Next Article in Journal
Implication of Cellular Senescence in Osteoarthritis: A Study on Equine Synovial Fluid Mesenchymal Stromal Cells
Next Article in Special Issue
Special Issue “Dietary Bioactive Components in Inflammatory Bowel Disease”
Previous Article in Journal
Potassium Dehydroandrograpolide Succinate Targets NRP1 Mediated VEGFR2/VE-Cadherin Signaling Pathway to Promote Endothelial Barrier Repair
Previous Article in Special Issue
Therapeutic Potential of Bioactive Components from Scutellaria baicalensis Georgi in Inflammatory Bowel Disease and Colorectal Cancer: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease

by
Alessia Arangia
1,†,
Ylenia Marino
1,†,
Daniela Impellizzeri
1,*,
Ramona D’Amico
1,*,
Salvatore Cuzzocrea
1,‡ and
Rosanna Di Paola
2,‡
1
Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
2
Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Salvatore Cuzzocrea and Rosanna Di Paola shared senior authorship.
Int. J. Mol. Sci. 2023, 24(4), 3111; https://doi.org/10.3390/ijms24043111
Submission received: 31 December 2022 / Revised: 30 January 2023 / Accepted: 1 February 2023 / Published: 4 February 2023
(This article belongs to the Special Issue Dietary Bioactive Components in Inflammatory Bowel Disease)

Abstract

:
In recent years, the phytoconstituents of foods in the Mediterranean diet (MD) have been the subject of several studies for their beneficial effects on human health. The traditional MD is described as a diet heavy in vegetable oils, fruits, nuts, and fish. The most studied element of MD is undoubtedly olive oil due precisely to its beneficial properties that make it an object of interest. Several studies have attributed these protective effects to hydroxytyrosol (HT), the main polyphenol contained in olive oil and leaves. HT has been shown to be able to modulate the oxidative and inflammatory process in numerous chronic disorders, including intestinal and gastrointestinal pathologies. To date, there is no paper that summarizes the role of HT in these disorders. This review provides an overview of the anti-inflammatory and antioxidant proprieties of HT against intestinal and gastrointestinal diseases.

1. Introduction

Over the last few years, the concept that food can act as medicine has been getting more and more acceptance by the scientific community. It is well known, in fact, that nutrition is capable of substantially modifying the risk profile of the population at both primary and secondary levels of prevention. Different types of diets have gained public attention, but the one that got the greatest interest is certainly the Mediterranean diet (MD). The MD was first described by Ancel Keys around the 1960s following the results of an epidemiological study [1]. In particular, Keys and colleagues demonstrated that olive-growing regions bordering the Mediterranean Sea had a reduced incidence of cardiovascular disease and cancer compared to other populations [1]. This observation has been confirmed by several subsequent studies, which provide strong evidence for the benefits of MD on cardiovascular health, including the reduction in the incidence of cardiovascular outcomes and risk factors, including obesity, hypertension, metabolic syndrome, and dyslipidemia [2,3,4,5]. Not only evidence from long-term observational studies but also clinical and preclinical studies demonstrated that the MD, closely linked to traditional areas of olive cultivation in the Mediterranean region, has been associated with low rates of chronic diseases, reductions in the incidence of some cancers, and neurodegenerative diseases; consequently, MD provides high adult life expectancy [2,6,7,8,9]. The traditional MD is described as a diet heavy in vegetable oils and low in saturated fat; it is commonly characterized by a high intake of fresh or minimally refined foods, such as cereals, vegetables, fruits, nuts, legumes, fish, olive oil, and red wine in moderation [9,10]. Individual foods and components of MD (e.g., extra-virgin olive oil and nuts) have numerous health-promoting properties [11,12,13,14,15,16], as they are rich in polyphenols, flavonoids, phenolic acids, and other bioactive compounds. Indeed, polyphenols are well-documented and can act as a free radical scavenger or metal chelator [17,18,19]. Thus, MD could trigger changes in signaling pathways and subsequent gene expression; in particular, it appears to act by modulating anti-inflammatory and antioxidant pathways [20,21,22]. Additionally, recent research has revealed that MD has antibacterial benefits in addition to lowering blood cholesterol levels and enhancing endothelial function [23,24].
It should be emphasized that MD is characterized by the regular intake of extra virgin olive oil (EVOO), whose significant beneficial effects are attributed to its phenolic constituents, fatty acid composition, and carotenoids such as lutein and beta-carotene [25]. One phenolic compound is hydroxytyrosol (HT), considered one of the most powerful antioxidant compounds among phenolic compounds from olive trees, followed by oleuropein, caffeic, and tyrosol (TYR) [25,26]. Due to its molecular structure (4-(2-hydroxyethyl)-1,2-benezediol), its regular consumption has several beneficial effects such as antioxidant, anti-inflammatory, antidiabetic, antimicrobial, anticancer, and as a protector of skin and eyes, etc. [27,28,29]. In this regard, the nutraceutical and food industries employ HT as a nutritional supplement due also to its high bioavailability and lack of toxicity [30,31]. Growing knowledge of the beneficial effect of HT has led researchers to investigate it as a therapeutic agent for chronic disorders, especially inflammatory bowel diseases (IBDs) [32]. IBDs are a group of immune-based disorders characterized by prolonged inflammation and severe intestinal dysbiosis resulting in severe damage to the gastrointestinal tract [32,33]. To date there are no papers that summarize the effects of HT in these pathologies; therefore, the objective of this review is to provide an overview of the anti-inflammatory and antioxidant effects of HT against intestinal and gastrointestinal diseases.

2. Hydroxytyrosol

2.1. Source of Hydroxytyrosol

HT is a polyphenol with potent antioxidant capacity due to its ortho-dihydroxy conformation in the aromatic ring (Figure 1). It is a member of the catechol class and is a primary alcohol. HT and TYR are obtained through the hydrolysis of oleuropein, which is the main constituent that gives olives their bitter taste. This process of oleuropein hydrolysis occurs during olive ripening, storage, and processing [34]. In fact, HT is the most abundant metabolite found in extra virgin olive oil, olives (Olea europea L.), and wine. What makes the study of HT interesting is its ability to induce the synthesis of antioxidant enzymes, as well as its ability to scavenge reactive oxygen species (ROS) [35]. Furthermore, Napolitano et al. have shown that HT is able to donate hydrogen ions, and this gives it a better ability to chelate iron compared to TYR [36].

2.2. Extraction and Purification of Hydroxytyrosol

A high concentration of HT is found in olive leaves; however, this is lost as a residue during oil production. To reduce industrial waste, the residue is used to obtain natural products used in the food, cosmetics, and pharmaceutical industries [26]. HT is obtained from vegetation water (i.e., waste water deriving from the processing of olive oil) or leaves through three steps. The first step consists of obtaining liquid rich in phenols which are extracted and purified. In the second step, an extract and mixture of HT with 3,4-dihydroxyphenlglycol are obtained, and HT acetate is produced. In the end, these components are made highly pure. In industrial processes, different sources of extraction are used; for example, olive waters are produced during processing by centrifuge/separation of the oil. The leaves, on the other hand, are dehydrated and undergo a process of hydroalcoholic extraction and hydrolysis. From this hydroalcoholic solution, oleuropein is obtained, which in turn is purified from its hydrolysis by HT [37]. Papageorgiou and colleagues described the purification processes of HT in detail [38]. Here, we summarize that HT can be recovered through solid–liquid extraction, acid hydrolysis, or liquid–liquid extraction with ethyl acetate [38].

2.3. Absorption, Distribution, Metabolism, and Excretion (ADME) of Hydroxytyrosol

Phenolic compounds are absorbed in a dose-dependent manner and undergo intestinal-hepatic metabolism. Specifically, HT is absorbed and excreted differently depending on how it is taken (aqueous or oily) [10]. The average absorption of HT is about 66% and occurs in the small intestine and colon by passive transport through the intestinal epithelium [25]. The uptake of HT present in wine is different. Indeed, Perez-Mana et al. have highlighted how the interaction between HT and ethanol may result in a change in dopamine metabolism that induces the production of HT instead of producing DOPAC (3,4-dihydroxyl phenylacetic acid) [10,39]. In any case, absorption is rapid, and a maximum plasma concentration is reached 5–10 min after intake, followed by a rapid decline. After absorption, HT binds to circulating lipoproteins, acting as an antioxidant and cardiovascular protector [40]. Furthermore, HT is easily distributed in various organs such as the kidney, liver, skeletal muscle, lungs, and heart. Not only, but HT is also able to cross the blood-brain barrier, thereby reaching the brain and interacting with dopaminergic pathways, as observed in a study by D’Angelo through radioactively labeling HT by intravenous administration in rats [41]. Regarding metabolism, HT suffers the first step inside enterocytes and subsequently in the liver [10]. Both steps are important because HT undergoes several modifications and transformations, presumably endowing it with its beneficial properties. Briefly, HT metabolism can be summarized according to three reactions: oxidation, methylation, and methylation-oxidation, as described by Ramírez-Tortose et al. [34]. Finally, HT and its metabolites are excreted through the urine within 6 h in humans and within 4 h in rats. Excretion could vary depending on the administration to humans or animals or even according to quantity or vehicle [10]. Obviously, all this affects the dosage of HT used for different pharmacological and nutraceutical purposes [34].

2.4. Toxicity and Dose Establishment

Due to its chemical properties, HT is used as a dietary supplement in the nutraceutical and food industries. In fact, it is an excellent scavenger and protects against lipid peroxidation. HT is the main component of olive oil studied to date and is responsible for numerous beneficial effects. Regarding its toxicity and safety profile, there are several studies done on olive mill wastewater and olive oil [10]. According to several studies based on the different doses of HT, no toxic effect of HT was observed both in acute and subchronic toxicity [27,34]. Therefore, based on the observations made, it was concluded that HT does not exhibit genotoxic and mutagenic effects, as stated in in vitro models by Aunon-Calles et al. [10]. In fact, it has been seen that HT has no toxic effects but has beneficial effects such as anti-apoptotic, antiproliferative, antioxidant, and anti-inflammatory activity and acts as a cardiovascular protector. So toxicity is not relevant, but the European Food Safety Authority suggests taking 5 mg of HT in order for it to perform its activity as an anti-inflammatory, antioxidant, and low-density lipoprotein (LDL) oxidation inhibiter [34].

2.5. Proprieties of Hydroxytyrosol

HT has numerous beneficial effects, as described earlier, is a nonmutagenic compound, is not genotoxic, and has a high safety profile. The main effects of HT are described below (Figure 2).

2.5.1. Antioxidant Proprieties and ROS Scavenging

HT exhibits a protective effect on the organism as it induces the production of antioxidant enzymes through modulation of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. This involves the translocation of Nrf2 into the nucleus resulting in the expression of cytoprotective genes such as heme oxygenase (HO-1) [10,42]. It also induces the synthesis of proteins that lead to DNA repair or phase II detoxifying enzymes [25]. Indeed, HT is able to increase the antioxidant network, such as superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT), and keep reduced glutathione (GSH) levels constant [43]. This allows ROS to be eliminated in the cellular environment [10]. Furthermore, in an in vitro study by Rietjens et al., it has been shown that HT stimulates the activation of signaling pathways that recognize the presence of ROS [44], and HT can protect against LDL oxidation even at low concentrations.

2.5.2. Anti-Inflammatory Proprieties

Inflammation is a form of protection against damaged cells and pathogens. Normally the inflammatory response involves the nuclear factor kappa-light-chain-enhancer pathway of activated B cells (NF-κB). HT may act as an anti-inflammatory agent by inhibiting nuclear translocation of NF-κB, thereby inhibiting the expression of genes involved in inflammation. In particular, HT blocks arachidonic acid (AA), cyclo-oxygenase, and lipoxygenase (COX and LOX), inhibits pro-inflammatory cytokines, such as TNF-α and IL-1β, and other enzymes involved in inflammation [26,45]. In addition, HT is able to suppress inducible nitric oxide synthase/nitric oxide. In a study by Scoditti et al. conducted on activated human monocytes, HT at appropriate concentrations suppresses the activation and expression of the enzymes MMP-9 and COX-2 [46]. Additionally, the lowered lymphocyte activity inhibited by the anti-inflammatory effects of HT could result in the development of novel ways for the treatment of inflammatory disorders [26].

2.5.3. Anticancer Proprieties

HT has antiproliferative and hence apoptotic effects via chelating metal ions and scavenging free radicals [10,27]. In addition, studies carried out in vitro on HL60 cell lines (leukemia cell lines) revealed that HT behaved as a pro-apoptotic by causing the release of cytochrome c from the mitochondrial intermembrane gap and activating caspase 8 [47,48]. In vitro studies demonstrated that HT could inhibit ERK1/2 to reduce cell growth in MCF-7 (Michigan Cancer Foundation-7) breast cancer cells [27]. Chemento et al. showed that HT induces apoptosis of ER-negative SL-BR-3 breast cancer by acting as an antagonist for the G-protein-coupled estrogen receptor (GPER), leading to apoptosis [49]. Rosignoli and colleagues demonstrated that HT plays a different role on different cancer cell lines of MCF-7 (breast carcinoma), LNCap, and PC3 (prostate carcinoma), SW480, and HCT116 (colon carcinoma), in that in the culture medium, it leads to the accumulation of hydrogen peroxide, acting as a prooxidant-anti-tumor [50]. In addition, HT has been seen to lead to the inhibition of signaling pathways such as Akt, STAT3, and EGFR in pancreatic, prostate, and thyroid cancer cell lines [50,51].

2.5.4. Antimicrobial Proprieties

In the past, microbial infections were cured by olive leaf extracts and olive oil. Properly in the leaves, there is a high concentration of HT. In fact, it has potent antioxidant activity against some microorganisms such as Escherichia coli, Candida albicans, Clostridium perfringens, Streptococcus mutans, and Salmonella enterica [27]. Furthermore, Bisignano et al. have highlighted that HT acts as an antibacterial, but it also has an antiparasitic effect [10]. The mode by which HT acts is to reduce ROS production, which is induced by microbial biofilms. There are contradictions regarding the effects of HT against bacteria and parasites, so further studies are needed.

2.5.5. Other Proprieties of Hydroxytyrosol

Finally, several studies have identified other protective effects of HT important for human health. It was demonstrated that HT limits the development of atherosclerotic plaque, protects against LDL oxidation, and reduces the risk of cardiovascular disease [10]. The cardioprotective effect carried out by HT leads to the inhibition of aging-related proteins in cardiac cells [52]. Furthermore, HT is also neuroprotective, as it can cross the BBB and diffuses freely into central neurons. Here, it appears to inhibit monoamine oxidase (MAO) in patients with Parkinson’s disease, protecting against neurodegeneration [6,53,54]. Finally, HT protects against UV radiation, thus having a dermoprotective effect. Skin is exposed daily to ultraviolet radiation (UVR), increasing the formation of free radicals. Zwane et al. showed that HT and its metabolites act as radical scavengers for skin cells, and in addition, HT significantly reduces DNA breakdown caused by ultraviolet B [10]. Therefore, HT may exert different roles in chronic diseases.

3. Role of Hydroxytyrosol in IBDs

IBD is a global healthcare problem with a sustained increasing incidence, affecting millions of people worldwide, all ages and genders [55,56]. It includes two major forms, Ulcerative Colitis, and Crohn’s Disease, which are distinct chronic inflammatory disorders of the gastrointestinal tract with differences in the pathology and clinical characteristics [57,58]. Ulcerative Colitis is typified by mucosal inflammation and is limited to the colon. In contrast, Crohn’s Disease can cause transmural inflammation and affect any part of the gastrointestinal tract (most commonly, the terminal ileum or the perianal region) in a non-continuous type [59,60]. It is unclear what causes IBD, but it is thought to be caused by a combination of factors, including genetic, environmental, and especially dietary variables [61,62,63]. The association between nutrition and IBD has been extensively studied. Dietary fat has been found to play a role in the IBD pathogenesis [64,65,66]; on the contrary, patients with an MD lifestyle, with a high intake of dietary fibers, such as fruits, vegetables, and especially EVOO, are associated with reduced risk for both Ulcerative Colitis and Crohn’s Disease [67,68,69,70]. This is probably because the underlying pathogenetic mechanism of IBD involves the alteration of the redox balance [71,72]; the increased oxidative stress leads to the propagation of inflammation and exacerbation of mucosal damage in the gastrointestinal tract [73,74,75]. Below, we report the in vitro and in vivo findings, along with clinical results, supporting the protective role of HT against IBD due to its antioxidant and anti-inflammatory activity.

3.1. Hydroxytyrosol in Ulcerative Colitis

Ulcerative colitis is one of the most common inflammatory bowel diseases [20], characterized by abdominal and/or rectal pain, diarrhea, bloody stools, weight loss, fever, and even rectal prolapse in severe cases. Ulcerative colitis can also progress to colon cancer [76,77]. It has been widely demonstrated that a dysfunction of the immune system underlies the development of colitis [78,79]. The gastrointestinal tract is the target of various harmful bacteria and substances, and immune cells usually mount an immune-inflammatory response that neutralizes them. However, in ulcerative colitis, this response does not occur [80,81]; this leads to damage to the intestinal mucosa and submucosa (in severe disease, the muscularis is also involved), with consequent loss of integrity of the intestinal barrier [82,83]. The immune response is usually mediated by macrophages and dendritic cells [84]. These cells have TLR receptors on the surface of the membrane, responsible both [85] for the differentiation of T-cells, which in turn stimulate the synthesis of proinflammatory cytokines, but they also induce the activation of the transcription factor NF-κB, further exacerbating the inflammatory process [84,86,87,88]. The key factors involved in the inflammatory cascade during ulcerative colitis are cytokines, such as tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin (IL) -2, IL-6, IL-8, which are also linked to chemokines and adhesion molecules [89,90]. HT has been shown to have protective and immunomodulatory effects against intestinal inflammation in several in vivo and in vitro experimental models [25,91,92]. In particular, HT appears to act by modulating the NF-κB signaling pathway, the consequent release of pro-inflammatory cytokines, and the expression of molecules downstream of the inflammatory cascade, such as COX-2 and iNOS [32,91,93,94,95,96,97]. Another study demonstrated that HT could exert anti-inflammatory effects in DSS-induced ulcerative colitis by inhibiting NLRP3 inflammasome activation and modulating gut microbiota in vivo [98]. Furthermore, ulcerative colitis is associated with a disequilibrium between the oxidant and antioxidant system, giving rise to the overproduction of ROS, which contributes to worsening the complications of the pathology [86,99,100]. In recent years, both in vivo and in vitro studies have been conducted confirming the antioxidant activity of HT also against ulcerative colitis. These effects are probably due to the fact that HT is able to promote Nrf2 nuclear localization [101,102]. The nrf2 signaling pathway is important for maintaining redox homeostasis and defending against ROS by inducing antioxidant enzyme synthesis [103,104]. Indeed, HT can also increase antioxidant enzyme expressions, such as SOD, CAT, and GPx, and at the same time diminish lipid peroxidation marker malondialdehyde (MDA) [93,101,105]. In addition, in vitro studies confirmed the antioxidant effect of HT using human intestinal Caco-2 cells [91,96,106]. Lastly, in a study by Elmaksoud et al., HT has been shown to modulate the apoptotic process by downregulation of the apoptotic gene Bax and upregulation of the anti-apoptotic gene Bcl2 [93]. Randomized crossover clinical trials have also been performed investigating the effect of HT, which is the main component of EVOO, in UC patients [94,107,108]. The promising results from the studies reproduce many of the effects seen in animal and in vitro studies, such as anti-inflammatory effects, gastrointestinal symptom reduction, and positive microbiota changes [32]. However, we must be cautious in translating these findings into a real-life clinical setting.

3.2. Hydroxytyrosol in Crohn’s Disease

As in ulcerative colitis, also in the development of Crohn’s disease, there is an incorrect regulation of the immune system and the response mediated by TLR receptors [109]. The NOD receptors of innate immunity are often found next to the TLR receptors, which under normal conditions, induce the release of NF-κB and cytokines to eliminate agents harmful to the intestinal mucosa [110,111]. In Crohn’s disease, however, these receptors induce a persistent chronic inflammatory condition that leads to excessive release of inflammatory cytokines such as IL-1β, IL-2, IL-3, IL-22, and TFN-α [109]. Furthermore, it is thought that there is also an alteration of the adaptive immune response and B-cells, but this mechanism is not yet fully understood. Certainly, it has been seen and confirmed that the inability of T-cells to undergo apoptosis results in T-cell accumulation and mucosal damage [112]. In the inflammatory environment, HT inhibits the release of TNF-α and proinflammatory mediators, and, in addition, HT also acts on T-cells, as demonstrated by Kawaguchi et al. [113,114]. Additionally, the commonly observed effects to elucidate the molecular basis for HT’s anti-inflammatory activity in Crohn’s disease were inhibition of p38/MAPK and NF-κB signaling pathways and reduction of iNOS expression and NO release [115,116,117]. These results were also confirmed in a study by Vezza et al. [95] that investigated the ex vivo organ cultures of mucosal explants from CD patients, showing a reduction of the proinflammatory mediators’ production. Furthermore, it should be emphasized that an alteration of the redox balance also occurs in Crohn’s disease with excessive production of ROS [73]. In this regard, HT has been defined as a strong antioxidant and free radical scavenger in several studies investigating Crohn’s disease [118,119]. To date, the clinical trials in the literature are preliminary data but are certainly promising at this early stage [40,120].

4. Role of HT in Other Gastrointestinal Diseases

4.1. Hydroxytyrosol in Gastric Ulcer

Gastric ulcer is a disease that affects many people around the world; in most cases, the underlying cause is infection sustained by Helicobacter pylori, long-term use of anti-inflammatory drugs, and other factors such as smoking, alcohol drinking, and dietary [121,122]. The causes mentioned above cause an imbalance in the normal mucosal barrier, altering the production of gastric mucus that acts as a protective layer for the underlying epithelium, from gastric juice and pepsin [123,124,125]. As we have already described above, HT also has an antimicrobial action. The reason why HT has been shown through in vitro studies to have beneficial effects on gastric ulcers as it acts against the proliferation of Helicobacter pylori. Not only that, HT was able to reduce the size of the ulcer as well [32,126,127]. Studies further confirmed that HT inhibits NF-κB transcription, thereby reducing the release of inflammatory cytokines, including TNF-α [128,128]. Furthermore, in an in vivo study by Arismendi Sosa et al., HT demonstrated not only antioxidant and antimicrobial effects but also improved gastric mucosal damage [127]. A number of clinical trials are being developed to demonstrate the anti-inflammatory and antimicrobial effects of HT on gastric ulcers [129].

4.2. Hydroxytyrosol in Colorectal Cancer

Colon-rectal cancer is a disease that affects many people around the world. It is a pathology characterized by a certain aggressiveness due to the ability of the tumor to proliferate and invade the surrounding tissues. Mutations and genetic modifications affecting DNA repair systems underlying the molecular mechanism of colorectal cancer development have been identified [130,131]. In recent years, higher consumption of olive oil, rich in polyphenols such as hydroxytyrosol, has been associated with low incidence and prevalence of cancer, including colorectal cancer [132,133,134,135]. The beneficial action of HT has been evaluated through in vitro studies on epithelial and carcinogenic cell lines: it has been demonstrated that HT was able to act on genes involved in apoptosis, such as Bax and Bcl-2, Caspase-3, and p53 [47,136]. Additionally, recent studies have evidenced that HT inhibits the cell proliferation of colon cancer cells by activating the estrogen receptor-β (ERβ) [51]. In fact, it has been proven that cancerous progression in human colon mucosa is characterized by a reduction in ERβ expression [133]. Finally, Hormozi et al. [47] demonstrated that HT upregulates antioxidative activity in the colorectal cancer cell line through a significant increase of antioxidant enzymes. However, in vivo studies on the therapeutic effects of HT on colorectal cancer in animal models can give better conclusions. Indeed clinical studies have shown how HT affects the mechanisms involved in the development of this type of cancer [137].

4.3. Hydroxytyrosol in Gastroesophageal Reflux Disease and Eosinophilic Esophagitis

Gastroesophageal reflux is a chronic and relapsing disorder occurring when the stomach contents reflux into the esophagus, causing troublesome symptoms that have a negative impact on the quality of life of patients. The cause that leads to the development of this disorder is the dysfunction of the esophageal sphincter, which does not close properly [138,139]. In addition, Barrett’s esophagus (development of columnar metaplasia in place of the normal squamous epithelium), esophageal strictures, and esophageal adenocarcinoma are conditions linked to gastrointestinal reflux [140,141]. Another pathology affecting the esophagus is eosinophilic esophagitis, which is often confused with gastroesophageal reflux. This is a chronic immune-mediated inflammatory disease of the esophagus, still poorly understood, that causes dysphagia, food impaction of the esophagus, and esophageal strictures [142]. Studies have shown that damage to the mucosa of the esophagus leads to a chronic inflammatory condition, with the release of proinflammatory cytokines and often also the formation of fibrotic tissue in both conditions [143,144]. To date, it has been shown that a balanced diet with a high intake of polyphenols can prevent the development of these disorders [145,146,147]. Last year, a double-blinded randomized-controlled trial assessed the efficacy of extracts from O. europaea leaves (rich in bioactive compounds such as HT and tyrosol) in gastrointestinal discomfort, showing a decrease in frequency and intensity of the main symptoms of this disorder, such as abdominal swelling, heartburn, and belching [148]. The protective effects may be due to the active compounds present in the extract, which exert a protective action on gastric mucosa together with the protection from oxidative damage and associated complications and through the modulation of the inflammatory response [148,149,150]. The results of the study by Malfa and colleagues [148] were positive, but further supporting studies are needed.

5. Conclusions

In conclusion, we can state that the main phytoconstituent of the olive tree, HT has excellent potential for use on human health. The various studies carried out so far have shown that HT has excellent bioavailability; in fact, it is easily absorbed and metabolized by the human body. Furthermore, it has been demonstrated, both in vitro and in vivo, that HT is not toxic, genotoxic, or mutagenic. In this review, we have highlighted the potential role of HT in modulating the molecular mechanisms underlying the development of IBDs and gastrointestinal diseases. This is made possible as HT has shown to have a strong antioxidant and free radical scavenger action, as it increases the activity of antioxidant enzymes and restores the oxidative balance. Among its main proprieties, HT also has an anti-inflammatory effect, mainly due to the inhibition of the NF-κB pathway and the release of inflammatory cytokines. Furthermore, it is important to underline that HT has an immunomodulatory and antimicrobial, but also pro-apoptotic and anti-proliferative action, which is why it appears to act against colorectal cancer. This could explain the beneficial effects that HT shows towards these multifactorial pathologies. Therefore, due to its beneficial properties, good safety profile, and numerous protective effects, HT should be further investigated in order to be used in clinical conditions.

Author Contributions

Conceptualization, A.A. and Y.M.; resources, D.I. and R.D.P.; writing—original draft preparation, A.A. and Y.M.; writing—review and editing, R.D.P. and S.C., visualization, R.D. and D.I.; supervision, R.D.P. and S.C.; project administration, S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Keys, A.; Menotti, A.; Karvonen, M.J.; Aravanis, C.; Blackburn, H.; Buzina, R.; Djordjevic, B.S.; Dontas, A.S.; Fidanza, F.; Keys, M.H.; et al. The diet and 15-year death rate in the seven countries study. Am. J. Epidemiol. 1986, 124, 903–915. [Google Scholar] [CrossRef] [PubMed]
  2. Guasch-Ferre, M.; Willett, W.C. The Mediterranean diet and health: A comprehensive overview. J. Intern. Med. 2021, 290, 549–566. [Google Scholar] [CrossRef]
  3. Dinu, M.; Pagliai, G.; Casini, A.; Sofi, F. Mediterranean diet and multiple health outcomes: An umbrella review of meta-analyses of observational studies and randomised trials. Eur. J. Clin. Nutr. 2018, 72, 30–43. [Google Scholar] [CrossRef]
  4. Becerra-Tomas, N.; Blanco Mejia, S.; Viguiliouk, E.; Khan, T.; Kendall, C.W.C.; Kahleova, H.; Rahelic, D.; Sievenpiper, J.L.; Salas-Salvado, J. Mediterranean diet, cardiovascular disease and mortality in diabetes: A systematic review and meta-analysis of prospective cohort studies and randomized clinical trials. Crit. Rev. Food Sci. Nutr. 2020, 60, 1207–1227. [Google Scholar] [CrossRef]
  5. Sanchez-Sanchez, M.L.; Garcia-Vigara, A.; Hidalgo-Mora, J.J.; Garcia-Perez, M.A.; Tarin, J.; Cano, A. Mediterranean diet and health: A systematic review of epidemiological studies and intervention trials. Maturitas 2020, 136, 25–37. [Google Scholar] [CrossRef] [PubMed]
  6. Cordaro, M.; Trovato Salinaro, A.; Siracusa, R.; D’Amico, R.; Impellizzeri, D.; Scuto, M.; Ontario, M.L.; Crea, R.; Cuzzocrea, S.; Di Paola, R.; et al. Hidrox((R)) Roles in Neuroprotection: Biochemical Links between Traumatic Brain Injury and Alzheimer’s Disease. Antioxidants 2021, 10, 818. [Google Scholar] [CrossRef]
  7. Ontario, M.L.; Siracusa, R.; Modafferi, S.; Scuto, M.; Sciuto, S.; Greco, V.; Bertuccio, M.P.; Trovato Salinaro, A.; Crea, R.; Calabrese, E.J.; et al. Potential prevention and treatment of neurodegenerative disorders by olive polyphenols and hidrox. Mech. Ageing. Dev. 2022, 203, 111637. [Google Scholar] [CrossRef] [PubMed]
  8. Takeda, Y.; Jamsransuren, D.; Matsuda, S.; Crea, R.; Ogawa, H. The SARS-CoV-2-Inactivating Activity of Hydroxytyrosol-Rich Aqueous Olive Pulp Extract (HIDROX((R))) and Its Use as a Virucidal Cream for Topical Application. Viruses 2021, 13, 232. [Google Scholar] [CrossRef]
  9. Riolo, R.; De Rosa, R.; Simonetta, I.; Tuttolomondo, A. Olive Oil in the Mediterranean Diet and Its Biochemical and Molecular Effects on Cardiovascular Health through an Analysis of Genetics and Epigenetics. Int. J. Mol. Sci. 2022, 23, 6002. [Google Scholar] [CrossRef]
  10. Robles-Almazan, M.; Pulido-Moran, M.; Moreno-Fernandez, J.; Ramirez-Tortosa, C.; Rodriguez-Garcia, C.; Quiles, J.L.; Ramirez-Tortosa, M. Hydroxytyrosol: Bioavailability, toxicity, and clinical applications. Food. Res. Int. 2018, 105, 654–667. [Google Scholar] [CrossRef] [PubMed]
  11. Fusco, R.; Cordaro, M.; Siracusa, R.; Peritore, A.F.; Gugliandolo, E.; Genovese, T.; D’Amico, R.; Crupi, R.; Smeriglio, A.; Mandalari, G.; et al. Consumption of Anacardium occidentale L. (Cashew Nuts) Inhibits Oxidative Stress through Modulation of the Nrf2/HO-1 and NF-kB Pathways. Molecules 2020, 25, 4426. [Google Scholar] [CrossRef] [PubMed]
  12. Cordaro, M.; Siracusa, R.; Fusco, R.; D’Amico, R.; Peritore, A.F.; Gugliandolo, E.; Genovese, T.; Scuto, M.; Crupi, R.; Mandalari, G.; et al. Cashew (Anacardium occidentale L.) Nuts Counteract Oxidative Stress and Inflammation in an Acute Experimental Model of Carrageenan-Induced Paw Edema. Antioxidants 2020, 9, 660. [Google Scholar] [CrossRef]
  13. Cordaro, M.; Fusco, R.; D’Amico, R.; Siracusa, R.; Peritore, A.F.; Gugliandolo, E.; Genovese, T.; Crupi, R.; Mandalari, G.; Cuzzocrea, S.; et al. Cashew (Anacardium occidentale L.) Nuts Modulate the Nrf2 and NLRP3 Pathways in Pancreas and Lung after Induction of Acute Pancreatitis by Cerulein. Antioxidants 2020, 9, 992. [Google Scholar] [CrossRef] [PubMed]
  14. Castejon, M.L.; Montoya, T.; Ortega-Vidal, J.; Altarejos, J.; Alarcon-de-la-Lastra, C. Ligstroside aglycon, an extra virgin olive oil secoiridoid, prevents inflammation by regulation of MAPKs, JAK/STAT, NF-kappaB, Nrf2/HO-1, and NLRP3 inflammasome signaling pathways in LPS-stimulated murine peritoneal macrophages. Food Funct. 2022, 13, 10200–10209. [Google Scholar] [CrossRef]
  15. Loganathan, R.; Nagapan, G.; Teng, K.T.; Voon, P.T.; Yap, S.Y.; Ng, Y.T.; Ng, T.K.W.; Choo, Y.M.; Ong, A.S.H.; Ong, S.H.; et al. Diets enriched with palm olein, cocoa butter, and extra virgin olive oil exhibited similar lipid response: A randomized controlled study in young healthy adults. Nutr. Res. 2022, 105, 113–125. [Google Scholar] [CrossRef] [PubMed]
  16. Abdoli, A.; Shahbazi, R.; Zoghi, G.; Davoodian, P.; Kheirandish, S.; Azad, M.; Kheirandish, M. The effect of topical olive oil dressing on the healing of grade 1 and 2 diabetic foot ulcers: An assessor-blind randomized controlled trial in type 2 diabetes patients. Diabetes Metab. Syndr. 2022, 16, 102678. [Google Scholar] [CrossRef]
  17. Gangwar, M.; Gautam, M.K.; Sharma, A.K.; Tripathi, Y.B.; Goel, R.K.; Nath, G. Antioxidant capacity and radical scavenging effect of polyphenol rich Mallotus philippenensis fruit extract on human erythrocytes: An in vitro study. Sci. World J. 2014, 2014, 279451. [Google Scholar] [CrossRef]
  18. Platzer, M.; Kiese, S.; Tybussek, T.; Herfellner, T.; Schneider, F.; Schweiggert-Weisz, U.; Eisner, P. Radical Scavenging Mechanisms of Phenolic Compounds: A Quantitative Structure-Property Relationship (QSPR) Study. Front. Nutr. 2022, 9, 882458. [Google Scholar] [CrossRef]
  19. Bertelli, A.; Biagi, M.; Corsini, M.; Baini, G.; Cappellucci, G.; Miraldi, E. Polyphenols: From Theory to Practice. Foods 2021, 10, 2595. [Google Scholar] [CrossRef]
  20. Siracusa, R.; Fusco, R.; Peritore, A.F.; Cordaro, M.; D’Amico, R.; Genovese, T.; Gugliandolo, E.; Crupi, R.; Smeriglio, A.; Mandalari, G.; et al. The Antioxidant and Anti-Inflammatory Properties of Anacardium occidentale L. Cashew Nuts in a Mouse Model of Colitis. Nutrients 2020, 12, 834. [Google Scholar] [CrossRef] [Green Version]
  21. Fusco, R.; Siracusa, R.; Peritore, A.F.; Gugliandolo, E.; Genovese, T.; D’Amico, R.; Cordaro, M.; Crupi, R.; Mandalari, G.; Impellizzeri, D.; et al. The Role of Cashew (Anacardium occidentale L.) Nuts on an Experimental Model of Painful Degenerative Joint Disease. Antioxidants 2020, 9, 511. [Google Scholar] [CrossRef]
  22. Costa, V.; Costa, M.; Videira, R.A.; Andrade, P.B.; Paiva-Martins, F. Anti-Inflammatory Activity of Olive Oil Polyphenols-The Role of Oleacein and Its Metabolites. Biomedicines 2022, 10, 2990. [Google Scholar] [CrossRef] [PubMed]
  23. Schwingshackl, L.; Morze, J.; Hoffmann, G. Mediterranean diet and health status: Active ingredients and pharmacological mechanisms. Br. J. Pharm. 2020, 177, 1241–1257. [Google Scholar] [CrossRef] [PubMed]
  24. D’Amico, R.; Cordaro, M.; Fusco, R.; Peritore, A.F.; Genovese, T.; Gugliandolo, E.; Crupi, R.; Mandalari, G.; Caccamo, D.; Cuzzocrea, S.; et al. Consumption of Cashew (Anacardium occidentale L.) Nuts Counteracts Oxidative Stress and Tissue Inflammation in Mild Hyperhomocysteinemia in Rats. Nutrients 2022, 14, 1474. [Google Scholar] [CrossRef] [PubMed]
  25. Karković Marković, A.; Torić, J.; Barbarić, M.; Jakobušić Brala, C. Hydroxytyrosol, Tyrosol and Derivatives and Their Potential Effects on Human Health. Molecules 2019, 24, 2001. [Google Scholar] [CrossRef] [PubMed]
  26. Martinez, L.; Ros, G.; Nieto, G. Hydroxytyrosol: Health Benefits and Use as Functional Ingredient in Meat. Medicines 2018, 5, 13. [Google Scholar] [CrossRef] [PubMed]
  27. Bertelli, M.; Kiani, A.K.; Paolacci, S.; Manara, E.; Kurti, D.; Dhuli, K.; Bushati, V.; Miertus, J.; Pangallo, D.; Baglivo, M.; et al. Hydroxytyrosol: A natural compound with promising pharmacological activities. J. Biotechnol. 2020, 309, 29–33. [Google Scholar] [CrossRef] [PubMed]
  28. D’Amico, R.; Cordaro, M.; Siracusa, R.; Impellizzeri, D.; Trovato Salinaro, A.; Scuto, M.; Ontario, M.L.; Crea, R.; Cuzzocrea, S.; Di Paola, R.; et al. Wnt/beta-Catenin Pathway in Experimental Model of Fibromyalgia: Role of Hidrox((R)). Biomedicines 2021, 9, 1683. [Google Scholar] [CrossRef]
  29. Martínez-Zamora, L.; Peñalver, R.; Ros, G.; Nieto, G. Olive Tree Derivatives and Hydroxytyrosol: Their Potential Effects on Human Health and Its Use as Functional Ingredient in Meat. Foods 2021, 10, 2611. [Google Scholar] [CrossRef]
  30. Piroddi, M.; Albini, A.; Fabiani, R.; Giovannelli, L.; Luceri, C.; Natella, F.; Rosignoli, P.; Rossi, T.; Taticchi, A.; Servili, M.; et al. Nutrigenomics of extra-virgin olive oil: A review. Biofactors 2017, 43, 17–41. [Google Scholar] [CrossRef]
  31. Tome-Carneiro, J.; Crespo, M.C.; Iglesias-Gutierrez, E.; Martin, R.; Gil-Zamorano, J.; Tomas-Zapico, C.; Burgos-Ramos, E.; Correa, C.; Gomez-Coronado, D.; Lasuncion, M.A.; et al. Hydroxytyrosol supplementation modulates the expression of miRNAs in rodents and in humans. J. Nutr. Biochem. 2016, 34, 146–155. [Google Scholar] [CrossRef] [PubMed]
  32. Vrdoljak, J.; Kumric, M.; Vilovic, M.; Martinovic, D.; Tomic, I.J.; Krnic, M.; Ticinovic Kurir, T.; Bozic, J. Effects of Olive Oil and Its Components on Intestinal Inflammation and Inflammatory Bowel Disease. Nutrients 2022, 14, 757. [Google Scholar] [CrossRef]
  33. Estevinho, M.M.; Leao Moreira, P.; Silva, I.; Laranjeira Correia, J.; Santiago, M.; Magro, F. A scoping review on early inflammatory bowel disease: Definitions, pathogenesis, and impact on clinical outcomes. Ther. Adv. Gastroenterol. 2022, 15, 17562848221142673. [Google Scholar] [CrossRef]
  34. Ramírez-Tortose, M.d.C.; Pulido-Moran, M.; Granados, S.; Gaforio, J.J.; Quiles, J.L. Chapter 20—Hydroxytyrosol as a Component of the Mediterranean Diet and Its Role in Disease Prevention. In The Mediterranean Diet; Preedy, V.R., Watson, R.R., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 205–215. [Google Scholar] [CrossRef]
  35. Gorzynik-Debicka, M.; Przychodzen, P.; Cappello, F.; Kuban-Jankowska, A.; Marino Gammazza, A.; Knap, N.; Wozniak, M.; Gorska-Ponikowska, M. Potential Health Benefits of Olive Oil and Plant Polyphenols. Int. J. Mol. Sci. 2018, 19, 686. [Google Scholar] [CrossRef] [PubMed]
  36. Gallardo-Fernández, M.; Gonzalez-Ramirez, M.; Cerezo, A.B.; Troncoso, A.M.; Garcia-Parrilla, M.C. Hydroxytyrosol in Foods: Analysis, Food Sources, EU Dietary Intake, and Potential Uses. Foods 2022, 11, 2355. [Google Scholar] [CrossRef]
  37. Nieto, G.; Martínez, L.; Castillo, J.; Ros, G. Hydroxytyrosol extracts, olive oil and walnuts as functional components in chicken sausages. J. Sci. Food Agric. 2017, 97, 3761–3771. [Google Scholar] [CrossRef] [PubMed]
  38. Papageorgiou, C.S.; Lyri, P.; Xintaropoulou, I.; Diamantopoulos, I.; Zagklis, D.P.; Paraskeva, C.A. High-Yield Production of a Rich-in-Hydroxytyrosol Extract from Olive (Olea europaea) Leaves. Antioxidants 2022, 11, 1042. [Google Scholar] [CrossRef]
  39. Dominguez-Perles, R.; Aunon, D.; Ferreres, F.; Gil-Izquierdo, A. Gender differences in plasma and urine metabolites from Sprague-Dawley rats after oral administration of normal and high doses of hydroxytyrosol, hydroxytyrosol acetate, and DOPAC. Eur. J. Nutr. 2017, 56, 215–224. [Google Scholar] [CrossRef]
  40. Fernández-Mar, M.I.; Mateos, R.; García-Parrilla, M.C.; Puertas, B.; Cantos-Villar, E. Bioactive compounds in wine: Resveratrol, hydroxytyrosol and melatonin: A review. Food Chem. 2012, 130, 797–813. [Google Scholar] [CrossRef]
  41. D’Angelo, S.; Manna, C.; Migliardi, V.; Mazzoni, O.; Morrica, P.; Capasso, G.; Pontoni, G.; Galletti, P.; Zappia, V. Pharmacokinetics and metabolism of hydroxytyrosol, a natural antioxidant from olive oil. Drug. Metab. Dispos. 2001, 29, 1492–1498. [Google Scholar]
  42. Cordaro, M.; Trovato Salinaro, A.; Siracusa, R.; D’Amico, R.; Impellizzeri, D.; Scuto, M.; Ontario, M.L.; Interdonato, L.; Crea, R.; Fusco, R.; et al. Hidrox((R)) and Endometriosis: Biochemical Evaluation of Oxidative Stress and Pain. Antioxidants 2021, 10, 720. [Google Scholar] [CrossRef] [PubMed]
  43. Fusco, R.; Salinaro, A.T.; Siracusa, R.; D’Amico, R.; Impellizzeri, D.; Scuto, M.; Ontario, M.L.; Crea, R.; Cordaro, M.; Cuzzocrea, S.; et al. Hidrox((R)) Counteracts Cyclophosphamide-Induced Male Infertility through NRF2 Pathways in a Mouse Model. Antioxidants 2021, 10, 778. [Google Scholar] [CrossRef] [PubMed]
  44. Rietjens, S.J.; Bast, A.; Haenen, G.R.M.M. New Insights into Controversies on the Antioxidant Potential of the Olive Oil Antioxidant Hydroxytyrosol. J. Agric. Food Chem. 2007, 55, 7609–7614. [Google Scholar] [CrossRef]
  45. D’Amico, R.; Trovato Salinaro, A.; Cordaro, M.; Fusco, R.; Impellizzeri, D.; Interdonato, L.; Scuto, M.; Ontario, M.L.; Crea, R.; Siracusa, R.; et al. Hidrox((R)) and Chronic Cystitis: Biochemical Evaluation of Inflammation, Oxidative Stress, and Pain. Antioxidants 2021, 10, 1046. [Google Scholar] [CrossRef] [PubMed]
  46. Ramírez-Expósito, M.J.; Martínez-Martos, J.M. Anti-Inflammatory and Antitumor Effects of Hydroxytyrosol but Not Oleuropein on Experimental Glioma In Vivo. A Putative Role for the Renin-Angiotensin System. Biomedicines 2018, 6, 11. [Google Scholar] [CrossRef] [PubMed]
  47. Hormozi, M.; Salehi Marzijerani, A.; Baharvand, P. Effects of Hydroxytyrosol on Expression of Apoptotic Genes and Activity of Antioxidant Enzymes in LS180 Cells. Cancer. Manag. Res. 2020, 12, 7913–7919. [Google Scholar] [CrossRef] [PubMed]
  48. Fabiani, R.; Rosignoli, P.; De Bartolomeo, A.; Fuccelli, R.; Morozzi, G. Inhibition of cell cycle progression by hydroxytyrosol is associated with upregulation of cyclin-dependent protein kinase inhibitors p21(WAF1/Cip1) and p27(Kip1) and with induction of differentiation in HL60 cells. J. Nutr. 2008, 138, 42–48. [Google Scholar] [CrossRef]
  49. Chimento, A.; Casaburi, I.; Rosano, C.; Avena, P.; De Luca, A.; Campana, C.; Martire, E.; Santolla, M.F.; Maggiolini, M.; Pezzi, V.; et al. Oleuropein and hydroxytyrosol activate GPER/GPR30-dependent pathways leading to apoptosis of ER-negative SKBR3 breast cancer cells. Mol. Nutr. Food Res. 2014, 58, 478–489. [Google Scholar] [CrossRef]
  50. Zubair, H.; Bhardwaj, A.; Ahmad, A.; Srivastava, S.K.; Khan, M.A.; Patel, G.K.; Singh, S.; Singh, A.P. Hydroxytyrosol Induces Apoptosis and Cell Cycle Arrest and Suppresses Multiple Oncogenic Signaling Pathways in Prostate Cancer Cells. Nutr. Cancer 2017, 69, 932–942. [Google Scholar] [CrossRef]
  51. Terzuoli, E.; Giachetti, A.; Ziche, M.; Donnini, S. Hydroxytyrosol, a product from olive oil, reduces colon cancer growth by enhancing epidermal growth factor receptor degradation. Mol. Nutr. Food Res. 2016, 60, 519–529. [Google Scholar] [CrossRef]
  52. De Pablos, R.M.; Espinosa-Oliva, A.M.; Hornedo-Ortega, R.; Cano, M.; Arguelles, S. Hydroxytyrosol protects from aging process via AMPK and autophagy; a review of its effects on cancer, metabolic syndrome, osteoporosis, immune-mediated and neurodegenerative diseases. Pharmacol. Res. 2019, 143, 58–72. [Google Scholar] [CrossRef]
  53. Goldstein, D.S.; Jinsmaa, Y.; Sullivan, P.; Holmes, C.; Kopin, I.J.; Sharabi, Y. 3,4-Dihydroxyphenylethanol (Hydroxytyrosol) Mitigates the Increase in Spontaneous Oxidation of Dopamine During Monoamine Oxidase Inhibition in PC12 Cells. Neurochem. Res. 2016, 41, 2173–2178. [Google Scholar] [CrossRef] [PubMed]
  54. De La Cruz Cortés, J.P.; Pérez de Algaba, I.; Martín-Aurioles, E.; Arrebola, M.M.; Ortega-Hombrados, L.; Rodríguez-Pérez, M.D.; Fernández-Prior, M.; Bermúdez-Oria, A.; Verdugo, C.; González-Correa, J.A. Extra Virgin Oil Polyphenols Improve the Protective Effects of Hydroxytyrosol in an In Vitro Model of Hypoxia-Reoxygenation of Rat Brain. Brain Sci. 2021, 11, 1133. [Google Scholar] [CrossRef] [PubMed]
  55. Ye, Y.; Pang, Z.; Chen, W.; Ju, S.; Zhou, C. The epidemiology and risk factors of inflammatory bowel disease. Int. J. Clin. Exp. Med. 2015, 8, 22529–22542. [Google Scholar] [PubMed]
  56. Di Paola, R.; Fusco, R.; Gugliandolo, E.; D’Amico, R.; Cordaro, M.; Impellizzeri, D.; Perretti, M.; Cuzzocrea, S. Formyl peptide receptor 1 signalling promotes experimental colitis in mice. Pharm. Res. 2019, 141, 591–601. [Google Scholar] [CrossRef] [PubMed]
  57. Seyedian, S.S.; Nokhostin, F.; Malamir, M.D. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. J. Med. Life 2019, 12, 113–122. [Google Scholar] [CrossRef]
  58. Yusuf, K.; Saha, S.; Umar, S. Health Benefits of Dietary Fiber for the Management of Inflammatory Bowel Disease. Biomedicines 2022, 10, 1242. [Google Scholar] [CrossRef] [PubMed]
  59. Kim, S.K.; Lee, H.S.; Kim, B.J.; Park, J.H.; Hwang, S.W.; Yang, D.H.; Ye, B.D.; Byeon, J.S.; Myung, S.J.; Yang, S.K.; et al. The Clinical Features of Inflammatory Bowel Disease in Patients with Obesity. Can. J. Gastroenterol. Hepatol. 2021, 2021, 9981482. [Google Scholar] [CrossRef]
  60. Li, S.; Zhang, F.; Zhang, Q. Pathological features-based targeted delivery strategies in IBD therapy: A mini review. Biomed. Pharm. 2022, 151, 113079. [Google Scholar] [CrossRef]
  61. Krela-Kazmierczak, I.; Zakerska-Banaszak, O.; Skrzypczak-Zielinska, M.; Lykowska-Szuber, L.; Szymczak-Tomczak, A.; Zawada, A.; Rychter, A.M.; Ratajczak, A.E.; Skoracka, K.; Skrzypczak, D.; et al. Where Do We Stand in the Behavioral Pathogenesis of Inflammatory Bowel Disease? The Western Dietary Pattern and Microbiota-A Narrative Review. Nutrients 2022, 14, 2520. [Google Scholar] [CrossRef]
  62. 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]
  63. Cui, G.; Yuan, A. A Systematic Review of Epidemiology and Risk Factors Associated With Chinese Inflammatory Bowel Disease. Front. Med. 2018, 5, 183. [Google Scholar] [CrossRef] [PubMed]
  64. Pieczynska, J.; Prescha, A.; Zablocka-Slowinska, K.; Neubauer, K.; Smereka, A.; Grajeta, H.; Biernat, J.; Paradowski, L. Occurrence of dietary risk factors in inflammatory bowel disease: Influence on the nutritional status of patients in clinical remission. Adv. Clin. Exp. Med. 2019, 28, 587–592. [Google Scholar] [CrossRef]
  65. Li, X.; Wei, X.; Sun, Y.; Du, J.; Li, X.; Xun, Z.; Li, Y.C. High-fat diet promotes experimental colitis by inducing oxidative stress in the colon. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G453–G462. [Google Scholar] [CrossRef]
  66. Lee, Y.H.; Kim, H.; Nam, S.; Chu, J.R.; Kim, J.H.; Lim, J.S.; Kim, S.E.; Sung, M.K. Protective Effects of High-Fat Diet against Murine Colitis in Association with Leptin Signaling and Gut Microbiome. Life 2022, 12, 972. [Google Scholar] [CrossRef]
  67. Quintana-Navarro, G.M.; Alcala-Diaz, J.F.; Lopez-Moreno, J.; Perez-Corral, I.; Leon-Acuna, A.; Torres-Pena, J.D.; Rangel-Zuniga, O.A.; Arenas de Larriva, A.P.; Corina, A.; Camargo, A.; et al. Long-term dietary adherence and changes in dietary intake in coronary patients after intervention with a Mediterranean diet or a low-fat diet: The CORDIOPREV randomized trial. Eur. J. Nutr. 2020, 59, 2099–2110. [Google Scholar] [CrossRef]
  68. Chicco, F.; Magri, S.; Cingolani, A.; Paduano, D.; Pesenti, M.; Zara, F.; Tumbarello, F.; Urru, E.; Melis, A.; Casula, L.; et al. Multidimensional Impact of Mediterranean Diet on IBD Patients. Inflamm. Bowel Dis. 2021, 27, 1–9. [Google Scholar] [CrossRef] [PubMed]
  69. Vrdoljak, J.; Vilovic, M.; Zivkovic, P.M.; Tadin Hadjina, I.; Rusic, D.; Bukic, J.; Borovac, J.A.; Bozic, J. Mediterranean Diet Adherence and Dietary Attitudes in Patients with Inflammatory Bowel Disease. Nutrients 2020, 12, 3429. [Google Scholar] [CrossRef]
  70. Strisciuglio, C.; Cenni, S.; Serra, M.R.; Dolce, P.; Kolacek, S.; Sila, S.; Trivic, I.; Bar Lev, M.R.; Shamir, R.; Kostovski, A.; et al. Diet and Pediatric Functional Gastrointestinal Disorders in Mediterranean Countries. Nutrients 2022, 14, 2335. [Google Scholar] [CrossRef] [PubMed]
  71. Kloska, M.; Mankowska-Wierzbicka, D.; Czlapka-Matyasik, M.; Dobrowolska, A.; Grzymislawski, M. Oxidative stress in etiopathogenesis of inflammatory bowel diseases. Postep. Biochem. 2020, 66, 143–150. [Google Scholar] [CrossRef]
  72. Rezaie, A.; Parker, R.D.; Abdollahi, M. Oxidative stress and pathogenesis of inflammatory bowel disease: An epiphenomenon or the cause? Dig. Dis. Sci. 2007, 52, 2015–2021. [Google Scholar] [CrossRef] [PubMed]
  73. Alemany-Cosme, E.; Saez-Gonzalez, E.; Moret, I.; Mateos, B.; Iborra, M.; Nos, P.; Sandoval, J.; Beltran, B. Oxidative Stress in the Pathogenesis of Crohn’s Disease and the Interconnection with Immunological Response, Microbiota, External Environmental Factors, and Epigenetics. Antioxidants 2021, 10, 64. [Google Scholar] [CrossRef]
  74. Gugliandolo, E.; Cordaro, M.; Siracusa, R.; D’Amico, R.; Peritore, A.F.; Genovese, T.; Impellizzeri, D.; Paola, R.D.; Crupi, R.; Cuzzocrea, S.; et al. Novel Combination of COX-2 Inhibitor and Antioxidant Therapy for Modulating Oxidative Stress Associated with Intestinal Ischemic Reperfusion Injury and Endotoxemia. Antioxidants 2020, 9, 930. [Google Scholar] [CrossRef] [PubMed]
  75. Cuzzocrea, S.; Mazzon, E.; Di Paola, R.; Patel, N.S.; Genovese, T.; Muia, C.; De Sarro, A.; Thiemermann, C. Erythropoietin reduces the development of experimental inflammatory bowel disease. J. Pharm. Exp. Ther. 2004, 311, 1272–1280. [Google Scholar] [CrossRef] [PubMed]
  76. Adams, S.M.; Close, E.D.; Shreenath, A.P. Ulcerative Colitis: Rapid Evidence Review. Am. Fam. Physician 2022, 105, 406–411. [Google Scholar]
  77. Li, S.; Wu, B.; Fu, W.; Reddivari, L. The anti-inflammatory effects of dietary anthocyanins against ulcerative colitis. Int. J. Mol. Sci. 2019, 20, 2588. [Google Scholar] [CrossRef] [PubMed]
  78. Kaluzna, A.; Olczyk, P.; Komosinska-Vassev, K. The Role of Innate and Adaptive Immune Cells in the Pathogenesis and Development of the Inflammatory Response in Ulcerative Colitis. J. Clin. Med. 2022, 11, 400. [Google Scholar] [CrossRef]
  79. Gao, X.; Cao, Q.; Cheng, Y.; Zhao, D.; Wang, Z.; Yang, H.; Wu, Q.; You, L.; Wang, Y.; Lin, Y.; et al. Chronic stress promotes colitis by disturbing the gut microbiota and triggering immune system response. Proc. Natl. Acad. Sci. USA 2018, 115, E2960–E2969. [Google Scholar] [CrossRef]
  80. Kucharzik, T.; Koletzko, S.; Kannengiesser, K.; Dignass, A. Ulcerative Colitis-Diagnostic and Therapeutic Algorithms. Dtsch. Arztebl. Int. 2020, 117, 564–574. [Google Scholar] [CrossRef]
  81. Du, L.; Ha, C. Epidemiology and Pathogenesis of Ulcerative Colitis. Gastroenterol. Clin. 2020, 49, 643–654. [Google Scholar] [CrossRef]
  82. Cordaro, M.; Scuto, M.; Siracusa, R.; D’Amico, R.; Filippo Peritore, A.; Gugliandolo, E.; Fusco, R.; Crupi, R.; Impellizzeri, D.; Pozzebon, M.; et al. Effect of N-palmitoylethanolamine-oxazoline on comorbid neuropsychiatric disturbance associated with inflammatory bowel disease. FASEB J. 2020, 34, 4085–4106. [Google Scholar] [CrossRef] [PubMed]
  83. Rapa, S.F.; Di Paola, R.; Cordaro, M.; Siracusa, R.; D’Amico, R.; Fusco, R.; Autore, G.; Cuzzocrea, S.; Stuppner, H.; Marzocco, S. Plumericin Protects against Experimental Inflammatory Bowel Disease by Restoring Intestinal Barrier Function and Reducing Apoptosis. Biomedicines 2021, 9, 67. [Google Scholar] [CrossRef] [PubMed]
  84. Segal, J.P.; LeBlanc, J.F.; Hart, A.L. Ulcerative colitis: An update. Clin. Med. 2021, 21, 135–139. [Google Scholar] [CrossRef] [PubMed]
  85. Bullard, B.M.; VanderVeen, B.N.; McDonald, S.J.; Cardaci, T.D.; Murphy, E.A. Cross talk between the gut microbiome and host immune response in ulcerative colitis: Nonpharmacological strategies to improve homeostasis. Am. J. Physiol. Gastrointest. Liver Physiol. 2022, 323, G554–G561. [Google Scholar] [CrossRef]
  86. Balmus, I.M.; Ciobica, A.; Trifan, A.; Stanciu, C. The implications of oxidative stress and antioxidant therapies in Inflammatory Bowel Disease: Clinical aspects and animal models. Saudi. J. Gastroenterol. Off. J. Saudi. Gastroenterol. Assoc. 2016, 22, 3. [Google Scholar] [CrossRef] [PubMed]
  87. Impellizzeri, D.; Fusco, R.; Genovese, T.; Cordaro, M.; D’Amico, R.; Trovato Salinaro, A.; Ontario, M.L.; Modafferi, S.; Cuzzocrea, S.; Di Paola, R.; et al. Coriolus Versicolor Downregulates TLR4/NF-kappaB Signaling Cascade in Dinitrobenzenesulfonic Acid-Treated Mice: A Possible Mechanism for the Anti-Colitis Effect. Antioxidants 2022, 11, 406. [Google Scholar] [CrossRef]
  88. Wang, Z.; Li, S.; Cao, Y.; Tian, X.; Zeng, R.; Liao, D.-F.; Cao, D. Oxidative stress and carbonyl lesions in ulcerative colitis and associated colorectal cancer. Oxidative Med. Cell Longev. 2016, 2016, 9875298. [Google Scholar] [CrossRef] [PubMed]
  89. Rapa, S.F.; Waltenberger, B.; Di Paola, R.; Adesso, S.; Siracusa, R.; Peritore, A.F.; D’Amico, R.; Autore, G.; Cuzzocrea, S.; Stuppner, H.; et al. Plumericin prevents intestinal inflammation and oxidative stress in vitro and in vivo. FASEB J. 2020, 34, 1576–1590. [Google Scholar] [CrossRef]
  90. Peritore, A.F.; D’Amico, R.; Cordaro, M.; Siracusa, R.; Fusco, R.; Gugliandolo, E.; Genovese, T.; Crupi, R.; Di Paola, R.; Cuzzocrea, S.; et al. PEA/Polydatin: Anti-Inflammatory and Antioxidant Approach to Counteract DNBS-Induced Colitis. Antioxidants 2021, 10, 464. [Google Scholar] [CrossRef]
  91. Reddy, K.V.K.; Naidu, K.A. Oleic acid, hydroxytyrosol and n-3 fatty acids collectively modulate colitis through reduction of oxidative stress and IL-8 synthesis; in vitro and in vivo studies. Int. Immunopharmacol. 2016, 35, 29–42. [Google Scholar] [CrossRef]
  92. Lee, S.H.; Kwon, J.E.; Cho, M.L. Immunological pathogenesis of inflammatory bowel disease. Intest. Res. 2018, 16, 26–42. [Google Scholar] [CrossRef] [Green Version]
  93. Elmaksoud, H.A.A.; Motawea, M.H.; Desoky, A.A.; Elharrif, M.G.; Ibrahimi, A. Hydroxytyrosol alleviate intestinal inflammation, oxidative stress and apoptosis resulted in ulcerative colitis. Biomed. Pharm. 2021, 142, 112073. [Google Scholar] [CrossRef]
  94. Morvaridi, M.; Jafarirad, S.; Seyedian, S.S.; Alavinejad, P.; Cheraghian, B. The effects of extra virgin olive oil and canola oil on inflammatory markers and gastrointestinal symptoms in patients with ulcerative colitis. Eur. J. Clin. Nutr. 2020, 74, 891–899. [Google Scholar] [CrossRef] [PubMed]
  95. Vezza, T.; Algieri, F.; Rodriguez-Nogales, A.; Garrido-Mesa, J.; Utrilla, M.P.; Talhaoui, N.; Gomez-Caravaca, A.M.; Segura-Carretero, A.; Rodriguez-Cabezas, M.E.; Monteleone, G.; et al. Immunomodulatory properties of Olea europaea leaf extract in intestinal inflammation. Mol. Nutr. Food Res. 2017, 61, 201601066. [Google Scholar] [CrossRef]
  96. Serreli, G.; Melis, M.P.; Corona, G.; Deiana, M. Modulation of LPS-induced nitric oxide production in intestinal cells by hydroxytyrosol and tyrosol metabolites: Insight into the mechanism of action. Food Chem. Toxicol. 2019, 125, 520–527. [Google Scholar] [CrossRef] [PubMed]
  97. Wark, G.; Samocha-Bonet, D.; Ghaly, S.; Danta, M. The Role of Diet in the Pathogenesis and Management of Inflammatory Bowel Disease: A Review. Nutrients 2021, 13, 135. [Google Scholar] [CrossRef] [PubMed]
  98. Miao, F. Hydroxytyrosol alleviates dextran sodium sulfate-induced colitis by inhibiting NLRP3 inflammasome activation and modulating gut microbiota in vivo. Nutrition 2022, 97, 111579. [Google Scholar] [CrossRef] [PubMed]
  99. Bourgonje, A.R.; Feelisch, M.; Faber, K.N.; Pasch, A.; Dijkstra, G.; van Goor, H. Oxidative Stress and Redox-Modulating Therapeutics in Inflammatory Bowel Disease. Trends Mol. Med. 2020, 26, 1034–1046. [Google Scholar] [CrossRef] [PubMed]
  100. Yuksel, M.; Ates, I.; Kaplan, M.; Arikan, M.F.; Ozin, Y.O.; Kilic, Z.M.Y.; Topcuoglu, C.; Kayacetin, E. Is Oxidative Stress Associated with Activation and Pathogenesis of Inflammatory Bowel Disease? J. Med. Biochem. 2017, 36, 341–348. [Google Scholar] [CrossRef]
  101. Wang, Q.; Wang, C.; Abdullah; Tian, W.; Qiu, Z.; Song, M.; Cao, Y.; Xiao, J. Hydroxytyrosol Alleviates Dextran Sulfate Sodium-Induced Colitis by Modulating Inflammatory Responses, Intestinal Barrier, and Microbiome. J. Agric. Food Chem. 2022, 70, 2241–2252. [Google Scholar] [CrossRef]
  102. Fuccelli, R.; Fabiani, R.; Rosignoli, P. Hydroxytyrosol Exerts Anti-Inflammatory and Anti-Oxidant Activities in a Mouse Model of Systemic Inflammation. Molecules 2018, 23, 3212. [Google Scholar] [CrossRef] [Green Version]
  103. Fusco, R.; Cordaro, M.; Siracusa, R.; D’Amico, R.; Genovese, T.; Gugliandolo, E.; Peritore, A.F.; Crupi, R.; Impellizzeri, D.; Cuzzocrea, S.; et al. Biochemical Evaluation of the Antioxidant Effects of Hydroxytyrosol on Pancreatitis-Associated Gut Injury. Antioxidants 2020, 9, 781. [Google Scholar] [CrossRef] [PubMed]
  104. Fusco, R.; Cordaro, M.; Siracusa, R.; Peritore, A.F.; D’Amico, R.; Licata, P.; Crupi, R.; Gugliandolo, E. Effects of Hydroxytyrosol against Lipopolysaccharide-Induced Inflammation and Oxidative Stress in Bovine Mammary Epithelial Cells: A Natural Therapeutic Tool for Bovine Mastitis. Antioxidants 2020, 9, 693. [Google Scholar] [CrossRef] [PubMed]
  105. Han, H.; Zhong, R.; Zhang, S.; Wang, M.; Wen, X.; Yi, B.; Zhao, Y.; Chen, L.; Zhang, H. Hydroxytyrosol attenuates diquat-induced oxidative stress by activating Nrf2 pathway and modulating colonic microbiota in mice. J. Nutr. Biochem. 2022, 113, 109256. [Google Scholar] [CrossRef] [PubMed]
  106. Atzeri, A.; Lucas, R.; Incani, A.; Penalver, P.; Zafra-Gomez, A.; Melis, M.P.; Pizzala, R.; Morales, J.C.; Deiana, M. Hydroxytyrosol and tyrosol sulfate metabolites protect against the oxidized cholesterol pro-oxidant effect in Caco-2 human enterocyte-like cells. Food Funct. 2016, 7, 337–346. [Google Scholar] [CrossRef] [PubMed]
  107. Martin-Pelaez, S.; Castaner, O.; Sola, R.; Motilva, M.J.; Castell, M.; Perez-Cano, F.J.; Fito, M. Influence of Phenol-Enriched Olive Oils on Human Intestinal Immune Function. Nutrients 2016, 8, 213. [Google Scholar] [CrossRef]
  108. Martin-Pelaez, S.; Mosele, J.I.; Pizarro, N.; Farras, M.; de la Torre, R.; Subirana, I.; Perez-Cano, F.J.; Castaner, O.; Sola, R.; Fernandez-Castillejo, S.; et al. Effect of virgin olive oil and thyme phenolic compounds on blood lipid profile: Implications of human gut microbiota. Eur. J. Nutr. 2017, 56, 119–131. [Google Scholar] [CrossRef]
  109. Torres, J.; Mehandru, S.; Colombel, J.-F.; Peyrin-Biroulet, L. Crohn’s disease. Lancet 2017, 389, 1741–1755. [Google Scholar] [CrossRef]
  110. Claes, A.K.; Zhou, J.Y.; Philpott, D.J. NOD-Like Receptors: Guardians of Intestinal Mucosal Barriers. Physiology 2015, 30, 241–250. [Google Scholar] [CrossRef]
  111. Fernandez-Garcia, V.; Gonzalez-Ramos, S.; Martin-Sanz, P.; Garcia-Del Portillo, F.; Laparra, J.M.; Bosca, L. NOD1 in the interplay between microbiota and gastrointestinal immune adaptations. Pharm. Res. 2021, 171, 105775. [Google Scholar] [CrossRef]
  112. Roda, G.; Chien Ng, S.; Kotze, P.G.; Argollo, M.; Panaccione, R.; Spinelli, A.; Kaser, A.; Peyrin-Biroulet, L.; Danese, S. Crohn’s disease. Nat. Rev. Dis. Prim. 2020, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  113. Kawaguchi, K.; Matsumoto, T.; Kumazawa, Y. Effects of antioxidant polyphenols on TNF-alpha-related diseases. Curr. Top. Med. Chem. 2011, 11, 1767–1779. [Google Scholar] [CrossRef]
  114. Khalili, H.; Chan, S.S.M.; Lochhead, P.; Ananthakrishnan, A.N.; Hart, A.R.; Chan, A.T. The role of diet in the aetiopathogenesis of inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 525–535. [Google Scholar] [CrossRef] [PubMed]
  115. Guina, T.; Deiana, M.; Calfapietra, S.; Cabboi, B.; Maina, M.; Tuberoso, C.I.; Leonarduzzi, G.; Gamba, P.; Gargiulo, S.; Testa, G.; et al. The role of p38 MAPK in the induction of intestinal inflammation by dietary oxysterols: Modulation by wine phenolics. Food Funct. 2015, 6, 1218–1228. [Google Scholar] [CrossRef]
  116. Serra, G.; Deiana, M.; Spencer, J.P.E.; Corona, G. Olive Oil Phenolics Prevent Oxysterol-Induced Proinflammatory Cytokine Secretion and Reactive Oxygen Species Production in Human Peripheral Blood Mononuclear Cells, Through Modulation of p38 and JNK Pathways. Mol. Nutr. Food Res. 2017, 61, 1700283. [Google Scholar] [CrossRef] [PubMed]
  117. Gu, P.; Feagins, L.A. Diet in the Pathogenesis and Management of Crohn’s Disease. Gastroenterol. Clin. 2022, 51, 319–335. [Google Scholar] [CrossRef]
  118. Papada, E.; Amerikanou, C.; Forbes, A.; Kaliora, A.C. Adherence to Mediterranean diet in Crohn’s disease. Eur. J. Nutr. 2020, 59, 1115–1121. [Google Scholar] [CrossRef] [PubMed]
  119. Feuerstein, J.D.; Cheifetz, A.S. Crohn Disease: Epidemiology, Diagnosis, and Management. Mayo Clin. Proc. 2017, 92, 1088–1103. [Google Scholar] [CrossRef] [PubMed]
  120. Rondanelli, M.; Lamburghini, S.; Faliva, M.A.; Peroni, G.; Riva, A.; Allegrini, P.; Spadaccini, D.; Gasparri, C.; Iannello, G.; Infantino, V. A food pyramid, based on a review of the emerging literature, for subjects with inflammatory bowel disease. Endocrinol. Diabetes. Y. Nutr. (Engl. Ed.) 2021, 68, 17–46. [Google Scholar]
  121. Gugliandolo, E.; Cordaro, M.; Fusco, R.; Peritore, A.F.; Siracusa, R.; Genovese, T.; D’Amico, R.; Impellizzeri, D.; Di Paola, R.; Cuzzocrea, S.; et al. Protective effect of snail secretion filtrate against ethanol-induced gastric ulcer in mice. Sci. Rep. 2021, 11, 3638. [Google Scholar] [CrossRef]
  122. Kauffman, G.L. Duodenal ulcer disease: Treatment by surgery, antibiotics, or both. Adv. Surg. 2000, 34, 121–135. [Google Scholar]
  123. Allen, A.; Pearson, J.P.; Blackburn, A.; Coan, R.M.; Hutton, D.A.; Mall, A.S. Pepsins and the mucus barrier in peptic ulcer disease. Scand. J. Gastroenterol. Suppl. 1988, 146, 50–57. [Google Scholar] [CrossRef]
  124. Ansari, A.Z.; Bhatia, N.Y.; Gharat, S.A.; Godad, A.P.; Doshi, G.M. Exploring Cytokines as Potential Target in Peptic Ulcer Disease: A Systematic Update. Endocr. Metab. Immune. Disord. Drug. Targets 2022, 23, 21–34. [Google Scholar] [CrossRef]
  125. Malik, T.F.; Gnanapandithan, K.; Singh, K. Peptic Ulcer Disease; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  126. Castro, M.; Romero, C.; de Castro, A.; Vargas, J.; Medina, E.; Millan, R.; Brenes, M. Assessment of Helicobacter pylori eradication by virgin olive oil. Helicobacter 2012, 17, 305–311. [Google Scholar] [CrossRef]
  127. Arismendi Sosa, A.C.; Mariani, M.L.; Vega, A.E.; Penissi, A.B. Extra virgin olive oil inhibits Helicobacter pylori growth in vitro and the development of mice gastric mucosa lesions in vivo. Front. Microbiol. 2022, 13, 961597. [Google Scholar] [CrossRef]
  128. Sangiovanni, E.; Colombo, E.; Fumagalli, M.; Abbiati, F.; Caruso, D.; Dell’Agli, M. Inhibition of NF- κB activity by minor polar components of extra-virgin olive oil at gastric level. Phytother. Res. 2012, 26, 1569–1571. [Google Scholar] [CrossRef]
  129. Reis, R.; Sipahi, H.; Zeybekoğlu, G.; Çelik, N.; Kırmızıbekmez, H.; Kaklıkkaya, N.; Aydın, A. Hydroxytyrosol: The phytochemical responsible for bioactivity of traditionally used olive pits. Euroasian. J. Hepato.-Gastroenterol. 2018, 8, 126. [Google Scholar]
  130. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [PubMed]
  131. Lucafò, M.; Curci, D.; Franzin, M.; Decorti, G.; Stocco, G. Inflammatory bowel disease and risk of colorectal cancer: An overview from pathophysiology to pharmacological prevention. Front. Pharmacol. 2021, 12, 772101. [Google Scholar] [CrossRef] [PubMed]
  132. Mattioli, A.V.; Serra, F.; Spatafora, F.; Toni, S.; Farinetti, A.; Gelmini, R. Polyphenols, Olive oil and Colonrectal cancer: The effect of Mediterranean Diet in the prevention. Acta Biomed. 2022, 92, e2021307. [Google Scholar] [CrossRef] [PubMed]
  133. Bernini, R.; Carastro, I.; Palmini, G.; Tanini, A.; Zonefrati, R.; Pinelli, P.; Brandi, M.L.; Romani, A. Lipophilization of Hydroxytyrosol-Enriched Fractions from Olea europaea L. Byproducts and Evaluation of the in Vitro Effects on a Model of Colorectal Cancer Cells. J. Agric. Food Chem. 2017, 65, 6506–6512. [Google Scholar] [CrossRef]
  134. Sain, A.; Sahu, S.; Naskar, D. Potential of olive oil and its phenolic compounds as therapeutic intervention against colorectal cancer: A comprehensive review. Br. J. Nutr. 2022, 128, 1257–1273. [Google Scholar] [CrossRef] [PubMed]
  135. Xie, P.; Cecchi, L.; Bellumori, M.; Balli, D.; Giovannelli, L.; Huang, L.; Mulinacci, N. Phenolic Compounds and Triterpenes in Different Olive Tissues and Olive Oil By-Products, and Cytotoxicity on Human Colorectal Cancer Cells: The Case of Frantoio, Moraiolo and Leccino Cultivars (Olea europaea L.). Foods 2021, 10, 2823. [Google Scholar] [CrossRef] [PubMed]
  136. Santarelli, R.; Pompili, C.; Gilardini Montani, M.S.; Evangelista, L.; Gonnella, R.; Cirone, M. 3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol) Counteracts ERK1/2 and mTOR Activation, Pro-Inflammatory Cytokine Release, Autophagy and Mitophagy Reduction Mediated by Benzo[a]pyrene in Primary Human Colonic Epithelial Cells. Pharmaceutics 2022, 14, 663. [Google Scholar] [CrossRef]
  137. Imran, M.; Nadeem, M.; Gilani, S.A.; Khan, S.; Sajid, M.W.; Amir, R.M. Antitumor perspectives of oleuropein and its metabolite hydroxytyrosol: Recent updates. J. Food Sci. 2018, 83, 1781–1791. [Google Scholar] [CrossRef]
  138. Yen, H.H.; Tsai, H.Y.; Wang, C.C.; Tsai, M.C.; Tseng, M.H. An Improved Endoscopic Automatic Classification Model for Gastroesophageal Reflux Disease Using Deep Learning Integrated Machine Learning. Diagnostics 2022, 12, 2827. [Google Scholar] [CrossRef] [PubMed]
  139. Kellerman, R.; Kintanar, T. Gastroesophageal Reflux Disease. Prim. Care. Clin. Off. Pract. 2017, 44, 561–573. [Google Scholar] [CrossRef]
  140. Wang, C.C.; Chiu, Y.C.; Chen, W.L.; Yang, T.W.; Tsai, M.C.; Tseng, M.H. A Deep Learning Model for Classification of Endoscopic Gastroesophageal Reflux Disease. Int. J. Env. Res. Public Health 2021, 18, 2428. [Google Scholar] [CrossRef]
  141. Gyawali, C.P.; Fass, R. Management of Gastroesophageal Reflux Disease. Gastroenterology 2018, 154, 302–318. [Google Scholar] [CrossRef]
  142. Liacouras, C.A.; Ruchelli, E. Eosinophilic esophagitis. Curr. Opin. Pediatr. 2004, 16, 560–566. [Google Scholar] [CrossRef]
  143. O’Shea, K.M.; Aceves, S.S.; Dellon, E.S.; Gupta, S.K.; Spergel, J.M.; Furuta, G.T.; Rothenberg, M.E. Pathophysiology of Eosinophilic Esophagitis. Gastroenterology 2018, 154, 333–345. [Google Scholar] [CrossRef] [PubMed]
  144. Rothenberg, M.E.; Mishra, A.; Collins, M.H.; Putnam, P.E. Pathogenesis and clinical features of eosinophilic esophagitis. J. Allergy. Clin. Immunol. 2001, 108, 891–894. [Google Scholar] [CrossRef] [PubMed]
  145. Sethi, S.; Richter, J.E. Diet and gastroesophageal reflux disease: Role in pathogenesis and management. Curr. Opin. Gastroenterol. 2017, 33, 107–111. [Google Scholar] [CrossRef]
  146. Cao, H.; Huang, X.; Zhi, X.; Han, C.; Li, L.; Li, Y. Association between tea consumption and gastroesophageal reflux disease: A meta-analysis. Medicine 2019, 98, e14173. [Google Scholar] [CrossRef]
  147. Roh, S.S.; Shin, M.R.; Shin, S.H.; Lee, J.Y.; Song, Y.O.; Woo, M.; Jeong, K.S.; Noh, J.S. Low-Molecular-Weight Oligonol, a Polyphenol Derived from Lychee Fruit, Attenuates Experimental Reflux Esophagitis and HCl/Ethanol-Induced Gastric Ulcer. J. Med. Food 2017, 20, 1214–1221. [Google Scholar] [CrossRef]
  148. Malfa, G.A.; Di Giacomo, C.; Cardia, L.; Sorbara, E.E.; Mannucci, C.; Calapai, G. A standardized extract of Opuntia ficus-indica (L.) Mill and Olea europaea L. improves gastrointestinal discomfort: A double-blinded randomized-controlled study. Phytother. Res. 2021, 35, 3756–3768. [Google Scholar] [CrossRef] [PubMed]
  149. Acar-Tek, N.; Agagunduz, D. Olive Leaf (Olea europaea L. folium): Potential Effects on Glycemia and Lipidemia. Ann. Nutr. Metab. 2020, 76, 10–15. [Google Scholar] [CrossRef]
  150. Romani, A.; Ieri, F.; Urciuoli, S.; Noce, A.; Marrone, G.; Nediani, C.; Bernini, R. Health Effects of Phenolic Compounds Found in Extra-Virgin Olive Oil, By-Products, and Leaf of Olea europaea L. Nutrients 2019, 11, 1776. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The chemical structure of hydroxytyrosol, a derivative of the hydrolysis of oleuropein contained in olive oil.
Figure 1. The chemical structure of hydroxytyrosol, a derivative of the hydrolysis of oleuropein contained in olive oil.
Ijms 24 03111 g001
Figure 2. Principal proprieties of hydroxytyrosol.
Figure 2. Principal proprieties of hydroxytyrosol.
Ijms 24 03111 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arangia, A.; Marino, Y.; Impellizzeri, D.; D’Amico, R.; Cuzzocrea, S.; Di Paola, R. Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease. Int. J. Mol. Sci. 2023, 24, 3111. https://doi.org/10.3390/ijms24043111

AMA Style

Arangia A, Marino Y, Impellizzeri D, D’Amico R, Cuzzocrea S, Di Paola R. Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease. International Journal of Molecular Sciences. 2023; 24(4):3111. https://doi.org/10.3390/ijms24043111

Chicago/Turabian Style

Arangia, Alessia, Ylenia Marino, Daniela Impellizzeri, Ramona D’Amico, Salvatore Cuzzocrea, and Rosanna Di Paola. 2023. "Hydroxytyrosol and Its Potential Uses on Intestinal and Gastrointestinal Disease" International Journal of Molecular Sciences 24, no. 4: 3111. https://doi.org/10.3390/ijms24043111

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