mechanisms of action and pharmacological properties of TRPA1 blockers remain understudied.

Conclusion: Acquire of new information about TRPA1 will help in the development of its modulators, which can become promising analgesics, anti-inflammatory drugs, bronchodilators, and agents for the treatment of cardiovascular diseases of new generations.

Keywords

TRPAj, transient receptor potential ankyrin, structure, activation of TRPAr

Introduction

Transient receptor potential ankyrin 1 (TRPA^ belongs to an extensive and diverse group of relatively non-selective cation channels permeable to such ions as potassium, sodium,

magnesium, and calcium and called transient receptor potential (TRP) channels (Samanta et al. 2018). TRP channels got their name from the trp gene of drosophila, first described by Cosens and Manning (1969). Deletion of this gene caused a transient abnormal response of photoreceptors to

Copyright Pyatigorskaya NV et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

light on an electroretinogram, when mutant flies, exposed to permanent exposure of bright light, stopped making a pho-totactic choice in a T-shaped maze, and restored their light sensitivity only some time later in dim ambient light (Co-sens and Manning 1969). The slow recovery of the mutant eye and the temporary blindness of the mutants led to the name of trp as a protein gene responsible for the "temporary (transient) receptor potential". The TRP family of channels is currently represented by eleven subfamilies, united on the basis of sequence homology into two groups, and each subfamily includes several members (Himmel and Cox 2020). The ankyrin subfamily, which currently includes 7 members, is widely represented in the organic world. At the same time, proteins of this subfamily can be both specific to a certain group of organisms, for example, HsTRPA, expressed only in hymenoptera, and widely represented in various taxa, for example, TRPA1 found in vertebrates, arthropods, mollusks, nematodes, and other organisms (Peng et al. 2015). In some cases, the corresponding proteins of various species are somewhat different in the quantitative and qualitative composition of amino acids (for example, rat TRPA1 contains 1125, and mouse - 1115 amino acid residues), which causes the species-specific action of some biologically active compounds (thioamines, caffeine) (Chen et al. 2008; Talavera et al. 2020). As a result of studying the activity of TRPAp it became possible to determine its role as a universal and polymodal sensor of various adverse factors: physical (high and low temperatures, mechanical stimuli, radiation), chemical (irritants and toxins), and biological (metabolites, toxic bacterial products) (Viana 2016). The presented review is aimed at highlighting the basic data on the structure, activation mechanisms and molecular determinants associated with the human TRPAj and their relationship with the therapeutic potential of TRPA1 blockers.

Materials and methods

The article presents an overview of the literature data and the results of research studies on the structure, function of the channel and their connection with the performed physiological functions, as well as the structure of activators and antagonists of TRPAj obtained from available and open sources for the period from 1998 to the present, with an emphasis on the last 10 years. Information was collected in such electronic resources as: PubMed, PubChem, Elibrary, Scopus, Web of Science, WIPO for the main search queries: "TRPAj", "TRPAj structure", "TRPAj functions", "activation of TRPAj", and "TRPAj antagonists".

Results and discussion

TRPA1 gene and expression

In humans, the TRPA1 gene is located in chromosome 8, band 8q21.11, and consists of 73635 bases and 29 exons, and an open mRNA reading frame of the gene encodes

a polypeptide consisting of 1119 amino acids with a molecular weight of 127.5 kDa (Talavera et al. 2020). The expression of the gene varies depending on the age of the organism and the type of cells. Although TRPAj was initially isolated from SV40 transformed/immortalized human fibroblasts, it is mainly expressed in C- or A-fibers of small-diameter sensory ganglia, including posterior horns of a spinal cord, trigeminal and nodular ganglia (Beskh-melnitsyna et al. 2016; Meents et al. 2019). TRPAj channel co-expresses with such nociceptive markers as tachykin-ins, substance P, neurokinin A and calcitonin gene-related peptide, for which their participation in neurogenic inflammatory reactions was determined (Choi and Nardo 2018). In the central nervous system, TRPA1 is expressed in hip-pocampal neurons, where it is apparently associated with the cannabinoid receptor CB1 in astrocytes, where it is involved in maintaining intracellular Ca2+ levels and regulation of inhibitory synapses (Araujo et al. 2017; Jiang et al. 2019). TRPAj expression was also shown in Schwann cells, colon cells and enterochromaffin cells, in all respiratory tract, keratinocytes, melanocytes, vascular endothelial cells, tooth pulp fibroblasts, and other cells (Souza et al. 2020). Thus, TRPA1 channels are mainly located either in nociceptors or in tissues exposed to the external environment or exogenous factors. This is mainly due to the fact that TRPAj acts as a universal sensor to diverse stimuli that adversely affect the vital activity of the organism.

Structure and molecular determinants of TRPA1

Like most other representatives of transient receptor potential channels, TRPAj has four levels of protein organization. The quaternary structure of ankyrin TRP channels is a homo- or heterotetramer formed by the integration of subunits consisting of 6 transmembrane domains (S1 - S6). According to Oakes and Domene (2019), su-bunits may belong to one or more subfamilies of TRP channels, for example, Cheng (2010) shows the functionally active integration of four TRPA1 subunits or two TRPAj and two TRPVj (TRPV is a vaniloid TRP channel, which is a structurally similar cation channel characterized by the presence of 6 ankyrin repeats, and for which the participation in thermo- and nociception was found). In the center of the quaternary protein structure, there is a negatively charged pore (P) capable of passing cations through, usually Ca2+ and Mg2+ (Fig. 1).

Each TRPA1 subunit, which is a tertiary organization of the protein structure, consists of 6 transmembrane domains containing a-helices. The fifth and sixth a-helices form reentrant pore loop consisting of the pore helix 1, 2 and intracellular N- and C-terminals, which are totally 78% of the polypeptide weight. At the same time, only 14% is the C-terminal, and the remaining 64% of the protein mass is the N-terminal containing 16 ankyrin repeats, the presence of significant number of which caused this protein to be named 'ankyrin-like with transmembrane domains protein 1: (ANKTM1) and to be renamed as the "ankyrin channel transient receptor potential 1" after it was found to be

Figure 1. Scheme of the quaternary structure of homotetrameric (left) and heterotetrameric (right) TRPA1 channel (top view). Note: P is the pore of the channel, S1-S6 are transmembrane domains.

related to the TRP channel family (Suo et al. 2020). It is noteworthy that later a new subfamily of the TRP channels called "non-mechanoreceptof' (TRPN) was discovered, the genes of which are widely expressed in animals, but are a pseudogene in amniotes, in particular, in humans, and contain about 28-29 ankyrin repeats in their structure (Schüler et al. 2015). Ankyrin (from Greek. äyKupa - anchor) repeats are an example of a protein motif consisting of 33 amino acid residues, forming a-helices connected by loops, which supposedly participate in protein-protein interactions and/or provide protein embedding in the phospholipid bilayer (Li et al. 2006).

The most detailed data on the TRPAj structure was obtained due to the electron cryomicroscopy, and mutational studies and studies of TRPAj chimeras made it possible to isolate clinically and pharmacologically significant elements of the protein. So, in the article of Paulsen et al. (2015) presented the results of a study of the structural features of a full-length human TRPAp using electron cryomicroscopy of single particles with a resolution of 4 Ä in the presence of various ligands. With the greatest accuracy, a three-dimensional structure was obtained for the Lys446-Thr1078 protein chain segment, without a detailed understanding of the structure of distal cytoplasmic fragments - up to 12 ankyrin repeats (AR1-12) from the N-terminal and after the coiled-coil domain of the C-ter-minal, as well as the region containing Cys665, linkers S1 - S2, S2 - S3, S3 - S4 and the connection between the third ß-strand, which is part of the ß-sheet and the coiled-coil domain (dotted line in Fig. 2).

Structural modeling of the ankyrin repeats showed that a quaternary protein structure is formed due to the interaction of the coiled-coil domain of the C-terminals of four subunits which placed in the center of the channel, below the ion-permeable pore, while the distal N-terminals are located outside, thereby participating in the formation of a structure similar to an anchor displayed in electronic photos in the form of a crescent. Myo-inositol hexaphosphate molecules also participate in maintaining the quaternary structure of the protein, acting as a stabilizing cofactor between the coiled-coil

domains of the C-terminals, due to the formation of ionic bonds with Leu1046 and Lys1050 of one and Leu1048 and Leu1052 of the other subunits (Paulsen et al. 2015).

Ankyrin repeats include several sites responsible for the specific sensitivity of the channel. So according to Za-yats et al. (2013), the EF-hand domain (Gly479 - Leu486 as part of 12th ankyrin repeat) is one of the sites responsible for the sensitivity of the channel to intracellular calcium concentration (according to Christensen et al. (2016), others include, for example, Asp915), Ser250, deletion of which reverses the thermal sensitivity of the channel from cold to heat, and to Glu179, involved in temperature perception and identified in the study of chimeric TRPAj of humans and rattlesnakes (Naziroglu and Braidy 2017). TRPA1 sensitivity sites to the oxygen level in the cell are also located at the N-terminal: with normal oxygen saturation, Pro394 is in a hydroxylated state, inhibiting the opening of the channel, which prolinghydroxylase is responsible for. But with hypoxia, the activity of pro-linghydroxylase decreases, which leads to an increase in the number of non-hydroxylated Pro394 TRPAj channels with increased activity (Miyake et al. 2016). However, hyperoxia leads to reversible oxidation of Cys633 and Cys856, changing the reactivity of the channel (Kannler et al. 2018). The involvement of Cys540 in the sensitivity of the channel to the concentration of active forms of nitrogen in the cell was also shown (Takahashi and Mori 2011).

A pre-S1 region accessible to water molecules is behind the ankyrin repeats and consists of a linker domain and a pre-S1 helix, which includes 4 amino acid residues responsible for the sensitivity of TRPAj to electrophilic irritants. These 4 reactive pendant radicals of the amino acids - Cys621, Cys641, Cys665 and Lys710 - form covalent links with various electrophilic agents, acting as toxins, irritants and signaling molecules of inflammation and pain, changing the conformational structure of the protein (Zhao et al. 2020). The TRP-like domain spatially links up with the pre-S1 region and follows the S6 segment. It is responsible for the "transmission" of the conformational signal from the covalently modified amino acid residues

Figure 2. The three-dimensional structure of TRPA1, a. A linear domain structure; b. A three-dimensional structure, a three-side view. Note: S1 - S6 - transmembrane segments, TRP - transient receptor potential, AR-12 - AR-16 - ankyrin repeats).

to the channel gates, which leads to their opening and intracellular cation influx (Paulsen et al. 2015).

For the intracellular C-terminus, sensitivity to calcium concentration by calmodulin has also been shown. The Leu992 - Asn1008 site binds to calcium-ion-activated calmodulin, resulting in sensitization and subsequent slow desensitization of the receptor (Zimova et al. 2020). It is noteworthy that heavy metal ions (Zn2+, Cu2+) also cause potentiation of TRPA1, but by interaction with His983 and Cys1021 of the C-terminal (Miura et al. 2013).

As noted earlier, TRPA1 is based on the four transmembrane subunits, each of which contains 6 transmembrane segments with the fifth and sixth domains connected by the reentrant pore loop consisting of the pore helix 1, 2. The second pore helix contains anionic amino acid residues (Glu920, Glu924, and Glu930) attracting cations and repulsing anions. The channel pore includes two constrictions called channel gates, the upper of which, with a diameter of 7-8 A, is formed by the residues Asp915 of four subunits, and the lower - by the hydrophobic residues Ile957 and Val961, forming a ring, presumably with a diameter of 6 A (Paulsen et al. 2015). Thus, the channel gate is wide enough to pass relatively large ions such as barium, calcium and magnesium. At the same time, activation of the channel by agonists causes reversible dilation of the channel pore, which leads to the transmission of even such large cations as the Yo-Pro fluorescent dye and meglumin (Chen et al. 2009). And binding to a positively charged non-selective ion channel blocker, ruthenium red,

which forms stable ion bonds in the channel pore and prevents the cations flow, was shown for the anionic upper channel gates (Asp915) and Glu920 of the second pore helix (Wu et al. 2017).

For the selective antagonist A-967079, interaction with Tyr874 of the S5 transmembrane segment and Phe909, which is part of the pore helix 1, as well as presumably with four more surrounding amino acid residues (Ser873, Leu881, Phe944, and Val948), was revealed, which prevents the opening of the channel gate (Paulsen et al. 2015). It is noteworthy that the supposed binding site of such general anesthetics as isoflurane and propofol overlaps with the binding site of antagonist A-967079 (Ton et al. 2020). And mutations in the key amino acids of the binding site of A-967079 do not influence the activity of another selective antagonist, HC-030031, which indicates the presence of other TRPA1 blocker binding sites, but their search by cryo-electron microscopy was unproductive (Paulsen et al. 2015). Further study of the molecular determinants of TRPA1 antagonists by chimeric studies and point mutations helped to isolate Asn855, which is part of the loop between the 4th and 5th transmembrane segments, as well as the C-terminal, significantly contributing to the inhibitory effect of HC-030031 (Gupta et al. 2016).

Other binding sites with biologically active compounds have been detected for the transmembrane backbone. It has been shown that TRPA1 is activated by binding essential oil compounds - menthol, thymol and carvacrol - with Ser873 and Thr874 in the S5 transmembrane segment

(Meents et al. 2016). And the amino acid residues Ser943 and Ile946, which are part of the S6 transmembrane domain, form bonds with thioamines that are antagonistic to human TRPAj, but cause activation of rat TRPAj when interacting with Ala946 and Met949 (Chen et al. 2008). The transmembrane part also includes five residues of cysteine and lysine, three of which (Cys727, Lys771, and Cys834) are in a lipophilic medium and, therefore, can be modified by hydrophobic electrophiles, including secondary messengers of inflammation and pain (Paulsen et al. 2015).

Thus, due to the presence of both selective (binding site of calmodulin, heavy metals) and non-selective sensory molecular determinants (reactive cysteine and lysine ami-no acid residues) described above, the ankyrin TRP channel serves a universal and labile sensor of diverse stimuli, whereas the molecular mechanisms of action and binding sites of selective TRPAj antagonists require further study.

Activation and post-activation effects of TRPAj

TRPAj can be activated by the various stimuli: physical, chemical and biological. Story et al. (2003) in their initial studies talked about the sensitivity of TRPAj to low temperatures (below 17 °C), but not in every instance it was possible to reproduce the activation of the channel by cold, so today the cold sensitivity of TRPAj is relatively controversial (Weyer-Menkhoff and Lotsch 2019; Weyer-Menk-hoff et al. 2019). It has also been shown that TRPAj has mechanosensitivity, manifested only in the oxidized form of protein, which may be a cause of mechanical allodynia in the inflammatory process, when reactive oxygen species accumulate (Moparthi and Zygmunt 2020). In human melanocytes, TRPA1 is indirectly activated by ultraviolet radiation, which leads to an increase in melanin synthesis and skin pigmentation (Bellono et al. 2013).

Moreover, TRPA1 is one of the main chemosensors in living organisms. Compounds agonists of TRPA1 are extremely diverse in nature. They can be divided into selective synthetic compounds discovered and used when studying the nociceptive properties of TRPA1 (for example, compound PF-4840154, described by Ryckmans et al. (2011), which is a derivative of piperidine-5-carboxa-mide, EC50=23 nM), nonelectrophilic compounds (menthol, thymol, carvacrol, nicotine, clotrimazole, nifedipine, diclofenac, etc.) and the most extensive group of elec-trophilic compounds forming covalent bonds with TRPA1 cysteine residues. Among electrophilic agonists, hydrop-hobic compounds can also be distinguished (4-oxo-2-no-nenal is a product of lipid peroxidation; 4,5-epoxyeico-satrienoic acid is an endogenous signaling molecule) and relatively hydrophilic irritants of natural origin, for example, allicin contained in garlic, allyl isothiocyana-te formed in mustard and horseradish; cinnamaldehyde, etc., as well as anthropogenic: formaldehyde, acrolein, crotonaldehyde, hydrogen peroxide, nitrogen oxides, heavy metals and endogenous compounds: methylglyoxal, reactive oxygen and nitrogen species (Cheng et al. 2010; Mukaiyama et al. 2020).

TRPA1 can also be activated by transmitting an in-tracellular signal from other receptors, including G-pro-tein-coupled receptors (GPCR) (Fig. 3).

Thus, it was found that bradykinin, a nonapeptide with a known algogenic effect, via the bradykinin B2 receptor activates or sensitizes TRPAj by phosphorylation of TRPAj by protein kinase A (PKA) or via the phospho-lipase pathway (PLC - phospholipase C, PIP2 - phosp-hatidylinositol diphosphate) via diacylglycerol (DAG) (Al-Shamlan and El- Hashim 2019). The phospholipase pathway of TRPA1 activation is also involved in the transmission of a signal from the protease-activated re-ceptor-2 (PAR2), interacting with trypsin and tryptase of mast cells, the synthesis of which increases in allergic reactions (Chen et al. 2016). The intracellular signal of prostaglandin E2 (PGE2), a mediator of inflammation, pain and edema interacting with prostaglandin receptors (EP), is also transmitted through the adenylate cyclase system (AC), which leads to the accumulation of cAMP and activation of PKA (Dall'Acqua et al. 2014). Sensi-tization of the TRPA1 channel can also occur through its phosphorylation by protein kinase C (PKC) activated by the bile acid receptor (TGR5) during its interaction with bile acids, which have shown a pruritogenic (itching) effect (Lieu et al. 2014). Itching can also be caused by an antimalarial drug chloroquine, and a peptide of the adrenal medulla of cattle (BAM8-22), which interact with MAS (derived from the surname Massey)-related GPCRs (MrgprA3 and MrgprC11), leading to the dissociation of G-protein, the disconnected p-subunit of which directly activates TRPAj (Wilson et al. 2011). Regulation of the ankyrin TRP is also carried out at the level of its gene. Thus, nerve growth factor (NGF), for which anti-inflammatory effects have been identified, suppresses a transcription of the TRPAj gene via phosphorylated mitogen-activated protein kinase p38 (MAPK) activated by the tyrosine kinase receptor A (TrkA) (Diogenes et al. 2007).

It is also known that TRPA1 can be activated by lipopo-lysaccharide (LPS), a component of the outer membrane of all gram-negative bacteria formed during their death, but the molecular mechanisms of this interaction are uncertain. It is assumed that LPS, embedded in the phospholipid bilayer, changes the tension of the membrane, which leads to the opening of the channel (Startek et al. 2018). Thus, TRPAj may be involved in the body's immune response to the bacterial pathogenic factors.

Summarizing the above, TRPA1 can be activated not only by the direct action of adverse stimuli of physical, chemical and biological nature, but also be regulated indirectly through more specific receptors of pro-inflammatory, algogenic and pruritogenic factors, thereby confirming its role as one of the main integral sensors of pain and inflammation.

Regardless of the factor and the mechanism of TRPAj activation, it leads to an intracellular influx of calcium, which in nociceptor terminals causes membrane depolarization and propagation of the action potential into

Figure 3. Intracellular regulation of TRPA1 activity. Note: NGF - nerve growth factor, TrkA - tyrosine kinase receptor A, p 38 MAPK - mitogen-activated protein kinase p38, PGE2 - prostaglandin E2, EP - prostaglandin receptor, Gs(q) - G-proteins, AC - adenylate cyclase, cAMP - cyclic adenosine monophosphate, PKA - protein kinase A, P - phosphate residue, BAM 8-22 - a peptide of the adrenal medulla of cattle, PAR2 - protease-activated receptor-2, B2 - a bradykinin receptor, PLC - phospholipase C, PIP2 - phosphatidylinositol diphosphate, DAG - diacylglycerol, TGR5 - a G protein-coupled bile acid receptor, PKC - protein kinase C.

the central nervous system, causing a sensation of pain. Pain can also be caused by dilation of cerebral vessels when calcium ions entering through the TRPA1 activate potassium channels. In turn, pain is aimed at avoiding the unfavorable factor that caused it. In barrier cells, intracellular calcium influx releases vasodilators, pro-inflammatory agents and algogens (for example, substance P and a peptide associated with the calcitonin gene), causing local vasodilation, edema, lymphocyte influx, aimed at preventing the spread of harmful agents throughout the body and activation of the body's defense mechanisms (Gouin et al. 2017). Thus, TRPA1 is the primary link in the signaling mechanism and protection of the body from adverse factors of the internal and external environment.

TRPA1 antagonists

Hyperstimulation of TRPA1, caused by a super-strong or a long-acting irritant, can lead to physiological and psychological disorders, manifesting as pain and inflammatory process. Today, there has been an active search for and

study of TRPA1 activity regulators that could be useful in the treatment of various diseases accompanied by pain and inflammation or caused by an inflammatory process, which is evidenced by a high patent activity (with over 30 patents since 2007) in this area. Analysis of available data on the TRPA1 antagonists has shown that they can be divided into two large groups: natural antagonists showing weak activity and selectivity, for example: terpenoids and phenols described in Hoag and Salerno patent (2020) and synthetic TRPA1 blockers, most diverse in their structure.

Historically, the first patented TRPA1 antagonists were commercially available Cambridge-5861528 and HC030031, which are representatives of an extensive group of fused pyrimidinediones, and AP 18, a representative of the oxime group (Patapoutian and Jegla 2007). These compounds are characterized by inhibitory concentrations in the micromolar range and low solubility, so at the moment they are used only in research. Further search for TRPA1 blockers was aimed at improving the structure of fused pyrimidinediones and the discovery of new classes of compounds. Thus, replacement of a fused

pyrrole ring with imidazole (WO 2009144548), thiophe-ne (WO 2010109334), isothiazole (WO 2010109328), furan and isoxazole (WO 2011114184) rings and a combination of various pendent groups reduced the inhibitory concentration to the nanomolar range, and appending a hydrophilic substituent in pendent groups with the formation of arylacetylimines (CN 103261201) increased the solubility of the derivatives (Chaudhari et al. 2009; Kumar et al. 2010a, b; Chaudhari et al. 2011; Sukers et al. 2015). In addition to arylacetylimines, pht-halimides (WO 2009118596) and quinazolinediones (US 20090325987), which together form a subgroup of arylacetamides (Muthuppalniappan 2009a, b), can be referred to compounds similar to pyrimidinedione-fu-sed ones, with TRPAj antagonistic activity. This subgroup, together with N-arylsulfonylacetamides, divided into cyclic (for example, WO 2016128529) and acyclic (for example, WO 2014135617), is part of an extensive group - acetamides (Fruttarolo et al. 2014; Estrada et al. 2016). Other groups of TRPAj blockers include 4-fluo-rine (WO 2014053694), carbonic acid derivatives (WO 2014056958), imidazopyridines (WO 2014076021), phenyl pyrazinopyrimidine diones (RU 2014147911), bi-cyclic oxadiazoles (WO 2018162607) and tetrahydrofu-ranil oxadiazoles (WO 2019182925), 3-aryloxy-3-aryl-propylamines (WO 2020035040) (Arvela et al. 2014; Bachmann et al. 2014; Brotherton-Pleiss et al. 2014; Kochkarov et al. 2016; Chen et al. 2019; Terrett et al. 2019; Wang et al. 2020).

Despite all the structural diversity of compounds with TRPA1 antagonistic activity in vitro, only a few compounds have been subjected to preclinical tests. This is due not only to the low solubility of most compounds, but also to the heterogeneity of human and rodent TRPA1 channels. For example, according to Chen et al. (2008) representatives of the AMG group (AMG 2504, AMG5445, AMG9090, etc.) exhibit a partial agonistic activity or do not interact with TRPA1 of rats.

The possibility of treatment of acute pain with TRPAj antagonists was shown in preclinical studies. In the study of Beskhmelnitsyna et al. (2018; 2019a; 2019b), the compound ZC02-0012 showed analgesic activity superior to that of ketorolac according to hot plate and acetic acid-induced writhing tests, as well as anti-inflammatory activity, comparable to that of diclofenac sodium. Intra-plantar injection of AR18 reduced mechanical hyperalgesia in wild-type mice and had no effect in mice with TRPAj genetic knockout (Petrus et al. 2007). In Wei et al study (2011), intrathecal administration of A-967079 or Chembridge-5861528 reduced secondary mechanosen-sitivity after injection of capsacin or formalin. In a number of works by David et al. (2008); Wei et al. (2009); Chen et al. (2011), it has been shown that the systematic administration of HC-030031 and Chembridge-5861528 reduces the response to mechanical stimuli in rats with neuropathy and inflammation. In 2018, Galderma SA patented a number of TRPA1 antagonists for the treatment of atopic dermatitis (Ouvry et al. 2018). In van

den Berg et al. study (2021), compound BI01305834 reduced bronchospasm in guinea pigs in response to the allergens. The antitussive effect of GRC 17536 was demonstrated in citric-acid-induced cough responses in guinea pigs (Mukhopadhyay et al. 2014). Li et al. (2019) showed the possibility of using HC-030031 in the treatment of cardiac dysfunction caused by myocardial infarction in mice. The selective TRPA1 blocker reduced cardiac fibrosis and apoptosis of cells after myocardial infarction and significantly increased angiogenesis in the border zone of infarction. Intraperitoneal or intrathecal administration of Chembridge-5861528 suppressed neuropathy caused by diabetes in the absence of noticeable side effects during single or long-term administration (Wei et al. 2010).

According to Chen and Terrett (2020), today only 5 compounds (CB-625, GRC 17536, 0DM-108, HX-100, and GDC-0334) reached the stage of clinical trials; however, as far as it is known from open sources, the development of all 5 molecules was discontinued mainly due to unsatisfactory pharmacokinetic parameters, and therefore the search for and study of new selective TRPA1 antagonists, which would be promising for further development of new analgesics, anti-inflammatory drugs, bronchodila-tors, and agents for the treatment of cardiovascular diseases, is an urgent unsolved problem.

Conclusion

Human TRPA1 is a recently discovered nonselective cation channel expressed mainly in nociceptor neurons and barrier cells. This channel has four levels of protein structure organization and stands out against the background of other related transmembrane proteins by an impressive intracellular N-terminal containing 16 ankyrin repeats. Accumulated data on the TRPA1 structure and activity, obtained by electron cryomicroscopy in mutational and chimeric studies, allowed us to select a number of molecular determinants and mechanisms indicating its role as a universal sensor of adverse factors of various (physical, chemical and biological) nature, which are converted into nerve impulses or neurohumoral local responses aimed at protection of the body from the effects of these factors. Despite the protective function of the TRPAj channel, an increase in its activity can lead to a number of pathological conditions characterized by pain and inflammation. Currently, there is an active search for and study of the mechanisms of action of TRPA1 antagonists, which, as shown in preclinical studies, can become effective agents in the treatment of pain, neuropathy, inflammatory processes, atopic dermatitis, cardiovascular diseases, and respiratory pathologies. Since 2007, various classes of compounds with TRPAj antagonistic activity in nanomolar concentrations have been synthesized and patented, but none of them is currently used as a drug, and the molecular mechanisms of action and properties of the discovered compounds have not been sufficiently studied.

Financial support Conflict of interests

This study hasn't had any support from outside organizations. The authors declare no conflict of interests.

References

■ Al-Shamlan F, El-Hashim AZ (2019) Bradykinin sensitizes the cough reflex via a B2 receptor dependent activation of TRPV1 and TRPA1 channels through metabolites of cyclooxygenase and 12-lipoxygenase. Respiratory Research 20(1): 110. https://doi. org/10.1186/s12931-019-1060-8 [PubMed] [PMC]

■ Araujo DSM, Miya-Coreixas VS, Pandolfo P, Calaza KC (2017) Cannabinoid receptors and TRPA1 on neuroprotection in a model of retinal ischemia. Experimental Eye Research 154: 116-125. https://doi.org/10.1016/j.exer.2016.11.015 [PubMed]

■ Arvela R, HeikkinEN T, Holm P, Prusis P, Roslund M, Salo H (2014) N-prop-2-ynyl carboxamide derivatives and their use as trpa1 antagonists. Patent WIPO (PCT), no. W0/2014/053694 A1.

■ Bachmann S, Erickson SD, Laine DI, Qian Y (2014) Imidazopyridine derivatives. Patent WIPO (PCT), no. W0/2014/076021 A1.

■ Bellono NW, Kammel LG, Zimmerman AL, Oancea E (2013) UV light phototransduction activates transient receptor potential A1 ion channels in human melanocytes. Proceedings of the National Academy of Sciences 110(6): 2383-2388. https://doi.org/10.1073/ pnas.1215555110 [PubMed] [PMC]

■ van den Berg MPM, Nijboer-Brinksma S, Bos IST, van den Berge M, Lamb D, van Faassen M, Kema IP, Gosens R, Kistemaker LEM (2021) The novel TRPA1 antagonist BI01305834 inhibits oval-bumin-induced bronchoconstriction in guinea pigs. Respiratory Research 22(1): 48. https://doi.org/10.1186/s12931-021-01638-7 [PubMed] [PMC]

■ Beskhmelnitsyna EA, Pokrovskii MV, Dolzhikov AA, Avtina TV, Zhernakova NI, Peresypkina AA (2019a) Study of the analgesic and anti-inflammatory activity of a new non-opioid analgesic based on a selective inhibitor of trpal ion channels. Kuban Scientific Medical Bulletin [Kubanskii Nauchnyy i Meditsinskii Vestnik] 26: 77-87. https://doi.org/10.25207/1608-6228-2019-26-1-77-87 [in Russian]

■ Beskhmelnitsyna EA, Pokrovskii MV, Kulikov AL, Peresypkina AA, Varavin EI (2019b) Study of anti-inflammatory activity of a new non-opioid analgesic on the basis of a selective inhibitor of TRPA1 ion channels. Anti-Inflammatory & Anti-Allergy Agents in Medicinal Chemistry 18(2): 110-125. https://doi.org/10.2174/1871 523018666190208123700 [PubMed]

■ Beskhmelnitsyna EA, Sernov LN, Avtina TV, Yakushev VI, Kostina DA, Martynova OV, Varavin II (2016) New pharmacological target for the treatment of pain syndrome. International Journal of Pharmacy and Technology 8(2): 14300-14306.

■ Beskhmelnitsyna EA, Kravchenko DV, Sernov LN, Dolzhikova IN, Avtina TV, Kulikov AL, Rozhnova DV, Yakushev VI, Martynov MA (2018) Search and evaluation of pharmacodynamic and pharmacoki-netic parameters of selective blocker of TRPA 1 ion channels from the group of substituted pyrazinopyrimidinones. Research Results in Pharmacology 4(3): 49-62. https://doi.org/10.3897/rrpharmacology.4.30303

■ Brotherton-Pleiss CE, Chen Z, Erickson SD, Kim K, Li H, Qian Y, So S-S, Wovkulich PM, Yi L (2014) Substituted phenylcarbamate compounds. Patent WIPO (PCT), no. W0/2014/4056958 A1.

■ Chaudhari SS, Thomas A, Patil NP, Deshmukh VG, Khairatkar-Joshi N, Mukhopadhyay I (2009) Imidazo [2,1-b] purine derivatives as trpal modulators. Patent WIPO (PCT), no. WO/2009/144548.

■ Chaudhari SS, Kumar S, Thomas A, Patil NP, Kadam AB, Wagh-mare NT, Khairatkar-Joshi N, Mukhopadhyay I (2011) Amides of heterocyclic compounds as trpal inhibitors. Patent WIPO (PCT), no. W0/2011/114184.

■ Chen D, Liu N, Li M, Liang S (2016) Blocking PAR2 alleviates bladder pain and hyperactivity via TRPA1 signal. Translational Neuroscience 7(1): 133-138. https://doi.org/10.1515/tnsci-2016-0020 [PubMed] [PMC]

■ Chen H, Terrett JA (2020) Transient receptor potential ankyrin 1 (TRPA1) antagonists: a patent review (2015-2019). Expert Opinion on Therapeutic Patents 30(9): 643-657. https://doi.org/10.1080/135 43776.2020.1797679 [PubMed]

■ Chen H, Safina B, Shore D, Terrett J, Villemure E, Volgraf M, Ward S, Lu A, Larouche-Gauthier R, Beaumier F, Dery M, Constantineau-Forget L, Grand-Maitre C, Lepissier L (2019) Oxadiazole transient receptor potential channel inhibitors. Patent WIPO (PCT), no. WO/2019/182925.

■ Chen J, Kim D, Bianchi BR, Cavanaugh EJ, Faltynek CR, Kym PR, Reilly RM (2009) Pore dilation occurs in TRPA1 but not in TRPM8 cannels. Molecular Pain 5: 3. https://doi.org/10.1186/1744-8069-5-3 [PubMed] [PMC]

■ Chen J, Zhang X-F, Kort ME, Huth JR, Sun C, Miesbauer LJ, Cassar SC, Neelands T, Scott VE, Moreland RB, Reilly RM, Hajduk PJ, Kym PR, Hutchins CW, Faltynek CR (2008) Molecular determinants of species-specific activation or blockade of TRPA1 channels. Journal of Neuroscience 28(19): 5063-5071. https://doi.org/10.1523/ JNEUROSCI.0047-08.2008 [PubMed] [PMC]

■ Chen Y, Yang C, Wang ZJ (2011) Proteinase-activated receptor 2 sensitizes transient receptor potential vanilloid 1, transient receptor potential vanilloid 4, and transient receptor potential ankyrin 1 in paclitaxel-induced neuropathic pain. Neuroscience 193: 440-451. https://doi.org/10.1016/j.neuroscience.2011.06.085 [PubMed]

■ Cheng W, Sun C, Zheng J (2010) Heteromerization of TRP channel subunits: extending functional diversity. Protein & Cell 1(9): 802-810. https://doi.org/10.1007/s13238-010-0108-9 [PubMed] [PMC]

■ Choi JE, Di Nardo A (2018) Skin neurogenic inflammation. Seminars in Immunopathology 40(3): 249-259. https://doi.org/10.1007/ s00281-018-0675-z [PubMed] [PMC]

■ Christensen AP, Akyuz N, Corey DP (2016) The outer pore and selectivity filter of TRPA1. PLoS ONE 11(11): e0166167. https:// doi.org/10.1371/journal.pone.0166167 [PubMed] [PMC]

■ Cosens DJ, Manning A (1969) Abnormal electroretinogram from a drosophila mutant. Nature 224(5216): 285-287. https://doi. org/10.1038/224285a0 [PubMed]

■ Dall'Acqua MC, Bonet IJM, Zampronio AR, Tambeli CH, Parada CA, Fischer L (2014) The contribution of transient receptor potential ankyrin 1 (TRPA1) to the in vivo nociceptive effects of prostaglandin

E2. Life Sciences 105(1-2): 7-13. https://doi.Org/10.1016/j. lfs.2014.02.031 [PubMed]

■ Diogenes A, Akopian AN, Hargreaves KM (2007) NGF up-regulates TRPA1: Implications for orofacial pain. Journal of Dental Research 86(6): 550-555. https://doi.org/10.1177/154405910708600612 [PubMed]

■ Estrada A, Volgraf M, Chen H, Kolesnikov A, Villemure E, Verma

V, Wang L, Shore D, Do S, Yuen P-W, Hu B, Wu G, Lin X, Lu A (2016) 1-(het)arylsulfonyl-(pyrrolidine or piperidine)-2-carboxam-ide derivatives and their use as trpa1 antagonists. Patent Canada, no. CA 2975196 A1.

■ Fruttarolo F, Pavani MG, Bencivenni S, Gatti R, Napoletano M (2014) Novel sulfonamide trpa1 receptor antagonists. Patent South Korea, no. KR 20150125716 A.

■ Gouin O, L'Herondelle K, Lebonvallet N, Le Gall-Ianotto C, Sakka M, Buhé V, Plée-Gautier E, Carré J-L, Lefeuvre L, Misery L, Le Garrec R (2017) TRPV1 and TRPA1 in cutaneous neurogenic and chronic inflammation: pro-inflammatory response induced by their activation and their sensitization. Protein & Cell 8(9): 644-661. https://doi.org/10.1007/s13238-017-0395-5 [PubMed] [PMC]

■ Gupta R, Saito S, Mori Y, Itoh SG, Okumura H, Tominaga M (2016) Structural basis of TRPA1 inhibition by HC-030031 utilizing species-specific differences. Scientific Reports 6: 37460. https://doi. org/10.1038/srep37460 [PubMed] [PMC]

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

■ Himmel NJ, Cox DN (2020) Transient receptor potential channels: current perspectives on evolution, structure, function and nomenclature. Proceedings of the Royal Society B: Biological Sciences 287(1933): 20201309. https://doi.org/10.1098/ rspb.2020.1309 [PubMed] [PMC]

■ Jiang L, Ma D, Grubb BD, Wang M (2019) ROS/TRPA1/CGRP signaling mediates cortical spreading depression. The Journal of Headache and Pain 20: 25. https://doi.org/10.1186/s10194-019-0978-z [PubMed] [PMC]

■ Kannler M, Lüling R, Yildirim AÖ, Gudermann T, Steinritz D, Dietrich A (2018) TRPA1 channels: expression in non-neuronal murine lung tissues and dispensability for hyperoxia-induced alveolar epithelial hyperplasia. Pflügers Archiv - European Journal of Physiology 470(8): 1231-1241. https://doi.org/10.1007/s00424-018-2148-6 [PubMed]

■ Kochkarov VI, Nikitin AV, Gureev VV, Korokin MV, Gudyrev OS, Pokrovskaya TG, Beskhmelnitsyna EA, Kravchenko DV, Yakushev

VI, Shabelnikova AS, Ivashchenko AA, Shafeev MA, Korokina LV, Pokrovsky MV (2016) Substituted pyrazinopyrimidinones as TRPA1 channel blockers, pharmaceutical composition, methods for their preparation and use. Patent Rissia, no. RU 2014147911A.

■ Kumar S, Thomas A, Margal S, Khairatkar-Joshi N, Mukhopadhyay I (2010a) Isothiazolo-pyrimidinedione derivatives as trpa1 modulators. Patent ARIPO, no. 2011005888 AO.

■ Kumar S, Thomas A, Waghmare NT, Margal S, Khairatkar-Joshi N, Mukhopadhyay I (2010b) Thienopyrimidinedione derivatives as trpa1 modulators. Patent WIPO (PCT), no. WO 2010109334 A3.

■ Li J, Mahajan A, Tsai M-D (2006) Ankyrin repeat: A unique motif mediating protein-protein interactions. Biochemistry 45(51): 1516815178. https://doi.org/10.1021/bi062188q [PubMed]

■ Li R, Liu R, Yan F, Zhuang X, Shi H, Gao X (2019) Inhibition of TRPA1 promotes cardiac repair in mice after myocardial infarction. Journal of Cardiovascular Pharmacology 75(3): 240-249. https:// doi.org/10.1097/FJC.0000000000000783 [PubMed]

■ Lieu T, Jayaweera G, Zhao P, Poole DP, Jensen D, Grace M, Mcln-tyre P, Bron R, Wilson YM, Krappitz M, Haerteis S, Korbmacher C, Steinhoff MS, Nassini R, Materazzi S, Geppetti P, Corvera CU, Bun-nett NW (2014) The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice. Gastroenterology 147(6): 1417-1428. https://doi.org/10.1053Zj.gastro.2014.08.042 [PubMed] [PMC]

■ Meents JE, Fischer MJM, McNaughton PA (2016) Agonist-induced sensitisation of the irritant receptor ion channel TRPA1: Agonist-induced sensitisation of TRPA1. The Journal of Physiology 594(22): 6643-6660. https://doi.org/10.1113/JP272237 [PubMed] [PMC]

■ Meents JE, Ciotu CI, Fischer MJM (2019) TRPA1: a molecular view. Journal of Neurophysiology 121(2): 427-443. https://doi. org/10.1152/jn.00524.2018 [PubMed]

■ Miura S, Takahashi K, Imagawa T, Uchida K, Saito S, Tominaga M, 0hta T (2013) Involvement of TRPA1 activation in acute pain induced by cadmium in mice. Molecular Pain 9: 7. https://doi. org/10.1186/1744-8069-9-7 [PubMed] [PMC]

■ Miyake T, Nakamura S, Zhao M, So K, Inoue K, Numata T, Takahashi N, Shirakawa H, Mori Y, Nakagawa T, Kaneko S (2016) Cold sensitivity of TRPA1 is unveiled by the prolyl hydroxylation blockade-induced sensitization to R0S. Nature Communications 7: 12840. https://doi.org/10.1038/ncomms12840 [PubMed] [PMC]

■ Moparthi L, Zygmunt PM (2020) Human TRPA1 is an inherently mechanosensitive bilayer-gated ion channel. Cell Calcium 91: 102255. https://doi.org/10.1016/j.ceca.2020.102255 [PubMed]

■ Mukaiyama M, Usui T, Nagumo Y (2020) Non-electrophilic TRPA1 agonists, menthol, carvacrol and clotrimazole, open epithelial tight junctions via TRPA1 activation. The Journal of Biochemistry 168(4): 407-415. https://doi.org/10.1093/jb/mvaa057 [PubMed]

■ Mukhopadhyay I, Kulkarni A, Aranake S, Karnik P, Shetty M, Thorat S, Ghosh I, Wale D, Bhosale V, Khairatkar-Joshi N (2014) Transient receptor potential ankyrin 1 receptor activation in vitro and in vivo by pro-tussive agents: GRC 17536 as a promising anti-tussive therapeutic. PLoS ONE 9: e97005. https://doi.org/10.1371/ journal.pone.0097005 [PubMed] [PMC]

■ Muthuppalniappan M, Kumar S, Thomas A, Khairatkar-Joshi N, Mukhopadhyay I (2009a) Phthalimide derivatives as trpa1 modulators. Patent WIPO (PCT), no. WO 2009118596 A3.

■ Muthuppalniappan M, Thomas A, Kumar S, Margal S, Khairatkar-Joshi N, Mukhopadhyay I, Gullapalli S (2009b) Quinazolinedione derivatives as trpa1 modulators. Patent United States, no. US 7951814 B2.

■ Naziroglu M, Braidy N (2017) Thermo-sensitive TRP channels: Novel targets for treating chemotherapy-induced peripheral pain. Frontiers in Physiology 8: 1040. https://doi.org/10.3389/ fphys.2017.01040 [PubMed] [PMC]

■ Oakes V, Domene C (2019) Combining structural data with computational methodologies to investigate structure-function relationships in TRP channels. Methods in Molecular Biology 1987: 65-82. https://doi.org/10.1007/978-1-4939-9446-5_5 [PubMed]

■ Ouvry G, Hacini-Rachinel F, Nonne C (2018) Trpa1 antagonists for use in the treatment of atopic dermatitis. Patent WIPO (PCT), no. WO 2018109155 A1.

■ Patapoutian A, Jegla TJ (2007) Methods and compositions for treating hyperalgesia. Patent WIPO (PCT), no. WO 2007098252 A2.

■ Paulsen CE, Armache J-P, Gao Y, Cheng Y, Julius D (2015) Structure of the TRPA1 ion channel suggests regulatory mechanisms. Nature 520(7548): 511-517. https://doi.org/10.1038/nature14367 [PubMed] [PMC]

■ Peng G, Shi X, Kadowaki T (2015) Evolution of TRP channels inferred by their classification in diverse animal species. Molecular Phylogenetics and Evolution 84: 145-157. https://doi.org/10.1016/j. ympev.2014.06.016 [PubMed]

■ Petrus M, Peier AM, Bandell M, Hwang SW, Huynh T, Olney N, Jegla T, Patapoutian A (2007) A role of TRPA1 in mechanical hyperalgesia is revealed by pharmacological inhibition. Molecular Pain 3: 40. https://doi.org/10.1186/1744-8069-3-40 [PubMed] [PMC]

■ Ryckmans T, Aubdool AA, Bodkin JV, Cox P, Brain SD, Dupont T, Fairman E, Hashizume Y, Ishii N, Kato T, Kitching L, Newman J, Omoto K, Rawson D, Strover J (2011) Design and pharmacological evaluation of PF-4840154, a non-electrophilic reference agonist of the TrpA1 channel. Bioorganic & Medicinal Chemistry Letters 21(16): 4857-4859. https://doi.org/10.1016/j.bmcl.2011.06.035 [PubMed]

■ Samanta A, Hughes TET, Moiseenkova-Bell VY (2018) Transient receptor potential (TRP) channels. Subcellular Biochemistry 87: 141-165. https://doi.org/10.1007/978-981-10-7757-9_6 [PubMed] [PMC]

■ Schüler A, Schmitz G, Reft A, Özbek S, Thurm U, Bornberg-Bauer E (2015) The rise and fall of TRP-N, an ancient family of mechanogat-ed ion channels, in metazoa. Genome Biology and Evolution 7(6): 1713-1727. https://doi.org/10.1093/gbe/evv091 [PubMed] [PMC]

■ Souza Monteiro de Araujo D, Nassini R, Geppetti P, De Logu F (2020) TRPA1 as a therapeutic target for nociceptive pain. Expert Opinion on Therapeutic Targets 24(10): 997-1008. https://doi.org/1 0.1080/14728222.2020.1815191 [PubMed] [PMC]

■ Startek JB, Talavera K, Voets T, Alpizar YA (2018) Differential interactions of bacterial lipopolysaccharides with lipid membranes: implications for TRPA1-mediated chemosensation. Scientific Reports 8(1): 12010. https://doi.org/10.1038/s41598-018-30534-2 [PubMed] [PMC]

■ Story GM, Peier AM, Reeve AJ, Eid SR, Mosbacher J, Hricik TR, Earley TJ, Hergarden AC, Andersson DA, Hwang SW, McIntyre P, Jegla T, Bevan S, Patapoutian A (2003) ANKTM1, a TRP-like channel expressed in nociceptive neurons, is activated by cold temperatures. Cell 112(6): 819-829. https://doi.org/10.1016/S0092-8674(03)00158-2 [PubMed]

■ Suo Y, Wang Z, Zubcevic L, Hsu AL, He Q, Borgnia MJ, Ji R-R, Lee S-Y (2020) Structural insights into electrophile irritant sensing by the human TRPA1 channel. Neuron 105(5): 882-894.e5. https://doi. org/10.1016/j.neuron.2019.11.023 [PubMed] [PMC]

■ Sukers K, Abraham T, Sachin SCA, Baipin PK, Venkata RY, Nilima K-J, Indranil M, Gilles J (2015) 2-amino-4-arylthiazole compounds as TRPA1 antagonists. Patent China, no. CN 103261201 B.

■ Takahashi N, Mori Y (2011) TRP channels as sensors and signal integrators of redox status changes. Frontiers in Pharmacology 2: 58. https://doi.org/10.3389/fphar.2011.00058 [PubMed] [PMC]

■ Talavera K, Startek JB, Alvarez-Collazo J, Boonen B, Alpizar YA, Sanchez A, Naert R, Nilius B (2020) Mammalian transient receptor potential TRPA1 channels: From structure to disease. Physiological Reviews 100(2): 725-803. https://doi.org/10.1152/ physrev.00005.2019 [PubMed]

■ Terrett JA, Chen H, Constantineau-Forget L, Larouche-Gauthier R, Lépissier L, Beaumier F, Déry M, Grand-Maître C, Sturino C, Vol-graf M, Villemure E (2019) Oxadiazole transient receptor potential channel inhibitors. Patent WIPO (PCT), no. WO 2019182925 A1.

■ Ton HT, Phan TX, Ahern GP (2020) Inhibition of ligand-gated TRPA1 by general anesthetics. Molecular Pharmacology 98(3): 185-191. https://doi.org/10.1124/mol.119.118851 [PubMed] [PMC]

■ Viana F (2016) TRPA1 channels: Molecular sentinels of cellular stress and tissue damage. The Journal of Physiology 594(15): 4151-4169. https://doi.org/10.1113/JP270935 [PubMed] [PMC]

■ Wang Y, Qu Z, Zhang L (2020) 3-aryloxyl-3-five-membered he-teroaryl propylamine compound and use thereof. Patent WIPO (PCT), no. WO 20035040 A1.

■ Wei H, Hämäläinen MM, Saarnilehto M, Koivisto A, Pertovaara A (2009) Attenuation of mechanical hypersensitivity by an antagonist of the TRPA1 ion channel in diabetic animals. Anesthesiology 111(1): 147-154. https://doi.org/10.1097/ALN.0b013e3181a1642b [PubMed]

■ Wei H, Chapman H, Saarnilehto M, Kuokkanen K, Koivisto A, Per-tovaara A (2010) Roles of cutaneous versus spinal TRPA1 channels in mechanical hypersensitivity in the diabetic or mustard oil-treated non-diabetic rat. Neuropharmacology 58(3): 578-584. https://doi. org/10.1016/j.neuropharm.2009.12.001 [PubMed]

■ Wei H, Koivisto A, Saarnilehto M, Chapman H, Kuokkanen K, Hao B, Huang J-L, Wang Y-X, Pertovaara A (2011) Spinal transient receptor potential ankyrin 1 channel contributes to central pain hypersensitivity in various pathophysiological conditions in the rat. Pain 152(3): 582-591. https://doi.org/10.1016/j.pain.2010.11.031 [PubMed]

■ Weyer-Menkhoff I, Lötsch J (2019) TRPA1 sensitization produces hyperalgesia to heat but not to cold stimuli in human volunteers. The Clinical Journal of Pain 35(4): 321-327. https://doi.org/10.1097/ AJP.0000000000000677 [PubMed]

■ Weyer-Menkhoff I, Pinter A, Schlierbach H, Schänzer A, Lötsch J (2019) Epidermal expression of human TRPM8, but not of TRPA1 ion channels, is associated with sensory responses to local skin cooling. Pain 160(12): 2699-2709. https://doi.org/10.1097/j. pain.0000000000001660 [PubMed]

■ Wilson SR, Gerhold KA, Bifolck-Fisher A, Liu Q, Patel KN, Dong X, Bautista DM (2011) TRPA1 is required for histamine-indepen-dent, Mas-related G protein-coupled receptor-mediated itch. Nature Neuroscience 14(5): 595-602. https://doi.org/10.1038/nn.2789 [PubMed] [PMC]

■ Wu W, Zhou H-R, Pestka JJ (2017) Potential roles for calcium-sensing receptor (CaSR) and transient receptor potential ankyrin-1 (TRPA1) in murine anorectic response to deoxynivalenol (vomitox-in). Archives of Toxicology 91(1): 495-507. https://doi.org/10.1007/ s00204-016-1687-x [PubMed]

■ Zayats V, Samad A, Minofar B, Roelofs KE, Stockner T, Ettrich R (2013) Regulation of the transient receptor potential channel TRPA1 by its N-terminal ankyrin repeat domain. Journal of Molecular Modeling 19(11): 4689-4700. https://doi.org/10.1007/s00894-012-1505-1 [PubMed]

■ Zhao J, Lin King JV, Paulsen CE, Cheng Y, Julius D (2020) Irritant-evoked activation and calcium modulation of the TRPA1 receptor. Nature 585(7823): 141-145. https://doi.org/10.1038/s41586-020-2480-9 [PubMed] [PMC]

■ Zimova L, Barvikova K, Macikova L, Vyklicka L, Sinica V, Barvik I, Vlachova V (2020) Proximal C-terminus serves as a signaling hub for TRPA1 channel regulation via its interacting molecules and supramolecular complexes. Frontiers in Physiology 11: 189. https://doi.org/10.3389/fphys.2020.00189 [PubMed] [PMC]

Author contribution

■ Natalia V. Pyatigorskaya, Doctor Habil. of Sciences in Pharmacy, Head of the Department of Industrial Pharmacy, e-mail: pyatigorskaya_n_v@staff.sechenov.ru, ORCID ID http://orcid.org/0000-0003-4901-4625.The author suggested the idea of the article, made contributions to the design of the article and participated in drafting the article.

■ Olga V. Filippova, Doctor Habil. of Sciences in Medicine, Professor of the Department of Industrial Pharmacy, e-mail: filippova_o_v_3@staff.sechenov.ru, ORCID ID http://orcid.org/0000-0001-9470-6335. The author participated in the scientific editing of the article.

■ Natalia S. Nikolenko, PhD in Pharmacy, Senior Lecturer of the Department of Industrial Pharmacy, e-mail: niko-lenko_n_s@staff.sechenov.ru, ORCID ID http://orcid.org/0000-0001-5567-8570. The author made contributions to the design of the article and participated in drafting the article.

■ Aleksey D. Kravchenko, postgraduate student of the Department of Industrial Pharmacy, e-mail: aleksej_kravchen-ko97@mail.ru, ORCID ID http://orcid.org/0000-0001-6476-0138. The author participated in the analysis of literature data and writing the article.

i Надоели баннеры? Вы всегда можете отключить рекламу.