Научная статья на тему 'CANNABINOIDS AND CANNABINOID-LIKE COMPOUNDS: BIOCHEMICAL CHARACTERIZATION AND PHARMACOLOGICAL PERSPECTIVES'

CANNABINOIDS AND CANNABINOID-LIKE COMPOUNDS: BIOCHEMICAL CHARACTERIZATION AND PHARMACOLOGICAL PERSPECTIVES Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
157
23
i Надоели баннеры? Вы всегда можете отключить рекламу.
Журнал
Russian Open Medical Journal
WOS
Scopus
ВАК
Ключевые слова
N-ACYLETHANOLAMINE / ANANDAMIDE / OLEOYLETHANOLAMINE / PALMITOYLETHANOLAMINE / DELTA(Δ)9-TETRAHYDROCANNABINOL / CANNABIDIOL / GPCRS / PPARS / TRPS

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Kytikova O.Yu., Denisenko Yu.K., Novgorodtseva T.P., Kovalenko I.S.

Publication interest in cannabinoids, including phytocannabinoids, endogenous cannabinoids, synthetic cannabinoids and cannabinomimetic compounds, is due to the therapeutic potential of these compounds in inflammatory pathology. Since recent years, scientific interest was focused on compounds with cannabinomimetic activity. The therapeutic use of phytocannabinoids and endocannabinoids is somewhat limited due to unresolved issues of dosing, toxicity and safety in humans, while cannabinoid-like compounds combine similar therapeutic effects with a high confirmed safety. Targets for endocannabinoids and phytocannabinoids are endocannabinoid receptors 1 and 2, G protein-coupled receptors (GPCRs), peroxisome proliferator-activated receptors (PPARs), and transient receptor potential ion channels (TRPs). Non-endocannabinoid N-acylethanolamines do not interact with cannabinoid receptors and exhibit agonist activity towards non-cannabinoid receptors, such as PPARs, GPCRs and TRPs. This literature review includes contemporary information on the biological activity, metabolism and pharmacological properties of cannabinoids and cannabinoid-like compounds, as well as their receptors. We established that only a few studies were devoted to the relationship of non-endocannabinoid N-acylethanolamines with non-cannabinoid receptors, such as PPARs, GPCRs, and also with TRPs. We have focused on issues that were insufficiently covered in the published sources in order to identify gaps in existing knowledge and determine the prospects for scientific research.

i Надоели баннеры? Вы всегда можете отключить рекламу.

Похожие темы научных работ по фундаментальной медицине , автор научной работы — Kytikova O.Yu., Denisenko Yu.K., Novgorodtseva T.P., Kovalenko I.S.

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

Текст научной работы на тему «CANNABINOIDS AND CANNABINOID-LIKE COMPOUNDS: BIOCHEMICAL CHARACTERIZATION AND PHARMACOLOGICAL PERSPECTIVES»

ISSN 2304-3415, Russian Open Medical Journal 1 of 10

2023. Volume 12. Issue 1 (March). Article CID e0107 Phamamionw DOI: 10.15275/rusomj.2023.0107_Pharmac0l0gy

Review

Cannabinoids and cannabinoid-like compounds: Biochemical characterization and pharmacological

perspectives

Oksana Yu. Kytikova, Yulia K. Denisenko, Tatyana P. Novgorodtseva, Ivan S. Kovalenko

Vladivostok Branch of Far Eastern Scientific Center for Physiology and Pathology of Respiration - Institute of Medical Climatology and

Rehabilitation Therapy, Vladivostok, Russia

Received 5 September 2022, Revised 11 October 2022, Accepted 23 November 2022 © 2022, Russian Open Medical Journal

Abstract: Publication interest in cannabinoids, including phytocannabinoids, endogenous cannabinoids, synthetic cannabinoids and cannabinomimetic compounds, is due to the therapeutic potential of these compounds in inflammatory pathology. Since recent years, scientific interest was focused on compounds with cannabinomimetic activity. The therapeutic use of phytocannabinoids and endocannabinoids is somewhat limited due to unresolved issues of dosing, toxicity and safety in humans, while cannabinoid-like compounds combine similar therapeutic effects with a high confirmed safety. Targets for endocannabinoids and phytocannabinoids are endocannabinoid receptors 1 and 2, G protein-coupled receptors (GPCRs), peroxisome proliferator-activated receptors (PPARs), and transient receptor potential ion channels (TRPs). Non-endocannabinoid N-acylethanolamines do not interact with cannabinoid receptors and exhibit agonist activity towards non-cannabinoid receptors, such as PPARs, GPCRs and TRPs. This literature review includes contemporary information on the biological activity, metabolism and pharmacological properties of cannabinoids and cannabinoid-like compounds, as well as their receptors. We established that only a few studies were devoted to the relationship of non-endocannabinoid N-acylethanolamines with non-cannabinoid receptors, such as PPARs, GPCRs, and also with TRPs. We have focused on issues that were insufficiently covered in the published sources in order to identify gaps in existing knowledge and determine the prospects for scientific research.

Keywords: N-acylethanolamine, anandamide, oleoylethanolamine, palmitoylethanolamine, delta(A)9-tetrahydrocannabinol, cannabidiol, GPCRs, PPARs, TRPs.

Cite as Kytikova OYu, Denisenko YuK, Novgorodtseva TP, Kovalenko IS. Cannabinoids and cannabinoid-like compounds: Biochemical characterization and pharmacological perspectives. Russian Open Medical Journal 2023; 12: e0107.

Correspondence to Oksana Yu. Kytikova. Address: 73g Russkaya St., Vladivostok 690105, Russia. Phone: +74232788201. E-mail: kytikova@yandex.ru.

Introduction

Interest in cannabinoids and cannabinoid-like compounds is due to their therapeutic potential for inflammatory diseases that are currently widespread worldwide [1-9].

Cannabinoids include natural cannabinoids, endogenous cannabinoids, synthetic cannabinoids, and cannabinomimetic compounds [6].

Natural cannabinoids, or phytocannabinoids, are found in plants of the hemp family (cannabis, derived from the Latin Cannabis sativa L.) [6]. The most studied phytocannabinoids are delta(A)9-tetrahydrocannabinol (THC) and cannabidiol (CBD), along with secondary cannabinoids [10].

Endogenous cannabinoids, or endocannabinoids, are lipids that are synthesized in the human body. First of all, these include N-arachidonoylethanolamine (AEA), also known as anandamide (ANA), and 2-arachidonoylglycerol (2-AG) [5, 6, 11, 12].

Compounds with cannabinomimetic activity include derivatives of saturated or monounsaturated fatty acids, such as N-palmitoylethanolamine (PEA), N-oleoylethanolamine (OEA), stearoylethanolamine, etc. [13].

Cannabinoids are also represented by synthetic substances with a structure and action similar to phytocannabinoids (classical cannabinoids) and by synthetic substances that differ from plant cannabinoids in structure, albeit similar in action (nonclassical cannabinoids) [6]. The latter include aminoalkylindoles, eicosanoids, quinolines, arylsulfonamides, etc. [13].

According to the international lipid classification displayed on the LIPID MAPS website (https://www.lipidmaps.org/), endocannabinoids include N-acylethanolamines (NAE) of saturated, monoenoic, and polyunsaturated fatty acids synthesized in the human body. Some cannabinomimetic compounds also belong to the class of NAE [14, 15]. For instance, NAE includes AEA, which contains a polyunsaturated fatty acid, as well as PEA and OEA, which contain saturated or monounsaturated fatty acids. As for 2-AG, it belongs to the monoacylglycerol group (MAG) and is a true endocannabinoid, while AEA is a byproduct of the main NAE: PEA and OEA. At the same time, the physiological role of PEA and OEA is less studied, compared with 2-AG.

Potential targets for endocannabinoids and phytocannabinoids are endocannabinoid receptors 1 and 2 (CB1, CB2) [16], orphan G

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

protein-coupled receptors, GPCRs) [17-19], peroxisome proliferator-activated receptors (PPARs) [20], and transient receptor potential ion channels (TRPs) [10, 11, 21, 22]. However, amides of saturated or monounsaturated fatty acids, such as PEA and OEA, are considered non-endocannabinoid NAE, since they do not interact with cannabinoid receptors and exhibit agonist activity towards non-cannabinoid receptors, such as PPARs, GPCRs, and TRPs [18].

CB1, CB2, and other GPCRs, along with PPARs and TRPs, are expressed in the digestive and respiratory tracts, skin, and many other organs and systems [16]. Consequently, the therapeutic potential of phytocannabinoids, endocannabinoids, and compounds with cannabinomimetic properties is actively studied in bronchopulmonary inflammatory diseases [1, 7], inflammatory diseases of bowel [4, 5] and skin [6, 8, 9], Alzheimer's disease [2], multiple sclerosis [3], chronic pain [23], obesity [24], and a number of other pathologies.

It should be noted that the therapeutic use of phytocannabinoids and endocannabinoids is rather limited due to the unresolved issues of their dosing, toxicity and safety for humans [21]. The non-endocannabinoid NAE (PEA and OEA) combine similar therapeutic effects with a higher confirmed safety [25]. These N-acylethanolamines have anti-inflammatory, analgesic and anorexic effects via non-endocannabinoid receptors [18].

The PubMed database was initially searched for articles published in English between January 1, 2017 and January 1, 2022. We used the following keywords: N-acylethanolamine, anandamide, oleoylethanolamine, palmitoylethanolamine, delta(A ^-tetrahydrocannabinol, cannabidiol, GPCRs, PPARs, TRPs (Table 1).

Publication interest in cannabinoids is steadily increasing. E.g., a search query with this keyword conducted in the PubMed database yielded 9,832 results over the past five years, while 2,431 (25%) of them were associated with the last year. Scientific interest in N-acylethanolamines is also constantly growing. A PubMed search with the N-acylethanolamine keyword revealed 230 articles between 2017 and 2022, 62 of which (27%) were from the last year. At the same time, a small number of studies was published on the relationship of PEA and OEA with GPCRs, PPARs and TRPs, as well as of all NAEs with GPCRs (GPR55 and GPR119).

Our review summarizes the latest data on the biological activity, metabolism, and anti-inflammatory effects of endocannabinoid and non-endocannabinoid N-acylethanolamines, as well as their receptors. We established that only a small number of studies was devoted to the relationship of non-endocannabinoid NAEs with non-cannabinoid receptors, such as PPARs, GPCRs, and TRPs. Emphasis was placed on issues that were insufficiently covered in the published sources in order to identify gaps in existing knowledge and determine the prospects for scientific research.

Endocannabinoids and their receptors

The endocannabinoid system consists of cannabinoid receptors (CB1 and CB2), endogenous cannabinoid neurotransmitters (AEA and 2-AG), and also includes transmembrane and intracellular transport systems, along with enzymes responsible for the synthesis and decomposition of neurotransmitters. These enzymes are represented by fatty acid amide hydrolase (FAAH) for AEA and monoacylglycerol lipase (MAGL) for 2-AG [26]. Alternative ways of endocannabinoid degradation include their oxidation with the cyclooxygenase, lipoxygenase and cytochrome P450 [5].

Table 1. Results of a systematic search of 2017-2022 publications on the research topic in the PubMed database

Keywords_Number of articles for the period of2017-2022

"N-acylethanolamine" and CB1 and CB2 7

"Palmitoylethanolamide" and CB1 and CB2 21

"Anandamide" and CB1 and CB2 144

"Oleoylethanolamine" and CB1 and CB2 3

"^-tetrahydrocannabinol" and CB1 and CB2 23

"Cannabidiol" and CB1 and CB2 159

"N-acylethanolamine" and peroxisome proliferator-activated receptor 25

"Palmitoylethanolamide" and peroxisome proliferator-activated receptor 70

"Anandamide" and peroxisome proliferator-activated receptor 30

"Oleoylethanolamine" and peroxisome proliferator-activated receptor 5

"^-tetrahydrocannabinol" and peroxisome proliferator-activated receptor 5

"Cannabidiol" and peroxisome proliferator-activated receptor 43

"N-acylethanolamine" and transient receptor potential channels 1

"Palmitoylethanolamide" and transient receptor potential channels 4

"Anandamide" and transient receptor potential channels 62

"Oleoylethanolamine" and transient receptor potential channels 2

"^-tetrahydrocannabinol" and transient receptor potential channels 1

"Cannabidiol" and transient receptor potential channels 52

"N-acylethanolamine" and GPR55 4

"Palmitoylethanolamide" and GPR55 12

"Anandamide" and GPR55 17

"Oleoylethanolamine" and GPR55 1

'^-tetrahydrocannabinol" and GPR55 3

"Cannabidiol" and GPR55 58

"N-acylethanolamine" and GPR119 1

"Palmitoylethanolamide" and GPR119 2

"Anandamide" and GPR119 2

"Oleoylethanolamine" and GPR119 1

^-tetrahydrocannabinol" and GPR119 3

"Cannabidiol" and GPR119 5

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

Table 2. Endocannabinoids and their receptors

Endocannabinoids/Receptors AEA 2-AG

CB1 partial agonist agonist

CB2 partial agonist primary endogenous agonist

GPCRs agonist agonist

TRPA1 agonist agonist

TRPV1 agonist agonist

TRPV2 agonist agonist

TRPV3 agonist agonist

TRPV4 agonist agonist

TRPM8 agonist agonist

PPAR-a agonist agonist

PPAR-ß/ö agonist agonist

PPAR-y agonist agonist

Table 3. Phytocannabinoids and their receptors

Phytocannabinoids/Receptors THC CBD

CB1 partial agonist / antagonist

agonist low affinity

CB2 partial antagonist

agonist low affinity

GPRs agonist inverse agonist

TRPA1 agonist agonist

TRPV1 agonist partial agonist

TRPV2 agonist agonist

TRPV3 agonist agonist

TRPV4 agonist agonist

TRPM8 agonist antagonist

PPAR-a - agonist

PPAR-ß/ö - agonist

PPAR-y agonist possible agonist

partial agonist at low concentration

5-HT1A - inverse agonist at higher

concentration

D2 dopamine receptor - partial agonist

Table 4. Non-endocannabinoid N-acylethanolamines and their receptors

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

N-acylethanolamines/Receptors PEA OEA

CB1 agonist -

CB2 - -

GPR55 agonist -

GPR19 agonist agonist

GPR40 - agonist

TRPA1 - -

TRPV1 agonist agonist

TRPV2 - -

TRPV3 - -

TRPV4 - -

TRPM8 - -

PPAR-a agonist -

PPAR-ß/ö - -

PPAR-y - -

Structurally, AEA and 2-AG are eicosanoids: 2-AG is involved in regulating the circulatory system, pain and inflammation (immune inflammation and neuroinflammation), while AEA is involved in thermoregulation and nociception.

Endocannabinoids and their receptors are presented in Table

2.

Endocannabinoids 2-AG and AEA are agonists of both cannabinoid receptors CB1 and CB2.

The CB1 expression (central type) prevails in the nociceptive areas of the central nervous system, albeit it is also detected in the liver, pancreas, muscle tissue and adipose tissue [27]. CB1 receptor

is responsible for the adaptive regulation of intracellular signal cascades, synaptic and cellular plasticity processes [28-30]. This receptor participates in the mechanisms of forming the anxiety, pain, addiction, inflammation, as well as regulation of metabolism [16].

CB2 (peripheral type) is expressed mainly on the immune system cells [30-32]. CB2 is involved in the implementation of the anti-inflammatory effect via reducing the production of proinflammatory cytokines and increasing the production of anti-inflammatory cytokines [33]. CB2 agonists inhibit recruitment of leukocytes and reduce the content of tumor necrosis factor-alpha (TNF-a), interleukins (IL-1p, IL-6, IL-18), monocyte chemoattractant protein-1 (MCP-1) and active forms of oxygen [31]. CB2 agonism leads to the activation of 5'-AMP-activated protein kinase (AMPK), stimulating oxidative phosphorylation and thereby the development of anti-inflammatory effect.

The latest scientific data indicate the existence of other receptors for cannabinoids associated with G proteins. These G protein-coupled receptors (GPCRs) are located in the central nervous system, intestines, liver, bones, skeletal muscles and adipose tissue: G protein-coupled receptor 18 (GPR18), G proteincoupled receptor 55 (GPR55), G-protein coupled receptor 119 (GPR119) and many others [17, 18].

GPR55 receptors are involved in glucose homeostasis, antiinflammatory and analgesic effects [34, 35]. They are localized in several areas of the brain associated with the development of pain, as well as in the spinal cord [36]. The role of the GPR55 in the pathogenesis of neuropathic pain remains controversial, since some data indicate that their activation causes nociception [34, 37], while other data imply the absence of such effect [38].

GPR119 consists of 335 amino acids and participates mainly in glucose homeostasis [35]. It is expressed in the spinal cord and some areas of the brain, but its involvement in the development of pain is unknown [34, 35]. The study by A. Zuniga-Romero et al. established that both GPR55 and GPR119 can be important targets for the treatment of neuropathic pain [34].

AEA and 2-AG endocannabinoids are also capable of interacting with other GPCRs (GPR3, GPR6, GPR12, GPR18), as well as with TRPs (TRPV1, TRPV2, TRPV3, TRPV4, TRPM8, TRPA1) and PPARs (PPAR-a, PPAR-P/5, PPAR-y) [5, 6, 12]. Besides, they exhibit affinity to serotonin receptors (5-HT1A, 5-HT3), as well as to adenosine and glycine receptors [39]. Anandamide was the first discovered endogenous agonist of TRPV1 and antagonist of TRPM8 [20, 40].

PPARs (PPAR-a, PPAR-P/5 u PPAR-y) play an important role in the regulation of cellular processes (differentiation, proliferation, apoptosis) and inflammatory responses [41-43]. These receptors are classified as nuclear transcription factors with anti-inflammatory activity [44, 45].

PPAR-a isomer is expressed largely in the liver, kidneys, heart, skeletal muscles, brown adipose tissue, as well as in epithelial cells, macrophages, lymphocytes and dendritic cells of the respiratory tract [46]. Activation of PPAR-a reduces the production of proinflammatory mediators (TNF-a, IL-1, IL-6, IL-8) and modulates the expression of adhesion molecules and chemotactic factors involved in the pathogenesis of inflammation [47]. PPAR-a is also capable of inducing the production of anti-inflammatory IL-10 [47].

PPAR-P/5 isomer (PPAR-5, PPAR-P, hNUCl, FAAR) is most pronounced in the brain, liver, skin, adipose tissue, keratinocytes

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

and skeletal muscles [48, 49]. It participates in oxidation of fatty acids and regulates the level of glucose in the blood, along with participating in the wound healing processes [50].

PPAR-y is expressed virtually in all tissues and cells, but mainly in adipose tissue, colon and spleen [51]. This receptor is a regulator of cellular homeostasis, energy metabolism [41] and a participant of inflammatory responses [44, 45].

Superfamily of transient receptor potential (TRP) ion channels combines conventional receptors of the following categories: canonical (TRPC 1-7), vanilloid (TRPV 1-6), polycystic (TRPP 1-3), mucolipin (TRPML 1-3), ankyrin 1 (TRPA 1), and melastatin (TRPM 1-8) [52, 53]. TRP channels are expressed mainly in neuronal cells, as well as in bronchial epithelium and endothelia, smooth muscle cells, and unmyelinated nociceptive C-type fibers of the lungs [54]. Thermosensitive TRPs (TRTRV1, TRPV2, TRPV4, TRPM3, TRPM8 u TRPA1) constitute the subfamily of TRPs, the representatives of which are activated when temperature changes [55].

Therefore, CB1, CB2, GPCRs (GPR3, GPR6, GPR12, GPR18), TRPs (TRPV1, TRPV2, TRPV3, TRPV4, TRPM8, TRPA1), PPARs (PPAR-a, PPARP/5, PPAR-y), 5-HT1A, 5-HT3, as well as adenosine and glycine receptors, are the targets for endocannabinoids.

Anti-inflammatory effects of endocannabinoids

The complex modes of the action of cannabinoids include their anti-inflammatory and antioxidant effects [23, 27, 28], as well as their ability to modulate immunological processes [22]. The levels of circulating endocannabinoids increase with a number of diseases, the pathogenetic mechanism of which is systemic inflammation [56]. Thus, modulation of endocannabinoid system can have therapeutic potential for multiple pathological conditions [57].

Although the most studied functions of endocannabinoids are associated with the central nervous system and immune processes [22], experimental studies of the last decades confirmed that the transmission of cannabinoid signals is involved in the maintenance of skin homeostasis. Hence. The disorders of skin homeostasis regulation contributes to the development of atopic dermatitis, psoriasis, scleroderma and other skin diseases [6, 8, 9]. CB1 and CB2 are located in keratinocytes, hair follicles, sebaceous glands, sensory neurons, immune system cells and skin fibroblasts [6]. In the experimental study, the CB2 agonist caused the epithelization of skin wounds via increasing the proliferation and migration of keratinocytes [58]. Similarly, in another experiment, the activation of the CB2 receptor limited the infiltration of neutrophils and macrophages and stimulated the proliferation of keratinocytes, thereby contributing to faster wound healing [59]. CB2 agonists lead to a reduction in the production of proinflammatory macrophages M1 and an increase in the number of anti-inflammatory macrophages M2 [59]. The recently developed local inhibitor of the endocannabinoid cell membrane transporter (WOL67-531) minimized allergic manifestations and itching of the skin in the model of atopic dermatitis [60].

CB1, CB2, GPR55 and PPAR-a receptors were revealed in the digestive tract of dogs [4]. Galiazzo et al. demonstrated that these receptors play a protective role in inflammatory intestinal diseases [4]. There are data on a change in the expression of the CB1, CB2 and levels of endocannabinoids in diverticulitis, irritable intestine, inflammatory intestine and colon cancer [5]. Endocannabinoids are capable of acting as double agonists of CB2 and PPAR-y, leveling off chronic inflammation [3].

In our earlier publication, we covered in detail the role of the endocannabinoid signal system in the pathophysiology of bronchial asthma and obesity [7]. In the bronchi of mice, PPAR-a, CB1 and CB2 receptors are localized [20], with which endocannabinoids interact. CB2 plays an important role in the migration of eosinophils into the respiratory tract [61]. Activation of this receptor type on mast cells has a direct anti-inflammatory effect. The CB2 agonist exhibited a strong anti-inflammatory activity mediated via inhibiting the leukocyte chemoattractant leukotriene B4, prostaglandin E2, thromboxane B2, and prostaglandin F2a [62]. A decrease in CB2 expression maintains chronic inflammation with chronic obstructive pulmonary disease and is accompanied by an increase in the level of proinflammatory cytokines (TNF-a and a transforming growth factor-beta (TGF-P) [63].

The dysfunction of TRP channels constitutes the pathogenetic basis of many inflammatory diseases [40, 53, 54]. The sensitivity of nociceptors is associated with the activity of these channels, the number of which increases significantly in the lungs of patients with bronchial asthma [64]. Chronic inflammation of the respiratory tract and mucus hypersecretion is associated with TRPV1 sensitization [64, 65].

Some studies demonstrated high potential of cannabinoids in the treatment of systemic respiratory infection [33]. For example, CB2 activation limited the infiltration of immune cells into the lungs of animals infected with respiratory syncytial virus and reduced the number of neutrophils and monocytes in the bronchoalveolar lavage. The effects were accompanied by a decrease in the IFN-y, macrophage inflammatory protein-1a (MIP-1a) and an increase in IL-10 [66].

The increased ratio of polyunsaturated fatty acids (PUFA), w-6/w-3, leads to an increase in the levels of AEA and 2-AG, CB1 activity in adipose tissue, pancreas, muscles and liver, and a disorder of the energy homeostasis regulation [67]. Activation of this type of receptors with endocannabinoids helps launching the processes of glucose absorption, adipogenesis, induction of lipogenesis processes, and a reduction in brown adipose tissue thermogenesis [7]. The use of CB1 antagonists is a promising strategy for the treatment of obesity [68].

Thus, the presented data demonstrate high anti-inflammatory potential of endocannabinoids.

Phytocannabinoids

Along with endocannabinoids, phytocannabinoids interact with cannabinoid receptors as well. Phytocannabinoids include THC and CBD, as well as secondary cannabinoids, such as cannabigerolic acid (CBGA) and cannabidiolic acid (CBDA) [10, 39]. Other Cannabis sativa L. compounds include cannabigerol (CBG), cannabichromene (CBC), tetrahydrocannabivarin (THCV), cannabicitran (CBT), cannabidivarin (CBDV) and cannabigerivarin (CBGV) [69]. The precursor to all plant cannabinoids is CBGA, which is converted to CBDA, cannabichromenic acid, and A-9-tetrahydrocannabinolic acid. These acids form free cannabinoids (CBC, CBD and THC). In addition, Cannabis sativa L. contains terpenes, which exhibit a synergistic effect when interacting with cannabinoids [70].

Structurally, THC and CBD are classical cannabinoids [71].

Phytocannabinoids and their receptors are summarized in Table 3.

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

THC binds predominantly to CB1 and, to a lesser extent, to CB2 [19, 31]. It actively influences TRPV2 and has a moderate modulating effect on TRPV3, TRPV4, TRPA1, and TRPM8 [20].

CBD exhibits even lower affinity for CB1 and CB2 receptors than THC. At low concentrations, it even acts as a CB1 and CB2 antagonist [21]. At low concentrations, CBD is a partial agonist of 5-HT1A receptors, while at higher concentrations, it is an inverse agonist [71], as well as a probable PPAR-y agonist [22]. Besides, CBD is an agonist for TRPV1-4 and TRPA1 and antagonist for TRPM8 [11].

Also, GPR55 and GPR18 are potential targets for phytocannabinoids [10]. THC was identified as a non-selective GPR55 agonist, while CBD blocked this receptor [10]. More recent studies have led to the identification of selective non-cannabinoid GPR55 ligands, including several molecular species of endogenous phospholipids and peptides [17].

CBG, CBDA, CBGA, CBDV, and CBGV act differently on CB1 and CB2 [10]. These phytocannabinoids are agonists; however, they can act as inverse agonists under certain conditions.

Anti-inflammatory effects of phytocannabinoids

THC and its butyl analog, along with A-8-tetrahydrocannabinol and A-9-tetrahydrocannabivarin, have a psychotropic effect. Contrariwise, CBD, lacking such effect, is capable of changing the action of psychotropic cannabinoids [32, 39]. The therapeutic efficacy of THC is associated with toxicity and a high risk of addiction. The pharmacological effects of THC are close to those of endocannabinoids [57].

An experiment involving the models of systemic inflammation (non-specific ulcerative colitis, Crohn's disease) demonstrated that THC was a suppressor of aberrant immune responses [72]. In addition, THC prevents the development of colorectal cancer associated with colitis. The anticarcinogenic and anti-inflammatory effects of THC are due to the activation of CB2 on immune cells and inhibition of the release of proinflammatory cytokines (IFNy, TNF-a, IL-17, IL-23 and IL-22) [72]. Interacting with CB2, THC activates the formation of TGF-P1, inducing the production of regulatory T cells (Treg), aimed at suppressing the synthesis of IFNy and TNFa. Suppression of antigen-presenting cells (APCs) by THC blocks the formation of IL-22 and IL-17. Endocannabinoid levels in patients with ulcerative colitis correlate with clinical parameters and depend on cannabis consumption [73].

Available published data suggest that phytocannabinoids may be effective for the treatment of inflammatory skin diseases [71, 74]. It is known that oral preparations for these diseases are effective only at high doses, causing pronounced side effects [74]. Selective CB2 agonists are effective in the treatment of systemic scleroderma and dermatomyositis. Sublingual administration of THC and CBD reduces pain in epidermolysis bullosa [71]. Topical application of phytocannabinoids is effective in atopic and contact dermatitis and psoriasis [74]. In experimental conditions, it was demonstrated that topical application of THC reduced the activity of allergic inflammation via blocking the production of proinflammatory mediators by keratinocytes [71]. Piomelli et al. demonstrated an ability of THC to inhibit the production of IFN-y, and the release of proinflammatory chemokines and cytokines independently of cannabinoid receptors [26].

CBD exhibits neuroprotective, antiepileptic, anxiolytic, antidepressant, anti-inflammatory, analgesic, and anticarcinogenic

properties [32, 75]. In experimental conditions, the effects of CBD depended on the dose, administration route, and the phase of the inflammatory process [39].

CBD suppresses the expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), both of which are the invasion pathways for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) localized in the epithelium of the oral cavity, lungs and intestines [76]. The benefits of using CB2 agonists to block the inflammatory response in patients infected with SARS-CoV-2 are discussed in some publications, which is associated with the ability of cannabinoid receptors to reduce the production of proinflammatory cytokines and immune cell proliferation [77]. CBD limits the severity and progression of coronavirus disease (COVID-19) by suppressing the expression of two key receptors for SARS-CoV-2 and a wide range of immunomodulatory and anti-inflammatory effects of this phytocannabinoid [78]. Activation of PPAR-y in resident alveolar macrophages limits pulmonary inflammation and promotes recovery from viral respiratory infections [51]. As a PPAR-y agonist, CBD also exhibits antiviral activity and is able to inhibit the development of pulmonary fibrosis [78]. TRP channels partially mediate the effects of CBD on reducing inflammation and pain [11].

It should be noted that a limitation to the clinical use of cannabinoids is the development of side effects (drowsiness, dizziness, speech disorders, memory impairment, drug addiction) at dosages that have therapeutic value [32]. It is likely that the conflicting effects observed in clinical trials using endocannabinoids may be due to the high heterogeneity of their receptors and the complexity of cannabinoid signaling [7]. While promising results from CBD have been identified, its effectiveness is limited by uncertainty regarding its regulatory status and safety in humans [21]. Most studies used significant differences in dosing regimens and administration routes [21]. This determines the high demand for non-endocannabinoid N-acylethanolamines (PEA and OEA), which combine similar effects with high safety [25]. These NAE do not bind to cannabinoid receptors, exerting anti-inflammatory, analgesic, and anorexic effects predominantly via PPARs, GPR55, GPR119, and TRPs [18].

Non-endocannabinoid N-acylethanolamines and their receptors

NAE and their receptors are summarized in Table 4. PEA is the most common NAE; it cannot be strictly classified as a classical endocannabinoid, albeit it is often studied in combination with AEA due to similar metabolic pathways of biosynthesis [20, 13]. The mechanism of PEA action is primarily mediated by activation of the PPAR-a nuclear receptor [25, 42]. The results of its interaction with GPR55 [79, 80], GPR119, and TRPV1 [13, 81] are presented.

OEA is a monounsaturated analog of AEA, acting independently of the cannabinoid pathway [82, 83]. OEA activates PPAR-a [81]. It was also demonstrated that OEA binds to GPR119 [84], GPR40 [85], and TRPV1 [86].

Anti-inflammatory effects of non-endocannabinoid N-acylethanolamines

The anti-inflammatory activity of PEA was first described by Ganley and Robinson in 1958 [87]. Further studies demonstrated

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

that PEA had a favorable effect on neuroinflammation and neurodegeneration, and also has analgesic activity [13, 20, 26].

Several in vitro and in vivo preclinical studies demonstrated that the biological effects of PEA were caused by its impact on the central and peripheral nervous systems [18, 20, 42, 88]. Currently, PEA is widely used to reduce pain and inflammation [89] and is seen as a promising alternative to exogenous CBD [25]. For example, products and supplements containing PEA (Levagen®, Levagen®+, Normast®, Glialia®, Adolene®, Visimast®, and Pelvilin®) are licensed in various countries as dietary supplements or nutraceuticals, or foods for special medical purposes (FSMPs) at the recommended daily dose of 1200 mg [13, 90, 91].

The anti-inflammatory activity of PEA is implemented through its interaction with PPAR-a, a key regulator of inflammation and pain, which was first demonstrated in 2005 by J. LoVerme et al. [92]. PEA is an endogenous PPAR-a agonist; consequently, blocking the decomposition of PEA by inhibitors of N-acylethanolamine acid amidase (NAAA) leads to activation of the PEA/PPAR-a signaling pathway and control of inflammation and pain [93]. Since chronic pain is associated with the release of NAAA in spinal cord cells, inhibition of this amidase represents a novel approach to the treatment of inflammation and pain [93]. Under experimental conditions, PPAR-a antagonists blocked the protective effects of PEA in neuroinflammation and neurodegeneration [88].

PEA caused an increase in PPAR-a activity during the development of inflammation in the experiment (P <0.0001), while the effects of PEA were blocked by the PPAR-a antagonist, GW6471 [94]. Interestingly, in some animal models, the anti-inflammatory activity of PEA was leveled off by the inverse agonist of the cannabinoid CB2 receptor, SR144528, while PEA did not interact with these receptors and retained its effects in mice lacking CB2 [92]. In addition, SR144528 also blocks the effects of the PPAR-a agonist GW7647. It should be noted that sometimes PPAR-a antagonists do not affect PEA activity, even though they are sensitive to AM630, which is an inverse agonist of the CB2 receptor [95]. Therefore, the effects of PEA mediated by CB2 need further studying. For example, in an experiment, PEA reduced itching and inflammation in skin diseases by activating not only PPAR-a, but also CB2 [96]. The involvement of CB2 in the implementation of the anti-inflammatory action of PEA is rather controversial [79].

In a randomized double-blind controlled trial, D.G. Couch et al. demonstrated that PEA reduced the permeability of the human gastrointestinal tract in vitro, ex vivo and in vivo [94]. This study evaluated the effect of PEA on the absorption of lactulose and mannitol in people taking aspirin. In vivo, aspirin increased the absorption of lactulose and mannitol, which was reduced by PEA (p<0.001). These results are applicable to diseases associated with increased intestinal permeability, such as inflammatory bowel diseases [94]. In an experimental model of colitis, it was demonstrated as well that exogenous administration of PEA reduced the severity of inflammation and intestinal permeability. It is noteworthy that these effects were accompanied by suppression of GPR55 mRNA, but not by changes in the activities of CB1, CB2, and PPAR-a [97].

The anti-inflammatory effect of PEA is also implemented via GPR55 [79, 80]. E.g., PEA prevents the development of atherosclerosis by promoting switching to the anti-inflammatory phenotype of macrophages due to the activation of GPR55 [79]. Recently, the neuroprotective role of GPR55 in stroke was

described both in vitro and in vivo [80]. Taken together, these data suggest that the neuroinflammation-leveling effects of PEA may be partially mediated by GPR55 activation.

B. Marichal-Cancino et al. proposed that PEA inhibited vasopressor responses to sympathetic stimulation and exogenously administered norepinephrine and induced hypotension through interactions with CB1, TRPV1, and possibly GPR55, but not with CB2 [98]. One of the studies discovered that PEA was also able to prevent the reduction in TRPV1 activity during inflammation (P<0.0001) [94].

Oleoylethanolamine (OEA) is anorectic N-acylethanolamine produced in the intestines from dietary fats. It modulates lipid metabolism and insulin secretion via the activation of PPAR-a and GPR119 [85]. The role of OEA in appetite regulation, fat metabolism, and energy homeostasis is well documented [81]. Endocannabinoids stimulate appetite, and lipid and glucose anabolism, thereby increasing body weight, while OEA has the opposite effect due to the activation of PPAR-a [81]. The molecular mechanisms underlying appetite suppression by this NAE remain to be clarified.

Supplements containing OEA mitigate risk factors associated with non-alcoholic fatty liver disease by increasing the level of PPAR-a gene expression [99]. Thus, the use of OEA leads to a decrease in serum levels of triglycerides, alanine aminotransferase (ALT), aspartate aminotransferase (AST), ALT/AST ratio, an increase in high-density lipoprotein cholesterol levels and an improvement in appetite. PPAR-a and GPR119 are important for improving metabolism after bariatric surgery conducted for the treatment of obesity in the experiment (vertical sleeve gastrectomy) [84].

This lipid not only regulates nutrition and body weight by stimulating lipolysis [85], but also has an antitumor effect [100]. There is evidence that OEA protects mice from acute ischemic brain injury by enhancing PPAR-a signaling [101]. During cerebral ischemia, a significant amount of NAE accumulates, but the role of this accumulation is not clear [81, 102]. OEA is used for oral administration in stroke using the nanoparticle-based drug delivery system (NDDS) [82, 83].

Further research is needed to specify the biological activities of PEA and OEA, as well as to develop therapeutic preparations to combat inflammatory diseases.

Conclusion

Over the 20-year period that passed since the discovery of endocannabinoids and cannabinoid receptors, numerous studies were carried out to examine the functions of the endocannabinoid system and explore the therapeutic potential of cannabinoids in various pathologies. However, with a wide range of biological effects, cannabinoids have a number of side effects that limit their therapeutic potential. The discovery of endocannabinoid-like molecules that are saturated or monounsaturated NAE and MAG, and have a similar mechanism of action to cannabinoids but lack their side effects, has shifted scientific interest towards investigating these molecules.

Use of cannabinoids and cannabinoid-like compounds in medicine is actively studied, but the relationship between these compounds and their receptors remains to be clarified. For instance, phytocannabinoids and cannabinoid N-acylethanolamines have similar anti-inflammatory, antimicrobial,

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

antioxidant and neuromodulatory effects in the course of their interaction with the same receptors: CB1, CB2, PPARs, GPCRs and TRPs. At the same time, non-endocannabinoid N-acylethanolamines do not interact with CB1 and CB2. A growing body of evidence suggests that lipid signaling mediators have similar receptors but act differently and have specific signaling.

PEA and OEA could be more therapeutically promising than cannabinoids. Therefore, further studies are needed to elucidate the molecular mechanisms underlying their biological activity.

Conflict of interest

We declare that we have no conflicts of interest. References

1. Fraguas-Sánchez AI, Martín-Sabroso C, Torres-Suárez AI. Insights into the effects of the endocannabinoid system in cancer: a review. Br J Pharmacol 2018; 175(13): 2566-2580. https://doi.org/10.1111/bph.14331.

2. Maurya N, Velmurugan BK. Therapeutic applications of cannabinoids. Chem Biol Interact 2018;293: 77-88. https://doi.org/10.1016/Ubi.2018.07.018.

3. García-Martín A, Garrido-Rodríguez M, Navarrete C, Caprioglio D, Palomares B, DeMesa J, et al. Cannabinoid derivatives acting as dual PPAR-y/CB2 agonists as therapeutic agents for systemic sclerosis. Biochem Pharmacol 2019; 163: 321-334. https://doi.org/10.1016/j.bcp.2019.02.029.

4. Galiazzo G, Giancola F, Stanzani A, Fracassi F, Bernardini C, Forni M, et al. Localization of cannabinoid receptors CB1, CB2, GPR55, and PPAR-a in the canine gastrointestinal tract. Histochem Cell Biol 2018; 150(2): 187-205. https://doi.org/10.1007/s00418-018-1684-7.

5. Lee Y, Jo J, Chung HY, Pothoulakis C, Im E. Endocannabinoids in the gastrointestinal tract. Am J Physiol Liver Physiol 2016; 311(4): G655-G666. https://doi.org/10.1152/ajpgi.00294.2015.

6. Scheau C, Badarau I.A, Mihai L.-G., Scheau A.E, Costache D.O, Constantin C., Calina D., Caruntu C. Cannabinoids in the pathophysiology of skin inflammation. Molecules 2020; 25(3): 652. https://doi.org/10.3390/molecules25030652.

7. Kytikova OY, Novgorodtseva TP, Denisenko YK, Antonyuk MV, Gvozdenko TA. The role of the endocannabinoid signaling system in the pathophysiology of asthma and obesity. Annals of the Russian academy of medical sciences 2019; 74(3): 200-209. Russian. https://doi.org/10.15690/vramn1133.

8. Tóth KF, Ádám D, Bíró T, Oláh A. Cannabinoid signaling in the skin: Therapeutic potential of the "c(ut)annabinoid" system. Molecules 2019; 24(5): 918. https://doi.org/10.3390/molecules24050918.

9. Cintosun A, Lara-Corrales I, Pope E. Mechanisms of cannabinoids and potential applicability to skin diseases. Clin Drug Investig 2020; 40(4): 293-304. https://doi.org/10.1007/s40261-020-00894-7.

10. Navarro G, Varani K, Lillo A, Vincenzi F, Rivas-Santisteban R, Raich I, et al. Pharmacological data of cannabidiol- and cannabigerol-type phytocannabinoids acting on cannabinoid CB1, CB2 and CB1/CB2 heteromer receptors. Pharmacol Res 2020; 159: 104940. https://doi.org/10.1016/j.phrs.2020.104940.

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

11. Etemad L, Karimi G, Alavi MS, Roohbakhsh A. Pharmacological effects of cannabidiol by transient receptor potential channels. Life Sci 2022; 300: 120582. https://doi.org/10.1016/j.lfs.2022.120582.

12. Morales P, Isawi I, Reggio PH. Towards a better understanding of the cannabinoid-related orphan receptors GPR3, GPR6, and GPR12. Drug Metab Rev 2018; 50(1): 74-93. https://doi.org/10.1080/03602532.2018.1428616.

13. Rankin L, Fowler CJ. The basal pharmacology of palmitoylethanolamide. Int J Mol Sci 2020; 21(21): 7942. https://doi.org/10.3390/ijms21217942.

14. Cannon AE, Chapman KD. Lipid signaling through G proteins. Trends Plant Sci 2021; 26(7): 720-728. https://doi.org/10.1016/j.tplants.2020.12.012.

15. Tsuboi K, Tai T, Yamashita R, Ali H, Watanabe T, Uyama T, et al. Involvement of acid ceramidase in the degradation of bioactive N-acylethanolamines. Biochem Biophys Acta Mol Cell Biol Lipids 2021; 1866(9): 158972. https://doi.org/10.1016/j.bbalip.2021.158972.

16. Graczyk M, Lewandowska AA, Dzierzanowski T. The therapeutic potential of cannabis in counteracting oxidative stress and inflammation. Molecules 2021; 26(15): 4551. https://doi.org/10.3390/molecules26154551.

17. Foster SR, Hauser AS, Vedel L, Strachan RT, Huang XP, Gavin AC, et al. Discovery of human signaling systems: Pairing peptides to g proteincoupled receptors. Cell 2019; 179(4): 895-908.e21. https://doi.org/10.1016/j.cell.2019.10.010.

18. Tsuboi K, Uyama T, Okamoto Y, Ueda N. Endocannabinoids and related N-acylethanolamines: Biological activities and metabolism. Inflame Regen 2018; 38: 28. https://doi.org/10.1186/s41232-018-0086-5.

19. Ng T, Gupta V. Tetrahydrocannabinol (THC). In: StatPearls. Treasure Island (FL): StatPearls Publishing. 2022. https://www.ncbi.nlm.nih.gov/books/NBK563174.

20. Petrosino S, Di Marzo V. The pharmacology of palmitoylethanolamide and first data on the therapeutic efficacy of some of its new formulations. Br J Pharmacol 2017; 174(11): 1349-1365. https://doi.org/10.1111/bph.13580.

21. Larsen C, Shahinas J. Dosage, efficacy and safety of cannabidiol administration in adults: A systematic review of human trials. J Clin Med Res 2020; 12(3): 129-141. https://doi.org/10.14740/jocmr4090.

22. Nichols JM, Kaplan BLF. Immune responses regulated by cannabidiol. Cannabis Cannabinoid Res 2020; 5(1): 12-31. https://doi.org/10.1089/can.2018.0073.

23. Mlost J, W^sik A, Starowicz K. Role of endocannabinoid system in dopamine signaling within the reward circuits affected by chronic pain. Pharmacol Res 2019; 143: 40-47. https://doi.org/10.1016/j.phrs.2019.02.029.

24. Ruiz de Azua I, Lutz B. Multiple endocannabinoid-mediated mechanisms in the regulation of energy homeostasis in brain and peripheral tissues. Cell Mol Life Sci 2019;76(7): 1341-1363. https://doi.org/10.1007/s00018-018-2994-6.

25. Clayton P, Subah S, Venkatesh R, Hill M, Bogoda N. Palmitoylethanolamide: A potential alternative to cannabidiol. J Diet Suppl 2021: 1-26. https://doi.org/10.1080/19390211.2021.2005733.

26. Piomelli D, Scalvini L, Fotio Y, Lodola A, Spadoni G, Tarzia G, et al. N-acylethanolamine acid amidase (NAAA): Structure, function, and inhibition. J Med Chem 2020; 63(14): 7475-7490. https://doi.org/10.1021/acs.jmedchem.0c00191.

27. Zou S, Kumar U. Cannabinoid receptors and the endocannabinoid system: Signaling and function in the central nervous system. Int J Mol Sci 2018; 19(3): 833. https://doi.org/10.3390/ijms19030833.

28. Lutz B. Neurobiology of cannabinoid receptor signaling. Dialogues Clin Neurosci 2020; 22(3): 207-222. https://doi.org/10.31887/dcns.2020.223/blutz.

29. Saldaña-Shumaker SL., Grenning AJ., Cunningham CW. Modern approaches to the development of synthetic cannabinoid receptor probes. Pharmacol Biochem Behav 2021; 203: 173119. https://doi.org/10.1016/j.pbb.2021.173119.

30. Silver RJ. The endocannabinoid system of animals. Animals (Basel) 2019; 9(9): 686. https://doi.org/10.3390/ani9090686.

31. Navarrete F, García-Gutiérrez MS, Gasparyan A, Navarro D, Manzanares J. CB2 Receptor involvement in the treatment of substance use disorders. Biomolecules 2021; 11(11): 1556. https://doi.org/10.3390/biom11111556.

32. Giorgi V, Marotto D, Batticciotto A, Atzeni F, Bongiovanni S, Sarzi-Puttini P. Cannabis and autoimmunity: Possible mechanisms of action.

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

Immunotargets Ther 2021; 10: 261-271.

https://doi.org/10.2147/itt.s267905.

33. Raj V, Park JG, Cho KH, Choi P, Kim T, Ham J, et al. Assessment of antiviral potencies of cannabinoids against SARS-CoV-2 using computational and in vitro approaches. Int J Biol Macromol 2021; 168: 474-485. https://doi.org/10.1016/j.ijbiomac.2020.12.020.

34. Zúñiga-Romero Á, Rivera-Plata Q, Arrieta J, Flores-Murrieta FJ, Rodríguez-Silverio J, Reyes-García JG, et al. GPR55 and GPR119 receptors contribute to the processing of neuropathic pain in rats. Pharmaceuticals (Basel) 2022; 15(1): 67. https://doi.org/10.3390/ph15010067.

35. Im D-S. GPR119 and GPR55 as receptors for fatty acid ethanolamides, oleoylethanolamide and palmitoylethanolamide. Int J Mol Sci 2021; 22(3): 1034. https://doi.org/10.3390/ijms22031034.

36. Chiocchetti R, Galiazzo G, Tagliavia C, Stanzani A, Giancola F, Menchetti M, et al. Cellular distribution of canonical and putative cannabinoid receptors in canine cervical dorsal root ganglia. Front Vet Sci 2019; 6: 313. https://doi.org/10.3389/fvets.2019.00313.

37. Armin S, Muenster S, Abood M, Benamar K. GPR55 in the brain and chronic neuropathic pain. Behav Brain Res 2021; 406: 113248. https://doi.org/10.1016/j.bbr.2021.113248.

38. Carey LM, Gutierrez T, Deng L, Lee WH, Mackie K, Hohmann AG. Inflammatory and neuropathic nociception is preserved in GPR55 knockout mice. Sci Rep 2017; 7(1): 944. https://doi.org/10.1038/s41598-017-01062-2.

39. García-Gutiérrez MS, Navarrete F, Gasparyan A, Austrich-Olivares A, Sala F, Manzanares J. Cannabidiol: A potential new alternative for the treatment of anxiety, depression, and psychotic disorders. Biomolecules 2020; 10(11): 1575. https://doi.org/10.3390/biom10111575.

40. Muller C, Morales P, Reggio PH. Cannabinoid ligands targeting TRP channels. Front Mol Neurosci 2019; 11: 487. https://doi.org/10.3389/fnmol.2018.00487.

41. Muralikumar S, Vetrivel U, Narayanasamy A, N Das U. Probing the intermolecular interactions of PPAR-y-LBD with polyunsaturated fatty acids and their anti-inflammatory metabolites to infer most potential binding moieties. Lipids Health Dis 2017; 16(1): 17. https://doi.org/10.1186/s12944-016-0404-3.

42. De Gregorio D, Manchia M, Carpiniello B, Valtorta F, Nobile M, Gobbi G, et al. Role of palmitoylethanolamide (PEA) in depression: Translational evidence: Special Section on "Translational and Neuroscience Studies in Affective Disorders". J Affect Disord 2018; 255: 195-200. https://doi.org/10.1016/j.jad.2018.10.117.

43. Li J, Guo C, Wu J. 15-Deoxy-ñ-1214-prostaglandin J2 (15d-PGJ2), an endogenous ligand of PPAR-y: Function and mechanism. PPAR Res 2019; 2019: 7242030. https://doi.org/10.1155/2019/7242030.

44. Banno A, Reddy AT, Lakshmi SP, Reddy RC. PPARs: Key regulators of airway inflammation and potential therapeutic targets in asthma. Nucl Receptor Res 2018; 5: 101306. https://doi.org/10.11131/2018/101306.

45. Nobs SP, Kopf M. PPAR-y in innate and adaptive lung immunity. J Leukoc Biol 2018;104(4): 737-741. https://doi.org/10.1002/jlb.3mr0118-034r.

46. Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol 2021;18(12): 809-823. https://doi.org/10.1038/s41569-021-00569-6.

47. Grabacka M, Pierzchalska M, Ptonka PM, Pierzchalski P. The role of PPAR alpha in the modulation of innate immunity. Int J Mol Sci 2021; 22(19): 10545. https://doi.org/10.3390/ijms221910545.

48. Sarre C, Contreras-Lopez R, Nernpermpisooth N, Barrere C, Bahraoui S, Terraza C, et al. Correction: PPARß/6 priming enhances the anti-apoptotic and therapeutic properties of mesenchymal stromal cells in myocardial ischemia-reperfusion injury. Stem Cell Res Ther 2022; 13(1): 338. https://doi.org/10.1186/s13287-022-03086-6.

49. Chen M, Jing D, Ye R, Yi J, Zhao Z. PPARß/6 accelerates bone regeneration in diabetic mellitus by enhancing AMPK/mTOR pathway-

mediated autophagy. Stem Cell Res Ther 2021;12(1): 566. https://doi.org/10.1186/s13287-021-02628-8.

50. Magadum A, Engel FB. PPARß/6: Linking metabolism to regeneration. Int J Mol Sci 2018; 19(7): 2013. https://doi.org/10.3390/ijms19072013.

51. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395(10223): 497-506. https://doi.org/10.1016/S0140-6736(20)30183-5.

52. Storch U, Forst AL, Pardatscher F, Erdogmus S, Philipp M, Gregoritza M, et al. Dynamic NHERF interaction with TRPC4/5 proteins is required for channel gating by diacylglycerol. Proc Natl Acad Sci USA 2017; 114: E37-E46. https://doi.org/10.1073/pnas.1612263114.

53. Rosenbaum T, Morales-Lázaro SL, Islas LD. TRP channels: A journey towards a molecular understanding of pain. Nat Rev Neurosci 2022; 23(10): 596-610. https://doi.org/10.1038/s41583-022-00611-7.

54. Reyes-García J, Carbajal-García A, Montaño LM. Transient receptor potential cation channel subfamily V (TRPV) and its importance in asthma. Eur J Pharmacol 2022;915: 174692. https://doi.org/10.1016/j.ejphar.2021.174692.

55. Lezama-García K, Mota-Rojas D, Pereira AMF, Martínez-Burnes J, Ghezzi M, Domínguez A, et al. Transient receptor potential (TRP) and thermoregulation in animals: Structural biology and neurophysiological aspects. Animals (Basel) 2022; 12(1): 106. https://doi.org/10.3390/ani12010106.

56. Hillard CJ. Circulating endocannabinoids: From whence do they come and where are they going? Neuropsychopharmacology 2018; 43(1): 155-172. https://doi.org/10.1038/npp.2017.130.

57. Corroon J, Felice JF. The endocannabinoid system and its modulation by cannabidiol (CBD). Altern Ther Health Med 2019;25(S2): 6-14. https://pubmed.ncbi.nlm.nih.gov/31202198.

58. Koyama S, Purk A, Kaur M, Soini HA, Novotny MV, Davis K, et al. Beta-caryophyllene enhances wound healing through multiple routes. PLoS One 2019; 14(12): e0216104. https://doi.org/10.1371/journal.pone.0216104.

59. Du Y, Ren P, Wang Q, Jiang SK, Zhang M, Li JY, et al. Cannabinoid 2 receptor attenuates inflammation during skin wound healing by inhibiting M1 macrophages rather than activating M2 macrophages. J Inflamm (Lond) 2018; 15: 25. https://doi.org/10.1186/s12950-018-0201-z.

60. Marsella R, Ahrens K, Sanford R, Trujillo A, Massre D, Soeberdt M, et al. Double blinded, vehicle controlled, crossover study on the efficacy of a topical endocannabinoid membrane transporter inhibitor in atopic beagles. Arch Dermatol Res 2019; 311(10): 795-800. https://doi.org/10.1007/s00403-019-01963-4.

61. Espinosa-Riquer ZP, Ibarra-Sánchez A, Vibhushan S, Bratti M, Charles N, Blank U, et al. TLR4 receptor induces 2-AG-dependent tolerance to lipopolysaccharide and trafficking of CB2 receptor in mast cells. J Immunol 2019; 202(8): 2360-2371. https://doi.org/10.4049/jimmunol.1800997.

62. Motwani MP, Bennett F, Norris PC, Maini AA, George MJ, Newson J, et al. Potent anti-inflammatory and pro-resolving effects of anabasum in a human model of self-resolving acute inflammation. Clin Pharmacol Ther 2018; 104(4): 675-686. https://doi.org/10.1002/cpt.980.

63. Novgorodtseva TP, Gvozdenk TA, Vitkina TI, Denisenko YK, Antonyuk MV, Knyshova VV. Regulatory signal mechanisms of systemic inflammation in respiratory pathology. Russ Open Med J 2019; 8: e0106. https://doi.org/10.15275/rusomj.2019.0106.

64. Aghazadeh Tabrizi M, Baraldi PG, Baraldi S, Gessi S, Merighi S, Borea PA. Medicinal chemistry, pharmacology, and clinical implications of trpv1 receptor antagonists. Med Res Rev 2017; 37(4): 936-983. https://doi.org/10.1002/med.21427.

65. Kytikova OYu, Novgorodtseva TP, Denisenko YuK, Naumov DE, Gvozdenko TA, Perelman YuM. Thermosensory transient receptor potential ion channels and asthma. Biomedicine 2021; 9 (7): 816. https://doi.org/10.3390/biomedicines9070816.

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

66. Tahamtan A, Samieipoor Y, Nayeri FS, Rahbarimanesh AA, Izadi A, Rashidi-Nezhad A, et al. Effects of cannabinoid receptor type 2 in respiratory syncytial virus infection in human subjects and mice. Virulence 2018; 9: 217-230. https://doi.org/10.1080/21505594.2017.1389369.

67. Freitas HR, Isaac AR, Malcher-Lopes R, Diaz BL, Trevenzoli IH, De Melo Reis RA. Polyunsaturated fatty acids and endocannabinoids in health and disease. Nutr Neurosci 2018; 21(10): 695-714. https://doi.org/10.1080/1028415x.2017.1347373.

68. Amato G, Khan NS, Maitra R. A patent update on cannabinoid receptor 1 antagonists (2015-2018). Expert Opin Ther Pat 2019; 29(4): 261-269. https://doi.org/10.1080/13543776.2019.1597851.

69. El Biali M, Broers B, Besson M, Demeules J. Cannabinoids and COVID-19. Med Cannabis Cannabinoids 2020; 3(2): 111-115. https://doi.org/10.1159/000510799.

70. Sommano SR, Chittasupho C, Ruksiriwanich W, Jantrawut P. The cannabis terpenes. Molecules 2020; 25(24): 5792. https://doi.org/10.3390/molecules25245792.

71. Sivesind TE, Maghfour J, Rietcheck H, Kamel K, Malik AS, Dellavalle RP. Cannabinoids for the treatment of dermatologic conditions. JID Innov 2022; 2(2): 100095. https://doi.org/10.1016/j.xjidi.2022.100095.

72. Becker W, Alrafas HR, Wilson K, Miranda K, Culpepper C, Chatzistamou I, et al. Activation of cannabinoid receptor 2 prevents colitis-associated colon cancer through myeloid cell de-activation upstream of IL-22 production. iScience 2020; 23(9): 101504. https://doi.org/10.1016/Msci.2020.101504.

73. Tartakover Matalon S, Azar S, Meiri D, Hadar R, Nemirovski A, Abu Jabal N, et al. Endocannabinoid levels in ulcerative colitis patients correlate with clinical parameters and are affected by cannabis consumption. Front Endocrinol (Lausanne) 2021; 12: 685289. https://doi.org/10.3389/fendo.2021.685289.

74. Martins AM, Gomes AL, Vilas Boas I, Marto J, Ribeiro HM. Cannabis-based products for the treatment of skin inflammatory diseases: A timely review. Pharmaceuticals (Basel) 2022; 15(2): 210. https://doi.org/10.3390/ph15020210.

75. Peng J, Fan M, An C, Ni F, Huang W, Luo J. A narrative review of molecular mechanism and therapeutic effect of cannabidiol (CBD). Basic Clin Pharmacol Toxicol 2022; 130(4): 439-456. https://doi.org/10.1111/bcpt.13710.

76. Wang B, Kovalchuk A, Li D, Rodriguez-Juarez R, Ilnytskyy Y, Kovalchuk I. In search of preventative strategies: Novel antiinflammatory high-CBD Cannabis sativa extracts modulate ACE2 expression in COVID-19 gateway tissues. Aging (Albany NY) 2020; 12(22): 2242522444. https://doi.org/10.18632/aging.202225.

77. Rossi F, Tortora C, Argenziano M, Di Paola A, Punzo F. Cannabinoid receptor type 2: A possible target in SARS-CoV-2 (CoV-19) infection? Int J Mol Sci 2020; 21(11): 3809. https://doi.org/10.3390/ijms21113809.

78. Esposito G, Pesce M, Seguella L, Sanseverino W, Lu J, Corpetti C, Sarnelli G. The potential of cannabidiol in the COVID-19 pandemic. Br J Pharmacol 2020; 177(21): 4967-4970. https://doi.org/10.1111/bph.15157.

79. Rinne P, Guillamat-Prats R, Rami M, Bindila L, Ring L, Lyytikäinen LP, et al. Palmitoylethanolamide promotes a proresolving macrophage phenotype and attenuates atherosclerotic plaque formation. Arterioscler Thromb Vasc Biol 2018; 38(11): 2562-2575. https://doi.org/10.1161/atvbaha.118.311185.

80. Hil JD, Zuluaga-Ramirez V, Gajghate S, Winfield M, Sriram U, Rom S, et al. Activation of GPR55 induces neuroprotection of hippocampal neurogenesis and immune responses of neural stem cells following chronic, systemic inflammation. Brain Behav Immun 2019; 76: 165181. https://doi.org/10.1016/j.bbi.2018.11.017.

81. Rahman SMK, Uyama T, Hussain Z, Ueda N. Roles of Endocannabinoids and endocannabinoid-like molecules in energy homeostasis and

metabolic regulation: A nutritional perspective. Annu Rev Nutr 2021; 41: 177-202. https://doi.org/10.1146/annurev-nutr-043020-090216.

82. Yang X, Xu L, Zhou J, Ge Y, Wu S, Huang J, et al. Integration of phospholipid-complex nanocarrier assembly with endogenous N-oleoylethanolamine for efficient stroke therapy. J Nanobiotechnology 2019; 17(1): 8. https://doi.org/10.1186/s12951-019-0442-x.

83. Yang X, Wu S. N-oleoylethanolamine - phosphatidylcholine complex loaded, DSPE-PEG integrated liposomes for efficient stroke. Drug Deliv 2021; 28(1): 2525-2533. https://doi.org/10.1080/10717544.2021.2008058.

84. Hutch CR, Trakimas DR, Roelofs K, Pressler J, Sorrell J, Cota D, et al. OEA signaling pathways and the metabolic benefits of vertical sleeve gastrectomy. Ann Surg 2020;271(3): 509-518. https://doi.org/10.1097/sla.0000000000003093.

85. Sihag J, Jones PJH. Oleoylethanolamide: The role of a bioactive lipid amide in modulating eating behaviour. Obes Rev 2018; 19(2): 178-197. https://doi.org/10.1111/obr.12630.

86. Li Y, Chen X, Nie Y, Tian Y, Xiao X, Yang F. Endocannabinoid activation of the TRPV1 ion channel is distinct from activation by capsaicin. J Biol Chem 2021; 297(3): 101022. https://doi.org/10.1016/j.jbc.2021.101022.

87. Ganley OH, Graessle OE, Robinson HJ. Anti-inflammatory activity of compounds obtained from egg yolk, peanut oil, and soybean lecithin. J Lab Clin Med 1958; 51(5): 709-714 https://pubmed.ncbi.nlm.nih.gov/13539486.

88. Cristiano C, Pirozzi C, Coretti L, Cavaliere G, Lama A, Russo R, et al. Palmitoylethanolamide counteracts autistic-like behaviours in BTBR T+tf/J mice: Contribution of central and peripheral mechanisms. Brain Behav Immun 2018; 74: 166-175. https://doi.org/10.1016/j.bbi.2018.09.003.

89. Giammusso B, Di Mauro R, Bernardini R. The efficacy of an association of palmitoylethanolamide and alpha-lipoic acid in patients with chronic prostatitis/chronic pelvic pain syndrome: A randomized clinical trial. Arch Ital Urol Androl 2017; 89(1): 17-21. https://doi.org/10.4081/aiua.2017.1.17.

90. Beggiato S, Tomasini MC, Ferraro L. Palmitoylethanolamide (PEA) as a potential therapeutic agent in Alzheimer's disease. Front Pharmacol 2019; 10: 821. https://doi.org/10.3389/fphar.2019.00821.

91. Briskey D, Mallard AR, Rao A. Increased absorption of palmitoylethanolamide using a novel dispersion technology system (LipiSperse). Nutraceuticals Food Sci 2020; 5(2): 3. https://doi.org/10.36648/nutraceuticals.5.2.3.

92. LoVerme J, Russo R, La Rana G, Fu J, Farthing J, Mattace-Raso G, et al. Rapid broad-spectrum analgesia through activation of peroxisome proliferator-activated receptor-a. J Pharmacol Exp Ther 2006; 319(3); 1051-1061. https://doi.org/10.1124/jpet.106.111385.

93. Malamas MS, Farah SI, Lamani M, Pelekoudas DN, Perry NT, Rajarshi G, et al. Design and synthesis of cyanamides as potent and selective N-acylethanolamine acid amidase inhibitors. Bioorg Med Chem 2020; 28(1): 115195. https://doi.org/10.1016/j.bmc.2019.115195.

94. Couch DG, Cook H, Ortori C, Barrett D, Lun J.N, O'Sullivan SE. Palmitoylethanolamide and cannabidiol prevent inflammation-induced hyperpermeability of the human gut in vitro and in vivo - A randomized, placebo-controlled, double-blind controlled trial. Inflamm Bowel Dis 2019; 25(6): 1006-1018. https://doi.org/10.1093/ibd/izz017.

95. Saliba SW, Jauch H, Gargouri B, Keil A, Hurrle T, Volz N, et al. Anti-neuroinflammatory effects of GPR55 antagonists in LPS-activated primary microglial cells. J Neuroinflammation 2018; 15(1): 322. https://doi.org/10.1186/s12974-018-1362-7.

96. Vaia M, Petrosino S, De Filippis D, Negro L, Guarino A, Carnuccio R, et al. Palmitoylethanolamide reduces inflammation and itch in a mouse model of contact allergic dermatitis. Eur J Pharmacol 2016; 791: 669674. https://doi.org/10.1016/j.ejphar.2016.10.005.

97. Borrelli F, Romano B, Petrosino S, Pagano E, Capasso R, Coppola D, et al. Palmitoylethanolamide, a naturally occurring lipid, is an orally

ISSN 2304-3415, Russian Open Medical Journal

2023. Volume 12. Issue 1 (March). Article CID e0107 DOI: 10.15275/rusomj.2023.0107_

Pharmacology

10 of 10

effective intestinal anti-inflammatory agent. Br J Pharmacol 2015; 172(1): 142-158. https://doi.org/10.1111/bph.12907.

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

98. Marichal-Cancino B, González-Hernández A, MaassenVanDenBrink A, Ramírez-San JE, Villalón CM. Potential mechanisms involved in palmitoylethanolamide-induced vasodepressor effects in rats. J Vasc Res 2020; 57(3): 152-163. https://doi.org/10.1159/000506158.

99. Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Hosseinzadeh-Attar MJ, Shakeri A, Asghari-Jafarabadi M, et al. Oleoylethanolamide supplementation in obese patients newly diagnosed with nonalcoholic fatty liver disease: Effects on metabolic parameters, anthropometric indices, and expression of PPAR-a, UCP1, and UCP2 genes. Pharmacol Res 2020; 156: 104770. https://doi.org/10.1016/j.phrs.2020.104770.

100.izgordü H, Sezer CV, Baygelebi K, Baloglu M, Kutlu HM. Cytotoxic Impact of N-oleoylethanolamine on bone cancer cells. Anticancer Agents Med Chem 2022; 22(6): 1119-1123. https://doi.org/10.2174/1871520621666210617091138.

101.Luo D, Zhang Y, Yuan X, Pan Y, Yang L, Zhao Y, et al. Oleoylethanolamide inhibits glial activation via modulating PPAR-a and promotes motor function recovery after brain ischemia. Pharmacol Res 2019; 141: 530-540. https://doi.org/10.1016/j.phrs.2019.01.027.

102.Zhou H, Yang WS, Li Y, Ren T, Peng L, Guo H, et al. Oleoylethanolamide attenuates apoptosis by inhibiting the TLR4/NF-KB and ERK1/2 signaling pathways in mice with acute ischemic stroke. Naunyn Schmiedebergs Arch Pharmacol 2017;390(1): 77-84. https://doi.org/10.1007/s00210-016-1309-4.

Oksana Yu. Kytikova - MD, DSc, Senior Researcher, Laboratory of Rehabilitative Treatment, Vladivostok Branch of Far Eastern Scientific Center for Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia. http://orcid.org/0000-0001-5018-0271.

Yulia K. Denisenko - DSc, Head of the Laboratory of Biomedical Research, Vladivostok Branch of Far Eastern Scientific Center for Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia. https://orcid.org/0000-0003-4130-8899.

Tatyana P. Novgorodtseva - DSc, Professor, Principal Researcher, Laboratory of Biomedical Research, Vladivostok Branch of Far Eastern Scientific Center for Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia. https://orcid.org/0000-0002-6058-201X.

Ivan S. Kovalenko - PhD student, Laboratory of Biomedical Research, Vladivostok Branch of Far Eastern Scientific Center for Physiology and Pathology of Respiration - Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia, http://orcid.org/0000-0002-7404-7501.

Authors:

i Надоели баннеры? Вы всегда можете отключить рекламу.