Protective Effects of Thymol on Inflammatory Bowel Disease: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Yao Liu.

Inflammatory bowel disease (IBD) is a gastrointestinal disease that involves chronic mucosal or submucosal lesions that affect tissue integrity. Although IBD is not life-threatening, it sometimes causes severe complications, such as colon cancer. The exact etiology of IBD remains unclear, but several risk factors, such as pathogen infection, stress, diet, age, and genetics, have been involved in the occurrence and aggravation of IBD. Immune system malfunction with the over-production of inflammatory cytokines and associated oxidative stress are the hallmarks of IBD. Dietary intervention and medical treatment suppressing abnormal inflammation and oxidative stress are recommended as potential therapies. Thymol, a natural monoterpene phenol that is mostly found in thyme, exhibits multiple biological functions as a potential adjuvant for IBD. The purpose of this review is to summarize current findings on the protective effect of thymol on intestinal health in the context of specific animal models of IBD, describe the role of thymol in the modulation of inflammation, oxidative stress, and gut microbiota against gastrointestinal disease, and discuss the potential mechanism for its pharmacological activity.

  • gastrointestinal disease
  • gut health
  • natural anti-oxidant

1. Introduction

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract (GIT) [1]. It is not fatal, but imposes a burden on global healthcare and greatly reduces the quality of life of patients. From 1990 to 2017, the number of IBD patients increased from 37 million to more than 68 million, and the global prevalence of IBD increased by 85.1% [2]. The United States has the highest age-standardized prevalence rate in the world, accounting for nearly a quarter of all global IBD patients in 2017. Among European countries, the United Kingdom has the highest age-standardized prevalence. The prevalence of IBD in the United States ranges from 252 to 439 cases per 100,000 people [3]. Compared with western countries, the incidence of IBD in Asia is relatively low, but it has risen from 0.54 to 3.44 per 100,000 individuals across eight Asian countries [4,5][4][5]. The prevalence and incidence of IBD are continuing to increase worldwide in different races and countries [6].
IBD includes Crohn’s disease (CD) and ulcerative colitis (UC). UC involves long-lasting inflammation and ulcers along the superficial lining of the large intestine (rectum and colon), while CD is characterized by the discontinuous transmural inflammation in deeper layers of most regions of the GIT [7,8][7][8]. The exact cause of IBD remains unknown, but malfunction in the immune system is its hallmark. In IBD patients, an abnormal immune response to pathogen infection or stress causes chronic inflammation in the GIT contributing to the incidence of the disease [9]. Some genetic factors involved in an inappropriate immune response are a reason for IBD [10]. Oxidative stress (OS) with excessive accumulation of reactive oxygen species (ROS) has been reported to correlate with intestinal chronic inflammation and incidence of IBD [11]. Moreover, alteration in the gut microbiota has been associated with the pathogenesis of IBD [12]. Unfortunately, there is currently no cure for IBD. Anti-inflammation medicines are used to relieve inflammation and even surgery is required to remove damaged parts of the GIT when patients suffer. Recently, advances in dietary regulation of inflammation, OS, and gut microbiota have resulted in dietary intervention against IBD incidence being recommended as a long-term prevention and therapy technique [13]. Currently, more research is focusing on natural plant extracts, such as phenol compounds, for the prevention and relief of IBD [14].
Thymol (2-isopropyl-5-methylphenol), a natural monoterpene phenol compound, is the major component of the essential oils extracted from plants of thyme species, such as Thymus vulgarisCoridothymus capitatus, and Origanum vulgare [15]. Thymol exhibits multiple biological and pharmacological properties, including anti-inflammation, anti-oxidation, anti-bacteria, anti-fungal, and anti-tumor potential [16]. A high dose of thymol up to 500 mg/kg diet has been shown to have no toxicity [17].

2. Pharmacokinetics and Pharmacological Properties of Thymol

Thymol is the secondary metabolite produced by the aromatization of γ-terpinene to p-cymene, followed by the hydroxylation of p-cymene in plants [19][18] (Figure 1), including Thymus zygis [20][19]Thymbra capitata [21][20]Thymus vulgaris [22][21]Satureja thymbra [23][22]Nigella sativa seeds [24][23], and Monarda didyma [25][24]. Pure thymol has low solubility in water, high volatility, and a strong bitter/irritating taste [26][25]; thus, it is usually encapsulated in electrospun nanofibers to enhance its water solubility and high temperature stability [27][26]. Emulsification is also used for thymol processing. Both encapsulation and emulsification have been shown to improve the anti-oxidant activity of thymol [28][27].
Figure 1. Thymol biosynthesis in plants. Thymol is synthesized by the hydroxylation of p-cymene, which originates from the aromatization of γ-terpinene.
Thymol administered orally can be rapidly absorbed in the stomach and small intestines, and then transported to various organs via the circulation system in animals [29][28]. Thymol can be metabolized to thymol sulfate and thymol glucuronide by sulfation and glucuronidation in the intestines, liver, kidney, and other organs, respectively [30][29]. Thymol and its metabolites reach the maximum concentration in the blood 30 min after oral administration and then are slowly eliminated in about 24 h [26][25]. They also present in the lungs, kidneys, mucosa of the small and large intestines, and other organs, suggesting that they may function directly in these organs. It is difficult to determine which compound is the active form of thymol, because thymol metabolites can be deconjugated to thymol locally and export its pharmacological activity in this way [31][30]. Recently, scientists have focused on the pharmacological activities of thymol. Thymol-containing plants have long been used in traditional Chinese medicine due to its pharmacological properties. Thymol acts as a potent inhibitor of the release of inflammatory cytokines, such as interleukin (IL)-6, IL-1β, and IL-8 [32][31]. Thymol enhances anti-oxidative capacity to alleviate OS in different tissues [33][32]. As a spectrum anti-bacterial agent, thymol reduces the activities of microorganisms belonging to the Enterobacteriaceae, Streptococcus, and Saccharomycetaceae families, involving membrane rupture, inhibition of biofilm formation, and other pathways [34,35][33][34]. Thymol shows anti-viral potential to inhibit virus colonization, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-298), by docking with the S1 receptor-binding domain of spike glycoprotein [36][35]. Thymol also exhibits other pharmacological functions, such as analgesia, anti-malarial, and anti-fungal properties [37,38,39,40,41][36][37][38][39][40]

3. Thymol Protects Intestinal Barrier Function against IBD

The intestinal barrier, mainly composed of intestinal epithelial cells (IECs) and immune cells, maintains the balance between the luminal contents and mucosa. The disturbance of this balance has been associated with gastrointestinal diseases, such as IBD [42][41]. Although the exact pathogenesis of IBD remains unclear, a “leaky gut” with impaired intestinal barrier function is the main feature. The intestinal barrier is the first line of defense against pathogen infection, and injury to the intestinal barrier aggravates the disease. Thus, understanding how thymol protects the intestinal barrier is important for relieving IBD. Thymol has exhibited a protective function for the intestinal barrier in both in vivo and in vitro studies, as illustrated in Table 1, and its specific mechanism is shown in Figure 2. Thymol attenuates weaning stress-induced diarrheal and intestinal barrier dysfunction in weanling pigs by reducing the serum diamine oxidase level, an indicator of intestinal integrity, and increasing the expression of the tight junction protein zonula occludens-1 (ZO-1) and occludins [43][42]. Thymol alleviates dextran sulfate sodium (DSS)-induced intestinal damage and increases tight junction claudin-3 expression [44][43]. Increased plasma endotoxin and D-lactic acid levels are markers of increased intestinal permeability. The latest study found that dietary thymol reduced the plasma endotoxin and D-lactic acid concentrations on days 7 and 14 post-weaning [45][44]. The intestinal mucus layer is the first line of defense maintaining bacterial symbiosis with the host and preventing bacterial penetration into epithelial cells [46][45]. Thymol increases mucus secretion to relieve ethanol-induced ulcer mucosal damage in rats [47][46]. In IPEC-J2 cells, thymol alleviates lipopolysaccharide (LPS)-induced decrease in trans-epithelial electrical resistance (TEER), indicating an increase in the integrity of the single cell layer [48][47]. In Caco-2 cells, thymol increases the integrity of the tight junction and up-regulates cyclooxygenase-1 (COX1) activity to maintain GIT homeostasis, which is beneficial for intestinal health [49][48]. In addition, thymol changes the expressions of 120 and 59 genes in the oxyntic and pyloric mucosa, respectively, which are associated with gastric epithelium proliferation and maturation activities in weaned pigs [29][28].
Figure 2. Action of thymol to protect intestinal barrier function. Pathogen infection in the intestinal lumen reduces mucus secretion and the expressions of tight junction proteins, and increases intestinal permeability, resulting in a “leaky gut”, which increases the risk of intestinal disease. Thymol has been shown to defend against pathogen invasion, promote mucus secretion, and enhance intestinal barrier integrity.
Table 1.
 Protective function of thymol on the intestinal barrier.

References

  1. Duerr, R.H.; Taylor, K.D.; Brant, S.R.; Rioux, J.D.; Silverberg, M.S.; Daly, M.J.; Steinhart, A.H.; Abraham, C.; Regueiro, M.; Griffiths, A.; et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006, 314, 1461–1463.
  2. Sudabeh Alatab, S.G.S. Kevin Ikuta, Homayoon Vahedi, Catherine Bisignano. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 5, 17–30.
  3. Kaser, A.; Zeissig, S.; Blumberg, R.S. Inflammatory bowel disease. Annu. Rev. Immunol. 2010, 28, 573–621.
  4. Loftus, E.V., Jr. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology 2004, 126, 1504–1517.
  5. Ng, S.C. Epidemiology of inflammatory bowel disease: Focus on Asia. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 363–372.
  6. Kaplan, G.G.; Ng, S.C. Understanding and Preventing the Global Increase of Inflammatory Bowel Disease. Gastroenterology 2017, 152, 313–321.
  7. Corridoni, D.; Arseneau, K.O.; Cominelli, F. Inflammatory bowel disease. Immunol. Lett. 2014, 161, 231–235.
  8. Walfish, A.; Sachar, D. Phenotype classification in IBD: Is there an impact on therapy? Inflamm. Bowel Dis. 2007, 13, 1573–1575.
  9. Geremia, A.; Biancheri, P.; Allan, P.; Corazza, G.R.; Di Sabatino, A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun. Rev. 2014, 13, 3–10.
  10. Van Limbergen, J.; Russell, R.K.; Nimmo, E.R.; Satsangi, J. The genetics of inflammatory bowel disease. Am. J. Gastroenterol. 2007, 102, 2820–2831.
  11. Zhao, Y.; Guo, Q.; Zhu, Q.; Tan, R.; Bai, D.; Bu, X.; Lin, B.; Zhao, K.; Pan, C.; Chen, H.; et al. Flavonoid VI-16 protects against DSS-induced colitis by inhibiting Txnip-dependent NLRP3 inflammasome activation in macrophages via reducing oxidative stress. Mucosal Immunol. 2019, 12, 1150–1163.
  12. Matsuoka, K.; Kanai, T. The gut microbiota and inflammatory bowel disease. Semin. Immunopathol. 2015, 37, 47–55.
  13. Lee, D.; Albenberg, L.; Compher, C.; Baldassano, R.; Piccoli, D.; Lewis, J.D.; Wu, G.D. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology 2015, 148, 1087–1106.
  14. Martin, D.A.; Bolling, B.W. A review of the efficacy of dietary polyphenols in experimental models of inflammatory bowel diseases. Food Funct. 2015, 6, 1773–1786.
  15. Hazzit, M.; Baaliouamer, A.; Faleiro, M.L.; Miguel, M.G. Composition of the essential oils of Thymus and Origanum species from Algeria and their antioxidant and antimicrobial activities. J. Agric. Food Chem. 2006, 54, 6314–6321.
  16. Salehi, B.; Mishra, A.P.; Shukla, I.; Sharifi-Rad, M.; Contreras, M.D.M.; Segura-Carretero, A.; Fathi, H.; Nasrabadi, N.N.; Kobarfard, F.; Sharifi-Rad, J. Thymol, thyme, and other plant sources: Health and potential uses. Phytother. Res. 2018, 32, 1688–1706.
  17. Geyikoglu, F.; Yilmaz, E.G.; Erol, H.S.; Koc, K.; Cerig, S.; Ozek, N.S.; Aysin, F. Hepatoprotective Role of Thymol in Drug-Induced Gastric Ulcer Model. Ann. Hepatol. 2018, 17, 980–991.
  18. Poulose, A.J.; Croteau, R. Biosynthesis of aromatic monoterpenes: Conversion of gamma-terpinene to p-cymene and thymol in Thymus vulgaris L. Arch. Biochem. Biophys. 1978, 187, 307–314.
  19. Ocaña, A.; Reglero, G. Effects of Thyme Extract Oils (from Thymus vulgaris, Thymus zygis, and Thymus hyemalis) on Cytokine Production and Gene Expression of oxLDL-Stimulated THP-1-Macrophages. J. Obes. 2012, 2012, 104706.
  20. Machado, M.; Dinis, A.M.; Salgueiro, L.; Cavaleiro, C.; Custódio, J.B.; Sousa Mdo, C. Anti-Giardia activity of phenolic-rich essential oils: Effects of Thymbra capitata, Origanum virens, Thymus zygis subsp. sylvestris, and Lippia graveolens on trophozoites growth, viability, adherence, and ultrastructure. Parasitol. Res. 2010, 106, 1205–1215.
  21. Nikolić, M.; Glamočlija, J.; Ferreira, I.C.F.R.; Calhelha, R.C.; Fernandes, Â.; Marković, T.; Marković, D.; Giweli, A.; Soković, M. Chemical composition, antimicrobial, antioxidant and antitumor activity of Thymus serpyllum L., Thymus algeriensis Boiss. and Reut and Thymus vulgaris L. essential oils. Ind. Crops Prod. 2014, 52, 183–190.
  22. Karousou, R.; Koureas, D.N.; Kokkini, S. Essential oil composition is related to the natural habitats: Coridothymus capitatus and Satureja thymbra in NATURA 2000 sites of Crete. Phytochemistry 2005, 66, 2668–2673.
  23. Maulidiani, M.; Sheikh, B.Y.; Mediani, A.; Wei, L.S.; Ismail, I.S.; Abas, F.; Lajis, N.H. Differentiation of Nigella sativa seeds from four different origins and their bioactivity correlations based on NMR-metabolomics approach. Phytochem. Lett. 2015, 13, 308–318.
  24. Laquale, S.; Avato, P.; Argentieri, M.P.; Bellardi, M.G.; D’Addabbo, T. Nematotoxic activity of essential oils from Monarda species. J. Pest. Sci. 2018, 91, 1115–1125.
  25. Nieddu, M.; Rassu, G.; Boatto, G.; Bosi, P.; Trevisi, P.; Giunchedi, P.; Carta, A.; Gavini, E. Improvement of thymol properties by complexation with cyclodextrins: In vitro and in vivo studies. Carbohydr. Polym. 2014, 102, 393–399.
  26. Celebioglu, A.; Yildiz, Z.I.; Uyar, T. Thymol/cyclodextrin inclusion complex nanofibrous webs: Enhanced water solubility, high thermal stability and antioxidant property of thymol. Food Res. Int. 2018, 106, 280–290.
  27. Sedaghat Doost, A.; Van Camp, J.; Dewettinck, K.; Van der Meeren, P. Production of thymol nanoemulsions stabilized using Quillaja Saponin as a biosurfactant: Antioxidant activity enhancement. Food Chem. 2019, 293, 134–143.
  28. Colombo, M.; Priori, D.; Gandolfi, G.; Boatto, G.; Nieddu, M.; Bosi, P.; Trevisi, P. Effect of free thymol on differential gene expression in gastric mucosa of the young pig. Animal 2014, 8, 786–791.
  29. Pisarčíková, J.; Oceľová, V.; Faix, Š.; Plachá, I.; Calderón, A.I. Identification and quantification of thymol metabolites in plasma, liver and duodenal wall of broiler chickens using UHPLC-ESI-QTOF-MS. Biomed. Chromatogr. 2017, 31, e3881.
  30. Lugarà, R.; Grześkowiak, Ł.; Zentek, J.; Meese, S.; Kreuzer, M.; Giller, K. A High-Energy Diet and Spirulina Supplementation during Pre-Gestation, Gestation, and Lactation do Not Affect the Reproductive and Lactational Performance of Primiparous Sows. Animals 2022, 12, 1171.
  31. Pandur, E.; Micalizzi, G.; Mondello, L.; Horvath, A.; Sipos, K.; Horvath, G. Antioxidant and Anti-Inflammatory Effects of Thyme (Thymus vulgaris L.) Essential Oils Prepared at Different Plant Phenophases on Pseudomonas aeruginosa LPS-Activated THP-1 Macrophages. Antioxidants 2022, 11, 1330.
  32. Ahmed, O.M.; Galaly, S.R.; Mostafa, M.M.A.; Eed, E.M.; Ali, T.M.; Fahmy, A.M.; Zaky, M.Y. Thyme Oil and Thymol Counter Doxorubicin-Induced Hepatotoxicity via Modulation of Inflammation, Apoptosis, and Oxidative Stress. Oxid. Med. Cell. Longev. 2022, 2022, 6702773.
  33. Yin, L.; Liang, C.; Wei, W.; Huang, S.; Ren, Y.; Geng, Y.; Huang, X.; Chen, D.; Guo, H.; Fang, J.; et al. The Antibacterial Activity of Thymol Against Drug-Resistant Streptococcus iniae and Its Protective Effect on Channel Catfish (Ictalurus punctatus). Front. Microbiol. 2022, 13, 914868.
  34. Yao, Z.; Feng, L.; Zhao, Y.; Zhang, X.; Chen, L.; Wang, L.; Zhang, Y.; Sun, Y.; Zhou, T.; Cao, J. Thymol Increases Sensitivity of Clinical Col-R Gram-Negative Bacteria to Colistin. Microbiol. Spectr. 2022, 10, e0018422.
  35. Kulkarni, S.A.; Nagarajan, S.K.; Ramesh, V.; Palaniyandi, V.; Selvam, S.P.; Madhavan, T. Computational evaluation of major components from plant essential oils as potent inhibitors of SARS-CoV-2 spike protein. J. Mol. Struct. 2020, 1221, 128823.
  36. Hassan, H.F.H.; Mansour, A.M.; Salama, S.A.; El-Sayed, E.M. The chemopreventive effect of thymol against dimethylhydrazine and/or high fat diet-induced colon cancer in rats: Relevance to NF-κB. Life Sci. 2021, 274, 119335.
  37. Mendes, S.S.; Bomfim, R.R.; Jesus, H.C.; Alves, P.B.; Blank, A.F.; Estevam, C.S.; Antoniolli, A.R.; Thomazzi, S.M. Evaluation of the analgesic and anti-inflammatory effects of the essential oil of Lippia gracilis leaves. J. Ethnopharmacol. 2010, 129, 391–397.
  38. Mohammadi, A.; Mahjoub, S.; Ghafarzadegan, K.; Nouri, H.R. Immunomodulatory effects of Thymol through modulation of redox status and trace element content in experimental model of asthma. Biomed. Pharmacother. 2018, 105, 856–861.
  39. Raghuvanshi, D.S.; Verma, N.; Singh, S.V.; Khare, S.; Pal, A.; Negi, A.S. Synthesis of thymol-based pyrazolines: An effort to perceive novel potent-antimalarials. Bioorg. Chem. 2019, 88, 102933.
  40. Robledo, N.; Vera, P.; López, L.; Yazdani-Pedram, M.; Tapia, C.; Abugoch, L. Thymol nanoemulsions incorporated in quinoa protein/chitosan edible films; antifungal effect in cherry tomatoes. Food Chem. 2018, 246, 211–219.
  41. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305.
  42. Wei, H.K.; Xue, H.X.; Zhou, Z.X.; Peng, J. A carvacrol-thymol blend decreased intestinal oxidative stress and influenced selected microbes without changing the messenger RNA levels of tight junction proteins in jejunal mucosa of weaning piglets. Animal 2017, 11, 193–201.
  43. Mueller, K.; Blum, N.M.; Mueller, A.S. Examination of the Anti-Inflammatory, Antioxidant, and Xenobiotic-Inducing Potential of Broccoli Extract and Various Essential Oils during a Mild DSS-Induced Colitis in Rats. ISRN Gastroenterol. 2013, 2013, 710856.
  44. Wang, Y.; Yang, Z.; Zhou, Y.; Tan, J.; Sun, H.; Sun, D.; Mu, Y.; Peng, J.; Wei, H. Effects of different amino acid levels and a carvacrol-thymol blend on growth performance and intestinal health of weaned pigs. J. Anim. Sci. Biotechnol. 2022, 13, 22.
  45. Fernández-Tomé, S.; Ortega Moreno, L.; Chaparro, M.; Gisbert, J.P. Gut Microbiota and Dietary Factors as Modulators of the Mucus Layer in Inflammatory Bowel Disease. Int. J. Mol. Sci. 2021, 22, 10224.
  46. Ribeiro, A.R.S.; Diniz, P.B.F.; Pinheiro, M.S.; Albuquerque-Júnior, R.L.C.; Thomazzi, S.M. Gastroprotective effects of thymol on acute and chronic ulcers in rats: The role of prostaglandins, ATP-sensitive K+ channels, and gastric mucus secretion. Chem. Biol. Interact. 2016, 244, 121–128.
  47. Omonijo, F.A.; Liu, S.; Hui, Q.; Zhang, H.; Lahaye, L.; Bodin, J.C.; Gong, J.; Nyachoti, M.; Yang, C. Thymol Improves Barrier Function and Attenuates Inflammatory Responses in Porcine Intestinal Epithelial Cells during Lipopolysaccharide (LPS)-Induced Inflammation. J. Agric. Food Chem. 2019, 67, 615–624.
  48. Putaala, H.; Nurminen, P.; Tiihonen, K. Effects of cinnamaldehyde and thymol on cytotoxicity, tight junction barrier resistance, and cyclooxygenase-1 and -2 expression in Caco-2 cells. J. Anim. Sci. 2017, 26, 274–284.
  49. Yin, D.; Du, E.; Yuan, J.; Gao, J.; Wang, Y.; Aggrey, S.E.; Guo, Y. Supplemental thymol and carvacrol increases ileum Lactobacillus population and reduces effect of necrotic enteritis caused by Clostridium perfringes in chickens. Sci. Rep. 2017, 7, 7334.
  50. Choy, M.C.; Visvanathan, K.; Cruz, P.D. An Overview of the Innate and Adaptive Immune System in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 2–13.
  51. Iliev, I.D.; Funari, V.A.; Taylor, K.D.; Nguyen, Q.; Reyes, C.N.; Strom, S.P.; Brown, J.; Becker, C.A.; Fleshner, P.R.; Dubinsky, M.; et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 2012, 336, 1314–1317.
  52. Roy, U.; Gálvez, E.J.C.; Iljazovic, A.; Lesker, T.R.; Błażejewski, A.J.; Pils, M.C.; Heise, U.; Huber, S.; Flavell, R.A.; Strowig, T. Distinct Microbial Communities Trigger Colitis Development upon Intestinal Barrier Damage via Innate or Adaptive Immune Cells. Cell. Rep. 2017, 21, 994–1008.
  53. Natarajan, K.; Abraham, P.; Kota, R.; Isaac, B. NF-κB-iNOS-COX2-TNF α inflammatory signaling pathway plays an important role in methotrexate induced small intestinal injury in rats. Food. Chem. Toxicol. 2018, 118, 766–783.
  54. Salim, S.Y.; Söderholm, J.D. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 362–381.
  55. Chauhan, A.K.; Kang, S.C. Thymol disrupts the membrane integrity of Salmonella ser. typhimurium in vitro and recovers infected macrophages from oxidative stress in an ex vivo model. Res. Microbiol. 2014, 165, 559–565.
  56. Peng, Y.; Zhang, X.; Zhang, T.; Grace, P.M.; Li, H.; Wang, Y.; Li, H.; Chen, H.; Watkins, L.R.; Hutchinson, M.R.; et al. Lovastatin inhibits Toll-like receptor 4 signaling in microglia by targeting its co-receptor myeloid differentiation protein 2 and attenuates neuropathic pain. Brain Behav. Immun. 2019, 82, 432–444.
  57. Khazdair, M.R.; Ghorani, V.; Alavinezhad, A.; Boskabady, M.H. Pharmacological effects of Zataria multiflora Boiss L. and its constituents focus on their anti-inflammatory, antioxidant, and immunomodulatory effects. Fundam. Clin. Pharmacol. 2018, 32, 26–50.
  58. Gholijani, N.; Gharagozloo, M.; Farjadian, S.; Amirghofran, Z. Modulatory effects of thymol and carvacrol on inflammatory transcription factors in lipopolysaccharide-treated macrophages. J. Immunotoxicol. 2016, 13, 157–164.
  59. Jin, B.R.; Chung, K.S.; Cheon, S.Y.; Lee, M.; Hwang, S.; Noh Hwang, S.; Rhee, K.J.; An, H.J. Rosmarinic acid suppresses colonic inflammation in dextran sulphate sodium (DSS)-induced mice via dual inhibition of NF-κB and STAT3 activation. Sci. Rep. 2017, 7, 46252.
  60. Chamanara, M.; Abdollahi, A.; Rezayat, S.M.; Ghazi-Khansari, M.; Dehpour, A.; Nassireslami, E.; Rashidian, A. Thymol reduces acetic acid-induced inflammatory response through inhibition of NF-kB signaling pathway in rat colon tissue. Inflammopharmacology 2019, 27, 1275–1283.
  61. Liu, D.-M.; Zhou, C.-Y.; Meng, X.-L.; Wang, P.; Li, W. Thymol exerts anti-inflammatory effect in dextran sulfate sodium-induced experimental murine colitis. Trop.J. Pharm. Res. 2018, 17, 1803–1810.
  62. Tahmasebi, P.; Abtahi Froushani, S.M.; Afzale Ahangaran, N. Thymol has beneficial effects on the experimental model of ulcerative colitis. Avicenna J. Phytomed. 2019, 9, 538–550.
  63. Waetzig, G.H.; Seegert, D.; Rosenstiel, P.; Nikolaus, S.; Schreiber, S. p38 Mitogen-Activated Protein Kinase Is Activated and Linked to TNF-α Signaling in Inflammatory Bowel Disease. J. Immunol. 2002, 168, 5342–5351.
  64. Liang, D.; Li, F.; Fu, Y.; Cao, Y.; Song, X.; Wang, T.; Wang, W.; Guo, M.; Zhou, E.; Li, D.; et al. Thymol inhibits LPS-stimulated inflammatory response via down-regulation of NF-κB and MAPK signaling pathways in mouse mammary epithelial cells. Inflammation 2014, 37, 214–222.
  65. Chen, J.; Li, D.-L.; Xie, L.-N.; Ma, Y.-R.; Wu, P.-P.; Li, C.; Liu, W.-F.; Zhang, K.; Zhou, R.-P.; Xu, X.-T.; et al. Synergistic anti-inflammatory effects of silibinin and thymol combination on LPS-induced RAW264.7 cells by inhibition of NF-κB and MAPK activation. Phytomedicine 2020, 78, 153309.
  66. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118.
  67. Namdari, H.; Izad, M.; Rezaei, F.; Amirghofran, Z. Thymol as a reciprocal regulator of T cell differentiation: Promotion of regulatory T cells and suppression of Th1/Th17 cells. Int. Immunopharmacol. 2019, 67, 417–426.
  68. Gholijani, N.; Amirghofran, Z. Effects of thymol and carvacrol on T-helper cell subset cytokines and their main transcription factors in ovalbumin-immunized mice. J. Immunotoxicol. 2016, 13, 729–737.
  69. Amirghofran, Z.; Ahmadi, H.; Karimi, M.H.; Kalantar, F.; Gholijani, N.; Malek-Hosseini, Z. In vitro inhibitory effects of thymol and carvacrol on dendritic cell activation and function. Pharm. Biol. 2016, 54, 1125–1132.
More
ScholarVision Creations