Next Article in Journal
Comprehensive Metabolomics and Machine Learning Identify Profound Oxidative Stress and Inflammation Signatures in Hypertensive Patients with Obstructive Sleep Apnea
Next Article in Special Issue
Effects of Quercetin on the Intestinal Microflora of Freshwater Dark Sleeper Odontobutis potamophila
Previous Article in Journal
Continuous Glucose Monitoring in Preterm Infants: The Role of Nutritional Management in Minimizing Glycemic Variability
Previous Article in Special Issue
Impact of Dietary Lavender Essential Oil on the Growth and Fatty Acid Profile of Breast Muscles, Antioxidant Activity, and Inflammatory Responses in Broiler Chickens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties

Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2022, 11(10), 1947; https://doi.org/10.3390/antiox11101947
Submission received: 25 July 2022 / Revised: 20 September 2022 / Accepted: 26 September 2022 / Published: 29 September 2022
(This article belongs to the Special Issue Dietary Antioxidants and Gut Health)

Abstract

:
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.

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]. 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]. 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 vulgaris, Coridothymus 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]. Thus, thymol is considered as a beneficial food supplement. Recent studies reported that thymol improves intestinal integrity and alleviates intestinal injury via the regulation of the immune response and oxidation-reduction homeostasis [18]. In this review, we discussed the potential of thymol as a natural anti-inflammatory, anti-oxidative, and anti-bacterial compound in the modulation of mucosal immunity, OS, and gut microbes for IBD treatment.

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] (Figure 1), including Thymus zygis [20], Thymbra capitata [21], Thymus vulgaris [22], Satureja thymbra [23], Nigella sativa seeds [24], and Monarda didyma [25]. Pure thymol has low solubility in water, high volatility, and a strong bitter/irritating taste [26]; thus, it is usually encapsulated in electrospun nanofibers to enhance its water solubility and high temperature stability [27]. Emulsification is also used for thymol processing. Both encapsulation and emulsification have been shown to improve the anti-oxidant activity of thymol [28].
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]. Thymol can be metabolized to thymol sulfate and thymol glucuronide by sulfation and glucuronidation in the intestines, liver, kidney, and other organs, respectively [30]. 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]. 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]. 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]. Thymol enhances anti-oxidative capacity to alleviate OS in different tissues [33]. 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]. 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]. Thymol also exhibits other pharmacological functions, such as analgesia, anti-malarial, and anti-fungal properties [37,38,39,40,41]. This review focuses on the therapeutic potential of thymol in IBD in terms of its pharmacological properties and discusses the underlying mechanisms.

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]. 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]. Thymol alleviates dextran sulfate sodium (DSS)-induced intestinal damage and increases tight junction claudin-3 expression [44]. 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]. The intestinal mucus layer is the first line of defense maintaining bacterial symbiosis with the host and preventing bacterial penetration into epithelial cells [46]. Thymol increases mucus secretion to relieve ethanol-induced ulcer mucosal damage in rats [47]. 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]. 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]. 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].

4. Thymol Alleviates Intestinal Inflammation in IBD

The dysregulation of innate and adaptive immune responses leads to chronic intestinal inflammation in IBD patients [51,52,53]. Transcription factor nuclear factor κB (NF-κB) is the key mediator regulating the inflammatory response. The activation of NF-κB signaling induces the expression of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), inductible nitric oxide synthase (iNOS), interleukin (IL-1β), and IL-6 [54]. The over-production and accumulation of inflammatory cytokines cause intestinal epithelial apoptosis and the disruption of intestinal homeostasis, resulting in the dysfunction of the intestinal epithelial barrier in IBD patients [55]. Currently, the suppression of inflammation is the mainstay of IBD treatment.
Thymol shows strong anti-inflammatory properties in in vivo and in vitro studies [56]. The anti-inflammatory properties of thymol are listed in Table 2 and a schematic diagram of thymol’s action is depicted in Figure 3. Toll-like receptors (TLRs) are the main sensors used to detect various dangerous signals and activate innate immune responses. The classic TLR signaling pathway activates NF-κB to regulate the expressions of a series of cytokines [57]. In mice and macrophages, thymol inhibits TLR4 expression and then inhibits the activation of NF-κB signaling, which reduces the production of inflammatory cytokines, such as TNF-α and IL-1β [58,59]. NF-κB is a master mediator of inflammatory responses. Inactive NF-κB binds to IκB, an inhibitory subunit of NF-κB, and presents in the cytoplasm. When activated by a variety of signals, such as cytokine receptors and pattern-recognition receptors (PRRs), IκB is phosphorylated and degraded to release RelA (p65) from the NF-κB complex. p65 is then translocated to the nucleus to induce pro-inflammatory cytokine expression as a transcription factor [60]. Thymol has been shown to inhibit NF-κB activation by reducing p65 translocation and abundance in the colons of acetic acid-induced colitis rats and in LPS-activated macrophages, respectively, with decreased cytokine production [61,62]. The activation of NF-κB induces the expressions of iNOS and COX-2, which further promote vigorous inflammation. In ulcerative colitis rats, thymol reduces the COX-2 expression and nitric oxide (NO) levels produced by iNOS in the rats’ colon [63]. These studies showed the anti-inflammatory function of thymol through the inhibition of the NF-κB signaling pathway.
Studies have reported that proteins in the MAPK family, such as JNK1/2, p38α, and ERK, are activated in the colonic mucosa of IBD patients [71]. The suppression of the MAPK signaling pathway is an approach for alleviating inflammation and thus IBD [71]. It has been reported that thymol inhibits p38 phosphorylation and interferes with the activation of the MAPK signaling pathway to maintain the immune balance [64]. Thymol also suppresses LPS-induced activation of p-p38, p-JNK, and p-ERK, and correspondingly inhibits the production of NO, IL-6, TNF-α, COX-2, and other inflammatory cytokines [72]. Therefore, thymol can also reduce the inflammatory response by inhibiting the MAPK signaling pathway.
Cytokines are synthesized and secreted by activated immune cells, such as macrophages, T cells, B cells, dendritic cells (DCs), and natural killer cells. Among them, the regulatory T cell (Treg) is essential to control autoimmunity. Treg is defined by the expression of CD4, CD25, and transcription factor forkhead box P3 (Foxp3) [73]. Thymol promotes the differentiation of naïve T cells to CD4+CD25+Foxp3+ Treg cells and induces Foxp3 expression [74]. Meanwhile, thymol also maintains the balance of the Th1/Tregs and Th17/Tregs ratios to prevent autoimmunity as a result of suppressed inflammation [74,75]. Additionally, thymol exerts inhibitory effects on DCs’ maturation and T cell activation [76]. Although thymol has influenced the immune cell population in some studies, more information is required on how thymol modulates immune cell differentiation and whether the changes in the immune cell population by thymol contribute to the relief of IBD.

5. Thymol Improves Anti-Oxidant Capacity in IBD

IBD has been also associated with intestinal OS [77]. OS is the result of the excessive accumulation of ROS caused by the imbalance of the oxidation and anti-oxidation systems in cells [78]. Generally, ROS include hydrogen peroxide (H2O2), superoxide anions (O2•−), and hydroxyl radicals (HO•). The accumulation of ROS alters the structure and function of cell contents, such as DNA and proteins, and causes a series of cellular dysfunctions, including the disruption of cell metabolism, disruption of cell cycles, dysregulation of the immune response, etc. In IBD patients, the overproduction of ROS destroys the cytoskeleton and interrupts tight junction proteins in the intestinal epithelium, resulting in increased epithelial permeability and intestinal barrier dysfunction [79]. In addition, OS has been reported to be positively correlated with the level of inflammation [80], and OS in the GIT exacerbates intestinal inflammation and IBD. Therefore, the relief of intestinal OS is another strategy for treating IBD [81].
Thymol has been shown to have anti-oxidative capacity in a variety of OS models in vitro and in vivo, as listed in Table 3. A schematic mechanism of thymol’s action is depicted in Figure 4. Thymol acts as a strong anti-oxidant. partially due to its phenolic hydroxyl groups, which directly neutralize free radicals [82]. 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS+) are commonly used free radicals to evaluate anti-oxidant activity. Thymol eliminates 50% of DPPH radicals and scavenges ABTS+ radicals as a direct anti-oxidant in vitro [83]. However, other studies have indicated that phenolic compounds exert an anti-oxidative function through the activation of anti-oxidation-related signaling pathways, rather than directly neutralizing free radicals by the phenolic hydroxyl group [84]. Studies indicated that dietary polyphenols activate the anti-oxidation-related nuclear factor-E2-related factor 2 (Nrf2) signaling pathway, which plays a pivotal role in regulating the expression of anti-oxidant genes and the activities of anti-oxidant enzymes [85]. Studies have indicated that thymol activates Nrf2 signaling in different tissues [86,87]. Importantly, the Nrf2 downstream target HO-1 catalyzes the degradation of heme into Fe2+, CO, and bilirubin. Heme enhances ROS formation in OS, while bilirubin is an anti-oxidant and protects tissues [88]. Thymol promotes HO-1 expression in the lungs of mice, accounting for its anti-oxidative capacity [87]. Moreover, HO-1-induced CO also performs anti-inflammation and anti-apoptosis functions, which support the mechanism of thymol alleviating inflammation and intestinal damage.
Nrf2 maintains the cellular redox balance and prevents oxidative damage by regulating anti-oxidant enzymes, such as GSH and GPX. As a result, thymol increases the activities of anti-oxidant enzymes to reduce ROS production. SOD reduces superoxide O2•− radicals by catalyzing them into O2 and H2O2. H2O2 is then transformed by either CAT or GPX in the peroxisome to generate water and O2. Thymol enhances the activities of anti-oxidant enzymes and alleviates OS in animal models of OS [89]. Thymol enhances the total anti-oxidative capacity and reduces the ROS level in rats challenged with imidacloprid [93]. Thymol reduces the MDA level, a byproduct of lipid peroxidation and a marker of OS, increases SOD activity, and ameliorates OS in mouse obesity models [95]. In in vitro studies, thymol ameliorates acetaminophen-induced OS in HepG2 cells by increasing the GPX and SOD activities and decreasing the MDA level [99]. In lung fibroblasts, thymol increases the enzyme activities of SOD, CAT, and GPX, leading to a reduction in ROS production [90]. In the Chang liver cell line, thymol inhibits ROS production by increasing the GPX level and decreasing the MDA level, alleviating t-butyl-hydroperoxide-induced oxidative damage. In addition to the aforementioned anti-oxidant enzymes, thymol reduces OS through the inhibition of free-radical-generating enzyme xanthine oxidase (XO) [100]. XO catalyzes the oxidation of xanthine and hypoxanthine with increased production of O2•− and H2O2 in purine metabolism [101]. Thymol binds to XO directly and inhibits XO activity to produce ROS [100]. Collectively, thymol relieves OS via directly scavenging free radicals by its phenolic hydroxyl group, directly inhibiting free-radical-producing enzymes, and indirectly activating the Nrf2-mediated signaling pathway.

6. Thymol Changes Gut Microbes and Prevents Pathogen Infection

Mammalian gut microbes are mainly composed of four phyla, including Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. Alterations in the gut microbiota are observed in patients with IBD compared with healthy individuals. Notably, the abundance of the phylum Firmicutes is reduced in the stool of UC patients [102]. Although there is a lack of clinical research about the thymol-modulated gut microbial structure in IBD, multiple animal studies have described increases in the proportional abundance of Firmicutes potentially against IBD pathogenesis due to thymol [103]. However, a direct causative relationship between IBD and dysbiosis has not been clearly established in humans. The change in gut microbes by thymol as a therapeutical mechanism requires more elucidative studies.
Currently, studies of the microbial etiology of IBD indicate that persistent pathogen infection, such as enterotoxic E. coli, causes a “leaky gut” and chronic inflammation, subsequently leading to the excessive translocation of intestinal bacteria and the dysbiosis between “beneficial” and “detrimental” bacteria, resulting in IBD [104]. The members of the Proteobacteria phylum, notably the Enterobacteriaceae E. coli, are increased in IBD patients relative to healthy individuals [105]. Several studies have indicated the anti-pathogen capacity of thymol in different animal infection models and bacterial cultures, as illustrated in Table 4, as the potential mechanism. Researchers found that thymol reduces the abundance of the detrimental bacteria E. coli in the GIT of pigs [43]. In another study, feeding pigs with microencapsulated thymol promoted the proliferation of beneficial bacteria in the colon and decreased the colonization of detrimental bacteria, such as Escherichia, Campylobacter, Treponema, and Streptococcus [106]. In chickens infected with C. perfringens, thymol inhibited C. perfringens proliferation and then alleviated intestinal damage and mortality [50]. In these studies, thymol also promoted the colonization of beneficial bacteria, such as Clostridium, Lactobacillus, and Bacteroides, to improve gut health. Additionally, thymol also exhibited a direct anti-bacterial effect inhibiting human pathogens, such as E. coli, Listeria monocytogenes, and C. perfringens, which are harmful to the body’s health [107,108,109,110].
Overall, thyme exhibits therapeutic potential to treat IBD through multiple mechanisms, including the intestinal barrier, inflammation, redox homeostasis, and gut microbiota. The deregulation of any one aspect influences others, which also renders IBD a complex and multifactor disease. As mentioned, a compromised gut barrier leads to the invasion of pathogens, leading to an imbalance in the gut microbiota and the activation of an immune response; uncontrolled inflammation causes enterocyte death and further aggravates intestine damage. Moreover, prolonged inflammation also leads to the accumulation of free radicals and imbalance of redox homeostasis, which exacerbate inflammation. Therefore, it is difficult to conclude which mechanism plays a pivotal role in mediating thymol action unless a dedicated molecular mechanism is illustrated.

7. Conclusions

Multiple factors, including the environment, microorganisms, and genetics, interact to promote the development and occurrence of IBD [112]. Environmental factors, such as diet, which lead to gut microbiota dysbiosis, alter host mucosal defenses, and induce intestinal OS, are deemed to be major potential risk factors for IBD [113]. Simultaneously, diet is also a double-edged sword that is beneficial to the prevention and relief of IBD. More recently, green tea polyphenols have been shown to benefit IBD patients by reversing gut dysbiosis [114]. Therefore, nutritional intervention should be further promoted as a low-cost, side-effect-free method for the prevention and treatment of IBD [115]. Thymol, a natural product derived from medicinal plants or herbs, is commonly used as a dietary supplement to prevent IBD by improving gut integrity, reducing gut OS, and modulating gut immune responses. In addition, thymol maintains a good intestinal microenvironment through its anti-bacterial properties. Currently, considering that the common drugs used for IBD, such as corticosteroids, mesalazine, and balasaladin, have relatively large side effects, herbal plant extracts, such as thymol, as natural anti-oxidants may represent promising substances in the complementary therapy of IBD, with special emphasis on prevention. However, there are still doubts regarding how thymol regulates mucosal immunity and the mechanism of treating IBD by improving OS in the intestinal tract. Meanwhile, there have been no clinical trials yet, so an effective method and dosage for people ingesting thymol need to be further studied. In conclusion, considering the health benefits of thymol and its non-toxicity, thymol is a potential drug and rational strategy for the relief of IBD.

Author Contributions

Writing—original draft preparation, Y.L. (Yao Liu) and H.Y.; writing—review and editing, B.Y., J.H., X.M., J.Y. and P.Z.; visualization, Z.H., Y.L. (Yuheng Luo), J.L. and A.W.; supervision and conceptualization, H.Y. and D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Sichuan Science and Technology Program under grants (2021YJ0195, 2021ZDZX0009), the National Key Research and Development Program of China (2021YFD1300201), and the China Agriculture Research System of MOF and MARA (CARS-35).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

There is no conflict of interest to declare.

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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar]
  3. Kaser, A.; Zeissig, S.; Blumberg, R.S. Inflammatory bowel disease. Annu. Rev. Immunol. 2010, 28, 573–621. [Google Scholar] [CrossRef] [PubMed]
  4. Loftus, E.V., Jr. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology 2004, 126, 1504–1517. [Google Scholar] [CrossRef]
  5. Ng, S.C. Epidemiology of inflammatory bowel disease: Focus on Asia. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 363–372. [Google Scholar] [CrossRef] [PubMed]
  6. Kaplan, G.G.; Ng, S.C. Understanding and Preventing the Global Increase of Inflammatory Bowel Disease. Gastroenterology 2017, 152, 313–321. [Google Scholar] [CrossRef]
  7. Corridoni, D.; Arseneau, K.O.; Cominelli, F. Inflammatory bowel disease. Immunol. Lett. 2014, 161, 231–235. [Google Scholar] [CrossRef]
  8. Walfish, A.; Sachar, D. Phenotype classification in IBD: Is there an impact on therapy? Inflamm. Bowel Dis. 2007, 13, 1573–1575. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Matsuoka, K.; Kanai, T. The gut microbiota and inflammatory bowel disease. Semin. Immunopathol. 2015, 37, 47–55. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  18. Du, E.; Wang, W.; Gan, L.; Li, Z.; Guo, S.; Guo, Y. Effects of thymol and carvacrol supplementation on intestinal integrity and immune responses of broiler chickens challenged with Clostridium perfringens. J. Anim. Sci. Biotechnol. 2016, 7, 19. [Google Scholar] [CrossRef]
  19. 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. [Google Scholar] [CrossRef]
  20. 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. [Google Scholar] [CrossRef]
  21. 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. [Google Scholar] [CrossRef]
  22. 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. [Google Scholar] [CrossRef]
  23. 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. [Google Scholar] [CrossRef] [PubMed]
  24. 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. [Google Scholar] [CrossRef]
  25. 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. [Google Scholar] [CrossRef]
  26. 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. [Google Scholar] [CrossRef]
  27. 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. [Google Scholar] [CrossRef] [PubMed]
  28. 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. [Google Scholar] [CrossRef]
  29. 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. [Google Scholar] [CrossRef]
  30. 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. [Google Scholar] [CrossRef]
  31. 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. [Google Scholar] [CrossRef] [PubMed]
  32. 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. [Google Scholar] [CrossRef] [PubMed]
  33. 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. [Google Scholar] [CrossRef]
  34. 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. [Google Scholar] [CrossRef] [PubMed]
  35. 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. [Google Scholar] [CrossRef] [PubMed]
  36. 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. [Google Scholar] [CrossRef]
  37. 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. [Google Scholar] [CrossRef]
  38. 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. [Google Scholar] [CrossRef]
  39. 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. [Google Scholar] [CrossRef]
  40. 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. [Google Scholar] [CrossRef]
  41. 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. [Google Scholar] [CrossRef] [PubMed]
  42. 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. [Google Scholar] [CrossRef]
  43. 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. [Google Scholar] [CrossRef] [PubMed]
  44. 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. [Google Scholar] [CrossRef] [PubMed]
  45. 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. [Google Scholar] [CrossRef] [PubMed]
  46. 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. [Google Scholar] [CrossRef] [PubMed]
  47. 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. [Google Scholar] [CrossRef]
  48. 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. [Google Scholar] [CrossRef]
  49. 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. [Google Scholar] [CrossRef]
  50. 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. [Google Scholar] [CrossRef]
  51. 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. [Google Scholar] [CrossRef] [PubMed]
  52. 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. [Google Scholar] [CrossRef] [PubMed]
  53. 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. [Google Scholar] [CrossRef] [PubMed]
  54. 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. [Google Scholar] [CrossRef]
  55. Salim, S.Y.; Söderholm, J.D. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 362–381. [Google Scholar] [CrossRef]
  56. 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. [Google Scholar] [CrossRef]
  57. 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. [Google Scholar] [CrossRef]
  58. 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. [Google Scholar] [CrossRef]
  59. 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. [Google Scholar] [CrossRef]
  60. 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. [Google Scholar] [CrossRef]
  61. 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. [Google Scholar] [CrossRef] [PubMed]
  62. 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. [Google Scholar] [CrossRef]
  63. 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. [Google Scholar]
  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. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Q.; Cheng, F.; Xu, Y.; Zhang, J.; Qi, J.; Liu, X.; Wang, R. Thymol alleviates lipopolysaccharide-stimulated inflammatory response via downregulation of RhoA-mediated NF-κB signalling pathway in human peritoneal mesothelial cells. Eur. J. Pharmacol. 2018, 833, 210–220. [Google Scholar] [CrossRef] [PubMed]
  66. Nagoor Meeran, M.F.; Jagadeesh, G.S.; Selvaraj, P. Thymol attenuates inflammation in isoproterenol induced myocardial infarcted rats by inhibiting the release of lysosomal enzymes and downregulating the expressions of proinflammatory cytokines. Eur. J. Pharmacol. 2015, 754, 153–161. [Google Scholar] [CrossRef] [PubMed]
  67. Khosravi, A.R.; Erle, D.J. Chitin-Induced Airway Epithelial Cell Innate Immune Responses Are Inhibited by Carvacrol/Thymol. PLoS ONE 2016, 11, e0159459. [Google Scholar] [CrossRef]
  68. Kwon, H.I.; Jeong, N.H.; Kim, S.Y.; Kim, M.H.; Son, J.H.; Jun, S.H.; Kim, S.; Jeon, H.; Kang, S.C.; Kim, S.H.; et al. Inhibitory effects of thymol on the cytotoxicity and inflammatory responses induced by Staphylococcus aureus extracellular vesicles in cultured keratinocytes. Microb. Pathog. 2019, 134, 103603. [Google Scholar] [CrossRef]
  69. Koc, K.; Cerig, S.; Ucar, S.; Colak, S.; Bakir, M.; Erol, H.S.; Yildirim, S.; Hosseinigouzdagani, M.; Simsek Ozek, N.; Aysin, F.; et al. Gastroprotective effects of oleuropein and thymol on indomethacin-induced gastric ulcer in Sprague-Dawley rats. Drug. Chem. Toxicol. 2020, 43, 441–453. [Google Scholar] [CrossRef]
  70. Kilic, K.; Sakat, M.S.; Yildirim, S.; Kandemir, F.M.; Gozeler, M.S.; Dortbudak, M.B.; Kucukler, S. The amendatory effect of hesperidin and thymol in allergic rhinitis: An ovalbumin-induced rat model. Eur. Arch. Otorhinolaryngol. 2019, 276, 407–415. [Google Scholar] [CrossRef]
  71. 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. [Google Scholar] [CrossRef] [PubMed]
  72. 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. [Google Scholar] [CrossRef]
  73. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef] [PubMed]
  74. 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. [Google Scholar] [CrossRef]
  75. 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. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar]
  77. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxid. Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef] [PubMed]
  78. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-Mediated Cellular Signaling. Oxid. Med. Cell. Longev. 2016, 2016, 4350965. [Google Scholar] [CrossRef] [PubMed]
  79. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed]
  80. Yan, H.; Jin, J.Q.; Yang, P.; Yu, B.; He, J.; Mao, X.B.; Yu, J.; Chen, D.W. Fermented soybean meal increases nutrient digestibility via the improvement of intestinal function, anti-oxidative capacity and immune function of weaned pigs. Animal 2022, 16, 100557. [Google Scholar] [CrossRef]
  81. Yao, J.; Wang, J.Y.; Liu, L.; Li, Y.X.; Xun, A.Y.; Zeng, W.S.; Jia, C.H.; Wei, X.X.; Feng, J.L.; Zhao, L.; et al. Anti-oxidant effects of resveratrol on mice with DSS-induced ulcerative colitis. Arch. Med. Res. 2010, 41, 288–294. [Google Scholar] [CrossRef] [PubMed]
  82. Gavaric, N.; Mozina, S.S.; Kladar, N.; Bozin, B. Chemical Profile, Antioxidant and Antibacterial Activity of Thyme and Oregano Essential Oils, Thymol and Carvacrol and Their Possible Synergism. J. Essent. Oil Bear. Plants. 2015, 18, 1013–1021. [Google Scholar] [CrossRef]
  83. Yu, Y.-M.; Chao, T.-Y.; Chang, W.-C.; Chang, M.J.; Lee, M.-F. Thymol reduces oxidative stress, aortic intimal thickening, and inflammation-related gene expression in hyperlipidemic rabbits. J. Food Drug Anal. 2016, 24, 556–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hollman, P.C.; Cassidy, A.; Comte, B.; Heinonen, M.; Richelle, M.; Richling, E.; Serafini, M.; Scalbert, A.; Sies, H.; Vidry, S. The biological relevance of direct antioxidant effects of polyphenols for cardiovascular health in humans is not established. J. Nutr. 2011, 141, 989S–1009S. [Google Scholar] [CrossRef]
  85. Croft, K.D. Dietary polyphenols: Antioxidants or not? Arch. Biochem. Biophys. 2016, 595, 120–124. [Google Scholar] [CrossRef]
  86. FangFang; Li, H.; Qin, T.; Li, M.; Ma, S. Thymol improves high-fat diet-induced cognitive deficits in mice via ameliorating brain insulin resistance and upregulating NRF2/HO-1 pathway. Metab. Brain Dis. 2017, 32, 385–393. [Google Scholar] [CrossRef]
  87. Yao, L.; Hou, G.; Wang, L.; Zuo, X.S.; Liu, Z. Protective effects of thymol on LPS-induced acute lung injury in mice. Microb. Pathog. 2018, 116, 8–12. [Google Scholar] [CrossRef]
  88. Bainbridge, S.A.; Smith, G.N. HO in pregnancy. Free Radic. Biol. Med. 2005, 38, 979–988. [Google Scholar] [CrossRef]
  89. El-Nekeety, A.A.; Mohamed, S.R.; Hathout, A.S.; Hassan, N.S.; Aly, S.E.; Abdel-Wahhab, M.A. Antioxidant properties of Thymus vulgaris oil against aflatoxin-induce oxidative stress in male rats. Toxicon 2011, 57, 984–991. [Google Scholar] [CrossRef]
  90. Archana, P.R.; Nageshwar Rao, B.; Ballal, M.; Satish Rao, B.S. Thymol, a naturally occurring monocyclic dietary phenolic compound protects Chinese hamster lung fibroblasts from radiation-induced cytotoxicity. Mutat. Res. 2009, 680, 70–77. [Google Scholar] [CrossRef]
  91. Archana, P.R.; Nageshwar Rao, B.; Satish Rao, B.S. Modulation of gamma ray-induced genotoxic effect by thymol, a monoterpene phenol derivative of cymene. Integr. Cancer Ther. 2011, 10, 374–383. [Google Scholar] [CrossRef] [PubMed]
  92. Khan, A.; Ahmad, A.; Ahmad Khan, L.; Padoa, C.J.; van Vuuren, S.; Manzoor, N. Effect of two monoterpene phenols on antioxidant defense system in Candida albicans. Microb. Pathog. 2015, 80, 50–56. [Google Scholar] [CrossRef] [PubMed]
  93. Saber, T.M.; Arisha, A.H.; Abo-Elmaaty, A.M.A.; Abdelgawad, F.E.; Metwally, M.M.M.; Saber, T.; Mansour, M.F. Thymol alleviates imidacloprid-induced testicular toxicity by modulating oxidative stress and expression of steroidogenesis and apoptosis-related genes in adult male rats. Ecotoxicol. Environ. Saf. 2021, 221, 112435. [Google Scholar] [CrossRef] [PubMed]
  94. Stojanović, N.M.; Mitić, K.V.; Randjelović, P.; Stevanović, M.; Stojiljković, N.; Ilić, S.; Tričković Vukić, D.; Sokolović, D.; Jevtović-Stoimenov, T.; Radulović, N.S. Thymol regulates the functions of immune cells in the rat peritoneal cavity after l-arginine-induced pancreatitis. Life Sci. 2021, 280, 119704. [Google Scholar] [CrossRef]
  95. Haque, M.R.; Ansari, S.H.; Najmi, A.K.; Ahmad, M.A. Monoterpene phenolic compound thymol prevents high fat diet induced obesity in murine model. Toxicol. Mech. Methods 2014, 24, 116–123. [Google Scholar] [CrossRef]
  96. Youdim, K.; Deans, S.G. Effect of thyme oil and thymol dietary supplementation on the antioxidant status and fatty acid composition of the ageing rat brain. Br. J. Nutr. 2000, 83, 87–93. [Google Scholar] [CrossRef]
  97. Güvenç, M.; Cellat, M.; Gökçek, İ.; Yavaş, İ.; Yurdagül Özsoy, Ş. Effects of thymol and carvacrol on sperm quality and oxidant/antioxidant balance in rats. Arch. Physiol. Biochem. 2019, 125, 396–403. [Google Scholar] [CrossRef]
  98. El-Sayed, E.M.; Abd-Allah, A.R.; Mansour, A.M.; El-Arabey, A.A. Thymol and carvacrol prevent cisplatin-induced nephrotoxicity by abrogation of oxidative stress, inflammation, and apoptosis in rats. J. Biochem. Mol. Toxicol. 2015, 29, 165–172. [Google Scholar]
  99. Palabiyik, S.S.; Karakus, E.; Halici, Z.; Cadirci, E.; Bayir, Y.; Ayaz, G.; Cinar, I. The protective effects of carvacrol and thymol against paracetamol-induced toxicity on human hepatocellular carcinoma cell lines (HepG2). Hum. Exp. Toxicol. 2016, 35, 1252–1263. [Google Scholar] [CrossRef]
  100. Abbasi, S.; Gharaghani, S.; Benvidi, A.; Rezaeinasab, M.; Saboury, A.A. An in-depth view of potential dual effect of thymol in inhibiting xanthine oxidase activity: Electrochemical measurements in combination with four way PARAFAC analysis and molecular docking insights. Int. J. Biol. Macromol. 2018, 119, 1298–1310. [Google Scholar] [CrossRef]
  101. Kostić, D.A.; Dimitrijević, D.S.; Stojanović, G.S.; Palić, I.R.; Đorđević, A.S.; Ickovski, J.D. Xanthine Oxidase: Isolation, Assays of Activity, and Inhibition. J. Chem. 2015, 2015, 294858. [Google Scholar] [CrossRef]
  102. Rehman, A.; Rausch, P.; Wang, J.; Skieceviciene, J.; Kiudelis, G.; Bhagalia, K.; Amarapurkar, D.; Kupcinskas, L.; Schreiber, S.; Rosenstiel, P.; et al. Geographical patterns of the standing and active human gut microbiome in health and IBD. Gut 2016, 65, 238–248. [Google Scholar] [CrossRef] [PubMed]
  103. Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, S.; Zhao, W.; Lan, P.; Mou, X. The microbiome in inflammatory bowel diseases: From pathogenesis to therapy. Protein Cell 2021, 12, 331–345. [Google Scholar] [CrossRef]
  105. Manichanh, C.; Rigottier-Gois, L.; Bonnaud, E.; Gloux, K.; Pelletier, E.; Frangeul, L.; Nalin, R.; Jarrin, C.; Chardon, P.; Marteau, P.; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–211. [Google Scholar] [CrossRef] [PubMed]
  106. Mo, K.; Li, J.; Liu, F.; Xu, Y.; Huang, X.; Ni, H. Superiority of Microencapsulated Essential Oils Compared With Common Essential Oils and Antibiotics: Effects on the Intestinal Health and Gut Microbiota of Weaning Piglet. Front. Nutr. 2021, 8, 808106. [Google Scholar] [CrossRef]
  107. Du, E.; Gan, L.; Li, Z.; Wang, W.; Liu, D.; Guo, Y. In vitro antibacterial activity of thymol and carvacrol and their effects on broiler chickens challenged with Clostridium perfringens. J. Anim. Sci. Biotechnol. 2015, 6, 58. [Google Scholar] [CrossRef]
  108. Chien, S.Y.; Sheen, S.; Sommers, C.H.; Sheen, L.Y. Modeling the Inactivation of Intestinal Pathogenic Escherichia coli O157:H7 and Uropathogenic E. coli in Ground Chicken by High Pressure Processing and Thymol. Front. Microbiol. 2016, 7, 920. [Google Scholar] [CrossRef]
  109. Shah, B.; Davidson, P.M.; Zhong, Q. Nanocapsular dispersion of thymol for enhanced dispersibility and increased antimicrobial effectiveness against Escherichia coli O157:H7 and Listeria monocytogenes in model food systems. Appl. Environ. Microbiol. 2012, 78, 8448–8453. [Google Scholar] [CrossRef]
  110. Varel, V.H.; Miller, D.N. Plant-derived oils reduce pathogens and gaseous emissions from stored cattle waste. Appl. Environ. Microbiol. 2001, 67, 1366–1370. [Google Scholar] [CrossRef]
  111. Qiao, J.; Shang, Z.; Liu, X.; Wang, K.; Wu, Z.; Wei, Q.; Li, H. Regulatory Effects of Combined Dietary Supplementation With Essential Oils and Organic Acids on Microbial Communities of Cobb Broilers. Front. Microbiol. 2021, 12, 814626. [Google Scholar] [CrossRef] [PubMed]
  112. Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef] [PubMed]
  113. Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Nikolopoulos, G.K.; Lytras, T.; Bonovas, S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Meta-analyses. Gastroenterology 2019, 157, 647–659.e644. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, Z.; Huang, S.; Li, T.; Li, N.; Han, D.; Zhang, B.; Xu, Z.Z.; Zhang, S.; Pang, J.; Wang, S.; et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome 2021, 9, 184. [Google Scholar] [CrossRef]
  115. Green, N.; Miller, T.; Suskind, D.; Lee, D. A Review of Dietary Therapy for IBD and a Vision for the Future. Nutrients 2019, 11, 947. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Thymol biosynthesis in plants. Thymol is synthesized by the hydroxylation of p-cymene, which originates from the aromatization of γ-terpinene.
Figure 1. Thymol biosynthesis in plants. Thymol is synthesized by the hydroxylation of p-cymene, which originates from the aromatization of γ-terpinene.
Antioxidants 11 01947 g001
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.
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.
Antioxidants 11 01947 g002
Figure 3. Mechanism of thymol in relieving inflammation. Cells receive various stimuli and activate IκB kinases (IκKs) through the TLR4 signaling pathway, and then IκB proteins are phosphorylated, ubiquitinated, and degraded, and NF-κB dimers are released. The NF-κB dimer is then further activated through various post-translational modifications and translocated into the nucleus to bind to target genes to promote pro-inflammatory gene transcription. In addition, by activating the MAPKs family, it can promote the expression of c-jun terminal kinases (JNK) 1/2, extracellular signal-regulated kinase (ERK), and p38 for activation, thereby promoting the nuclear entry of activator protein 1 (AP-1) to regulate the expression of pro-inflammatory genes. Thymol inhibits the dissociation of the IκB protein and NF-κB dimer and the activation of the mitogen-activated protein kinase (MAPK) signaling pathway to relieve inflammation.
Figure 3. Mechanism of thymol in relieving inflammation. Cells receive various stimuli and activate IκB kinases (IκKs) through the TLR4 signaling pathway, and then IκB proteins are phosphorylated, ubiquitinated, and degraded, and NF-κB dimers are released. The NF-κB dimer is then further activated through various post-translational modifications and translocated into the nucleus to bind to target genes to promote pro-inflammatory gene transcription. In addition, by activating the MAPKs family, it can promote the expression of c-jun terminal kinases (JNK) 1/2, extracellular signal-regulated kinase (ERK), and p38 for activation, thereby promoting the nuclear entry of activator protein 1 (AP-1) to regulate the expression of pro-inflammatory genes. Thymol inhibits the dissociation of the IκB protein and NF-κB dimer and the activation of the mitogen-activated protein kinase (MAPK) signaling pathway to relieve inflammation.
Antioxidants 11 01947 g003
Figure 4. Mechanism of thymol scavenging free radicals. XO catalyzes the oxidation of xanthine and hypoxanthine with increased production of O2•− in purine metabolism or O2•− is generated in the mitochondrial inner membrane through the respiratory electron transport chain. The former enters the peroxisome and the latter is catalyzed by SOD in the mitochondria to generate H2O2. CAT and GPX in the peroxisome catalyze H2O2 to generate H2O and O2, and H2O2 in the cytoplasm is affected by Fe2+ to generate OH·. Elevated levels of free radicals can lead to oxidative stress. Thymol reduces ROS by directly scavenging free radicals, combining with XO, or increasing anti-oxidant enzyme activity.
Figure 4. Mechanism of thymol scavenging free radicals. XO catalyzes the oxidation of xanthine and hypoxanthine with increased production of O2•− in purine metabolism or O2•− is generated in the mitochondrial inner membrane through the respiratory electron transport chain. The former enters the peroxisome and the latter is catalyzed by SOD in the mitochondria to generate H2O2. CAT and GPX in the peroxisome catalyze H2O2 to generate H2O and O2, and H2O2 in the cytoplasm is affected by Fe2+ to generate OH·. Elevated levels of free radicals can lead to oxidative stress. Thymol reduces ROS by directly scavenging free radicals, combining with XO, or increasing anti-oxidant enzyme activity.
Antioxidants 11 01947 g004
Table 1. Protective function of thymol on the intestinal barrier.
Table 1. Protective function of thymol on the intestinal barrier.
ModelThymol DoseEffectsRef.
In vivo
Weaned pigs50 mg/kg dietEnrichment of 120 and 59 gene sets in oxyntic
and pyloric mucosa↓
[29]
Ethanol-induced acute ulcer10–100 mg/kg dietMucosal damage↓
Amount of mucus↑
[47]
Chicken infected with
Clostridium perfringens
30 mg/kg dietIntestinal lesions and mortality↓
Lactobacillus salivarius and L. johnsonii
L. crispatus, L. agilis, and Escherichia coli
[50]
Weaned pigs100 mg/kg dietExpression of ZO-1 and occludins in jejunal mucosa↓
Enterococcus genus and E. coli
Plasma diamine oxidase concentration↓
Weaning-induced intestinal OS↓
[43]
In vitro
IPEC-J250 μMTEER↑
Cell permeability↓
ZO-1and actin staining↑
[48]
Caco-2 cells15 mg/LCOX1 transcription↑
COX1:COX2 ratio↑
TEER↑
[49]
↑ indicates for rising; ↓ indicates for descending.
Table 2. Summary of the effect of thymol in different inflammation models.
Table 2. Summary of the effect of thymol in different inflammation models.
ModelThymol DoseEffectsRef.
In vitro
Mouse macrophages challenged with LPS20 mg/mLIL-1β expression↓[59]
Mouse mammary epithelial cells
challenged with LPS
10–40 μg/mLTNF-α, IL-6, iNOS, and COX-2 expression↓
Phosphorylation of IκBα, NF-κBp65↓
[64]
Human peritoneal mesothelial cell
line challenged with LPS
10–40 μg/mLTLR4 expression↓
NF-κB p65, IκK, and IκBα phosphorylation↓
TNF-α, IL-6 expression↓
[65]
IPEC-J2 cells challenged with LPS10–100 µMIL-8 secretion↓[66]
Chitin-induced airway epithelial cells30 µg/mLTLR4 is inhibited
IL-25 and IL-33 release↓
[67]
HaCaT cells challenged with
Staphylococcus aureus
512 µg/mLIL-1β, IL-6, and IL-8 expression↓
Phosphorylation of p65 and IκBα↓
[68]
In vivo
DSS-induced mouse colitis30–60 mg/kg dietNO, TNF-α, IL-1β, IL-6 expression↓
Phosphorylation of IκBα and NF-κBp65↓
[62]
Indomethacin-induced rat gastric ulcer75–500 mg/kg dietTNF-α, iNOS levels↓[17]
Indomethacin-induced rat gastric ulcer50–500 mg/kg dietROS, eNOS, TNF-α, caspase-3 levels↓
Prostaglandin E2 (PGE2) levels↑
[69]
Rat ulcerative colitis100 mg/kg dietCOX-2, IL-6, IL-1β and TNF-α expression↓
mRNA level of NF-κB p65↓
Myeloperoxidase (MPO) activity, NO, and malondialdehyde (MDA) level↓
[63]
Ovalbumin-induced rat allergic rhinitis20 mg/kg dietPlasma IL-5, IL-13, IgE levels↓
TNF-α expression in the nasal mucosa of rats↓
[70]
Clostridium perfringens infection-induced
chicken necrotic enteritis
15–60 mg/kg dietTLR2 and TNF-α level in ileum↓[18]
↑ indicates for rising; ↓ indicates for descending.
Table 3. Anti-oxidant properties of thymol.
Table 3. Anti-oxidant properties of thymol.
ModelThymol DoseResponseRef.
In vitro
Tert-butyl hydroperoxide-induced OS in Chang cells12.5–50 µg/mLROS generation and MDA level↓
Glutathione (GSH) level↑
[89]
Radiation-induced cytotoxicity
in lung fibroblast (V79) cells
0–100 µg/mLRadiation-induced lipid peroxidation↓
GSH, catalase (CAT), and superoxide dismutase (SOD)↑
[90]
Chinese hamster lung fibroblast cells (V79)0–100 µg/mLRadiation-induced genotoxicity and apoptosis↓[91]
Candida albicans5–20 µg/mLActivity of CAT, glutathione peroxidase (GPX)↑[92]
in vivo
Imidacloprid-induced testicular toxicity30 mg/kg dietCAT, SOD, and GSH↑
MDA↓
[93]
L-arginine-induced acute pancreatitis50–100 mg/kg dietMPO and O2•−[94]
High fat diet-induced obesity in mice14 mg/kg dietSOD and CAT activity in serum↑
MDA level in serum↓
[95]
Ovalbumin-induced asthma in mice8–32 mg/kg dietMDA level↓[39]
Wistar rats42.5 mg/kg dietGSH-Px activity↑
Brain total anti-oxidant status↑
Proportions of docosahexaenoic acid (DHA)↑
[96]
Rats10–20 mg/kg dietMDA levels in testicles, liver, and kidney↓
Oxidative damage↓
Anti-oxidant levels and GSH levels↑
[97]
Cisplatin-induced nephrotoxicity in rats20 mg/kg dietGSH, SOD, and CAT levels in kidney↑
Caspase-3 and MDA levels in kidney↓
[98]
↑ indicates for rising; ↓ indicates for descending.
Table 4. Thymol prevents pathogen infection.
Table 4. Thymol prevents pathogen infection.
ModelThymol DoseEffectsRef.
In vivo
Weaning pigs2% thymol dietColon probiotic bacteria↑
Potential pathogens↓
[106]
Cobb broilers150 mg/kg dietClostridium, Bacteroides, Lactobacillus
Proteobacteria↓
[111]
Pathogen culture medium
Ground chicken100–200 ppmE. coli O157:H7↓[108]
Meat medium46.875–6000 μg/mLE. coli, Salmonella, and C. perfringens[107]
Apple cider and milk0.5 g/LE. coli O157:H7 and L. monocytogenes[109]
Cattle waste6.7 mMFecal coliforms↓[110]
↑ indicates for rising; ↓ indicates for descending.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, Y.; Yan, H.; Yu, B.; He, J.; Mao, X.; Yu, J.; Zheng, P.; Huang, Z.; Luo, Y.; Luo, J.; et al. Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties. Antioxidants 2022, 11, 1947. https://doi.org/10.3390/antiox11101947

AMA Style

Liu Y, Yan H, Yu B, He J, Mao X, Yu J, Zheng P, Huang Z, Luo Y, Luo J, et al. Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties. Antioxidants. 2022; 11(10):1947. https://doi.org/10.3390/antiox11101947

Chicago/Turabian Style

Liu, Yao, Hui Yan, Bing Yu, Jun He, Xiangbing Mao, Jie Yu, Ping Zheng, Zhiqing Huang, Yuheng Luo, Junqiu Luo, and et al. 2022. "Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties" Antioxidants 11, no. 10: 1947. https://doi.org/10.3390/antiox11101947

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

Article Metrics

Back to TopTop