Next Article in Journal
Standardized Sanguisorba officinalis L. Extract Inhibits Adipogenesis and Promotes Thermogenesis via Reducing Oxidative Stress
Previous Article in Journal
The Influence of 5′,8-Cyclo-2′-Deoxyguanosine on ds-DNA Charge Transfer Depends on Its Diastereomeric Form: A Theoretical Study
Previous Article in Special Issue
The Chelating Ability of Plant Polyphenols Can Affect Iron Homeostasis and Gut Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dietary Phenolic Compounds: Their Health Benefits and Association with the Gut Microbiota

1
Department of Nutrition, Faculty of Health Sciences, Kio University, Kitakatsuragi-gun, Nara 635-0832, Japan
2
Department of Immunology, Nara Medical University, Kashihara, Nara 634-8521, Japan
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(4), 880; https://doi.org/10.3390/antiox12040880
Submission received: 27 February 2023 / Revised: 23 March 2023 / Accepted: 24 March 2023 / Published: 4 April 2023

Abstract

:
Oxidative stress causes various diseases, such as type II diabetes and dyslipidemia, while antioxidants in foods may prevent a number of diseases and delay aging by exerting their effects in vivo. Phenolic compounds are phytochemicals such as flavonoids which consist of flavonols, flavones, flavanonols, flavanones, anthocyanidins, isoflavones, lignans, stilbenoids, curcuminoids, phenolic acids, and tannins. They have phenolic hydroxyl groups in their molecular structures. These compounds are present in most plants, are abundant in nature, and contribute to the bitterness and color of various foods. Dietary phenolic compounds, such as quercetin in onions and sesamin in sesame, exhibit antioxidant activity and help prevent cell aging and diseases. In addition, other kinds of compounds, such as tannins, have larger molecular weights, and many unexplained aspects still exist. The antioxidant activities of phenolic compounds may be beneficial for human health. On the other hand, metabolism by intestinal bacteria changes the structures of these compounds with antioxidant properties, and the resulting metabolites exert their effects in vivo. In recent years, it has become possible to analyze the composition of the intestinal microbiota. The augmentation of the intestinal microbiota by the intake of phenolic compounds has been implicated in disease prevention and symptom recovery. Furthermore, the “brain–gut axis”, which is a communication system between the gut microbiome and brain, is attracting increasing attention, and research has revealed that the gut microbiota and dietary phenolic compounds affect brain homeostasis. In this review, we discuss the usefulness of dietary phenolic compounds with antioxidant activities against some diseases, their biotransformation by the gut microbiota, the augmentation of the intestinal microflora, and their effects on the brain–gut axis.

1. Introduction

Phenolic compounds are components that contribute to the bitterness, astringency, and pigmentation of most plants. In addition to providing color to flowers, the physiological role of these compounds in plants is to confer biological protection against damage caused by ultraviolet rays, feeding by insects and herbivores, and pathogenic microorganisms. The type of phenolic compounds is dependent on its chemical structure [1] and includes well-known “catechins”, “isoflavones”, and “anthocyanins”. Phenolic compounds and their analogs have a wide variety of molecular sizes and structures (Figure 1).
Previous studies on the antioxidant activity of phenolic compounds confirmed their role in the detoxification of excess reactive oxygen species (ROS) and the prevention of lifestyle-related diseases. The biological effects of phenolic compounds depend on the amount consumed and their digestion, absorption, and bioavailability. The majority of these compounds are not absorbed in the small intestine and reach the colon, in which glycosides are hydrolyzed and degraded by intestinal bacteria, generating various catabolites [2]. These catabolites have been found to contribute to human health.
Among health issues, lifestyle diseases and neurodegenerative diseases are of great concern. As a dietary method that contributes to health, there is a ketogenic diet that mainly consists of lipids which is useful for Alzheimer’s disease relief [3] or prevention of obesity and diabetes [4]. In addition to these kinds of diet, dietary phenolic compounds and their catabolites also have health benefits in cardiovascular diseases [5], rheumatoid arthritis [6,7], depression [8,9], and eye diseases [10].
Research on intestinal bacteria has evolved in the past 20 years. The types and composition of bacteria that make up the intestinal flora may be investigated using a 16S rRNA-based metagenomic analysis. The type and composition of intestinal bacteria change under different disease states or with damage, which affects the regulation of metabolism and the immune system by these bacteria. In recent years, it has become possible to investigate the mechanisms by which the ingestion of phenolic compounds derived from various foods change the composition of intestinal bacteria and also their effects on the body. Dysbiosis of the intestinal microbiota is attracting attention as one of the pathogenic mechanisms of neurodegenerative diseases [11,12]. In the past decade, oxidative stress, inflammation, and impaired autophagy have been identified as pathogenetic factors for neurodegenerative diseases, such as Parkinson’s disease, Alzheimer’s disease, and amyotrophic lateral sclerosis [13,14,15,16]. Phenolic compounds, which are expected to exert antioxidant effects in vivo, may be involved in the attenuation or prevention of neurodegenerative diseases.
In our recent study in mice, administration of persimmon-derived tannin, a type of phenolic compound, suppressed the symptoms of Mycobacterium Avium Complex (MAC) infection [17], and decreased the severity of ulcerative colitis [18]. Furthermore, it is expected that persimmon-derived tannin is degraded by intestinal bacteria and the catabolites showed antioxidant activities in vivo [19]. In this review, we summarized the findings of studies in which the administration of phenolic compounds augmented the intestinal flora in vivo and exerted beneficial effects on health. Furthermore, we discussed some phenolic compounds that are indigestible and those with active substances that currently remain unknown.

2. Flavan-3-Ols

Flavan-3-ols (flavanols) are a group of flavonoids that have a 2-phenyl-3,4-dihydro-2H-chromen-3-ol skeleton. Dietary flavan-3-ols are abundant in cocoa, tea, apples, grapes (including red wine), berries, plums, apricots, and nuts. Flavan-3-ols are complex flavonoids in which monomers, such as catechins and epicatechins, make up units to form oligomers and polymers. They are components of proanthocyanidins, and many analogs exist in nature. Catechins, major dietary monomers, are abundant in tea leaves, and many studies have investigated their antioxidant properties [20,21]. Unlike other classes of flavonoids, flavan-3-ols are not present in a glycosylated form in foods [22] and monomeric flavan-3-ols are quickly absorbed in the small intestine. The galloylation and polymerization of flavan-3-ols were shown to significantly delay intestinal absorption [23]. Therefore, when oligomers and polymers reach the colon, they need to be metabolized by the colonic microbiota to provide health benefits.
The mechanisms underlying the antioxidant effects of monomers have been reported [24]. The antioxidant capacity of flavan-3-ol monomers is exerted through phenolic hydroxyl groups that trap ROS and the chelation of iron ions to prevent lipid peroxidation [25,26]. By indirectly employing antioxidant pathways, flavan-3-ols regulate the synthesis of antioxidant-related enzymes and the signaling pathways of oxidative stress [27]. However, the mechanisms of action of oligomers and polymers remain unclear.

2.1. Dietary Source and Metabolism of Flavan-3-Ols

2.1.1. Tea

Tea is a major source of catechins. Various types of tea are available from the Camellia sinensis (L.) plant, depending on the harvesting and processing of its leaves. Green tea is unfermented tea; black tea is completely fermented tea; white tea and oolong tea are tea types with different degrees of fermentation [28]. There are five main types of catechins present in tea: (+)-catechin, (−)-epicatechin (EC), (−)-epigallocatechin (EGC), (−)-epicatechin gallate (ECG), and (−)-epigallocatechin gallate (EGCG) (Figure 2) [29,30]. EGCG is the most abundant catechin in unfermented teas (green tea and white tea) [31]. During the fermentation of black tea, catechins are oxidized by polyphenol oxidase to complex structures, such as theaflavin dimers and thearubigin polymers [32]. Tea phenolic compounds and their metabolites possess antibacterial properties against pathogenic bacteria, such as Clostridium perfringens, C. difficile, Escherichia coli, Salmonella, and Pseudomonas, and enhance the activities of probiotics, including Bifidobacterium and Lactobacillus species, thereby improving the overall balance of intestinal microbes [33,34]. The products of the intestinal bacterial catabolism of major tea catechins are shown in Figure 3 [35].
Theaflavins and theasinensins are catechin dimers that are not absorbed in the small intestine to the same extent as catechin; they reach the large intestine and are metabolized by intestinal bacteria enzymes [36,37,38]. Four theaflavins exist in black tea: theaflavin (TF), theaflavin-3-gallate (TF3G), theaflavin-3′-gallate (TF3′G), and theaflavin-3,3′-digallate (TFDG) (Figure 2), with TFDG being the most abundant [39]. TFDG alters the composition of the intestinal flora, similar to EGCG; however, the metabolic profile was significantly different [38]. The accumulation of further findings from in vivo studies is expected. Theasinensins are also catechin dimers with two galloyl groups; five theasinensins in fermented tea have been identified and named theasinensins A, B, C, D, and E (Figure 2) [40]. Theasinensin A is the most abundant among the five compounds [37]. The galloyl group is easily removed by intestinal bacteria and decomposed into theasinensin C. However, the progression of the subsequent reaction is slower than that of EGCG, and the whole picture remains unclear. In vivo studies are needed on these compounds, and the findings obtained will contribute to human health [37].

2.1.2. Cocoa

Cocoa is generally produced by fermenting and roasting the seeds of Theobroma cacao and then pulverizing the cocoa cake obtained by removing the fat content. Although flavan-3-ols are relatively abundant in cocoa, its components vary depending on the type of cacao, place of origin, time of harvest, and processing of cocoa [41,42,43]. Cocoa flavan-3-ols, along with (+)-catechin and procyanidin B1 and B2 (Figure 4), as well as trace amounts of other flavanols [44], mostly exist as EC.
EC and procyanidin B1 in cocoa powder are metabolized in the intestines (Figure 5) [45]. Phenolic compounds in cocoa are metabolized in both the small and large intestine to produce metabolites that affect human health. [41,45,46].

2.2. Health Benefits of Flavan-3-Ols

2.2.1. Tea

A well-established causal relationship has been reported between the intake of EC and the regulation of cardiovascular function [47,48]. EC is rapidly absorbed, and its metabolites are excreted in the urine 72 h after consumption [49]. Although EC does not affect the composition of the microbial flora [50], EC phase II and gut microbiota metabolites may induce complex nutrigenomic/epigenomic changes that regulate the function of brain endothelial cells [49,51]. In other words, the metabolites of EC may reduce the risk of neurodegenerative diseases by maintaining the integrity of cerebrovascular endothelial cells, suggesting that the intake of EC contributes to improvements in cognitive ability [51].
The ingestion of tea reportedly attenuates alcoholic liver disease [52]. The administration of tea extract has been shown to activate antioxidant enzymes in the liver, change the intestinal flora, and promote liver function [53,54]. Although some types of teas promote liver function, others exert the opposite effects; therefore, further research on this subject is required [52,53].
EGCG is the major catechin found in unfermented tea [28] and exhibits the highest antioxidant activity among the four catechin monomers in vitro [30]. EGCG may attenuate non-alcoholic fatty liver disease (NAFLD) by regulating the interaction between the gut microbiota and bile acids [55].
NAFLD is closely associated with the gastrointestinal microflora and its dysbiosis [56,57]; therefore, further research on the treatment and prevention of NAFLD is needed. EGCG reportedly prevents the occurrence of NAFLD by regulating the intestinal flora. Akkermansia muciniphila, belonging to the phylum Verrucomicrobia, has been implicated in obesity, glucose metabolism, and intestinal immunity [58]. The abundance of the genus Akkermansia has been shown to increase with the intake of phenolic compounds and exerts anti-obesity effects [59]. Furthermore, EGCG intake increased the abundance of the genus Akkermansia in mice compared to a high-fat diet [55].
Inflammatory bowel disease (IBD) is an inflammatory disease that collectively refers to ulcerative colitis (UC) and Crohn’s disease, which are generally considered to have unknown (non-specific) etiologies. Catechins exhibit anti-inflammatory, antioxidant, and antibacterial activities, which may improve the abnormal condition of intestinal bacteria caused by IBD [60,61,62,63]. However, depending on the doses of catechin examined, conflicting findings have been reported; therefore, further research on this subject is needed [27].
Catechins in tea are metabolized into phenyl-γ-valerolactones by the action of intestinal bacteria as shown in Figure 3. Phenyl-γ-valerolactones regulate cellular proteolysis and exert neuroprotective effects [64]. In cell lines, EGCG, EGC, and ECG have been reported to inhibit amyloid-β-induced inflammation and neurotoxicity [65,66,67,68]. Animal studies also revealed the beneficial effects of EGCG on neurodegeneration in animal models of Alzheimer’s disease [69] and Parkinson’s disease [70,71]. Furthermore, EGCG was shown to affect hypoxia-induced neuroinflammation in cell lines [72]. Based on these findings, the intake of catechin may be effective against neurodegenerative diseases. However, there are many issues that need to be considered in clinical studies on humans, such as intake as food or supplements, dietary habits, and regional characteristics, and thus, further research is necessary.

2.2.2. Cocoa

Cocoa powder has been shown to affect the gut microbiota by changing their metabolites and promoting the growth of Lactobacillus and Bifidobacterium groups in pigs [73] Flavanols in cocoa may function as prebiotics to maintain intestinal immunomodulation by regulating the gut microbiota [74,75,76]. The ingestion of cocoa powder was previously suggested to change the intestinal flora of the diabetic Zucker rat model, by strengthening the intestinal barrier and ameliorating colonic inflammation, thereby attenuating diabetes [77]. Cocoa powder was also shown to down-regulate inflammation markers and suppress inflammation-related colon carcinogenesis; therefore, its consumption may be promising for the prevention of intestinal inflammation and related cancers [78]. Cocoa flavanols also exert endothelium-dependent vasodilatory effects [79], suggesting their potential to ameliorate cardiovascular diseases [80].
Flavan-3-ols derived from cacao are metabolized into phenyl-γ-valerolactones by the action of intestinal bacteria, similar to the above-described tea catechins (Figure 5). Therefore, they may be effective against neurodegenerative diseases [81,82,83].
However, difficulties are associated with investigating the effects of cocoa flavan-3-ols in vivo due to the selection of an appropriate dose and their complex relationship with the intestinal flora [84]. Since cocoa powder also contains dietary fiber and alkaloids, such as theobromine, further studies on its effects on human health are warranted.

3. Condensed Tannins

Tannin is a general term for astringent plant components that exist widely throughout the plant kingdom and have been traditionally used to tan leather. There are two types of tannins, one of which is hydrolyzed tannins which are polymers of ellagic acids or gallic acids, and the other is condensed tannins which are polymers of catechins. They are hydrolyzed or decomposed under specific conditions and produce low molecular weight phenolic compounds. The astringent skin of chestnuts and walnuts contain hydrolyzed tannins and astringent persimmons contain condensed tannins. Red wine also contains condensed tannins, but the degree of polymerization of catechins are altered depending on the degree of fermentation and the manufacturing method. In this chapter, we will focus on condensed tannins which are a component of astringent persimmon fruits.

3.1. Dietary Source and Metabolism of Tannins

Astringent Persimmon

Astringent persimmon fruits (Diospyros kaki Thunb.) contain large quantities of kaki tannin, a type of condensed tannin, such as EC, EGC, ECG, and EGCG (Figure 1) [85]. However, the structure of kaki tannin has not yet been clarified. Soluble kaki tannins in astringent persimmon fruits are converted into insoluble kaki tannins via dehydration, and dried persimmons lose their bitterness and have a sweet taste. Moreover, kaki tannins are reportedly non-hydrolyzable and non-digestible, but exhibit high antioxidant activity [86,87].

3.2. Health Benefits of Tannins

Astringent Persimmon

Kaki tannin has the property of binding with bile acids [87] and the effect of lowering cholesterol and ameliorating glucose metabolism [88,89]. Kaki tannins have also been reported to reshape the gut microbiota in rats fed a high-cholesterol diet [90].
Mycobacterium avium complex (MAC) is the most common nontuberculous mycobacterium that causes chronic pulmonary infections in immunodeficient individuals. Kaki tannins, used as a dietary supplement, reduce the symptoms of pulmonary MAC infection [17], suggesting an impact on mucosal immune inflammation, including that of the gut, through their anti-inflammatory effects and changes to the gut microbial composition. Moreover, kaki tannins may need to be digested and/or fermented into smaller molecules in vivo prior to their absorption into the body in order to exert their beneficial effects. The artificial digestion of the non-extracted residues of dried persimmons containing kaki tannins suggested that intestinal bacteria degraded the tannins into lower molecular weight fragments [19].
UC is a chronic IBD induced by the dysregulation of the immune response in the intestinal mucosa. The pathogenesis of UC was less severe in a mouse model fed kaki tannins than in a control diet group [18]. Furthermore, the gene expression of an inflammatory cytokine (IL-1β) and chemokine (CXCL1) was significantly decreased in the tannin diet group. An analysis of the composition of the fecal microbiota of mice employing 16S ribosomal RNA gene sequencing revealed that a treatment with DSS significantly increased the abundance of the phylum Enterobacteriaceae in the control diet group, whereas it was significantly suppressed in the kaki tannin diet group.
Dietary supplementation with kaki tannins ameliorated the pathogenesis of MAC disease and DSS-induced colitis by suppressing the inflammatory response and changing the composition of the microbiota. However, further studies are needed to establish the optimal method of administration, select the appropriate concentration of kaki tannin, and elucidate the detailed chemical structures of the decomposed tannins. Although tannins have been shown to promote lipid metabolism in animal experiments [87,91,92], and similar findings were obtained for humans [93], the relationship between these findings and gut bacteria remains unclear. Therefore, human clinical trials are needed in the future to assess the health benefits of tannins.

4. Flavonols

Flavonols, a subclass of flavonoids with a 3-hydroxyflavone skeleton, are widely present in plants [22]. Typical flavonols include myricetin (in grapes and berries), kaempferol (in tea, broccoli, and ginger), rutin (in asparagus and buckwheat), and quercetin (Figure 6). Quercetin is a representative flavonol that has been extensively examined and is present in vegetables and fruits, such as onions, broccoli, and apples. Flavonols generally exist in a glycosidic form and are deglycosylated and absorbed in the small intestine. After absorption, they are rapidly metabolized by phase II enzymes in the liver and circulate as methyl, glucuronide, and sulfate metabolites [94,95].

4.1. Dietary Sources and Metabolism of Flavonols

4.1.1. Onions

Onions (Allium cepa L.) are used as an ingredient in various dishes. They are rich in flavanols, the most abundant of which is quercetin [96,97]. Quercetin (an aglycone) is mostly present in the outer skin and quercetin 4′-glucoside and quercetin 3,4′-diglucoside in the bulbs, which are generally edible [98,99]. Figure 7 shows the quercetin catabolites produced by intestinal bacteria and phase II enzymes in the liver. Quercetin derivatives in onions increase their bioavailability through cooking processes, such as baking, frying, and grilling [100].

4.1.2. Buckwheat

Buckwheat is widely grown in Asia, Europe, and the Americas. Both common buckwheat (Fagopyrum esculentum Moench) and tartary buckwheat (F. tataricum (L.) Gilib.) are used as food sources, and the antioxidant activity of tartary buckwheat is higher than that of common buckwheat [101]. Rutin is the main flavonol in buckwheat, accounting for 90% of all phenolic compounds [102]. Rutin is a glycoside composed of flavonol aglycone quercetin along with disaccharide rutinose (Figure 6), and rutin is converted to quercetin by rutinosidase contained in seeds during grain milling [103]. Buckwheat is a potential gluten-free diet for people with gluten sensitivities and has been noted for its antioxidant properties and other health benefits [104].

4.2. Health Benefits of Flavonols

4.2.1. Onions

Quercetin exhibits antioxidant, anti-inflammatory, and anti-osteoporotic activities [95,105]. The administration of quercetin and quercetin glycosides extracted from onion skin to rats on a high-fat diet increased serum antioxidant activity and significantly increased enzyme activity derived from intestinal bacteria [106]. In other words, quercetin effectively reduced the intestinal flora abnormalities induced by the high-fat diet. However, in human clinical studies, the administration of onion peel extracts to obese patients with hypertension did not attenuate their symptoms [107]. Similarly, in clinical studies on hypertension and rheumatoid arthritis, the administration of quercetin did not exert beneficial effects [108,109,110,111,112]. Based on the beneficial effects of onion peel observed in animal and cell culture experiments, clinical studies need to be performed on humans under various conditions, particularly obesity.
Quercetin glycosides are catabolized to produce phenolic acids by intestinal bacteria [113]. Among the phenolic acids derived from quercetin glycosides, 3,4-dihydroxyphenylacetic acid is the most effective at scavenging free radicals and inducing phase II enzymes [114]. Moreover, 3,4-dihydroxyphenylacetic acid significantly inhibits hydrogen peroxide-induced cytotoxicity [114,115]. Quercetin has been implicated in the attenuation of insulin resistance and atherosclerosis in obesity-related diseases [116,117,118,119]. It was found to promote intestinal homeostasis by changing the intestinal flora [120,121] and also plays a role in the prevention and treatment of inflammatory bowel disease [122,123].
A previous study demonstrated that quercetin and rutin effectively suppressed the aggregation of amyloid-β in cell lines, and thus, they are expected to be effective against Alzheimer’s disease [124]. Quercetin has potential in the treatment of Alzheimer’s disease in cell lines [125,126,127] and was effective in a mouse model of Alzheimer’s disease [128]. Therapeutic effects have been suggested in animal models of Parkinson’s disease, and quercetin may be effective against neurodegenerative diseases [129,130]. In addition, the combined use of quercetin and piperine (a type of alkaloid), which is a component of pepper, appeared to exert neuroprotective effects [131,132].
Although cell cultures and animal experiments have provided important findings, few clinical experiments have been conducted in humans to date; therefore, future research and verification are required.

4.2.2. Buckwheat

Rutin and quercetin contained in tartary buckwheat regulate gut microbiota and are involved in lipid metabolism [133]. Rutin had little effect on attenuating obesity but tended to decrease fat deposition in the liver [133]. Phenolic compounds extracted from tartary buckwheat bran showed dose-dependent anticancer activity against human breast cancer MDA-MB-231 cells [134]. Further research is needed regarding the anticancer properties of rutin in humans [135]. It has been suggested that rutin has the potential to inhibit major proteases of SARS-CoV-2 in vitro [136,137].
Rutin and quercetin interact with buckwheat proteins and starch [138]. The presence of phenolic compounds such as rutin and quercetin reduces the digestibility of proteins and starches and allows them to be absorbed slowly [139,140]. While this is not a favorable outcome in terms of natural nutrient uptake, it also has some desirable consequences related to diabetes and lipid metabolism [141,142,143]. Concerning cardio-metabolic disease, meta-analyses have not yet yielded consistent results regarding the usefulness of phenolic compounds, such as rutin [144]. Recent studies suggest that buckwheat has inhibitory effects on Alzheimer’s disease and other neurological disorders [145], but it is not yet clear whether rutin is responsible for this effect [146]. Therefore, further research is needed.

5. Isoflavones

Isoflavones are flavonoids with 3-phenylchromone as the basic skeleton (Figure 8). They are abundant in plants of the legume family (Fabaceae), such as soybeans and kudzu. Isoflavones bind to estrogen receptors in the body and exert a number of effects because their chemical structures are similar to estrogen [147]. They may be beneficial, but also detrimental [148]. For example, while isoflavones are expected to effectively prevent osteoporosis, breast cancer, and prostate cancer, they also increase the risk of the onset and recurrence of breast cancer [148]. Glycosides are not easily absorbed in the small intestine and must be converted into aglycones, such as genistein and daidzein, to function in vivo [149,150].

5.1. Dietary Source and Metabolism of Isoflavones

Soybeans

Soybeans (Glycine max (L.) Merr.) are the most abundant source of isoflavones [151]. Many isoflavones, such as genistin and daidzin, are present in food (Figure 8). In the small intestine, lactase-phlorizin hydrolase and cytosolic β-glucosidase hydrolyze monoglucuronides to form aglycones [152,153]. The absorbed isoflavone aglycones are mainly metabolized to glucuronides and sulfates by endogenous phase I and phase II enzymes. Isoflavones are excreted into the intestines via the enterohepatic circulation, and unabsorbed isoflavones reach the colon and are metabolized to form the metabolite, equol, and other metabolites by intestinal bacteria [154] (Figure 9). Numerous studies have identified equol-producing bacteria; however, findings on the production of equol have been inconsistent because it is markedly affected by the diet of the host [154]. A previous study reported that 25–30% of the Western population possessed equol-producing gut bacteria, whereas they were detected in 50–60% of the Asian population [155].

5.2. Health Benefits of Isoflavones

Soybeans

Soybeans are rich in isoflavones, particularly genistin and daidzin [151]. Isoflavones are phytoestrogens, such as the female hormone 17-β-estradiol, which are less active than hormones, but exhibit estrogenic activity [156]. Therefore, the intake of isoflavones is expected to alleviate menopausal symptoms in women, increase bone formation, and reduce the incidence of cardiovascular disease. Equol is a metabolite of daidzin/daidzein formed by intestinal bacteria (Figure 9). It is more stable and more easily absorbed than daidzein [157] and exhibits stronger estrogenic activity than other isoflavones and isoflavone-derived metabolites [158,159,160,161]. Isoflavone aglycones and glycosides are both catabolized by enzymes of the intestinal microbiota to produce high levels of antioxidant substances, such as equol. A correlation has been reported between soybean intake and the attenuation of menopausal symptoms [162].
The intake of soy isoflavones has been suggested to reduce bone resorption, prevent some types of cancers, and improve learning [163,164,165,166]. These health effects are attributed to equol produced from soy isoflavones by the action of the intestinal microbiota. Therefore, these effects may be observed in individuals who produce equol in their intestines. Furthermore, the human gut microbiome is highly individualized, and its effects are inconsistent. This inconsistency poses a major challenge when considering the effects of isoflavones on humans. Adverse effects associated with the intake of soy isoflavones, including endometriosis, dysmenorrhea, and secondary infertility, have also been reported, and symptoms were ameliorated by the discontinuation of intake [167].
Isoflavones and their metabolites exert their effects by binding to the estrogen receptor (ER) and transmitting cell signals. However, isoflavones are agonists that activate ER as well as antagonists that inhibit it, which modulates estrogen signaling. Therefore, they may act as an endocrine disruptor, with more than just beneficial effects [167].
Animal studies showed that genistein, a soy isoflavone, was effective for the treatment of neurodegenerative diseases, such as Alzheimer’s disease [168,169,170] and Parkinson’s disease [171]. Early oral genistein therapy appeared to ameliorate the severity of disease in multiple sclerosis model mice [172].
In the future, we anticipate further advances in this field that will verify the effects of isoflavones and their metabolites on humans.

6. Phenylpropanoids

Phenylpropanoids, also called lignoids, are compounds that have a C6-C3 skeleton with a C3 group attached to an aromatic ring. Monomers include caffeic acid, which is widely present in plants, and chlorogenic acid (an ester of caffeic and quinic acid), which is abundant in green coffee beans. Sesamin is a dimer, also known as lignan, and is abundant in sesame seeds. Chlorogenic acid may be ingested from food. Figure 10 shows the chemical structures of the major phenylpropanoids.

6.1. Dietary Source and Metabolism of Phenylpropanoids

6.1.1. Coffee

Coffee is one of the most consumed beverages in the world. It contains at least 30 types of chlorogenic acids [173]. The term “chlorogenic acids” refers to a group of phenolic compounds, of which approximately 400 have been discovered to date [174]. 5-O-caffeoylquinic acid is the main chlorogenic acid found in green coffee beans. Although the type and concentration of chlorogenic acids vary depending on the type of coffee bean, the roasting process, and extraction method, the beneficial health effects of coffee are related to its chlorogenic acid content, whether green or roasted. The high antioxidant activity of coffee is attributed to the amount of chlorogenic acid present [175]. Figure 11 shows the main chlorogenic acids found in coffee [176]. Approximately 30% of these chlorogenic acids are absorbed in the stomach or small intestine, while the remainder are transferred to the large intestine, in which they are metabolized into dihydroferulic acid, its 4-O-sulfate, and dihydrocaffeic acid-3-O-sulfate by intestinal bacteria [177,178].

6.1.2. Sesame

Sesame (Sesamum indicum L.) is an edible seed and source of high-quality edible oil. Sesame oil exhibits antioxidant activity and possesses health-promoting properties because it contains vitamin E and lignans [179,180]. The major lignans in sesame are sesamin and sesamolin, which are formed by the dimerization of two phenylpropanoids [181]. Sesamin and sesamolin exhibit weak antioxidant activities in vitro because they do not have phenolic hydroxyl groups [182]; however, they possess antioxidant properties after being metabolized in vivo to form hydroxyl groups [183,184] (Figure 12).

6.2. Health Benefits of Phenylpropanoids

6.2.1. Coffee

Chlorogenic acids exhibit antioxidant activity [185,186,187] and anti-obesity activity in vivo [188,189,190]. Daily coffee consumption reduces the risk of type 2 diabetes [191]. Chlorogenic acid from coffee possesses prebiotic properties in vivo [192,193]. Therefore, the daily consumption of coffee may contribute to the prevention of obesity and lifestyle-related diseases.
Coffee consumption has been suggested to reduce the risk of developing neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and dementia; however, coffee contains a wide variety of components and their interactions need to be investigated [194]. Since chlorogenic acid was shown to exert neuroprotective effects against Parkinson’s disease [195,196,197] and Alzheimer’s disease [198] in animal experiments, it is expected to exert similar effects in humans. More data needs to be collected because the bioavailability of active ingredients markedly varies between individuals.

6.2.2. Sesame

The lignans in sesame have a number of health benefits, including anticancer activity, reducing the risk of cardiovascular diseases, and anti-inflammatory effects [199,200]. They are converted into enterolignans by intestinal bacteria and exert their effects as phytoestrogens [201]. Sesame lignans have been shown to inhibit L-tryptophan indole-lyase (TIL) produced by intestinal bacteria and suppress the production of indoxyl sulfate, a uremic toxin, catalyzed by TIL [202]. The inhibition of TIL by sesame lignans has potential as a strategy to prevent and treat chronic kidney diseases. Although sesaminol triglucoside, a sesame lignan glycoside, did not inhibit TIL, it induced significant increases in Lactobacillus and Bifidobacterium and changed the intestinal microbial environment [203]. Sesamin may also augment the intestinal environment by increasing the abundance of beneficial genera of bacteria, including Lactobacillus and Bifidobacterium, in the intestinal flora [204]. Moreover, sesamin reportedly promoted the adhesion of epithelial colonocytes and probiotics [204].
Sesamin, sesamolin, and sesamol exert neuroprotective effects and are expected to be effective against neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease [205,206,207,208,209]. Sesamin and sesamolin are phenylpropanoid dimers, as shown in Figure 12, which differ in structure from the phenylpropanoid monomer sesamol. Sesamin and sesamolin have both been shown to reduce amyloid-β toxicity, whereas sesamol did not [209]. However, sesamol ameliorated scopolamine-induced cholinergic disorders [205], remodeled the intestinal microbiota, significantly increased the content of short-chain fatty acids, and attenuated cognitive deficits [206]. Although structure–activity relationships warrant further investigation, these sesame lignans have neuroprotective potential.
Collectively, these findings support the potential of sesame lignans to contribute to human health; however, only a few studies have been conducted in this area of clinical research.

7. Stilbenoids

Stilbenoids are derivatives of stilbene, an aromatic hydrocarbon called 1,2-diphenylethene. Major stilbenoids are shown in Figure 13. Resveratrol is a type of stilbenoid that is present in many plant food materials, such as grapes, cranberries, red currants, and peanut skin, as well as in their processed products [210]. As a stilbenoid phenolic compound, resveratrol has been extensively studied.

7.1. Dietary Source and Metabolism of Stilbenoids

Grapes and Wine

Resveratrol, a stilbenoid found in many plants, possesses antifungal and antibacterial properties. The food sources that contain resveratrol are grapes, wine [210], and grape seed oil [211]. Resveratrol, in its native state, is present at low amounts in humans, with only 1–8% being detected in serum. Although 75% is absorbed, it is rapidly metabolized [212,213]. Resveratrol undergoes glucuronidation and sulfation in the liver and duodenum to form resveratrol-3-glucuronide (R3G) and resveratrol-3-sulfate (R3S), respectively [214,215] (Figure 14). Moreover, the intestinal flora metabolizes resveratrol to dihydroresveratrol (DHR); however, this metabolism differs among individuals [216]. Resveratrol also crosses the blood–brain barrier due to the absence of phenolic degradation products by intestinal bacteria [217]. Therefore, resveratrol may suppress neurodegeneration in the central nervous system [218], and many studies have investigated its effects on the nervous system.

7.2. Health Benefits of Stilbenoids

Grapes and Wine

Moderate wine consumption has been suggested to exert beneficial effects on health. This is commonly known as “the French paradox” because of the low incidence of coronary artery disease despite the consumption of high saturated fats by the French population [219,220].
Resveratrol has been shown to modulate and promote intestinal barrier function in mice, suggesting its potential to augment the intestinal flora [221,222]. Resveratrol prevented obesity and attenuated NAFLD and NASH by modulating the intestinal flora, maintaining intestinal barrier integrity, and suppressing intestinal inflammation in animal models [223,224,225,226]. Furthermore, the administration of resveratrol reportedly affected the intestinal flora and steroid metabolism in middle-aged men with metabolic syndrome [214,227,228,229]; however, the underlying mechanisms have not yet been elucidated. Red wine consumption reduced the risk of coronary heart disease and prevented obesity through the beneficial effects of phenolic compounds in red wine, particularly resveratrol [230,231]. Moreover, as reported in animal studies, resveratrol augmented the intestinal flora; however, further research is needed to confirm its effects in humans. Resveratrol also functions as a phytoestrogen, suggesting that its effects differ in males and females. Resveratrol may be used to treat diabetic complications during pregnancy, endometriosis, and dysmenorrhea [232].
Animal models using grape seed oil have demonstrated wound healing activity [233,234], efficacy against ulcerative colitis [235], protection against carbon tetrachloride-induced liver inflammation [236]. In cell lines, pancreatic β-cell apoptosis induced by hyperglycemia was reduced [237]. In human clinical trials, a milky lotion containing grapeseed oil was found to be effective in treating skin problems on the cheeks [238], and the use of grapeseed oil as massage oil was effective in reducing the physiological edema of pregnancy [239]. Oral administration of grape seed oil suppressed serum triglycerides in humans [240].
The protective effects of resveratrol against neurodegeneration have been extensively examined in cell lines and animals. It may also play a role in the treatment and prevention of Alzheimer’s disease [241,242,243,244], Parkinson’s disease [245,246,247], Huntington’s disease [248], multiple sclerosis [249], and amyotrophic lateral sclerosis [250]. However, it has also been suggested to exacerbate multiple sclerosis [251].

8. Curcuminoids

Curcuminoids are lipophilic phenolic compounds with a diarylheptanoid structure and are the yellow pigment components of turmeric.

8.1. Dietary Source and Metabolism of Curcuminoids

Turmeric

Turmeric is a spice prepared from the underground stems of Curcuma longa L. It contains curcuminoids, such as curcumin, demethoxycurcumin, and bisdemethoxy-curcumin (Figure 15). Curcumin is the most abundant curcuminoid in turmeric [252] and contains phenolic hydroxyl groups in its chemical structure; therefore, it functions as a potent antioxidant that suppresses the production of ROS [253].
Due to its insolubility in water, curcumin is poorly absorbed in the gastrointestinal tract and thus, has low bioavailability [254]. It reaches the large intestine and is biotransformed, as shown in Figure 16, by phase I and phase II enzymes and enzymes derived from intestinal bacteria. The resulting metabolites exhibit anti-inflammatory and antioxidant activities [255,256].

8.2. Health Benefits of Curcuminoids

Turmeric

Although turmeric is used as a spice in many dishes, its consumption per person is low. Many human clinical trials have examined the effects of curcumin supplements. Since the amount of curcumin consumed may be an important factor, the accumulation of further findings is necessary.
Curcumin exhibits anti-inflammatory, antibacterial, and anti-tumor activities [257,258,259,260,261] and also interferes with cancer-associated signaling pathways by targeting proteins and modulating gene expression [262,263]. In human clinical trials, the administration of curcumin capsules to patients with colorectal cancer reduced inflammation and oxidative stress in malignant colorectal epithelial cells. It also attenuated inflammation in patients with UC and gastrointestinal disorders [264,265,266,267].
Recent studies on curcumin and intestinal bacteria in animals reported that curcumin reduced cholesterol levels [268], ameliorated the pathology of UC [269,270], and promoted a favorable response to acute myeloid leukemia drugs [271]. Metabolites produced by the actions of intestinal bacteria may be responsible for these effects, and, in some cases, they may also be attributed to changes in the diversity of intestinal bacteria and flora. However, these effects were not observed under some conditions, and thus, further research is required to elucidate the underlying mechanisms [272]. Curcumin was previously shown to be effective against neurodegenerative diseases in many cell lines and animal studies [273,274,275,276,277]. It is also undergoing clinical trials for depression. Although curcumin may be useful in the treatment of depression, the confirmation of its therapeutic efficacy requires a multi-mechanistic approach due to the pathophysiological complexity of depression [278,279].

9. Other Phenolic Compounds: Dietary Sources, Metabolism, and Health Benefits

9.1. Protocatechuic Acid

Protocatechuic acid, a ubiquitous natural phenolic compound in plants, exerts diverse pharmacological effects, including antioxidant, antibacterial, antiviral, anticancer, anti-inflammatory, anti-aging, and anti-arteriosclerotic activities [280,281]. Protocatechuic acid is found not only in fruits and vegetables, but also in the herbal medicine Duzhong (Eucommia ulmoides Oliv.) [282]. Protocatechuic acid is also contained in oregano, which is used as a type of spice. After its ingestion, protocatechuic acid is absorbed through the intestinal epithelium, sulfated or glucuronylated through conjugation processes by phase II enzymes primarily in the liver, and then circulated throughout the body [283,284]. Protocatechuic acid is also produced in vivo as a metabolite via the degradation of phenolic compounds, particularly flavonoids, by the intestinal flora [285]. Figure 17 shows the degradation pathway of the production of protocatechuic acid from cyanidin [286].
Protocatechuic acid, a metabolite of various phenolic compounds, regulates oxidative stress and inflammatory responses. Furthermore, protocatechuic acid increases the energy expenditure of brown adipose tissue, which may reduce NAFLD [287], acts as an antidepressant [288], and inhibits the progression of neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease [286]. In addition, protocatechuic acid has been shown to affect the diversity and composition of the gut microbiota [286]. However, most of these findings were obtained from animal studies or cell culture experiments. Very few clinical trials have been conducted to date. Therefore, further animal experiments and clinical trials are required to establish whether protocatechuic acid can be applied to humans [281].

9.2. Ellagic Acid

Ellagic acid, an antioxidant, is a naturally occurring phenolic lactone compound that is abundant in strawberries, raspberries, cranberries, and walnuts [289,290]. It polymerizes with gallic acid to form glycoside ellagitannins. The hydrolyzable tannin ellagitannin is readily hydrolyzed in the gastrointestinal tract to produce ellagic acid. Ellagic acid is metabolized by intestinal bacteria into urolithin (Figure 18), which exhibits strong antioxidant activity and enhances the immune system.
Ellagic acid has been shown to change the composition of the gut microbiota, and is converted to urolithins by gut bacteria, and alleviates oxidative stress and inflammatory diseases in the gastrointestinal tract of animals [292].
It also changed the intestinal flora and ameliorated C. perfringen-induced enteritis in animal experiments [293]. However, only a few clinical trials have been conducted to date. The ingestion of ellagic acid from foods, such as fermented raspberry juice [294] or Arbutus unedo [291] may be beneficial for human health. Ellagic acid was also shown to be effective against cognitive impairment and multiple sclerosis [295,296], suggesting its efficacy in the treatment of neurodegenerative diseases. However, further animal experiments and clinical trials are needed in the future.

10. Conclusions

In this review, we introduced compounds that may attenuate some diseases through the involvement of phenolic compounds that exhibit antioxidant activities. Target phenolic compounds must be absorbed to exert their effects, and this requires the cleavage of the sugar of a glycoside. The glycoside is then converted into an aglycone that is subsequently metabolized by phase I and phase II enzymes in the small intestine and liver before circulating in the body. Unabsorbed phenolic compounds undergo biotransformation by intestinal bacteria, after which they are absorbed and circulated in the body. These metabolites exert antioxidant and anti-inflammatory effects.
Although phenolic compounds have been extensively examined in animal and cell culture studies in the last decade, the number of human clinical trials has been insufficient. Research on their effects in humans requires a great deal of effort because detailed planning and massive data collection are required due to large individual differences. Dietary ingredients are safe for consumption, but do not exert immediate effects. Further research on the nutrients present in the daily diet and their beneficial effects is warranted and may provide insights into the prevention or attenuation of diseases. Table 1 summarizes the studies introduced in this review that showed contributions to health. We hope that the efforts and achievements of researchers to date will lead to further advances in this field.

Author Contributions

Conceptualization, T.I. and Y.M.; Methodology, Y.M. and S.-i.K.; Validation, Y.M. and T.I.; Investigation, Y.M.; Resources, Y.M., M.K., and T.I.; data curation, Y.M.; writing—original draft preparation, Y.M.; writing—review and editing, S.-i.K. and T.I.; visualization, Y.M.; supervision, M.K. and T.I.; Project administration, T.I.; funding acquisition, T.I. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by grant obtainted for JSPS KAKENHI, Grant Number JP21K11587.

Institutional Review Board Statement

Animal experiments, cell culture experiments, and clinical trials conducted in the cited studies were approved by the Ethics Committee of each institution and published in journals or books.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Glevitzky, I.; Dumitrel, G.A.; Glevitzky, M.; Pasca, B.; Otrisal, P.; Bungau, S.; Cioca, G.; Pantis, C.; Popa, M. Statistical Analysis of the Relationship Between Antioxidant Activity and the Structure of Flavonoid Compounds. Rev. Chim. 2019, 70, 3103–3107. [Google Scholar] [CrossRef]
  2. Selma, M.V.; Espín, J.C.; Tomás-Barberán, F.A. Interaction between Phenolics and Gut Microbiota: Role in Human Health. J. Agric. Food Chem. 2009, 57, 6485–6501. [Google Scholar] [CrossRef] [PubMed]
  3. Uddin, M.S.; Kabir, M.T.; Tewari, D.; Al Mamun, A.; Barreto, G.E.; Bungau, S.G.; Bin-Jumah, M.N.; Abdel-Daim, M.M.; Ashraf, G.M. Emerging Therapeutic Promise of Ketogenic Diet to Attenuate Neuropathological Alterations in Alzheimer’s Disease. Mol. Neurobiol. 2020, 57, 4961–4977. [Google Scholar] [CrossRef]
  4. Kumar, S.; Behl, T.; Sachdeva, M.; Sehgal, A.; Kumari, S.; Kumar, A.; Kaur, G.; Yadav, H.N.; Bungau, S. Implicating the Effect of Ketogenic Diet as a Preventive Measure to Obesity and Diabetes Mellitus. Life Sci. 2021, 264, 118661. [Google Scholar] [CrossRef] [PubMed]
  5. Behl, T.; Bungau, S.; Kumar, K.; Zengin, G.; Khan, F.; Kumar, A.; Kaur, R.; Venkatachalam, T.; Tit, D.M.; Vesa, C.M.; et al. Pleotropic Effects of Polyphenols in Cardiovascular System. Biomed. Pharmacother. 2020, 130, 110714. [Google Scholar] [CrossRef] [PubMed]
  6. Behl, T.; Upadhyay, T.; Singh, S.; Chigurupati, S.; Alsubayiel, A.M.; Mani, V.; Vargas-De-la-cruz, C.; Uivarosan, D.; Bustea, C.; Sava, C.; et al. Polyphenols Targeting MAPK Mediated Oxidative Stress and Inflammation in Rheumatoid Arthritis. Molecules 2021, 26, 6570. [Google Scholar] [CrossRef] [PubMed]
  7. Behl, T.; Mehta, K.; Sehgal, A.; Singh, S.; Sharma, N.; Ahmadi, A.; Arora, S.; Bungau, S. Exploring the Role of Polyphenols in Rheumatoid Arthritis. Crit. Rev. Food Sci. Nutr. 2022, 62, 5372–5393. [Google Scholar] [CrossRef]
  8. Kabra, A.; Garg, R.; Brimson, J.; Živković, J.; Almawash, S.; Ayaz, M.; Nawaz, A.; Hassan, S.S.U.; Bungau, S. Mechanistic Insights into the Role of Plant Polyphenols and Their Nano-Formulations in the Management of Depression. Front. Pharm. 2022, 13, 1046599. [Google Scholar] [CrossRef]
  9. Behl, T.; Rana, T.; Alotaibi, G.H.; Shamsuzzaman, M.; Naqvi, M.; Sehgal, A.; Singh, S.; Sharma, N.; Almoshari, Y.; Abdellatif, A.A.H.; et al. Polyphenols Inhibiting MAPK Signalling Pathway Mediated Oxidative Stress and Inflammation in Depression. Biomed. Pharmacother. 2022, 146, 112545. [Google Scholar] [CrossRef]
  10. Bungau, S.; Abdel-Daim, M.M.; Tit, D.M.; Ghanem, E.; Sato, S.; Maruyama-Inoue, M.; Yamane, S.; Kadonosono, K. Health Benefits of Polyphenols and Carotenoids in Age-Related Eye Diseases. Oxid. Med. Cell Longev. 2019, 2019, 9783429. [Google Scholar] [CrossRef]
  11. Carabotti, M.; Scirocco, A.; Antonietta Maselli, M.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  12. Fisette, A.; Sergi, D.; Breton-Morin, A.; Descôteaux, S.; Martinoli, M.-G. New Insights on the Role of Bioactive Food Derivatives in Neurodegeneration and Neuroprotection. Curr. Pharm. Des. 2022, 28, 3068–3081. [Google Scholar] [CrossRef] [PubMed]
  13. Kumar, R.; Amruthanjali, T.; Singothu, S.; Singh, S.B.; Bhandari, V. Uncoupling Proteins as a Therapeutic Target for the Development of New Era Drugs against Neurodegenerative Disorder. Biomed. Pharmacother. 2022, 147, 112656. [Google Scholar] [CrossRef] [PubMed]
  14. He, L.; Wang, J.; Yang, Y.; Li, J.; Tu, H. Mitochondrial Sirtuins in Parkinson’s Disease. Neurochem. Res. 2022, 47, 1491–1502. [Google Scholar] [CrossRef] [PubMed]
  15. Rodrigo, R.; Brito, R.; González-Montero, J.; Benedetti, V. Antioxidants in Human Disease: Potential Therapeutic Opportunities Clinical Pharmacology and Translational Medicine. Clin. Pharm. Transl. Med. 2017, 1, 44–53. [Google Scholar]
  16. Thanan, R.; Oikawa, S.; Hiraku, Y.; Ohnishi, S.; Ma, N.; Pinlaor, S.; Yongvanit, P.; Kawanishi, S.; Murata, M. Oxidative Stress and Its Significant Roles in Neurodegenerative Diseases and Cancer. Int. J. Mol. Sci. 2014, 16, 193–217. [Google Scholar] [CrossRef] [Green Version]
  17. Matsumura, Y.; Kitabatake, M.; Ouji-Sageshima, N.; Yasui, S.; Mochida, N.; Nakano, R.; Kasahara, K.; Tomoda, K.; Yano, H.; Kayano, S.-i.; et al. Persimmon-Derived Tannin Has Bacteriostatic and Anti-Inflammatory Activity in a Murine Model of Mycobacterium Avium Complex (MAC) Disease. PLoS ONE 2017, 12, e0183489. [Google Scholar] [CrossRef] [Green Version]
  18. Kitabatake, M.; Matsumura, Y.; Ouji-Sageshima, N.; Nishioka, T.; Hara, A.; Kayano, S.-i.; Ito, T. Persimmon-Derived Tannin Ameliorates the Pathogenesis of Ulcerative Colitis in a Murine Model through Inhibition of the Inflammatory Response and Alteration of Microbiota. Sci. Rep. 2021, 11, 7286. [Google Scholar] [CrossRef]
  19. Matsumura, Y.; Ito, T.; Yano, H.; Kita, E.; Mikasa, K.; Okada, M.; Furutani, A.; Murono, Y.; Shibata, M.; Nishii, Y.; et al. Antioxidant Potential in Non-Extractable Fractions of Dried Persimmon (Diospyros kaki Thunb.). Food Chem. 2016, 202, 99–103. [Google Scholar] [CrossRef]
  20. Bors, W.; Heller, W.; Michel, C.; Saran, M. Flavonoids as Antioxidants: Determination of Radical-Scavenging Efficiencies. Methods Enzymol. 1990, 186, 343–355. [Google Scholar]
  21. Nanjo, F.; Honda, M.; Okushio, K.; Matsumoto, N.; Ishigaki, F.; Ishigami, T.; Hara, Y. Effects of Dietary Tea Catechins on Alpha-Tocopherol Levels, Lipid Peroxidation, and Erythrocyte Deformability in Rats Fed on High Palm Oil and Perilla Oil Diets. Biol. Pharm. Bull. 1993, 16, 1156–1159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food Sources and Bioavailability. Am J Clin Nutr 2004, 79, 727–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Donovan, J.L.; Manach, C.; Faulks, R.M.; Kroon, P. A In Plant Secondary Metabolites: Occurrence, Structure and Role in the Human Diet; Blackwell Pub: Oxford, UK, 2006; ISBN 9781405125093. [Google Scholar]
  24. Bernatoniene, J.; Kopustinskiene, D.M. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 23, 965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Youn, H.S.; Lee, J.Y.; Saitoh, S.I.; Miyake, K.; Kang, K.W.; Choi, Y.J.; Hwang, D.H. Suppression of MyD88- and TRIF-Dependent Signaling Pathways of Toll-like Receptor by (-)-Epigallocatechin-3-Gallate, a Polyphenol Component of Green Tea. Biochem. Pharm. 2006, 72, 850–859. [Google Scholar] [CrossRef]
  26. Fraga, C.G.; Galleano, M.; Verstraeten, S.V.; Oteiza, P.I. Basic Biochemical Mechanisms behind the Health Benefits of Polyphenols. Mol. Asp. Med. 2010, 31, 435–445. [Google Scholar] [CrossRef]
  27. Fan, F.Y.; Sang, L.X.; Jiang, M. Catechins and Their Therapeutic Benefits to Inflammatory Bowel Disease. Molecules 2017, 22, 484. [Google Scholar] [CrossRef] [Green Version]
  28. Dias, T.R.; Tomás, G.; Teixeira, N.F.; Alves, M.G.; Oliveira, P.F.; Silva, B.M. White Tea (Camellia sinensis (L.)): Antioxidant Properties And Beneficial Health Effects. Int. J. Food Sci. Nutr. Diet. 2013, 2, 19–26. [Google Scholar] [CrossRef]
  29. Hattori, M.; Kusumoto, I.T.; Namba, T.; Ishigami, T.; Hara, Y. Effect of Tea Polyphenols on Glucan Synthesis by Glucosyltransferase from Streptococcus Mutans. Chem. Pharm. Bull. 1990, 38, 717–720. [Google Scholar] [CrossRef] [Green Version]
  30. Matsuzaki, T.; Hara, Y. Antioxidative Activity of Tea Leaf Catechins. Nippon Nogeikagaku Kaishi 1985, 59, 129–134. [Google Scholar] [CrossRef] [Green Version]
  31. Dias, T.R.; Alves, M.G.; Casal, S.; Oliveira, P.F.; Silva, B.M. Promising Potential of Dietary (Poly)Phenolic Compounds in the Prevention and Treatment of Diabetes Mellitus. Curr. Med. Chem. 2017, 24, 334–354. [Google Scholar] [CrossRef]
  32. Moderno, P.M.; Carvalho, M.; Silva, B.M. Recent Patents on Camellia Sinensis: Source of Health Promoting Compounds. Recent Pat. Food Nutr. Agric. 2009, 1, 182–192. [Google Scholar] [CrossRef] [PubMed]
  33. Cho, Y.S.; Schiller, N.L.; Kahng, H.Y.; Oh, K.H. Cellular Responses and Proteomic Analysis of Escherichia Coli Exposed to Green Tea Polyphenols. Curr. Microbiol. 2007, 55, 501–506. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, H.C.; Jenner, A.M.; Low, C.S.; Lee, Y.K. Effect of Tea Phenolics and Their Aromatic Fecal Bacterial Metabolites on Intestinal Microbiota. Res. Microbiol. 2006, 157, 876–884. [Google Scholar] [CrossRef] [PubMed]
  35. Landau, J.M.; Lambert, J.D.; Yang, C.S. Green Tea. In Nutritional Oncology, 2nd, ed.; Academic Press: Cambridge, MA, USA, 2006; Chapter 35; pp. 597–606. [Google Scholar] [CrossRef]
  36. Pereira-Caro, G.; Moreno-Rojas, J.M.; Brindani, N.; del Rio, D.; Lean, M.E.J.; Hara, Y.; Crozier, A. Bioavailability of Black Tea Theaflavins: Absorption, Metabolism, and Colonic Catabolism. J. Agric. Food Chem. 2017, 65, 5365–5374. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, Z.; de Bruijn, W.J.C.; Sanders, M.G.; Wang, S.; Bruins, M.E.; Vincken, J.P. Insights in the Recalcitrance of Theasinensin A to Human Gut Microbial Degradation. J. Agric. Food Chem. 2021, 69, 2477–2484. [Google Scholar] [CrossRef]
  38. Liu, Z.; de Bruijn, W.J.C.; Bruins, M.E.; Vincken, J.P. Microbial Metabolism of Theaflavin-3,3′-Digallate and Its Gut Microbiota Composition Modulatory Effects. J. Agric. Food Chem. 2021, 69, 232–245. [Google Scholar] [CrossRef]
  39. Mulder, T.P.J.; van Platerink, C.J.; Schuyl, P.J.W.; van Amelsvoort, J.M.M. Analysis of Theaflavins in Biological Fluids Using Liquid Chromatography-Electrospray Mass Spectrometry. J. Chromatogr. B Biomed. Sci. Appl. 2001, 760, 271–279. [Google Scholar] [CrossRef]
  40. Hashimoto, F.; Nonaka, G.; Nishioka, I. Tannins and Related Compounds. LXIX.: Isolation and Structure Elucidation of B, B’-Linked Bisflavanoids, Theasinensins D-G and Oolongtheanin from Oolong Tea. (2). Chem. Pharm. Bull. 1988, 36, 1676–1684. [Google Scholar] [CrossRef] [Green Version]
  41. Sorrenti, V.; Ali, S.; Mancin, L.; Davinelli, S.; Paoli, A.; Scapagnini, G. Cocoa Polyphenols and Gut Microbiota Interplay: Bioavailability, Prebiotic Effect, and Impact on Human Health. Nutrients 2020, 12, 1908. [Google Scholar] [CrossRef]
  42. Flores, M.E.J. Cocoa Flavanols: Natural Agents with Attenuating Effects on Metabolic Syndrome Risk Factors. Nutrients 2019, 11, 751. [Google Scholar] [CrossRef] [Green Version]
  43. Natsume, M.; Osakabe, N.; Yamagishi, M.; Takizawa, T.; Nakamura, T.; Miyatake, H.; Hatano, T.; Yoshida, T. Analyses of Polyphenols in Cacao Liquor, Cocoa, and Chocolate by Normal-Phase and Reversed-Phase HPLC. Biosci. Biotechnol. Biochem. 2000, 64, 2581–2587. [Google Scholar] [CrossRef] [PubMed]
  44. Borchers, A.T.; Keen, C.L.; Hannum, S.M.; Gershwin, M.E. Cocoa and Chocolate: Composition, Bioavailability, and Health Implications. J. Med. Food 2000, 3, 77–105. [Google Scholar] [CrossRef]
  45. Gómez-Juaristi, M.; Sarria, B.; Martínez-López, S.; Clemente, L.B.; Mateos, R. Flavanol Bioavailability in Two Cocoa Products with Different Phenolic Content. A Comparative Study in Humans. Nutrients 2019, 11, 1441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Maldonado-Mateus, L.Y.; Perez-Burillo, S.; Lerma-Aguilera, A.; Hinojosa-Nogueira, D.; Ruíz-Pérez, S.; Gosalbes, M.J.; Francino, M.P.; Rufián-Henares, J.Á.; Pastoriza De La Cueva, S. Effect of Roasting Conditions on Cocoa Bioactivity and Gut Microbiota Modulation. Food Funct. 2021, 12, 9680–9692. [Google Scholar] [CrossRef] [PubMed]
  47. Loke, W.M.; Hodgson, J.M.; Proudfoot, J.M.; Mckinley, A.J.; Puddey, I.B.; Croft, K.D. Pure Dietary Flavonoids Quercetin and (−)-Epicatechin Augment Nitric Oxide Products and Reduce Endothelin-1 Acutely in Healthy Men. Am. J. Clin. Nutr. 2008, 88, 1018–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Schroeter, H.; Heiss, C.; Balzer, J.; Kleinbongard, P.; Keen, C.L.; Hollenberg, N.K.; Sies, H.; Kwik-Uribe, C.; Schmitz, H.H.; Kelm, M. (−)-Epicatechin Mediates Beneficial Effects of Flavanol-Rich Cocoa on Vascular Function in Humans. Proc. Natl. Acad. Sci. USA 2006, 103, 1024–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ottaviani, J.I.; Borges, G.; Momma, T.Y.; Spencer, J.P.E.; Keen, C.L.; Crozier, A.; Schroeter, H. The Metabolome of [2-14C](−)-Epicatechin in Humans: Implications for the Assessment of Efficacy, Safety, and Mechanisms of Action of Polyphenolic Bioactives. Sci. Rep. 2016, 6, 29034. [Google Scholar] [CrossRef] [Green Version]
  50. Cremonini, E.; Wang, Z.; Bettaieb, A.; Adamo, A.M.; Daveri, E.; Mills, D.A.; Kalanetra, K.M.; Haj, F.G.; Karakas, S.; Oteiza, P.I. (−)-Epicatechin Protects the Intestinal Barrier from High Fat Diet-Induced Permeabilization: Implications for Steatosis and Insulin Resistance. Redox Biol. 2018, 14, 588–599. [Google Scholar] [CrossRef]
  51. Corral-Jara, K.F.; Nuthikattu, S.; Rutledge, J.; Villablanca, A.; Fong, R.; Heiss, C.; Ottaviani, J.I.; Milenkovic, D. Structurally Related (−)-Epicatechin Metabolites and Gut Microbiota Derived Metabolites Exert Genomic Modifications via VEGF Signaling Pathways in Brain Microvascular Endothelial Cells under Lipotoxic Conditions: Integrated Multi-Omic Study. J. Proteom. 2022, 263, 104603. [Google Scholar] [CrossRef]
  52. Li, B.Y.; Li, H.Y.; Zhou, D.D.; Huang, S.Y.; Luo, M.; Gan, R.Y.; Mao, Q.Q.; Saimaiti, A.; Shang, A.; Li, H. bin Effects of Different Green Tea Extracts on Chronic Alcohol Induced-Fatty Liver Disease by Ameliorating Oxidative Stress and Inflammation in Mice. Oxid. Med. Cell Longev. 2021, 2021, 5188205. [Google Scholar] [CrossRef]
  53. Zhao, L.; Wang, S.; Zhang, N.; Zhou, J.; Mehmood, A.; Raka, R.N.; Zhou, F.; Zhao, L. The Beneficial Effects of Natural Extracts and Bioactive Compounds on the Gut-Liver Axis: A Promising Intervention for Alcoholic Liver Disease. Antioxidants 2022, 11, 1211. [Google Scholar] [CrossRef] [PubMed]
  54. Li, B.; Mao, Q.; Zhou, D.; Luo, M.; Gan, R.; Li, H.; Huang, S.; Saimaiti, A.; Shang, A.; Li, H. Effects of Tea against Alcoholic Fatty Liver Disease by Modulating Gut Microbiota in Chronic Alcohol-Exposed Mice. Foods 2021, 10, 1232. [Google Scholar] [CrossRef] [PubMed]
  55. Naito, Y.; Ushiroda, C.; Mizushima, K.; Inoue, R.; Yasukawa, Z.; Abe, A.; Takagi, T.; Gastroenterology, M. Epigallocatechin-3-Gallate (EGCG) Attenuates Non-Alcoholic Fatty Liver Disease via Modulating the Interaction between Gut Microbiota and Bile Acids. J. Clin. Biochem. Nutr 2020, 67, 2–9. [Google Scholar] [CrossRef] [PubMed]
  56. Sharma, S.P.; Suk, K.T.; Kim, D.J. Significance of Gut Microbiota in Alcoholic and Non-Alcoholic Fatty Liver Diseases. World J. Gastroenterol. 2021, 27, 6161–6179. [Google Scholar] [CrossRef] [PubMed]
  57. He, L.H.; Yao, D.H.; Wang, L.Y.; Zhang, L.; Bai, X.L. Gut Microbiome-Mediated Alteration of Immunity, Inflammation, and Metabolism Involved in the Regulation of Non-Alcoholic Fatty Liver Disease. Front. Microbiol. 2021, 12, 761836. [Google Scholar] [CrossRef]
  58. Zhou, K. Strategies to Promote Abundance of Akkermansia Muciniphila, an Emerging Probiotics in the Gut, Evidence from Dietary Intervention Studies. J. Funct. Foods 2017, 33, 194–201. [Google Scholar] [CrossRef]
  59. Roopchand, D.E.; Carmody, R.N.; Kuhn, P.; Moskal, K.; Rojas-Silva, P.; Turnbaugh, P.J.; Raskin, I. Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia Muciniphila and Attenuate High-Fat Diet-Induced Metabolic Syndrome. Diabetes 2015, 64, 2847–2858. [Google Scholar] [CrossRef] [Green Version]
  60. Dryden, G.W.; Lam, A.; Beatty, K.; Qazzaz, H.H.; McClain, C.J. A Pilot Study to Evaluate the Safety and Efficacy of an Oral Dose of (−)-Epigallocatechin-3-Gallate–Rich Polyphenon E in Patients with Mild to Moderate Ulcerative Colitis. Inflamm. Bowel. Dis. 2013, 19, 1904–1912. [Google Scholar] [CrossRef]
  61. Vasconcelos, P.C.D.P.; Seito, L.N.; di Stasi, L.C.; Akiko Hiruma-Lima, C.; Pellizzon, C.H. Epicatechin Used in the Treatment of Intestinal Inflammatory Disease: An Analysis by Experimental Models. Evid.-Based Complement. Altern. Med. 2012, 2012, 508902. [Google Scholar] [CrossRef] [Green Version]
  62. Brückner, M.; Westphal, S.; Domschke, W.; Kucharzik, T.; Lügering, A. Green Tea Polyphenol Epigallocatechin-3-Gallate Shows Therapeutic Antioxidative Effects in a Murine Model of Colitis. J. Crohns Colitis 2012, 6, 226–235. [Google Scholar] [CrossRef] [Green Version]
  63. Rodríguez-Ramiro, I.; Martín, M.Á.; Ramos, S.; Bravo, L.; Goya, L. Comparative Effects of Dietary Flavanols on Antioxidant Defenses and Their Response to Oxidant-Induced Stress on Caco2 Cells. Eur. J. Nutr. 2011, 50, 313–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cecarini, V.; Cuccioloni, M.; Zheng, Y.; Bonfili, L.; Gong, C.; Angeletti, M.; Mena, P.; del Rio, D.; Eleuteri, A.M. Flavan-3-Ol Microbial Metabolites Modulate Proteolysis in Neuronal Cells Reducing Amyloid-Beta (1-42) Levels. Mol. Nutr. Food Res. 2021, 65, 2100380. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, T.; Yang, Y.; Zhu, S.; Lu, Y.; Zhu, L.; Wang, Y.; Wang, X. Inhibition of Aβ Aggregates in Alzheimer’s Disease by Epigallocatechin and Epicatechin-3-Gallate from Green Tea. Bioorg. Chem. 2020, 105, 104382. [Google Scholar] [CrossRef]
  66. Zhong, X.; Liu, M.; Yao, W.; Du, K.; He, M.; Jin, X.; Jiao, L.; Ma, G.; Wei, B.; Wei, M. Epigallocatechin-3-Gallate Attenuates Microglial Inflammation and Neurotoxicity by Suppressing the Activation of Canonical and Noncanonical Inflammasome via TLR4/NF-ΚB Pathway. Mol. Nutr. Food Res. 2019, 63, 1801230. [Google Scholar] [CrossRef]
  67. Yamamoto, N.; Shibata, M.; Ishikuro, R.; Tanida, M.; Taniguchi, Y.; Ikeda-Matsuo, Y.; Sobue, K. Epigallocatechin Gallate Induces Extracellular Degradation of Amyloid β-Protein by Increasing Neprilysin Secretion from Astrocytes through Activation of ERK and PI3K Pathways. Neuroscience 2017, 362, 70–78. [Google Scholar] [CrossRef]
  68. Cheng-Chung Wei, J.; Huang, H.C.; Chen, W.J.; Huang, C.N.; Peng, C.H.; Lin, C.L. Epigallocatechin Gallate Attenuates Amyloid β-Induced Inflammation and Neurotoxicity in EOC 13.31 Microglia. Eur. J. Pharm. 2016, 770, 16–24. [Google Scholar] [CrossRef]
  69. Bao, J.; Liu, W.; Zhou, H.Y.; Gui, Y.R.; Yang, Y.H.; Wu, M.J.; Xiao, Y.F.; Shang, J.T.; Long, G.F.; Shu, X.J. Epigallocatechin-3-Gallate Alleviates Cognitive Deficits in APP/PS1 Mice. Curr. Med. Sci. 2020, 40, 18–27. [Google Scholar] [CrossRef] [PubMed]
  70. Xu, Q.; Langley, M.; Kanthasamy, A.G.; Reddy, M.B. Epigallocatechin Gallate Has a Neurorescue Effect in a Mouse Model of Parkinson Disease. J. Nutr. 2017, 147, 1926–1931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Sergi, C.M. Epigallocatechin Gallate for Parkinson’s Disease. Clin. Exp. Pharm. Physiol. 2022, 49, 1029–1041. [Google Scholar] [CrossRef]
  72. Kim, S.R.; Seong, K.J.; Kim, W.J.; Jung, J.Y. Epigallocatechin Gallate Protects against Hypoxia-Induced Inflammation in Microglia via NF-ΚB Suppression and Nrf-2/HO-1 Activation. Int. J. Mol. Sci. 2022, 23, 4004. [Google Scholar] [CrossRef]
  73. Jang, S.; Sun, J.; Chen, P.; Lakshman, S.; Molokin, A.; Harnly, J.M.; Vinyard, B.T.; Urban, J.F.; Davis, C.D.; Solano-Aguilar, G. Flavanol-Enriched Cocoa Powder Alters the Intestinal Microbiota, Tissue and Fluid Metabolite Profiles, and Intestinal Gene Expression in Pigs. J. Nutr. 2016, 146, 673–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Tzounis, X.; Rodriguez-Mateos, A.; Vulevic, J.; Gibson, G.R.; Kwik-Uribe, C.; Spencer, J.P.E. Prebiotic Evaluation of Cocoa-Derived Flavanols in Healthy Humans by Using a Randomized, Controlled, Double-Blind, Crossover Intervention Study. Am. J. Clin. Nutr. 2011, 93, 62–72. [Google Scholar] [CrossRef] [Green Version]
  75. Pérez-Cano, F.J.; Massot-Cladera, M.; Franch, À.; Castellote, C.; Castell, M. The Effects of Cocoa on the Immune System. Front. Pharm. 2013, 4, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Massot-Cladera, M.; Pérez-Berezo, T.; Franch, A.; Castell, M.; Pérez-Cano, F.J. Cocoa Modulatory Effect on Rat Faecal Microbiota and Colonic Crosstalk. Arch. Biochem. Biophys. 2012, 527, 105–112. [Google Scholar] [CrossRef]
  77. Álvarez-Cilleros, D.; Ramos, S.; López-Oliva, M.E.; Escrivá, F.; Álvarez, C.; Fernández-Millán, E.; Martín, M.Á. Cocoa Diet Modulates Gut Microbiota Composition and Improves Intestinal Health in Zucker Diabetic Rats. Food Res. Int. 2020, 132, 109058. [Google Scholar] [CrossRef]
  78. Rodríguez-Ramiro, I.; Ramos, S.; López-Oliva, E.; Agis-Torres, A.; Bravo, L.; Goya, L.; Martín, M.A. Cocoa Polyphenols Prevent Inflammation in the Colon of Azoxymethane-Treated Rats and in TNF-α-Stimulated Caco-2 Cells. Br. J. Nutr. 2013, 110, 206–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Agostoni, C.; Bresson, J.-L.; Fairweather-Tait, S.; Flynn, A.; Golly, I.; Korhonen, H.; Lagiou, P.; Løvik, M.; Marchelli, R.; Martin, A.; et al. Scientific Opinion on the Substantiation of a Health Claim Related to Cocoa Flavanols and Maintenance of Normal Endothelium-dependent Vasodilation Pursuant to Article 13(5) of Regulation (EC) No 1924/2006. EFSA J. 2012, 10, 2809. [Google Scholar] [CrossRef] [Green Version]
  80. Sesso, H.D.; Manson, J.E.; Aragaki, A.K.; Rist, P.M.; Johnson, L.G.; Friedenberg, G.; Copeland, T.; Clar, A.; Mora, S.; Moorthy, M.V.; et al. Effect of Cocoa Flavanol Supplementation for the Prevention of Cardiovascular Disease Events: The COcoa Supplement and Multivitamin Outcomes Study (COSMOS) Randomized Clinical Trial. Am. J. Clin. Nutr. 2022, 115, 1490–1500. [Google Scholar] [CrossRef]
  81. Dubner, L.; Wang, J.; Ho, L.; Ward, L.; Pasinetti, G.M. Recommendations for Development of New Standardized Forms of Cocoa Breeds and Cocoa Extract Processing for the Prevention of Alzheimer’s Disease: Role of Cocoa in Promotion of Cognitive Resilience and Healthy Brain Aging. J. Alzheimer’s Dis. 2015, 48, 879–889. [Google Scholar] [CrossRef]
  82. Wang, J.; Varghese, M.; Ono, K.; Yamada, M.; Levine, S.; Tzavaras, N.; Gong, B.; Hurst, W.J.; Blitzer, R.D.; Pasinetti, G.M. Cocoa Extracts Reduce Oligomerization of Amyloid-β: Implications for Cognitive Improvement in Alzheimer’s Disease. J. Alzheimer’s Dis. 2014, 41, 643–650. [Google Scholar] [CrossRef]
  83. Cimini, A.; Gentile, R.; D’Angelo, B.; Benedetti, E.; Cristiano, L.; Avantaggiati, M.L.; Giordano, A.; Ferri, C.; Desideri, G. Cocoa Powder Triggers Neuroprotective and Preventive Effects in a Human Alzheimer’s Disease Model by Modulating BDNF Signaling Pathway. J. Cell Biochem. 2013, 114, 2209–2220. [Google Scholar] [CrossRef] [Green Version]
  84. Oracz, J.; Nebesny, E.; Zyzelewicz, D.; Budryn, G.; Luzak, B. Bioavailability and Metabolism of Selected Cocoa Bioactive Compounds: A Comprehensive Review. Crit. Rev. Food Sci. Nutr. 2020, 60, 1947–1985. [Google Scholar] [CrossRef] [PubMed]
  85. Matsuo, T.; Ito, S. The Chemical Structure of Kaki-Tannin from Immature Fruit of the Persimmon (Diospyros kaki, L.). Agric. Biol. Chem. 1978, 42, 1637–1643. [Google Scholar]
  86. Serrano, J.; Puupponen-Pimiä, R.; Dauer, A.; Aura, A.M.; Saura-Calixto, F. Tannins: Current Knowledge of Food Sources, Intake, Bioavailability and Biological Effects. Mol. Nutr. Food Res. 2009, 53, S310–S329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Matsumoto, K.; Kadowaki, A.; Ozaki, N.; Takenaka, M.; Ono, H.; Yokoyama, S.I.; Gato, N. Bile Acid-Binding Ability of Kaki-Tannin from Young Fruits of Persimmon (Diospyros kaki) In Vitro and In Vivo. Phytother. Res. 2011, 25, 624–628. [Google Scholar] [CrossRef]
  88. Nishida, S.; Katsumi, N.; Matsumoto, K. Prevention of the Rise in Plasma Cholesterol and Glucose Levels by Kaki-tannin and Characterization of Its Bile Acid Binding Capacity. J. Sci. Food Agric. 2021, 101, 2117–2124. [Google Scholar] [CrossRef]
  89. Li, K.; Yao, F.; Du, J.; Deng, X.; Li, C. Persimmon Tannin Decreased the Glycemic Response through Decreasing the Digestibility of Starch and Inhibiting α-Amylase, α-Glucosidase, and Intestinal Glucose Uptake. J. Agric. Food Chem. 2018, 66, 1629–1637. [Google Scholar] [CrossRef]
  90. Zhu, W.; Lin, K.; Li, K.; Deng, X.; Li, C. Reshaped Fecal Gut Microbiota Composition by the Intake of High Molecular Weight Persimmon Tannin in Normal and High-Cholesterol Diet-Fed Rats. Food Funct. 2018, 9, 541–551. [Google Scholar] [CrossRef]
  91. Gorinstein, S.; Bartnikowska, E.; Kulasek, G.; Zemser, M.; Trakhtenberg, S. Dietary Persimmon Improves Lipid Metabolism in Rats Fed Diets Containing Cholesterol. J. Nutr. 1998, 128, 2023–2027. [Google Scholar] [CrossRef] [Green Version]
  92. Gorinstein, S.; Kulasek, G.W.; Bartnikowska, E.; Leontowicz, M.; Zemser, M.; Morawiec, M.; Trakhtenberg, S. The Effects of Diets, Supplemented with Either Whole Persimmon or Phenol-Free Persimmon, on Rats Fed Cholesterol. Food Chem. 2000, 70, 303–308. [Google Scholar] [CrossRef]
  93. Suzuki, T.; Moriguchi, Y.; Ozaki, Y.; Kometani, T.; Fukuda, M. Effects of Kaki-Tannin on Reducing Serum LDL Cholesterol Levels in Volunteers with Borderline and Mild Hyper-LDL Cholestrolemia—A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Comparison Trial. Jpn Pharm. 2022, 50, 237–246. [Google Scholar]
  94. Shabbir, U.; Rubab, M.; Daliri, E.B.M.; Chelliah, R.; Javed, A.; Oh, D.H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  95. Murota, K.; Nakamura, Y.; Uehara, M. Flavonoid Metabolism: The Interaction of Metabolites and Gut Microbiota. Biosci. Biotechnol. Biochem. 2018, 82, 600–610. [Google Scholar] [CrossRef] [Green Version]
  96. Slimestad, R.; Fossen, T.; Vågen, I.M. Onions: A Source of Unique Dietary Flavonoids. J. Agric. Food Chem. 2007, 55, 10067–10080. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, E.J.; Patil, B.S.; Yoo, K.S. Antioxidants of 15 Onions with White, Yellow, and Red Colors and Their Relationship with Pungency, Anthocyanin, and Quercetin. LWT Food Sci. Technol. 2015, 63, 108–114. [Google Scholar] [CrossRef]
  98. Benítez, V.; Mollá, E.; Martín-Cabrejas, M.A.; Aguilera, Y.; López-Andréu, F.J.; Cools, K.; Terry, L.A.; Esteban, R.M. Characterization of Industrial Onion Wastes (Allium Cepa, L.): Dietary Fibre and Bioactive Compounds. Plant Foods Hum. Nutr. 2011, 66, 48–57. [Google Scholar] [CrossRef] [Green Version]
  99. Sharma, K.; Asnin, L.; Ko, E.Y.; Lee, E.T.; Park, S.W. Phytochemical Composition of Onion during Long-Term Storage. Acta Agric. Scand B Soil Plant Sci. 2015, 65, 150–160. [Google Scholar] [CrossRef]
  100. Cattivelli, A.; Conte, A.; Martini, S.; Tagliazucchi, D. Influence of Cooking Methods on Onion Phenolic Compounds Bioaccessibility. Foods 2021, 10, 1023. [Google Scholar] [CrossRef]
  101. Sinkovič, L.; Kokalj Sinkovič, D.; Meglič, V. Milling Fractions Composition of Common (Fagopyrum Esculentum Moench) and Tartary (Fagopyrum tataricum (L.) Gaertn.) Buckwheat. Food Chem. 2021, 365, 130459. [Google Scholar] [CrossRef]
  102. Sytar, O.; Biel, W.; Smetanska, I.; Brestic, M. Bioactive Compounds and Their Biofunctional Properties of Different Buckwheat Germplasms for Food Processing. In Buckwheat Germplasm in the World; Academic Press: London, UK, 2018. [Google Scholar] [CrossRef]
  103. Yasuda, T.; Masaki, K.; Kashiwagi, T. An Enzyme Degrading Rutin in Tartary Buckwheat Seeds. Nippon Shokuhin Kogyo Gakkaishi 1992, 39, 994–1000. [Google Scholar] [CrossRef]
  104. Giménez-Bastida, J.A.; Zieliński, H. Buckwheat as a Functional Food and Its Effects on Health. J. Agric. Food Chem. 2015, 63, 7896–7913. [Google Scholar] [CrossRef] [PubMed]
  105. Formica, J.V; Regelson, W. Review of the Biology of Quercetin and Related Bioflavonoids. Food Chem. Toxic 1995, 33, 1061–1080. [Google Scholar] [CrossRef] [PubMed]
  106. Grzelak-Błaszczyk, K.; Milala, J.; Kosmala, M.; Kołodziejczyk, K.; Sójka, M.; Czarnecki, A.; Klewicki, R.; Juśkiewicz, J.; Fotschki, B.; Jurgoński, A. Onion Quercetin Monoglycosides Alter Microbial Activity and Increase Antioxidant Capacity. J. Nutr. Biochem. 2018, 56, 81–88. [Google Scholar] [CrossRef] [PubMed]
  107. Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Stehle, P.; et al. No Effects of Quercetin from Onion Skin Extract on Serum Leptin and Adiponectin Concentrations in Overweight-to-Obese Patients with (Pre-)Hypertension: A Randomized Double-Blinded, Placebo-Controlled Crossover Trial. Eur. J. Nutr. 2017, 56, 2265–2275. [Google Scholar] [CrossRef]
  108. Dower, J.I.; Geleijnse, J.M.; Gijsbers, L.; Schalkwijk, C.; Kromhout, D.; Hollman, P.C. Supplementation of the Pure Flavonoids Epicatechin and Quercetin Affects Some Biomarkers of Endothelial Dysfunction and Inflammation in (Pre)Hypertensive Adults: A Randomized Double-Blind, Placebo-Controlled, Crossover Trial. J. Nutr. 2015, 145, 1459–1463. [Google Scholar] [CrossRef] [Green Version]
  109. Lee, K.H.; Park, E.; Lee, H.J.; Kim, M.O.; Cha, Y.J.; Kim, J.M.; Lee, H.; Shin, M.J. Effects of Daily Quercetin-Rich Supplementation on Cardiometabolic Risks in Male Smokers. Nutr. Res. Pract. 2011, 5, 28–33. [Google Scholar] [CrossRef] [Green Version]
  110. Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does Quercetin Improve Cardiovascular Risk Factors and Inflammatory Biomarkers in Women with Type 2 Diabetes: A Double-Blind Randomized Controlled Clinical Trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar]
  111. Rezvan, N.; Moini, A.; Janani, L.; Mohammad, K.; Saedisomeolia, A.; Nourbakhsh, M.; Gorgani-Firuzjaee, S.; Mazaherioun, M.; Hosseinzadeh-Attar, M.J. Effects of Quercetin on Adiponectin-Mediated Insulin Sensitivity in Polycystic Ovary Syndrome: A Randomized Placebo-Controlled Double-Blind Clinical Trial. Horm. Metab. Res. 2017, 49, 115–121. [Google Scholar] [CrossRef]
  112. Javadi, F.; Eghtesadi, S.; Ahmadzadeh, A.; Aryaeian, N.; Zabihiyeganeh, M.; Foroushani, A.R.; Jazayeri, S. The Effect of Quercetin on Plasma Oxidative Status, C-Reactive Protein and Blood Pressure in Women with Rheumatoid Arthritis. Int. J. Prev. Med. 2014, 5, 293–301. [Google Scholar]
  113. Mullen, W.; Rouanet, J.-M.; Auger, C.; Teissèdre, P.-L.; Caldwell, S.T.; Hartley, R.C.; Lean, M.E.J.; Edwards, C.A.; Crozier, A. Bioavailability of [2-14C]Quercetin-4′-Glucoside in Rats. J. Agric. Food Chem. 2008, 56, 12127–12137. [Google Scholar] [CrossRef]
  114. Tang, Y.; Nakashima, S.; Saiki, S.; Myoi, Y.; Abe, N.; Kuwazuru, S.; Zhu, B.; Ashida, H.; Murata, Y.; Nakamura, Y. 3,4-Dihydroxyphenylacetic Acid Is a Predominant Biologically-Active Catabolite of Quercetin Glycosides. Food Res. Int. 2016, 89, 716–723. [Google Scholar] [CrossRef] [PubMed]
  115. Verzelloni, E.; Pellacani, C.; Tagliazucchi, D.; Tagliaferri, S.; Calani, L.; Costa, L.G.; Brighenti, F.; Borges, G.; Crozier, A.; Conte, A.; et al. Antiglycative and Neuroprotective Activity of Colon-Derived Polyphenol Catabolites. Mol. Nutr. Food Res. 2011, 55, S35–S43. [Google Scholar] [CrossRef] [PubMed]
  116. Carlsen, I.; Frøkiaer, J.; Nørregaard, R. Quercetin Attenuates Cyclooxygenase-2 Expression in Response to Acute Ureteral Obstruction. Am. J. Physiol. Ren. Physiol. 2015, 308, F1297–F1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Yang, Y.; Chen, G.; Yang, Q.; Ye, J.; Cai, X.; Tsering, P.; Cheng, X.; Hu, C.; Zhang, S.; Cao, P. Gut Microbiota Drives the Attenuation of Dextran Sulphate Sodium-Induced Colitis by Huangqin Decoction. Oncotarget 2017, 8, 48863–48874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Forney, L.A.; Lenard, N.R.; Stewart, L.K.; Henagan, T.M. Dietary Quercetin Attenuates Adipose Tissue Expansion and Inflammation and Alters Adipocyte Morphology in a Tissue-Specific Manner. Int. J. Mol. Sci. 2018, 19, 895. [Google Scholar] [CrossRef] [Green Version]
  119. Overman, A.; Chuang, C.C.; McIntosh, M. Quercetin Attenuates Inflammation in Human Macrophages and Adipocytes Exposed to Macrophage-Conditioned Media. Int. J. Obes. 2011, 35, 1165–1172. [Google Scholar] [CrossRef] [Green Version]
  120. Ju, S.; Ge, Y.; Li, P.; Tian, X.; Wang, H.; Zheng, X.; Ju, S. Dietary Quercetin Ameliorates Experimental Colitis in Mouse by Remodeling the Function of Colonic Macrophages via a Heme Oxygenase-1-Dependent Pathway. Cell Cycle 2018, 17, 53–63. [Google Scholar] [CrossRef] [Green Version]
  121. Shi, T.; Bian, X.; Yao, Z.; Wang, Y.; Gao, W.; Guo, C. Quercetin Improves Gut Dysbiosis in Antibiotic-Treated Mice. Food Funct. 2020, 11, 8003–8013. [Google Scholar] [CrossRef]
  122. Lin, R.; Piao, M.; Song, Y. Dietary Quercetin Increases Colonic Microbial Diversity and Attenuates Colitis Severity in Citrobacter Rodentium-Infected Mice. Front. Microbiol. 2019, 10, 1092. [Google Scholar] [CrossRef] [Green Version]
  123. Sato, S.; Mukai, Y. Modulation of Chronic Inflammation by Quercetin: The Beneficial Effects on Obesity. J. Inflamm. Res. 2020, 13, 421–431. [Google Scholar] [CrossRef]
  124. Jiménez-Aliaga, K.; Bermejo-Bescós, P.; Benedí, J.; Martín-Aragón, S. Quercetin and Rutin Exhibit Antiamyloidogenic and Fibril-Disaggregating Effects in Vitro and Potent Antioxidant Activity in APPswe Cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
  125. Yu, X.; Li, Y.; Mu, X. Effect of Quercetin on PC12 Alzheimer’s Disease Cell Model Induced by A β 25-35 and Its Mechanism Based on Sirtuin1/Nrf2/HO-1 Pathway. Biomed. Res. Int. 2020, 2020, 8210578. [Google Scholar] [CrossRef] [PubMed]
  126. Jiang, W.; Luo, T.; Li, S.; Zhou, Y.; Shen, X.Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin Protects against Okadaic Acid-Induced Injury via MAPK and PI3K/Akt/GSK3β Signaling Pathways in HT22 Hippocampal Neurons. PLoS ONE 2016, 11, e0152371. [Google Scholar] [CrossRef] [PubMed]
  127. Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and Flavones as BACE-1 Inhibitors: Structure-Activity Relationship in Cell-Free, Cell-Based and in Silico Studies Reveal Novel Pharmacophore Features. Biochim. Biophys. Acta Gen. Subj. 2008, 1780, 819–825. [Google Scholar] [CrossRef]
  128. Nakagawa, T.; Ohta, K. Quercetin Regulates the Integrated Stress Response to Improve Memory. Int. J. Mol. Sci. 2019, 20, 2761. [Google Scholar] [CrossRef] [Green Version]
  129. Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular Mechanisms Underlying Protective Effects of Quercetin against Mitochondrial Dysfunction and Progressive Dopaminergic Neurodegeneration in Cell Culture and MitoPark Transgenic Mouse Models of Parkinson’s Disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef] [Green Version]
  130. El-Horany, H.E.; El-Latif, R.N.A.; ElBatsh, M.M.; Emam, M.N. Ameliorative Effect of Quercetin on Neurochemical and Behavioral Deficits in Rotenone Rat Model of Parkinson’s Disease: Modulating Autophagy (Quercetin on Experimental Parkinson’s Disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef] [PubMed]
  131. Sharma, S.; Raj, K.; Singh, S. Neuroprotective Effect of Quercetin in Combination with Piperine Against Rotenone- and Iron Supplement–Induced Parkinson’s Disease in Experimental Rats. Neurotox Res. 2020, 37, 198–209. [Google Scholar] [CrossRef]
  132. Singh, S.; Jamwal, S.; Kumar, P. Neuroprotective Potential of Quercetin in Combination with Piperine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Neurotoxicity. Neural Regen Res. 2017, 12, 1137–1144. [Google Scholar] [CrossRef]
  133. Peng, L.; Zhang, Q.; Zhang, Y.; Yao, Z.; Song, P.; Wei, L.; Zhao, G.; Yan, Z. Effect of Tartary Buckwheat, Rutin, and Quercetin on Lipid Metabolism in Rats during High Dietary Fat Intake. Food Sci. Nutr. 2020, 8, 199–213. [Google Scholar] [CrossRef]
  134. Li, F.; Zhang, X.; Li, Y.; Lu, K.; Yin, R.; Ming, J. Phenolics Extracted from Tartary (Fagopyrum tartaricum, L. Gaerth) Buckwheat Bran Exhibit Antioxidant Activity, and an Antiproliferative Effect on Human Breast Cancer MDA-MB-231 Cells through the P38/MAP Kinase Pathway. Food Funct. 2017, 8, 177–188. [Google Scholar] [CrossRef] [PubMed]
  135. Perk, A.A.; Shatynska-mytsyk, I.; Gerçek, Y.C.; Boztas, K.; Yazgan, M.; Fayyaz, S.; Farooqi, A.A. Rutin Mediated Targeting of Signaling Machinery in Cancer Cells. Cancer Cell Int. 2014, 14, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kumari, A.; Rajput, V.S.; Nagpal, P.; Kukrety, H.; Grover, S.; Grover, A. Dual Inhibition of SARS-CoV-2 Spike and Main Protease through a Repurposed Drug, Rutin. J. Biomol. Struct. Dyn. 2022, 40, 4987–4999. [Google Scholar] [CrossRef] [PubMed]
  137. Rahman, F.; Tabrez, S.; Ali, R.; Alqahtani, A.S.; Ahmed, M.Z.; Rub, A. Molecular Docking Analysis of Rutin Reveals Possible Inhibition of SARS-CoV-2 Vital Proteins. J. Tradit. Complement. Med. 2021, 11, 173–179. [Google Scholar] [CrossRef] [PubMed]
  138. Kreft, I.; Germ, M.; Golob, A.; Vombergar, B.; Bonafaccia, F.; Luthar, Z. Impact of Rutin and Other Phenolic Substances on the Digestibility of Buckwheat Grain Metabolites. Int. J. Mol. Sci. 2022, 23, 3923. [Google Scholar] [CrossRef] [PubMed]
  139. Cirkovic Velickovic, T.D.; Stanic-Vucinic, D.J. The Role of Dietary Phenolic Compounds in Protein Digestion and Processing Technologies to Improve Their Antinutritive Properties. Compr. Rev. Food Sci. Food Saf. 2018, 17, 82–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Wang, L.; Wang, L.; Wang, T.; Li, Z.; Gao, Y.; Cui, S.W.; Qiu, J. Comparison of Quercetin and Rutin Inhibitory Influence on Tartary Buckwheat Starch Digestion in Vitro and Their Differences in Binding Sites with the Digestive Enzyme. Food Chem. 2022, 367, 130762. [Google Scholar] [CrossRef] [PubMed]
  141. Ikeda, K.; Kishida, M. Digestibility of Proteins in Buckwheat Seed. Fagopyrum 1993, 13, 21–24. [Google Scholar]
  142. Zhang, C.; Zhang, R.; Li, Y.M.; Liang, N.; Zhao, Y.; Zhu, H.; He, Z.; Liu, J.; Hao, W.; Jiao, R.; et al. Cholesterol-Lowering Activity of Tartary Buckwheat Protein. J. Agric. Food Chem. 2017, 65, 1900–1906. [Google Scholar] [CrossRef]
  143. Bao, T.; Wang, Y.; Li, Y.T.; Gowd, V.; Niu, X.H.; Yang, H.Y.; Chen, L.S.; Chen, W.; Sun, C.D. Antioxidant and Antidiabetic Properties of Tartary Buckwheat Rice Flavonoids after in Vitro Digestion. J. Zhejiang Univ. Sci. B 2016, 17, 941–951. [Google Scholar] [CrossRef] [Green Version]
  144. Llanaj, E.; Ahanchi, N.S.; Dizdari, H.; Taneri, P.E.; Niehot, C.D.; Wehrli, F.; Khatami, F.; Raeisi-Dehkordi, H.; Kastrati, L.; Bano, A.; et al. Buckwheat and Cardiometabolic Health: A Systematic Review and Meta-Analysis. J. Pers. Med. 2022, 12, 1940. [Google Scholar] [CrossRef] [PubMed]
  145. Enogieru, A.B.; Haylett, W.; Hiss, D.C.; Bardien, S.; Ekpo, O.E. Rutin as a Potent Antioxidant: Implications for Neurodegenerative Disorders. Oxid. Med. Cell Longev. 2018, 2018, 6241017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Noreen, S.; Rizwan, B.; Khan, M.; Farooq, S. Health Benefits of Buckwheat (Fagopyrum Esculentum), Potential Remedy for Diseases, Rare to Cancer: A Mini Review. Infect. Disord. Drug. Targets 2021, 21, e170721189478. [Google Scholar] [CrossRef] [PubMed]
  147. Křížová, L.; Dadáková, K.; Kašparovská, J.; Kašparovský, T. Isoflavones. Molecules 2019, 24, 1076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Kim, I.-S. Current Perspectives on the Beneficial Effects of Soybean Isoflavones and Their Metabolites for Humans. Antioxidants 2021, 10, 1064. [Google Scholar] [CrossRef]
  149. Shinkaruk, S.; Carreau, C.; Flouriot, G.; Bennetau-Pelissero, C.; Potier, M. Comparative Effects of R- and S-Equol and Implication of Transactivation Functions (AF-1 and AF-2) in Estrogen Receptor-Induced Transcriptional Activity. Nutrients 2010, 2, 340–354. [Google Scholar] [CrossRef] [Green Version]
  150. Setchell, K.D.R.; Brown, N.M.; Desai, P.; Zimmer-Nechemias, L.; Wolfe, B.E.; Brashear, W.T.; Kirschner, A.S.; Cassidy, A.; Heubi, J.E. Bioavailability of Pure Isoflavones in Healthy Humans and Analysis of Commercial Soy Isoflavone Supplements. J. Nutr. 2001, 131, 1362S–1375S. [Google Scholar] [CrossRef] [Green Version]
  151. Aguiar, C.L.; Baptista, A.S.; Alencar, S.M.; Haddad, R.; Eberlin, M.N. Analysis of Isoflavonoids from Leguminous Plant Extracts by RPHPLC/DAD and Electrospray Ionization Mass Spectrometry. Int. J. Food Sci. Nutr. 2007, 58, 116–124. [Google Scholar] [CrossRef]
  152. Day, A.J.; DuPont, M.S.; Ridley, S.; Rhodes, M.; Rhodes, M.J.C.; Morgan, M.R.A.; Williamson, G. Deglycosylation of Flavonoid and Isoflavonoid Glycosides by Human Small Intestine and Liver β-Glucosidase Activity. FEBS Lett. 1998, 436, 71–75. [Google Scholar] [CrossRef] [Green Version]
  153. Day, A.J.; Cañada, F.J.; Díaz, J.C.; Kroon, P.A.; Mclauchlan, R.; Faulds, C.B.; Plumb, G.W.; Morgan, M.R.A.; Williamson, G. Dietary Flavonoid and Isoflavone Glycosides Are Hydrolysed by the Lactase Site of Lactase Phlorizin Hydrolase. FEBS Lett. 2000, 468, 166–170. [Google Scholar] [CrossRef] [Green Version]
  154. Mayo, B.; Vázquez, L.; Flórez, A.B. Equol: A Bacterial Metabolite from the Daidzein Isoflavone and Its Presumed Beneficial Health Effects. Nutrients 2019, 11, 2231. [Google Scholar] [CrossRef] [Green Version]
  155. Setchell, K.D.R.; Clerici, C. Equol: History, Chemistry, and Formation. J. Nutr. 2010, 140, 1355S–1362S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Vitale, D.C.; Piazza, C.; Melilli, B.; Drago, F.; Salomone, S. Isoflavones: Estrogenic Activity, Biological Effect and Bioavailability. Eur. J. Drug Metab. Pharm. 2013, 38, 15–25. [Google Scholar] [CrossRef] [PubMed]
  157. Setchell, D.R.K.; Faughnan, M.S.; Avades, T.; Zimmer-Nechemias, L.; Brown, N.M.; Wolfe, B.E.; Brashear, W.T.; Desai, P.; Oldfield, M.F.; Botting, N.P.; et al. Comparing the Pharmacokinetics of Daidzein and Genistein with the Use of 13 C-Labeled Tracers in Premenopausal Women. Am. J. Clin. Nutr. 2003, 77, 411–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Setchell, K.D.; Clerici, C.; Lephart, E.D.; Cole, S.J.; Heenan, C.; Castellani, D.; Wolfe, B.E.; Nechemias-Zimmer, L.; Brown, N.M.; Lund, T.D.; et al. S-Equol, a Potent Ligand for Estrogen Receptor β, Is the Exclusive Enantiomeric Form of the Soy Isoflavone Metabolite Produced by Human Intestinal Bacterial Flora. Am. J. Clin. Nutr. 2005, 81, 1072–1079. [Google Scholar] [CrossRef] [Green Version]
  159. Jackson, R.L.; Greiwe, J.S.; Schwen, R.J. Emerging Evidence of the Health Benefits of S-Equol, an Estrogen Receptor β Agonist. Nutr. Rev. 2011, 69, 432–448. [Google Scholar] [CrossRef]
  160. Wei, X.J.; Wu, J.; Ni, Y.D.; Lu, L.Z.; Zhao, R.Q. Antioxidant Effect of a Phytoestrogen Equol on Cultured Muscle Cells of Embryonic Broilers. Vitr. Cell Dev. Biol. Anim. 2011, 47, 735–741. [Google Scholar] [CrossRef]
  161. Choi, E.J.; Kim, G.H. The Antioxidant Activity of Daidzein Metabolites, O-Desmethylangolensin and Equol, in HepG2 Cells. Mol. Med. Rep. 2014, 9, 328–332. [Google Scholar] [CrossRef] [Green Version]
  162. Messina, M. Soy and Health Update: Evaluation of the Clinical and Epidemiologic Literature. Nutrients 2016, 8, 754. [Google Scholar] [CrossRef] [Green Version]
  163. Harland, J.I.; Haffner, T.A. Systematic Review, Meta-Analysis and Regression of Randomised Controlled Trials Reporting an Association between an Intake of circa 25 g Soya Protein per Day and Blood Cholesterol. Atherosclerosis 2008, 200, 13–27. [Google Scholar] [CrossRef]
  164. Wei, P.; Liu, M.; Chen, Y.; Chen, D.-C.; De-Cai, C. Systematic Review of Soy Isoflavone Supplements on Osteoporosis in Women. Asian Pac. J. Trop Med. 2012, 5, 243–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Jing, Z.; Wei-Jie, Y. Effects of Soy Protein Containing Isoflavones in Patients with Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Clin. Nutr. 2016, 35, 117–124. [Google Scholar] [CrossRef] [PubMed]
  166. Jayachandran, M.; Xu, B. An Insight into the Health Benefits of Fermented Soy Products. Food Chem. 2019, 271, 362–371. [Google Scholar] [CrossRef] [PubMed]
  167. Mueller, S.O.; Simon, S.; Chae, K.; Metzler, M.; Korach, K.S. Phytoestrogens and Their Human Metabolites Show Distinct Agonistic and Antagonistic Properties on Estrogen Receptor α (ERα) and ERβ in Human Cells. Toxicol. Sci. 2004, 80, 14–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Uddin, M.S.; Kabir, M.T. Emerging Signal Regulating Potential of Genistein against Alzheimer’s Disease: A Promising Molecule of Interest. Front. Cell Dev. Biol. 2019, 7, 197. [Google Scholar] [CrossRef]
  169. Lu, C.; Wang, Y.; Wang, D.; Zhang, L.; Lv, J.; Jiang, N.; Fan, B.; Liu, X.; Wang, F. Neuroprotective Effects of Soy Isoflavones on Scopolamine-Induced Amnesia in Mice. Nutrients 2018, 10, 853. [Google Scholar] [CrossRef] [Green Version]
  170. Ye, S.; Wang, T.T.; Cai, B.; Wang, Y.; Li, J.; Zhan, J.X.; Shen, G.M. Genistein Protects Hippocampal Neurons against Injury by Regulating Calcium/Calmodulin Dependent Protein Kinase IV Protein Levels in Alzheimer’s Disease Model Rats. Neural Regen. Res. 2017, 12, 1479–1484. [Google Scholar] [CrossRef]
  171. Arbabi, E.; Hamidi, G.; Talaei, S.A.; Salami, M. Estrogen Agonist Genistein Differentially Influences the Cognitive and Motor Disorders in an Ovariectomized Animal Model of Parkinsonism. Iran J. Basic. Med. Sci. 2016, 19, 1285–1290. [Google Scholar] [CrossRef]
  172. Razeghi Jahromi, S.; Rafi Arrefhosseini, S.; Ghaemi, A.; Alizadeh, A.; Sabetghadam, F.; Togha, M.; Jahromi, R.S. Effect of Oral Genistein Administration in Early and Late Phases of Allergic Encephalomyelitis. Iran J. Basic Med. Sci. 2014, 17, 509–515. [Google Scholar]
  173. Franca Adriana, S.; Oliveira Leandro, S. Coffee and Its By-Products as Sources of Bioactive Compounds; Massey, J.L., Ed.; Nova Science Publishers, Inc.: New York, NY, USA, 2016; ISBN 978-1-63484-714-8. [Google Scholar]
  174. Clifford, M.N.; Jaganath, I.B.; Ludwig, I.A.; Crozier, A. Chlorogenic Acids and the Acyl-Quinic Acids: Discovery, Biosynthesis, Bioavailability and Bioactivity. Nat. Prod. Rep. 2017, 34, 1391–1421. [Google Scholar] [CrossRef] [Green Version]
  175. Perrone, D.; Farah, A.; Donangelo, C.M.; de Paulis, T.; Martin, P.R. Comprehensive Analysis of Major and Minor Chlorogenic Acids and Lactones in Economically Relevant Brazilian Coffee Cultivars. Food Chem. 2008, 106, 859–867. [Google Scholar] [CrossRef]
  176. Clifford, M.N. Chemical and Physical Aspects of Green Coffee and Coffee Products. In Coffee: Botany, Biochemistry and Production of Beans and Beverage; Croom Helm: London, UK, 1985; Chapter 13; pp. 305–374. ISBN 978-1-4615-6659-5. [Google Scholar]
  177. Stalmach, A.; Steiling, H.; Williamson, G.; Crozier, A. Bioavailability of Chlorogenic Acids Following Acute Ingestion of Coffee by Humans with an Ileostomy. Arch. Biochem. Biophys. 2010, 501, 98–105. [Google Scholar] [CrossRef]
  178. Olthof, M.R.; Hollman, P.C.H.; Katan, M.B. Chlorogenic Acid and Caffeic Acid Are Absorbed in Humans. J. Nutr. 2001, 131, 66–71. [Google Scholar] [CrossRef] [Green Version]
  179. Langyan, S.; Yadava, P.; Sharma, S.; Gupta, N.C.; Bansal, R.; Yadav, R.; Kalia, S.; Kumar, A. Food and Nutraceutical Functions of Sesame Oil: An Underutilized Crop for Nutritional and Health Benefits. Food Chem. 2022, 389, 132990. [Google Scholar] [CrossRef]
  180. Pathak, N.; Rai, A.K.; Kumari, R.; Bhat, K. v. Value Addition in Sesame: A Perspective on Bioactive Components for Enhancing Utility and Profitability. Pharm. Rev. 2014, 8, 147–155. [Google Scholar] [CrossRef] [Green Version]
  181. Andargie, M.; Vinas, M.; Rathgeb, A.; Möller, E.; Karlovsky, P. Lignans of Sesame (Sesamum indicum, L.): A Comprehensive Review. Molecules 2021, 26, 883. [Google Scholar] [CrossRef]
  182. Wan, Y.; Li, H.; Fu, G.; Chen, X.; Chen, F.; Xie, M. The Relationship of Antioxidant Components and Antioxidant Activity of Sesame Seed Oil. J. Sci. Food Agric. 2015, 95, 2571–2578. [Google Scholar] [CrossRef]
  183. Nakai, M.; Harada, M.; Nakahara, K.; Akimoto, K.; Shibata, H.; Miki, W.; Kiso, Y. Novel Antioxidative Metabolites in Rat Liver with Ingested Sesamin. J. Agric. Food Chem. 2003, 51, 1666–1670. [Google Scholar] [CrossRef]
  184. Kang, M.-H.; Naito, M.; Tsujihara, N.; Osawa, T. Nutrient Metabolism Sesamolin Inhibits Lipid Peroxidation in Rat Liver and Kidney 1. J. Nutr. 1998, 128, 1018–1022. [Google Scholar] [CrossRef] [Green Version]
  185. Cha, J.W.; Piao, M.J.; Kim, K.C.; Yao, C.W.; Zheng, J.; Kim, S.M.; Hyun, C.L.; Ahn, Y.S.; Hyun, J.W. The Polyphenol Chlorogenic Acid Attenuates UVB-Mediated Oxidative Stress in Human HaCaT Keratinocytes. Biomol. Ther. 2014, 22, 136–142. [Google Scholar] [CrossRef] [Green Version]
  186. Zang, L.-Y.; Cosma, G.; Gardner, H.; Castranova, V.; Vallyathan, V. Effect of Chlorogenic Acid on Hydroxyl Radical. Mol. Cell. Biochem. 2003, 247, 205–210. [Google Scholar] [CrossRef] [PubMed]
  187. Kono, Y.; Kobayashi, K.; Tagawa, S.; Adachi, K.; Ueda, A.; Sawa, Y.; Shibata, H. Antioxidant Activity of Polyphenolics in Diets Rate Constants of Reactions of Chlorogenic Acid and Caffeic Acid with Reactive Species of Oxygen and Nitrogen. Biochim. Biophys. Acta 1997, 1335, 335–342. [Google Scholar] [CrossRef] [PubMed]
  188. He, X.; Zheng, S.; Sheng, Y.; Miao, T.; Xu, J.; Xu, W.; Huang, K.; Zhao, C. Chlorogenic Acid Ameliorates Obesity by Preventing Energy Balance Shift in High-Fat Diet Induced Obese Mice. J. Sci. Food Agric. 2021, 101, 631–637. [Google Scholar] [CrossRef] [PubMed]
  189. Ye, X.; Liu, Y.; Hu, J.; Gao, Y.; Ma, Y.; Wen, D. Chlorogenic Acid-Induced Gut Microbiota Improves Metabolic Endotoxemia. Front. Endocrinol. 2021, 12, 762691. [Google Scholar] [CrossRef] [PubMed]
  190. Wang, Z.; Lam, K.L.; Hu, J.; Ge, S.; Zhou, A.; Zheng, B.; Zeng, S.; Lin, S. Chlorogenic Acid Alleviates Obesity and Modulates Gut Microbiota in High-Fat-Fed Mice. Food Sci. Nutr. 2019, 7, 579–588. [Google Scholar] [CrossRef]
  191. Huxley, R.; Man Ying Lee, C.; Barzi, F.; Timmermeister, L.; Czernichow, S.; Perkovic, V.; Grobbee, D.E.; Batty, D.; Woodward, M. Coffee, Decaffeinated Coffee, and Tea Consumption in Relation to Incident Type 2 Diabetes Mellitus. Arch. Intern. Med. 2009, 169, 2053–2063. [Google Scholar] [CrossRef]
  192. Mills, C.E.; Tzounis, X.; Oruna-Concha, M.J.; Mottram, D.S.; Gibson, G.R.; Spencer, J.P.E. In Vitro Colonic Metabolism of Coffee and Chlorogenic Acid Results in Selective Changes in Human Faecal Microbiota Growth. Br. J. Nutr. 2015, 113, 1220–1227. [Google Scholar] [CrossRef] [Green Version]
  193. Sales, A.L.; de Paula, J.; Mellinger Silva, C.; Cruz, A.; Lemos Miguel, M.A.; Farah, A. Effects of Regular and Decaffeinated Roasted Coffee (Coffea arabica and Coffea canephora) Extracts and Bioactive Compounds on in Vitro Probiotic Bacterial Growth. Food Funct. 2020, 11, 1410–1424. [Google Scholar] [CrossRef]
  194. Socała, K.; Szopa, A.; Serefko, A.; Poleszak, E.; Wlaź, P. Neuroprotective Effects of Coffee Bioactive Compounds: A Review. Int. J. Mol. Sci. 2020, 22, 107. [Google Scholar] [CrossRef]
  195. Singh, S.S.; Rai, S.N.; Birla, H.; Zahra, W.; Rathore, A.S.; Dilnashin, H.; Singh, R.; Singh, S.P. Neuroprotective Effect of Chlorogenic Acid on Mitochondrial Dysfunction-Mediated Apoptotic Death of Da Neurons in a Parkinsonian Mouse Model. Oxid. Med. Cell Longev. 2020, 2020, 6571484. [Google Scholar] [CrossRef]
  196. Miyazaki, I.; Isooka, N.; Wada, K.; Kikuoka, R.; Kitamura, Y.; Asanuma, M. Effects of Enteric Environmental Modification by Coffee Components on Neurodegeneration in Rotenone-Treated Mice. Cells 2019, 8, 221. [Google Scholar] [CrossRef] [Green Version]
  197. Singh, S.S.; Rai, S.N.; Birla, H.; Zahra, W.; Kumar, G.; Gedda, M.R.; Tiwari, N.; Patnaik, R.; Singh, R.K.; Singh, S.P. Effect of Chlorogenic Acid Supplementation in MPTP-Intoxicated Mouse. Front. Pharm. 2018, 9, 757. [Google Scholar] [CrossRef] [Green Version]
  198. Gao, L.; Li, X.; Meng, S.; Ma, T.; Wan, L.; Xu, S. Chlorogenic Acid Alleviates Aβ25-35-Induced Autophagy and Cognitive Impairment via the MTOR/TFEB Signaling Pathway. Drug. Des. Devel. 2020, 14, 1705–1716. [Google Scholar] [CrossRef]
  199. Kamal-Eldin, A.; Moazzami, A.; Washi, S. Sesame Seed Lignans: Potent Physiological Modulators and Possible Ingredients in Functional Foods & Nutraceuticals. Recent Pat. Food Nutr. Agric. 2011, 3, 17–29. [Google Scholar] [CrossRef]
  200. Dalibalta, S.; Majdalawieh, A.F.; Manjikian, H. Health Benefits of Sesamin on Cardiovascular Disease and Its Associated Risk Factors. Saudi Pharm. J. 2020, 28, 1276–1289. [Google Scholar] [CrossRef]
  201. Wu, W.-H.; Kang, Y.-P.; Wang, N.-H.; Jou, H.-J.; Wang, T.-A. Sesame Ingestion Affects Sex Hormones, Antioxidant Status, and Blood Lipids in Postmenopausal Women. J. Nutr. 2006, 136, 1270–1275. [Google Scholar] [CrossRef] [Green Version]
  202. Oikawa, D.; Yamashita, S.; Takahashi, S.; Waki, T.; Kikuchi, K.; Abe, T.; Katayama, T.; Nakayama, T. (+)-Sesamin, a Sesame Lignan, is a Potent Inhibitor of Gut Bacterial Tryptophan Indole-Lyase That Is a Key Enzyme in Chronic Kidney Disease Pathogenesis. Biochem. Biophys. Res. Commun. 2022, 590, 158–162. [Google Scholar] [CrossRef]
  203. Zhu, X.; Zhang, X.; Sun, Y.; Su, D.; Sun, Y.; Hu, B.; Zeng, X. Purification and Fermentation in Vitro of Sesaminol Triglucoside from Sesame Cake by Human Intestinal Microbiota. J. Agric. Food Chem. 2013, 61, 1868–1877. [Google Scholar] [CrossRef]
  204. Wang, M.; Liu, P.; Kong, L.; Xu, N.; Lei, H. Promotive Effects of Sesamin on Proliferation and Adhesion of Intestinal Probiotics and Its Mechanism of Action. Food Chem. Toxicol. 2021, 149, 112049. [Google Scholar] [CrossRef]
  205. Yun, D.; Wang, Y.; Zhang, Y.; Jia, M.; Xie, T.; Zhao, Y.; Yang, C.; Chen, W.; Guo, R.; Liu, X.; et al. Sesamol Attenuates Scopolamine-Induced Cholinergic Disorders, Neuroinflammation, and Cognitive Deficits in Mice. J. Agric. Food Chem. 2022, 70, 13602–13614. [Google Scholar] [CrossRef]
  206. Liu, Q.; Xie, T.; Xi, Y.; Li, L.; Mo, F.; Liu, X.; Liu, Z.; Gao, J.-M.; Yuan, T. Sesamol Attenuates Amyloid Peptide Accumulation and Cognitive Deficits in APP/PS1 Mice: The Mediating Role of the Gut–Brain Axis. J. Agric. Food Chem. 2021, 69, 12717–12729. [Google Scholar] [CrossRef]
  207. Ruankham, W.; Suwanjang, W.; Wongchitrat, P.; Prachayasittikul, V.; Prachayasittikul, S.; Phopin, K. Sesamin and Sesamol Attenuate H2O2-Induced Oxidative Stress on Human Neuronal Cells via the SIRT1-SIRT3-FOXO3a Signaling Pathway. Nutr. Neurosci. 2021, 24, 90–101. [Google Scholar] [CrossRef]
  208. Kongtawelert, P.; Kaewmool, C.; Phitak, T.; Phimphilai, M.; Pothacharoen, P.; Shwe, T.H. Sesamin Protects against Neurotoxicity via Inhibition of Microglial Activation under High Glucose Circumstances through Modulating P38 and JNK Signaling Pathways. Sci. Rep. 2022, 12, 11296. [Google Scholar] [CrossRef]
  209. Keowkase, R.; Shoomarom, N.; Bunargin, W.; Sitthithaworn, W.; Weerapreeyakul, N. Sesamin and Sesamolin Reduce Amyloid-β Toxicity in a Transgenic Caenorhabditis Elegans. Biomed. Pharmacother. 2018, 107, 656–664. [Google Scholar] [CrossRef]
  210. Rothwell, J.A.; Perez-Jimenez, J.; Neveu, V.; Medina-Remón, A.; M’hiri, N.; García-Lobato, P.; Manach, C.; Knox, C.; Eisner, R.; Wishart, D.S.; et al. Phenol-Explorer 3.0: A Major Update of the Phenol-Explorer Database to Incorporate Data on the Effects of Food Processing on Polyphenol Content. Database 2013, 2013, bat070. [Google Scholar] [CrossRef]
  211. Gitea, M.A.; Bungau, S.G.; Gitea, D.; Pasca, B.M.; Purza, A.L.; Radu, A.-F. Evaluation of the Phytochemistry–Therapeutic Activity Relationship for Grape Seeds Oil. Life 2023, 13, 178. [Google Scholar] [CrossRef]
  212. Walle, T. Bioavailability of Resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
  213. Francioso, A.; Mastromarino, P.; Masci, A.; d’Erme, M.; Mosca, L. Chemistry, Stability and Bioavailability of Resveratrol. Med. Chem. 2014, 10, 237–245. [Google Scholar] [CrossRef]
  214. Wenzel, E.; Somoza, V. Metabolism and Bioavailability of Trans-Resveratrol. Mol. Nutr. Food Res. 2005, 49, 472–481. [Google Scholar] [CrossRef]
  215. Wu, B.; Basu, S.; Meng, S.; Wang, X.; Zhang, S.; Hu, M. Regioselective Sulfation and Glucuronidation of Phenolics: Insights into the Structural Basis of Conjugation. Curr. Drug Metab. 2011, 12, 900–916. [Google Scholar] [CrossRef]
  216. Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.A.P.; Kulling, S.E. In Vivo and in Vitro Metabolism of Trans-Resveratrol by Human Gut Microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef] [Green Version]
  217. El-Mohsen, M.A.; Bayele, H.; Kuhnle, G.; Gibson, G.; Debnam, E.; Srai, S.K.; Rice-Evans, C.; Spencer, J.P.E. Distribution of [H]Trans-Resveratrol in Rat Tissues Following Oral Administration. Br. J. Nutr. 2006, 96, 62. [Google Scholar] [CrossRef] [Green Version]
  218. Renaud, J.; Martinoli, M.-G. Resveratrol as a Protective Molecule for Neuroinflammation: A Review of Mechanisms. Curr. Pharm. Biotechnol. 2014, 15, 318–329. [Google Scholar] [CrossRef]
  219. Vidavalur, R.; Otani, H.; Singal, P.K.; Maulik, N. Significance of Wine and Resveratrol in Cardiovascular Disease: French Paradox Revisited. Exp. Clin. Cardiol. 2006, 11, 217–225. [Google Scholar]
  220. Renaud, S.; de Lorgeril, M. Wine, Alcohol, Platelets, and the French Paradox for Coronary Heart Disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef]
  221. Song, X.; Liu, L.; Peng, S.; Liu, T.; Chen, Y.; Jia, R.; Zou, Y.; Li, L.; Zhao, X.; Liang, X.; et al. Resveratrol Regulates Intestinal Barrier Function in Cyclophosphamide-Induced Immunosuppressed Mice. J. Sci. Food Agric. 2022, 102, 1205–1215. [Google Scholar] [CrossRef]
  222. Yao, M.; Fei, Y.; Zhang, S.; Qiu, B.; Zhu, L.; Li, F.; Berglund, B.; Xiao, H.; Li, L. Gut Microbiota Composition in Relation to the Metabolism of Oral Administrated Resveratrol. Nutrients 2022, 14, 1013. [Google Scholar] [CrossRef]
  223. Wang, P.; Li, D.; Ke, W.; Liang, D.; Hu, X.; Chen, F. Resveratrol-Induced Gut Microbiota Reduces Obesity in High-Fat Diet-Fed Mice. Int. J. Obes. 2020, 44, 213–225. [Google Scholar] [CrossRef]
  224. Wang, P.; Wang, J.; Li, D.; Ke, W.; Chen, F.; Hu, X. Targeting the Gut Microbiota with Resveratrol: A Demonstration of Novel Evidence for the Management of Hepatic Steatosis. J. Nutr. Biochem. 2020, 81, 108363. [Google Scholar] [CrossRef]
  225. Chen, M.; Hou, P.; Zhou, M.; Ren, Q.; Wang, X.; Huang, L.; Hui, S.; Yi, L.; Mi, M. Resveratrol Attenuates High-Fat Diet-Induced Non-Alcoholic Steatohepatitis by Maintaining Gut Barrier Integrity and Inhibiting Gut Inflammation through Regulation of the Endocannabinoid System. Clin. Nutr. 2020, 39, 1264–1275. [Google Scholar] [CrossRef]
  226. Zhang, B.; Xu, Y.; Lv, H.; Pang, W.; Wang, J.; Ma, H.; Wang, S. Intestinal Pharmacokinetics of Resveratrol and Regulatory Effects of Resveratrol Metabolites on Gut Barrier and Gut Microbiota. Food Chem. 2021, 357, 129532. [Google Scholar] [CrossRef]
  227. Korsholm, A.S.; Kjær, T.N.; Ornstrup, M.J.; Pedersen, S.B. Comprehensive Metabolomic Analysis in Blood, Urine, Fat, and Muscle in Men with Metabolic Syndrome: A Randomized, Placebo-Controlled Clinical Trial on the Effects of Resveratrol after Four Months’ Treatment. Int. J. Mol. Sci. 2017, 18, 554. [Google Scholar] [CrossRef] [Green Version]
  228. Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R.H.; van de Weijer, T.; Goossens, G.H.; Hoeks, J.; van der Krieken, S.; Ryu, D.; Kersten, S.; et al. Calorie Restriction-like Effects of 30 Days of Resveratrol Supplementation on Energy Metabolism and Metabolic Profile in Obese Humans. Cell Metab. 2011, 14, 612–622. [Google Scholar] [CrossRef] [Green Version]
  229. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High Absorption but Very Low Bioavailability of Oral Resveratrol in Humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
  230. Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; di Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef] [Green Version]
  231. Golan, R.; Shelef, I.; Shemesh, E.; Henkin, Y.; Schwarzfuchs, D.; Gepner, Y.; Harman-Boehm, I.; Witkow, S.; Friger, M.; Chassidim, Y.; et al. Effects of Initiating Moderate Wine Intake on Abdominal Adipose Tissue in Adults with Type 2 Diabetes: A 2-Year Randomized Controlled Trial. Public Health Nutr. 2017, 20, 549–555. [Google Scholar] [CrossRef]
  232. Novakovic, R.; Rajkovic, J.; Gostimirovic, M.; Gojkovic-Bukarica, L.; Radunovic, N. Resveratrol and Reproductive Health. Life 2022, 12, 294. [Google Scholar] [CrossRef]
  233. Shivananda Nayak, B.; Dan Ramdath, D.; Marshall, J.R.; Isitor, G.; Xue, S.; Shi, J. Wound-Healing Properties of the Oils of Vitis Vinifera and Vaccinium Macrocarpon. Phytother. Res. 2011, 25, 1201–1208. [Google Scholar] [CrossRef]
  234. Al-Warhi, T.; Zahran, E.M.; Selim, S.; Al-Sanea, M.M.; Ghoneim, M.M.; Maher, S.A.; Mostafa, Y.A.; Alsenani, F.; Elrehany, M.A.; Almuhayawi, M.S.; et al. Antioxidant and Wound Healing Potential of Vitis Vinifera Seeds Supported by Phytochemical Characterization and Docking Studies. Antioxidants 2022, 11, 881. [Google Scholar] [CrossRef]
  235. Niknami, E.; Sajjadi, S.-E.; Talebi, A.; Minaiyan, M. Protective Effect of Vitis Vinifera (Black Grape) Seed Extract and Oil on Acetic Acid-Induced Colitis in Rats. Int. J. Prev. Med. 2020, 11, 102. [Google Scholar] [CrossRef]
  236. Ismail, A.F.M.; Salem, A.A.M.; Eassawy, M.M.T. Hepatoprotective Effect of Grape Seed Oil against Carbon Tetrachloride Induced Oxidative Stress in Liver of γ-Irradiated Rat. J. Photochem. Photobiol. B 2016, 160, 1–10. [Google Scholar] [CrossRef] [PubMed]
  237. Lai, X.; Kang, X.; Zeng, L.; Li, J.; Yang, Y.; Liu, D. The Protective Effects and Genetic Pathways of Thorn Grape Seeds Oil against High Glucose-Induced Apoptosis in Pancreatic β-Cells. BMC Complement. Altern Med. 2014, 14, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Sharif, A.; Akhtar, N.; Khan, M.S.; Menaa, A.; Menaa, B.; Khan, B.A.; Menaa, F. Formulation and Evaluation on Human Skin of a Water-in-Oil Emulsion Containing Muscat Hamburg Black Grape Seed Extract. Int. J. Cosmet. Sci. 2015, 37, 253–258. [Google Scholar] [CrossRef] [PubMed]
  239. Navaee, M.; Rakhshkhorshid, M. Comparing the Effect of Foot Massage with Grape Seed Oil and Sweet Almond Oil on Physiological Leg Edema in Primigravidae: A Randomized Clinical Trial. Evid. Based Complement. Altern. Med. 2020, 2020, 6835814. [Google Scholar] [CrossRef] [PubMed]
  240. Kaseb, F.; Biregani, A.N. Effects of Olive Oil and Grape Seed Oil on Lipid Profile and Blood Pressure in Patients with Hyperlipidemia: A Randomized Clinical Trial. Food Nutr. Sci. 2016, 07, 682–688. [Google Scholar] [CrossRef] [Green Version]
  241. Al-Edresi, S.; Alsalahat, I.; Freeman, S.; Aojula, H.; Penny, J. Resveratrol-Mediated Cleavage of Amyloid Β1–42 Peptide: Potential Relevance to Alzheimer’s Disease. Neurobiol. Aging 2020, 94, 24–33. [Google Scholar] [CrossRef]
  242. Xie, J.; Li, X.; Zhou, Y.; Wu, J.; Tan, Y.; Ma, X.; Zhao, Y.; Liu, X.; Zhao, Y. Resveratrol Abrogates Hypoxia-Induced Up-Regulation of Exosomal Amyloid-β Partially by Inhibiting CD147. Neurochem. Res. 2019, 44, 1113–1126. [Google Scholar] [CrossRef]
  243. Corpas, R.; Griñán-Ferré, C.; Rodríguez-Farré, E.; Pallàs, M.; Sanfeliu, C. Resveratrol Induces Brain Resilience Against Alzheimer Neurodegeneration Through Proteostasis Enhancement. Mol. Neurobiol. 2019, 56, 1502–1516. [Google Scholar] [CrossRef] [Green Version]
  244. Chiang, M.C.; Nicol, C.J.; Cheng, Y.C. Resveratrol Activation of AMPK-Dependent Pathways Is Neuroprotective in Human Neural Stem Cells against Amyloid-Beta-Induced Inflammation and Oxidative Stress. Neurochem. Int. 2018, 115, 1–10. [Google Scholar] [CrossRef]
  245. Xia, D.; Sui, R.; Zhang, Z. Administration of Resveratrol Improved Parkinson’s Disease-like Phenotype by Suppressing Apoptosis of Neurons via Modulating the MALAT1/MiR-129/SNCA Signaling Pathway. J. Cell Biochem. 2019, 120, 4942–4951. [Google Scholar] [CrossRef]
  246. Liu, Q.; Zhu, D.; Jiang, P.; Tang, X.; Lang, Q.; Yu, Q.; Zhang, S.; Che, Y.; Feng, X. Resveratrol Synergizes with Low Doses of L-DOPA to Improve MPTP-Induced Parkinson Disease in Mice. Behav. Brain Res. 2019, 367, 10–18. [Google Scholar] [CrossRef] [PubMed]
  247. Wang, Z.H.; Zhang, J.L.; Duan, Y.L.; Zhang, Q.S.; Li, G.F.; Zheng, D.L. MicroRNA-214 Participates in the Neuroprotective Effect of Resveratrol via Inhibiting α-Synuclein Expression in MPTP-Induced Parkinson’s Disease Mouse. Biomed. Pharmacother. 2015, 74, 252–256. [Google Scholar] [CrossRef] [PubMed]
  248. Naia, L.; Rosenstock, T.R.; Oliveira, A.M.; Oliveira-Sousa, S.I.; Caldeira, G.L.; Carmo, C.; Laço, M.N.; Hayden, M.R.; Oliveira, C.R.; Rego, A.C. Comparative Mitochondrial-Based Protective Effects of Resveratrol and Nicotinamide in Huntington’s Disease Models. Mol. Neurobiol. 2017, 54, 5385–5399. [Google Scholar] [CrossRef] [PubMed]
  249. Ghaiad, H.R.; Nooh, M.M.; El-Sawalhi, M.M.; Shaheen, A.A. Resveratrol Promotes Remyelination in Cuprizone Model of Multiple Sclerosis: Biochemical and Histological Study. Mol. Neurobiol. 2017, 54, 3219–3229. [Google Scholar] [CrossRef] [PubMed]
  250. Mancuso, R.; del Valle, J.; Modol, L.; Martinez, A.; Granado-Serrano, A.B.; Ramirez-Núñez, O.; Pallás, M.; Portero-Otin, M.; Osta, R.; Navarro, X. Resveratrol Improves Motoneuron Function and Extends Survival in SOD1G93A ALS Mice. Neurotherapeutics 2014, 11, 419–432. [Google Scholar] [CrossRef] [Green Version]
  251. Sato, F.; Martinez, N.E.; Shahid, M.; Rose, J.W.; Carlson, N.G.; Tsunoda, I. Resveratrol Exacerbates Both Autoimmune and Viral Models of Multiple Sclerosis. Am. J. Pathol. 2013, 183, 1390–1396. [Google Scholar] [CrossRef] [Green Version]
  252. Bresciani, L.; Favari, C.; Calani, L.; Francinelli, V.; Riva, A.; Petrangolini, G.; Allegrini, P.; Mena, P.; del Rio, D. The Effect of Formulation of Curcuminoids on Their Metabolism by Human Colonic Microbiota. Molecules 2020, 25, 940. [Google Scholar] [CrossRef] [Green Version]
  253. Chikara, S.; Nagaprashantha, L.D.; Singhal, J.; Horne, D.; Awasthi, S.; Singhal, S.S. Oxidative Stress and Dietary Phytochemicals: Role in Cancer Chemoprevention and Treatment. Cancer Lett. 2018, 413, 122–134. [Google Scholar] [CrossRef]
  254. Stohs, S.J.; Chen, O.; Ray, S.D.; Ji, J.; Bucci, L.R.; Preuss, H.G. Highly Bioavailable Forms of Curcumin and Promising Avenues for Curcumin-Based Research and Application: A Review. Molecules 2020, 25, 1397. [Google Scholar] [CrossRef] [Green Version]
  255. Zam, W. Gut Microbiota as a Prospective Therapeutic Target for Curcumin: A Review of Mutual Influence. J. Nutr. Metab. 2018, 2018, 1367984. [Google Scholar] [CrossRef]
  256. Tan, S.; Rupasinghe, T.W.T.; Tull, D.L.; Boughton, B.; Oliver, C.; McSweeny, C.; Gras, S.L.; Augustin, M.A. Degradation of Curcuminoids by in Vitro Pure Culture Fermentation. J. Agric. Food Chem. 2014, 62, 11005–11015. [Google Scholar] [CrossRef] [PubMed]
  257. Naeini, M.B.; Momtazi, A.A.; Jaafari, M.R.; Johnston, T.P.; Barreto, G.; Banach, M.; Sahebkar, A. Antitumor Effects of Curcumin: A Lipid Perspective. J. Cell Physiol. 2019, 234, 14743–14758. [Google Scholar] [CrossRef]
  258. Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Kishor Roy, N.; Prasad, S.; Aggarwal, B.B. Curcumin, the Golden Nutraceutical: Multitargeting for Multiple Chronic Diseases. Br. J. Pharmacol. 2017, 174, 1325–1348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  259. Eke-Okoro, U.J.; Raffa, R.B.; Pergolizzi, J.V.; Breve, F.; Taylor, R. Curcumin in Turmeric: Basic and Clinical Evidence for a Potential Role in Analgesia. J. Clin. Pharm. 2018, 43, 460–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Chen, C.Y.; Kao, C.L.; Liu, C.M. The Cancer Prevention, Anti-Inflammatory and Anti-Oxidation of Bioactive Phytochemicals Targeting the TLR4 Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 2729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  261. Burge, K.; Gunasekaran, A.; Eckert, J.; Chaaban, H. Curcumin and Intestinal Inflammatory Diseases: Molecular Mechanisms of Protection. Int. J. Mol. Sci. 2019, 20, 1912. [Google Scholar] [CrossRef] [Green Version]
  262. Carlos-Reyes, Á.; López-González, J.S.; Meneses-Flores, M.; Gallardo-Rincón, D.; Ruíz-García, E.; Marchat, L.A.; Astudillo-De La Vega, H.; Hernández De La Cruz, O.N.; López-Camarillo, C. Dietary Compounds as Epigenetic Modulating Agents in Cancer. Front. Genet. 2019, 10, 79. [Google Scholar] [CrossRef] [Green Version]
  263. Bahrami, A.; Amerizadeh, F.; ShahidSales, S.; Khazaei, M.; Ghayour-Mobarhan, M.; Sadeghnia, H.R.; Maftouh, M.; Hassanian, S.M.; Avan, A. Therapeutic Potential of Targeting Wnt/β-Catenin Pathway in Treatment of Colorectal Cancer: Rational and Progress. J. Cell. Biochem. 2017, 118, 1979–1983. [Google Scholar] [CrossRef]
  264. Adiwidjaja, J.; McLachlan, A.J.; Boddy, A.V. Curcumin as a Clinically-Promising Anti-Cancer Agent: Pharmacokinetics and Drug Interactions. Expert Opin. Drug Metab. Toxicol. 2017, 13, 953–972. [Google Scholar] [CrossRef]
  265. Hanai, H.; Iida, T.; Takeuchi, K.; Watanabe, F.; Maruyama, Y.; Andoh, A.; Tsujikawa, T.; Fujiyama, Y.; Mitsuyama, K.; Sata, M.; et al. Curcumin Maintenance Therapy for Ulcerative Colitis: Randomized, Multicenter, Double-Blind, Placebo-Controlled Trial. Clin. Gastroenterol. Hepatol. 2006, 4, 1502–1506. [Google Scholar] [CrossRef]
  266. Koosirirat, C.; Linpisarn, S.; Changsom, D.; Chawansuntati, K.; Wipasa, J. Investigation of the Anti-Inflammatory Effect of Curcuma Longa in Helicobacter Pylori-Infected Patients. Int. Immunopharmacol. 2010, 10, 815–818. [Google Scholar] [CrossRef] [PubMed]
  267. Di Mario, F.; Cavallaro, L.G.; Nouvenne, A.; Stefani, N.; Cavestro, G.M.; Iori, V.; Maino, M.; Comparato, G.; Fanigliulo, L.; Morana, E.; et al. A Curcumin-Based 1-Week Triple Therapy for Eradication of Helicobacter Pylori Infection: Something to Learn from Failure? Helicobacter 2007, 12, 238–243. [Google Scholar] [CrossRef] [PubMed]
  268. Hong, T.; Zou, J.; Jiang, X.; Yang, J.; Cao, Z.; He, Y.; Feng, D. Curcumin Supplementation Ameliorates Bile Cholesterol Supersaturation in Hamsters by Modulating Gut Microbiota and Cholesterol Absorption. Nutrients 2022, 14, 1828. [Google Scholar] [CrossRef] [PubMed]
  269. Xiao, Q.P.; Zhong, Y.B.; Kang, Z.P.; Huang, J.Q.; Fang, W.Y.; Wei, S.Y.; Long, J.; Li, S.S.; Zhao, H.M.; Liu, D.Y. Curcumin Regulates the Homeostasis of Th17/Treg and Improves the Composition of Gut Microbiota in Type 2 Diabetic Mice with Colitis. Phytother. Res. 2022, 36, 1708–1723. [Google Scholar] [CrossRef] [PubMed]
  270. Guo, X.; Xu, Y.; Geng, R.; Qiu, J.; He, X. Curcumin Alleviates Dextran Sulfate Sodium-Induced Colitis in Mice Through Regulating Gut Microbiota. Mol. Nutr. Food Res. 2022, 66, e2100943. [Google Scholar] [CrossRef]
  271. Liu, J.; Luo, W.; Chen, Q.; Chen, X.; Zhou, G.; Sun, H. Curcumin Sensitizes Response to Cytarabine in Acute Myeloid Leukemia by Regulating Intestinal Microbiota. Cancer Chemother. Pharm. 2022, 89, 243–253. [Google Scholar] [CrossRef]
  272. Lopresti, A.L.; Smith, S.J.; Rea, A.; Michel, S. Efficacy of a Curcumin Extract (CurcugenTM) on Gastrointestinal Symptoms and Intestinal Microbiota in Adults with Self-Reported Digestive Complaints: A Randomised, Double-Blind, Placebo-Controlled Study. BMC Complement. Med. 2021, 21, 40. [Google Scholar] [CrossRef]
  273. Liu, Z.J.; Li, Z.H.; Liu, L.; Tang, W.X.; Wang, Y.; Dong, M.R.; Xiao, C. Curcumin Attenuates Beta-Amyloid-Induced Neuroinflammation via Activation of Peroxisome Proliferator-Activated Receptor-Gamma Function in a Rat Model of Alzheimer’s Disease. Front Pharm. 2016, 7, 261. [Google Scholar] [CrossRef] [Green Version]
  274. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin Inhibits Formation of Amyloid β Oligomers and Fibrils, Binds Plaques, and Reduces Amyloid In Vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef] [Green Version]
  275. Sang, Q.; Liu, X.; Wang, L.; Qi, L.; Sun, W.; Wang, W.; Sun, Y.; Zhang, H. Curcumin Protects an SH-SY5Y Cell Model of Parkinson’s Disease against Toxic Injury by Regulating HSP90. Cell. Physiol. Biochem. 2018, 51, 681–691. [Google Scholar] [CrossRef]
  276. Sandhir, R.; Yadav, A.; Mehrotra, A.; Sunkaria, A.; Singh, A.; Sharma, S. Curcumin Nanoparticles Attenuate Neurochemical and Neurobehavioral Deficits in Experimental Model of Huntington’s Disease. Neuromolecular Med. 2014, 16, 106–118. [Google Scholar] [CrossRef] [PubMed]
  277. Chico, L.; Ienco, E.C.; Bisordi, C.; lo Gerfo, A.; Petrozzi, L.; Petrucci, A.; Mancuso, M.; Siciliano, G. Amyotrophic Lateral Sclerosis and Oxidative Stress: A Double-Blind Therapeutic Trial After Curcumin Supplementation. CNS Neurol Disord. Drug Targets 2018, 17, 767–779. [Google Scholar] [CrossRef] [PubMed]
  278. Ramaholimihaso, T.; Bouazzaoui, F.; Kaladjian, A. Curcumin in Depression: Potential Mechanisms of Action and Current Evidence—A Narrative Review. Front. Psychiatry 2020, 11, 572533. [Google Scholar] [CrossRef] [PubMed]
  279. Fusar-Poli, L.; Vozza, L.; Gabbiadini, A.; Vanella, A.; Concas, I.; Tinacci, S.; Petralia, A.; Signorelli, M.S.; Aguglia, E. Curcumin for Depression: A Meta-Analysis. Crit. Rev. Food Sci. Nutr. 2020, 60, 2643–2653. [Google Scholar] [CrossRef] [PubMed]
  280. Kakkar, S.; Bais, S. A Review on Protocatechuic Acid and Its Pharmacological Potential. ISRN Pharm. 2014, 2014, 952943. [Google Scholar] [CrossRef] [Green Version]
  281. Song, J.; He, Y.; Luo, C.; Feng, B.; Ran, F.; Xu, H.; Ci, Z.; Xu, R.; Han, L.; Zhang, D. New Progress in the Pharmacology of Protocatechuic Acid: A Compound Ingested in Daily Foods and Herbs Frequently and Heavily. Pharm. Res. 2020, 161, 105109. [Google Scholar] [CrossRef]
  282. Hung, M.Y.; Fu, T.Y.C.; Shih, P.H.; Lee, C.P.; Yen, G.C. Du-Zhong (Eucommia ulmoides Oliv.) Leaves Inhibits CCl4-Induced Hepatic Damage in Rats. Food Chem. Toxicol. 2006, 44, 1424–1431. [Google Scholar] [CrossRef]
  283. del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (Poly)Phenolics in Human Health: Structures, Bioavailability, and Evidence of Protective Effects Against Chronic Diseases. Antioxid. Redox Signal 2013, 18, 1818–1892. [Google Scholar] [CrossRef] [Green Version]
  284. Crozier, A.; Del Rio, D.; Clifford, M.N. Bioavailability of Dietary Flavonoids and Phenolic Compounds. Mol. Asp. Med. 2010, 31, 446–467. [Google Scholar] [CrossRef]
  285. Masella, R.; Santangelo, C.; D’Archivio, M.; LiVolti, G.; Giovannini, C.; Galvano, F. Protocatechuic Acid and Human Disease Prevention: Biological Activities and Molecular Mechanisms. Curr. Med. Chem. 2012, 19, 2901–2917. [Google Scholar] [CrossRef]
  286. Krzysztoforska, K.; Mirowska-Guzel, D.; Widy-Tyszkiewicz, E. Pharmacological Effects of Protocatechuic Acid and Its Therapeutic Potential in Neurodegenerative Diseases: Review on the Basis of in Vitro and in Vivo Studies in Rodents and Humans. Nutr. Neurosci. 2019, 22, 72–82. [Google Scholar] [CrossRef] [PubMed]
  287. Gao, Y.; Tian, R.; Liu, H.; Xue, H.; Zhang, R.; Han, S.; Ji, L.; Huang, W.; Zhan, J.; You, Y. Research Progress on Intervention Effect and Mechanism of Protocatechuic Acid on Nonalcoholic Fatty Liver Disease. Crit. Rev. Food Sci. Nutr. 2021, 62, 9053–9075. [Google Scholar] [CrossRef] [PubMed]
  288. Thakare, V.N.; Lakade, S.H.; Mahajan, M.P.; Kulkarni, Y.P.; Dhakane, V.D.; Harde, M.T.; Patel, B.M. Protocatechuic Acid Attenuates Chronic Unpredictable Mild Stress Induced-Behavioral and Biochemical Alterations in Mice. Eur. J. Pharm. 2021, 898, 173992. [Google Scholar] [CrossRef] [PubMed]
  289. Larrosa, M.; García-Conesa, M.T.; Espín, J.C.; Tomás-Barberán, F.A. Ellagitannins, Ellagic Acid and Vascular Health. Mol. Asp. Med. 2010, 31, 513–539. [Google Scholar] [CrossRef] [PubMed]
  290. Kilic, I.; Yeşiloğlu, Y.; Bayrak, Y. Spectroscopic Studies on the Antioxidant Activity of Ellagic Acid. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2014, 130, 447–452. [Google Scholar] [CrossRef]
  291. Selma, M.V.; Beltrán, D.; Luna, M.C.; Romo-Vaquero, M.; García-Villalba, R.; Mira, A.; Espín, J.C.; Tomás-Barberán, F.A. Isolation of Human Intestinal Bacteria Capable of Producing the Bioactive Metabolite Isourolithin a from Ellagic Acid. Front. Microbiol. 2017, 8, 1521. [Google Scholar] [CrossRef] [Green Version]
  292. Tang, Y.; Zhang, X.; Wang, Y.; Guo, Y.; Zhu, P.; Li, G.; Zhang, J.; Ma, Q.; Zhao, L. Dietary Ellagic Acid Ameliorated Clostridium Perfringens-Induced Subclinical Necrotic Enteritis in Broilers via Regulating Inflammation and Cecal Microbiota. J. Anim Sci. Biotechnol. 2022, 13, 47. [Google Scholar] [CrossRef]
  293. Wu, T.; Chu, X.; Cheng, Y.; Tang, S.; Zogona, D.; Pan, S.; Xu, X. Modulation of Gut Microbiota by Lactobacillus Casei Fermented Raspberry Juice In Vitro and In Vivo. Foods 2021, 10, 3055. [Google Scholar] [CrossRef]
  294. Mosele, J.I.; Macià, A.; Romero, M.P.; Motilva, M.J. Stability and Metabolism of Arbutus Unedo Bioactive Compounds (Phenolics and Antioxidants) under in Vitro Digestion and Colonic Fermentation. Food Chem. 2016, 201, 120–130. [Google Scholar] [CrossRef]
  295. Kiasalari, Z.; Afshin-Majd, S.; Baluchnejadmojarad, T.; Azadi-Ahmadabadi, E.; Esmaeil-Jamaat, E.; Fahanik-Babaei, J.; Fakour, M.; Fereidouni, F.; Ghasemi-Tarie, R.; Jalalzade-Ogvar, S.; et al. Ellagic Acid Ameliorates Neuroinflammation and Demyelination in Experimental Autoimmune Encephalomyelitis: Involvement of NLRP3 and Pyroptosis. J. Chem. Neuroanat. 2021, 111, 101891. [Google Scholar] [CrossRef]
  296. Farbood, Y.; Sarkaki, A.; Dianat, M.; Khodadadi, A.; Haddad, M.K.; Mashhadizadeh, S. Ellagic Acid Prevents Cognitive and Hippocampal Long-Term Potentiation Deficits and Brain Inflammation in Rat with Traumatic Brain Injury. Life Sci. 2015, 124, 120–127. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representative phenolic compounds.
Figure 1. Representative phenolic compounds.
Antioxidants 12 00880 g001
Figure 2. Chemical structures of catechins, theaflavins, and theasinensin A–E.
Figure 2. Chemical structures of catechins, theaflavins, and theasinensin A–E.
Antioxidants 12 00880 g002
Figure 3. Schematic diagram of the biotransformation of main tea catechins. Modified from [35].
Figure 3. Schematic diagram of the biotransformation of main tea catechins. Modified from [35].
Antioxidants 12 00880 g003
Figure 4. Chemical structures of main flavan-3-ols in cocoa powder.
Figure 4. Chemical structures of main flavan-3-ols in cocoa powder.
Antioxidants 12 00880 g004
Figure 5. Biotransformation pathways of main flavonols in humans. Quoted from [45].
Figure 5. Biotransformation pathways of main flavonols in humans. Quoted from [45].
Antioxidants 12 00880 g005
Figure 6. Chemical structures of major flavonols.
Figure 6. Chemical structures of major flavonols.
Antioxidants 12 00880 g006
Figure 7. Quercetin catabolites by intestinal bacteria.
Figure 7. Quercetin catabolites by intestinal bacteria.
Antioxidants 12 00880 g007
Figure 8. Chemical structure of the main isoflavones and isoflavone glycosides.
Figure 8. Chemical structure of the main isoflavones and isoflavone glycosides.
Antioxidants 12 00880 g008
Figure 9. Metabolism of the isoflavone glucoside daidzin by the human gut microbiota and biosynthesis pathway of equol. Modified from [154].
Figure 9. Metabolism of the isoflavone glucoside daidzin by the human gut microbiota and biosynthesis pathway of equol. Modified from [154].
Antioxidants 12 00880 g009
Figure 10. Chemical structures of major phenylpropanoids.
Figure 10. Chemical structures of major phenylpropanoids.
Antioxidants 12 00880 g010
Figure 11. Main chlorogenic acids present in coffee. The numbers in the figure are necessary to indicate where the caffeoyl group or feruoyl group is attached.
Figure 11. Main chlorogenic acids present in coffee. The numbers in the figure are necessary to indicate where the caffeoyl group or feruoyl group is attached.
Antioxidants 12 00880 g011
Figure 12. Biotransformation of sesamin and sesamolin.
Figure 12. Biotransformation of sesamin and sesamolin.
Antioxidants 12 00880 g012
Figure 13. Chemical structures of major stilbenoids.
Figure 13. Chemical structures of major stilbenoids.
Antioxidants 12 00880 g013
Figure 14. Chemical structures of resveratrol metabolites.
Figure 14. Chemical structures of resveratrol metabolites.
Antioxidants 12 00880 g014
Figure 15. Chemical structures of curcuminoids.
Figure 15. Chemical structures of curcuminoids.
Antioxidants 12 00880 g015
Figure 16. Biotransformation of curcumin.
Figure 16. Biotransformation of curcumin.
Antioxidants 12 00880 g016
Figure 17. Major metabolic pathway of the anthocyanin cyanidin-O-β-glucoside in humans. The presence of intestinal bacteria accelerates the formation of protocatechuic acid through cleavage of the C-ring shown in red letters in figure. Modified from [286].
Figure 17. Major metabolic pathway of the anthocyanin cyanidin-O-β-glucoside in humans. The presence of intestinal bacteria accelerates the formation of protocatechuic acid through cleavage of the C-ring shown in red letters in figure. Modified from [286].
Antioxidants 12 00880 g017
Figure 18. Schematic representation of the production of microbial metabolites from ellagic acid. Modified from [291].
Figure 18. Schematic representation of the production of microbial metabolites from ellagic acid. Modified from [291].
Antioxidants 12 00880 g018
Table 1. Salutary effects of phenolic compounds.
Table 1. Salutary effects of phenolic compounds.
Dietary Phenolic Compound Source/CompoundDiseaseStudy ResultsReference(s)
polyphenolscardiovascular diseasedatabase-linked survey of preclinical trials and clinical trials on polyphenols for the treatment of cardiovascular diseaseBehl et al., 2020 [5]
polyphenolsrheumatoid Arthritisefficacy of polyphenols to mitigate rheumatoid arthritis by inhibiting the MAPK signaling pathwayBehl et al., 2021 [6]
polyphenolsrheumatoid Arthritisa review of preclinical and clinical data on various pathways involved in rheumatoid arthritis and polyphenols as therapeutic agentsBehl et al., 2022 [7]
plant polyphenolsdepressiona review of the chemical, pharmacological, and neurological evidence for the potential of polyphenols in depressionKabra et al., 2022 [8]
polyphenolsdepressiona review of polyphenols that inhibit oxidative stress and inflammation through signaling pathways in depressionBehl et al., 2022 [9]
polyphenols
carotenoids
eye diseasea review of the health benefits of polyphenols and carotenoids for the prevention and treatment of age-related eye diseasesBungau et al., 2019 [10]
quercetin, ECarteriosclerosisaugmentation of nitric oxide status and attenuation of endothelin-1 concentration in plasma of healthy men Loke et al., 2008 [47]
cocoa/ECcardiovascular diseaseacute elevations in levels of circulating nitric oxide species, an enhanced flow-mediated vasodilation response of conduit arteries, and an augmented microcirculationSchroeter et al., 2006 [48]
ECbrain endothelial dysfunction, neurodegenerative disordersregulated protein expression and gene expression in brain endothelial cellsCorral-Jara et al., 2022 [51]
green tea extractsalcoholic fatty liver diseaseattenuation of triacylglycerol levels in serum and liver and aminotransferase activities in miceLi et al., 2021 [52]
tea extractsalcoholic fatty liver diseaseprevention of liver steatosis, decrease in oxidative stress and inflammation, modulation of gut microbiotaLi et al., 2021 [54]
green teaalcoholic fatty liver diseaseamelioration of alcoholic liver disease by activation of Akkermansia muciniphilaZhao et al., 2022 [53]
EGCGnon-alcoholic fatty liver diseaseinhibited the increase in histological fatty deposits and triglyceride accumulation in the liver induced by high fat diet, improved intestinal dysbiosis, and involved in sirtuin genesNaito et al., 2020 [55]
concord grape polyphenolsobesityincrease in the growth of Akkermansia muciniphila and decrease in the proportion of Firmicutes to BacteroidetesRoopchand et al., 2015 [59]
EGCGulcerative colitisthe active treatment remission rate was 53.3% (8 of 15) compared with 0% (0 of 4) for placeboDryden et al., 2013 [60]
ECacute and chronic colitisattenuation of COX-2 expression and increase in cell proliferation, repair of the epithelium by stimulating the expression of EGFVasconcelos et al., 2012 [61]
EGCG and piperineulcerative colitisincreased bioavailability, decreased colonic histological damage and MDA levels, and increased antioxidant enzyme activityBrückner et al., 2012 [62]
EGC and ECGAlzheimer’s diseaseattenuation of amyloid-β aggregation, reduced ROS production, less neurotoxicity to neuronsChen et al., 2020 [65]
EGCGAlzheimer’s diseasenegative regulation of microglial inflammation and neurotoxicityZhong et al., 2019 [66]
EGCGAlzheimer’s diseaseactivated ERK-and PI3K-mediated pathways in astrocytes and accelerated amyloid-β degradationYamamoto et al., 2017 [67]
EGCGAlzheimer’s diseaseinhibition of neuroinflammatory response in microglia, protection from indirect neurotoxicityCheng-Chung Wei et al., 2016 [68]
EGCGAlzheimer’s diseaseattenuation of cognitive deficits in APP/PS1 miceBao et al., 2020 [69]
EGCGParkinson’s diseasemodulation of the substantia nigra iron transport protein ferroportin, attenuation of oxidative stress, neuroprotective effectsXu et al., 2017 [70]
EGCGParkinson’s diseaseinhibition of substantia nigra neurodegeneration, neuroprotective effectSergi 2022 [71]
EGCGhypoxia-induced neuroinflammationprotection of microglia by disabling the NF-κB pathway and activating the Nrf-2/HO-1 pathwayKim et al., 2022 [72]
flavanol-enriched cocoa powderamelioration of intestinal environmentenhanced the abundance of Lactobacillus and Bifidobacterium species, modulated markers of local gut immunityJang et al., 2016 [73]
cocoa flavanolsdisorder of the intestinal environmentgrowth of select gut microflora in humans Tzounis et al., 2011 [74]
cocoadisorder of the intestinal environmentimproved gut-associated lymphoid tissue function by modulating IgA secretion and gut microbiotaPérez-Cano et al., 2013 [75]
cocoadeterioration of the intestinal immune systemdifferential TLR patterns, attenuation of intestinal IgA secretion and IgA-coating bacteriaMassot-Cladera et al., 2012 [76]
cocoadiabetes mellitusamelioration of intestinal flora, barrier integrity, and the inflammatory status of the intestineÁlvarez-Cilleros et al., 2020 [77]
cocoainflammation-related colon carcinogenesisattenuation of NF-κB, pro-inflammatory enzyme expression, and inducible NO synthase expressionRodríguez-Ramiro et al., 2013 [78]
cocoa flavanolscoronary artery diseasemaintenance of normal endothelium-dependent vasodilationAgostoni C. et al., 2012 [79]
cocoa extractcardiovascular disease among older adultslowered risk of total cardiovascular eventsSesso et al., 2022 [80]
cocoa extractAlzheimer’s diseasemodification of the physical structure of amyloid-β oligomersDubner et al., 2015 [81]
cocoa extractAlzheimer’s diseaseattenuation of amyloid-β oligomerizationWang et al., 2014 [82]
cocoa extractAlzheimer’s diseaseneuroprotection by activating the brain-derived neurotrophic factor survival pathwayCimini et al., 2013 [83]
kaki tanninmetabolic syndromestrong binding capacity for bile acidsMatsumoto et al., 2011 [87]
kaki tanninhypercholesterolemiacholesterol lowering effect and glucose metabolism amelioration by the ability of kaki tannin to bind bile acidsNishida et al., 2021 [88]
kaki tanninpostprandial hyperglycemiakaki tannins limited starch digestion and inhibited glucose uptake and transport, thereby alleviating postprandial hyperglycemiaLi et al., 2018 [89]
kaki tannindisruption of intestinal florareshaped fecal gut microbiotaZhu et al., 2018 [90]
kaki tanninMycobacterium avium complex (MAC) diseasebacteriostatic effect on MAC, attenuation of pulmonary granuloma formation, suppression of pro-inflammatory cytokine expressionMatsumura et al., 2017 [17]
kaki tanninulcerative colitisdecreased disease activity and colonic inflammation, changed microbiota composition and immune responseKitabatake et al., 2021 [18]
dry persimmondyslipidemialipid-lowering and antioxidant propertiesGorinstein et al., 1998 [91], Gorinstein et al., 2000 [92]
kaki tanninhyper-LDL cholesterolemiaattenuation of serum LDL cholesterol levels in humansSuzuki et al., 2022 [93]
quercetin and isoflavonesosteoporosiselucidation of metabolic pathways by intestinal microbiota, amelioration of bioavailabilityMurota et al., 2018 [95]
quercetin/red onionobesity and insulin resistanceadipose tissue remodelingForney et al., 2018 [118]
quercetin/grape powderobesity and insulin resistanceprevented macrophage inflammation and adipocyte macrophage-mediated insulin resistanceOverman et al., 2011 [119]
quercetinkidney disease due to atheroembolismattenuation of COX-2 induction by stressCarlsen et al., 2015 [116]
quercetinobesity-related diseasesantioxidant, anti-inflammatory, and antifibrotic effects on insulin resistance and atherosclerosisSato et al., 2020 [123]
quercetincolitisrebalanced the pro-inflammatory, anti-inflammatory, and bactericidal function of enteric macrophagesJu et al., 2018 [120]
quercetindisruption of intestinal florarestoration of gut microbiota in mice after antibiotic treatmentShi et al., 2020 [121]
quercetinC. rodentium-induced colitismodification of gut microbiota and suppression of proinflammatory cytokines in Citrobacter rodentium-induced colitis miceLin et al., 2019 [122]
quercetin and rutinAlzheimer’s diseaseanti-amyloidogenic and fibril-disaggregating effectsJiménez-Aliaga et al., 2011 [124]
quercetinAlzheimer’s diseasepromotion of viability and proliferation of Alzheimer’s disease model cells, increase in expression of sirtuin 1/Nrf2/HO-1 and antioxidant-related enzymesYu et al., 2020 [125]
quercetinAlzheimer’s diseaseinhibition of tau protein hyperphosphorylation and oxidative stress, inhibition of PI3K/Akt/GSK3β, MAPK, and NF-κB p65 in a cell line of mouse hippocampal neuronsJiang et al., 2016 [126]
quercetinAlzheimer’s diseaseinhibition of BACE-1 (Beta-site APP Cleaving Enzyme-1, β-secretase), attenuation of amyloid-β peptide levelsShimmyo et al., 2008 [127]
quercetinAlzheimer’s diseasetargeted integrated stress response signaling, suppressed amyloid-β (Aβ) production and prevented cognitive impairment in a mouse modelNakagawa et al., 2019 [128]
quercetinParkinson’s diseaseactivation of the PKD1–Akt cell survival signaling axis, neuroprotective signaling in a dopaminergic neuronal modelAy et al., 2017 [129]
quercetinParkinson’s diseasesignificant attenuation of rotenone-induced behavioral impairment, augment of autophagy, attenuation of ER stress-induced apoptosis with attenuated oxidative stressEl-Horany et al., 2016 [130]
quercetin with piperineParkinson’s diseaseattenuation of movement disorders and biochemical and neurotransmitter changesSharma et al., 2020 [131]
quercetin with piperineParkinson’s diseasesignificantly amelioration of MPTP-induced behavioral abnormalities in rats, reversal of the abnormal alterations of neurotransmitters in the striatumSingh et al., 2017 [132]
buckwheatHypercholesterolemia,
neurodegenerative disease, cancer, inflammation, diabetes,
hypertension
buckwheat as a food and its effects on healthGiménez-Bastida et al., 2015 [104]
quercetin, rutin/buckwheat dyslipidemia, metabolic syndromes,quercetin reduced obesity due to high-fat diet,
rutin, quercetin, and tartary buckwheat shaped specific structures of the intestinal microbiota
Peng et al., 2020 [133]
phenolic compounds/tartary buckwheathuman breast cancerinhibitory ability of phenolic compounds on breast cancer cell proliferationLi et al., 2017 [134]
rutincancerregulation of molecular networks and signaling mechanisms in cancer cells by rutinPerk et al., 2014 [135]
rutinCOVID-19conformational change upon binding of rutin and SARS-CoV-2 spike proteinKumari et al., 2022 [136]
Rahman et al., 2021 [137]
rutin, quercetin/buckwheatpostprandial rise in blood sugar, diabetes,
hypercholesterolemia
the rutin and phenolic compounds contained in buckwheat inhibited the action of digestive enzymes, suppressing the sudden rise in postprandial blood sugar levels and lowering cholesterolKreft et al., 2022 [138]
Cirkovic Velickovic et al., 2018 [139]
Wang et al., 2022 [140]
Ikeda et al., 1993 [141]
Zhang et al., 2017 [142]
Bao et al., 2016 [143]
buckwheatcardiovascular disease,
dyslipidemia
review and meta-analysis on buckwheat and cardiometabolic healthLlanaj et al., 2022 [144]
rutinneurodegenerative diseasea review of the neuroprotective mechanisms of rutinEnogieru et al., 2018 [145]
buckwheathypercholesterolemia, inflammation, neurodegenerative disease, cancer, diabetes, hypertension, celiac diseasehealth benefits of buckwheat, potential remedy for diseasesNoreen et al., 2021 [146]
isoflavonea wide range of hormonal disordersclassification, structure, and occurrence, with their metabolism, biological, and health effects in humans and animals, and their utilization and potential risksKřížová et al., 2019 [147]
isoflavone and metabolitescardiovascular diseases, metabolic syndromes, osteoporosis, diabetes, brain-related diseases, etc.the latest research trends that have shown substantial interest in the biological efficacy of isoflavones in humans and plants, and their related mechanismsKim 2021 [148]
isoflavonessome hormone-dependent diseases effects of isoflavones on chemoprevention of breast cancer, prostate cancer, and cardiovascular osteoporosis and alleviation of osteoporosis and postmenopausal symptomsVitale et al., 2013 [156]
S-equolvasomotor symptoms, osteoporosis, prostate cancer, cardiovascular diseasesummary of studies demonstrating effects of isoflavone supplements on menopausal symptoms, bone, prostate cancer, and cardiovascular biomarkersJackson et al., 2011 [159]
isoflavone/soybeansbreast, thyroid, and uterus of postmenopausal womena review of key studies related to soy, with a focus on clinical and epidemiological studiesMessina 2016 [162]
soy proteinblood cholesterol attenuation of total and LDL cholesterolHarland et al., 2008 [163]
soy isoflavonesosteoporosissignificant increase in bone density, decrease in urinary deoxypyridinoline, a marker of bone resorptionWei et al., 2012 [164]
dietary soychronic kidney diseasesignificantly reduced serum creatinine, serum phosphorus, CRP, and proteinuria; no significant change was found in creatinine clearance and glomerular filtration rateJing et al., 2016 [165]
fermented soy productsdiabetes mellitus, blood pressure, cardiac disorders, and cancer-related issuesattenuation of serum levels of total cholesterol, low-density lipoprotein (LDL), and triglycerides, maintenance of bone health and prevention of osteoporosis and maintenance of normal endothelial functionJayachandran et al., 2019 [166]
genisteinAlzheimer’s diseasedirectly targeted amyloid-β and tau to regulate intracellular signaling pathways involved in neuronal death in the brainUddin et al., 2019 [168]
soy isoflavonesAlzheimer’s diseaseneuroprotective effects on scopolamine-induced memory impairment, enhancement of cholinergic function, suppression of oxidative stress and activation of ERK/CREB/BDNF signalingLu et al., 2018 [169]
genisteinAlzheimer’s diseaseregulated CAMK4 to regulate tau hyperphosphorylationYe et al., 2017 [170]
genisteinParkinson’s diseaseneuroprotective effect on dopaminergic neuronsArbabi et al., 2016 [171]
genisteinearly phases of allergic encephalomyelitis, multiple sclerosisdecreased cell cytotoxicityRazeghi Jahromi et al., 2014 [172]
sesamediabetes mellitus, hypercholesterolemia, osteoarthritis, some types of cancerdetailed research on sesame oil contents, health effects, nutraceuticals, oil quality, and value addition strategiesLangyan et al., 2022 [179]
sesamefree radical-related diseasesNutraceutical, pharmacological, traditional, and industrial value of sesame seeds with respect to bioactive components that have high antioxidant activityPathak et al., 2014 [180]
chlorogenic acidobesity and associated glucose intoleranceattenuation of food intake, elevation of body temperature, increase in heat dissipation and activation of brown adipose tissueHe et al., 2021 [188]
chlorogenic acidobesity and obesity-related metabolic endotoxemiasuppression of body weight gain, attenuation of relative weight of fat, amelioration of intestinal barrier integrity, prevention of impaired glucose metabolism and endotoxemia, significant alteration of intestinal microbiota compositionYe et al., 2021 [189]
chlorogenic acidhigh-fat diet-induced obesityattenuation of plasma lipids, alteration of adipose tissue-associated gene expression, reversal of gut microbiota dysbiosisWang et al., 2019 [190]
coffeetype 2 diabetes mellitusattenuation of diabetes risk in humansHuxley et al., 2009 [191]
coffeedisruption of intestinal floraincrease in the growth of Bifidobacterium spp and Clostridium coccoides-Eubacterium rectale groupMills et al., 2015 [192]
coffeedisruption of intestinal floracoffee consumption can selectively improve the growth of probiotic strains, thus exerting a prebiotic effectSales et al., 2020 [193]
chlorogenic acidParkinson’s diseaseactivation of Akt/ERK signaling in the mitochondrial intrinsic apoptotic pathway, neuroprotection against MPTP-induced toxicity in a Parkinson’s disease mouse modelSingh et al., 2020 [195]
caffeic acid, chlorogenic acidParkinson’s diseaseprotection of rotenone-induced neurodegeneration of both nigral dopaminergic and enteric neurons, upregulation of metallothioneinMiyazaki et al., 2019 [196]
chlorogenic acidParkinson’s diseaseattenuation of oxidative stress and neuroinflammation in MPTP-poisoned miceSingh et al., 2018 [197]
chlorogenic acidAlzheimer’s diseaseattenuation of cognitive deficits in APP/PS1 mice by activation of the mTOR/TFEB signaling pathwayGao et al., 2020 [198]
sesaminvariety of cardiovascular diseasesattenuation of cardiovascular disease effects on RAS/MAPK, PI3K/AKT, ERK1/2, p38, p53, IL-6, TNFα, and NF-κB signaling networksDalibalta et al., 2020 [200]
sesameclimacteric disorderamelioration of blood lipid, antioxidant, and sex hormone statusWu et al., 2006 [201]
sesaminchronic kidney diseasesuppression of uremic toxin production by inhibition of bacterial L-tryptophan indole-lyaseOikawa et al., 2022 [202]
sesamindisruption of intestinal floraincrease in the adhesive index of probiotics, up-regulation of the adhesive protein (β-cadherin and E-cadherin) expressionWang et al., 2021 [204]
sesamolAlzheimer’s diseaseattenuation of SCOP-induced cognitive dysfunction via balancing the cholinergic system and reducing neuroinflammation and oxidative stressYun et al., 2022 [205]
sesamolAlzheimer’s diseaseattenuation of Alzheimer’s disease-related cognitive impairment and neuroinflammatory response by mediating the gut microbe–SCFA–brain axisLiu et al., 2021 [206]
sesamin, sesamolAlzheimer’s disease, Parkinson’s disease, Huntington’s diseaseactivation of SIRT1/SIRT3/FOXO3a expression, inhibition of BAX (pro-apoptotic protein) and upregulation of BCL-2 (anti-apoptotic protein)Ruankham et al., 2021 [207]
sesamindiabetes-induced neurodegenerative diseasesattenuation of microglial activation by high glucose, reduction of inflammatory response and neurotoxicityKongtawelert et al., 2022 [208]
sesamin, sesamolin, sesamolAlzheimer’s diseasesesamin protected against Aβ toxicity by reducing toxic Aβ oligomers, sesamin and sesamolin ameliorated amyloid-β-induced deficits in chemotactic behavior, anti-amyloid-β toxic activity and structure–activity relationship of sesame lignansKeowkase et al., 2018 [209]
resveratrolneuroinflammatory diseaseprevention of self-destruction of nerve cellsRenaud et al., 2014 [218]
resveratrol/red winecardiovascular disease, lung cancer, prostate cancereffect of red wine on cardiovascular morbidity and mortalityVidavalur et al., 2006 [219]
red winecoronary heart diseaseinhibition of platelet reactivity by wine (alcohol)Renaud et al., 1992 [220]
resveratrolintestinal dysfunctionregulation of intestinal barrier function under immunosuppressionSong et al., 2022 [221]
resveratrolcolitisactivation of metabolism by intestinal microbiota, modification of intestinal microbiotaYao et al., 2022 [222]
resveratrolobesityamelioration of intestinal flora, regulation of lipid metabolism, recovery of intestinal barrier function, amelioration of insulin sensitivityWang et al., 2020 [223]
resveratrolNAFLDamelioration of insulin resistance, amelioration of intestinal barrier function and intestinal microbiota composition, amelioration of lipid metabolismWang et al., 2020 [224]
resveratrolNAFLDinhibition of high-fat diet-induced elevation in cannabinoid receptor type 1 (CB1) mRNA expression, inhibition of colonic CB2 mRNA levels, and maintenance of intestinal barrier integrityChen et al., 2020 [225]
resveratrolmetabolic and intestinal diseaseupregulation of mRNA expression of tight junction and mucin-associated proteins, maintenance of intestinal barrierZhang et al., 2021 [226]
resveratrolmetabolic syndromeregulation of intestinal bacterial composition and metabolism and alteration of steroid metabolism in middle-aged menKorsholm et al., 2017 [227]
resveratrolobesitymetabolic activation and amelioration of mitochondrial respiration to muscle fatty acid-derived substrates and caloric restriction-like effect in obese menTimmers et al., 2011 [228]
resveratrolcardiovascular disease and a variety of cancersaccumulation of resveratrol in epithelial cells along the aerodigestive tract and presence of potentially active resveratrol metabolitesWalle et al., 2004 [229]
red winecoronary heart diseasechanges in lipid profiles, attenuation of insulin resistance, and decrease in oxidative stressCastaldo et al., 2019 [230]
wineobesityconsuming moderate amounts of wine as part of a Mediterranean diet did not promote weight gain or abdominal obesity.Golan et al., 2017 [231]
resveratrolpregnancy-related complicationseffects of resveratrol on embryogenesis and spermatogenesis mediated by several mechanismsNovakovic et al., 2022 [232]
grape seed oilwoundwound-healing properties of the oils of Vitis vinifera and Vaccinium macrocarpon in animal modelShivananda Nayak et al., 2011 [233]
Al-Warhi et al., 2022 [234]
grape seed oilulcerative colitisoral administration of grape seed oil and grape seed extract showed anti-inflammatory effect and effect on ulcerative colitisNiknami et al., 2020 [235]
grape seed oilacute liver injurygrape seed oil suppressed inflammation and protected the liver against acute liver injury caused by oxidative stressIsmail et al., 2016 [236]
grape seed oildiabetes mellitusseed oil of Vitis davidii Foex. protected pancreatic β-cells from anti-glucose-induced apoptosis and maintained insulin secretionLai et al., 2014 [237]
grape seed oilerythema of the skinthe application of a cream milky lotion containing grape seed oil was found to ameliorate the skin’s moisture content, sebum content, and erythemaSharif et al., 2015 [238]
grape seed oilphysiological leg edema in primigravidaephysiological edema in pregnancy was suppressed with foot massage using grape seed oilNavaee et al., 2020 [239]
grape seed oilhyperlipidemiablood triglycerides were suppressed by oral administration of grapeseed oil for 6 weeksKaseb et al., 2016 [240]
resveratrolAlzheimer’s diseasesignificant attenuation of cytotoxicity of amyloid-β1-42 peptide against SH-SY5Y human neuroblastoma cells, neuroprotective effectAl-Edresi et al., 2020 [241]
resveratrolhypoxia, Alzheimer’s diseaseprevention of hypoxia-induced upregulation of total amyloid and exosomal amyloid-β by inhibiting CD147Xie et al., 2019 [242]
resveratrolAlzheimer’s diseaseupregulation of the SIRT1 pathway, induction of cognitive enhancement and neuroprotection against amyloid and tau pathologiesCorpas et al., 2019 [243]
resveratrolAlzheimer’s diseaseactivation of AMPK-dependent signaling by resveratrol rescued amyloid-β-mediated neurotoxicity in hNSCs.Chiang et al., 2018 [244]
resveratrolParkinson’s diseaseregulation of the MALAT1/miR-129/SNCA signaling pathwayXia et al., 2019 [245]
resveratrolParkinson’s diseaseattenuation of MPTP-induced loss of dopaminergic neurons, attenuation of astroglial activation in the nigrostriatal pathway, attenuation of motor dysfunction in MPTP-treated miceLiu et al., 2019 [246]
resveratrolParkinson’s diseaseneuroprotective effects of regulation of α-synuclein expression upon loss of miR-214 in Parkinson’s diseaseWang et al., 2015 [247]
resveratrolHuntington’s diseaseimproved motor coordination and learning, enhanced expression of mitochondrial-encoded electron transport chain genes in YAC128 miceNaia et al., 2017 [248]
resveratrolmultiple sclerosispromoted remyelination effect of resveratrolGhaiad et al., 2017 [249]
resveratrolamyotrophic lateral sclerosis (ALS)increase in mitochondrial biogenesis in the SOD1(G93A) spinal cord, increase in expression and activation of Sirtuin 1 and AMPK in the ventral spinal cordMancuso et al., 2014 [250]
curcumincancerpotential of curcumin to influence lipogenic pathways that regulate human cancer cell metabolismNaeini et al., 2019 [257]
curcuminvarious chronic diseases including various types of cancers, diabetes, obesity, cardiovascular, pulmonary, neurological, and autoimmune diseasesAnti-inflammatory activity through the suppression of numerous cells signaling pathways including NF-κB, STAT3, Nrf2, ROS, and COX-2, Kunnumakkara et al., 2017 [258]
curcumincancerinhibition of activation of Toll-like receptor 4 (TLR4) signaling pathway associated with inflammatory response and cancer progressionChen et al., 2018 [260]
curcuminintestinal inflammatory diseases, such as Crohn’s disease, ulcerative colitis, and necrotizing enterocolitisimproved intestinal barrier function, regulated the gut microbiota, exhibited antioxidant and anti-inflammatory effectsBurge et al., 2019 [261]
curcumincancerpotent antitumor activity by reversing epigenetic changes associated with oncogene activation and tumor suppressor gene inactivationCarlos-Reyes et al., 2019 [262]
curcumincolorectal adenomaregulation of the Wnt/β-catenin pathway associated with colorectal cancerBahrami et al., 2017 [263]
curcumincolorectal cancerdisruption of tumor growth signaling such as COX-2 enzyme expression, attenuation of NF-kB signaling, suppression of EGFR phosphorylation, inhibition of angiogenesis, and apoptosis of malignant cellsAdiwidjaja et al., 2017 [264]
curcuminulcerative colitisreduced recurrence rates and maintained remission in patients with quiescent ulcerative colitisHanai et al., 2006 [265]
curcuminHelicobacter pylori-infected gastritisalthough treatment of H. pylori-infected patients with curcumin did not alter levels of inflammatory cytokine mRNA expression and had limited anti-bactericidal effect, it improved common symptoms in the patientsKoosirirat et al., 2010 [266]
curcuminHelicobacter pylori-infected gastritissignificant amelioration of dyspeptic symptoms and attenuation of serologic signs of gastric inflammation were observed in H. pylori-positive patients with functional dyspepsia despite the lack of eradication of H. pyloriMario et al., 2007 [267]
curcumingallstone diseasedefense against biliary cholesterol supersaturation by modulating intestinal microbiota and inhibiting intestinal cholesterol absorptionHong et al., 2022 [268]
curcuminulcerative colitis complicated by diabetes mellituseffectively alleviated colitis in mice with type 2 diabetes by restoring Th17/Treg homeostasis and improving gut microbiota compositionXiao et al., 2022 [269]
curcuminintestinal inflammatory diseasesenhancement of the intestinal barrier, attenuation of intestinal apoptosis by suppressing the caspase-3 pathway, reduction in intestinal inflammation by inhibiting the MAPK/NFκB/STAT3 pathway, and amelioration of gut bacteria involved in colitisGuo et al., 2022 [270]
curcuminacute myeloid leukemiapromoted responses to cytarabine through modulation of the microbiota, highlighting the importance of enhancing gut integrity in chemoresistance therapyLiu et al., 2022 [271]
curcuminirritable bowel syndromesignificant improvement in gastrointestinal symptom rating scale and stress scale indicatorsLopresti et al., 2021 [272]
curcuminAlzheimer’s diseaseeffects of curcumin-activated PPARγ on anti-neuroinflammatory and neuroprotective effects in Alzheimer’s diseaseLiu et al., 2016 [273]
curcuminAlzheimer’s diseaseblocked amyloid-β aggregation and fibril formation in vitro and in vivo by directly binding curcumin to small beta-amyloid speciesYang et al., 2005 [274]
curcuminParkinson’s diseaseeffective inhibition of the toxic effects of MPP+ on SH-SY5Y cells, greatly attenuating the adverse effects of MPP+ on dopaminergic neurons via upregulation of HSP90Sang et al., 2018 [275]
curcumin/encapsulatedHuntington’s diseaseamelioration of mitochondrial dysfunction and significant enhancement in neuromotor coordinationSandhir et al., 2014 [276]
curcuminamyotrophic lateral sclerosis (ALS)amelioration of aerobic metabolism and oxidative damage, and slowed disease progressionChico et al., 2018 [277]
curcuminmajor depressive disorderpotency to modulate neurotransmitter levels, inflammatory pathways, excitotoxicity, neuroplasticity, hypothalamic–pituitary–adrenal disorders, insulin resistance, oxidative and nitrosative stress, and the endocannabinoid systemRamaholimihaso et al., 2020 [278]
protocatechuic acidcancer, hyperlipidemia, diabetespotential to agent of antioxidant, antibacterial, anticancer, antihyperlipidemic, antidiabetic, and anti-inflammatoryKakkar et al., 2014 [280]
protocatechuic acidneurodegenerative disease, tumors, osteoporosis, liver disease, kidney disease, metabolic syndromeregulation of oxidative stress and inflammatory responses via multiple signaling pathwaysSong et al., 2020 [281]
protocatechuic acid/Du-Zhongchronic hepatotoxicityattenuation of liver lesions incidenceHung et al., 2006 [282]
protocatechuic acidAlzheimer’s disease, Parkinson’s diseaseinhibition of β-amyloid plaque accumulation and tau hyperphosphorylation in brain tissueKrzysztoforska et al., 2019 [286]
protocatechuic acidNAFLDregulation of glucose and lipid metabolism, oxidative stress, inflammation, gut microbiota, and metabolites, increase in energy expenditure of brown adipose tissueGao et al., 2021 [287]
protocatechuic aciddepressionmaintained brain-derived neurotrophic factor levels and modulated oxidative stress responses, cytokine systems, and antioxidant defense systems in miceThakare et al., 2021 [288]
ellagic acidinflammatory disease,
neurodegenerative diseases
discovery of a novel bacterial strain capable of converting ellagic acid to isourolithin A with anti-inflammatory, anti-carcinogenic, cardioprotective, and neuroprotective propertiesSelma et al., 2017 [292]
ellagic acidsubclinical necrotic enteritis of broiler caused by Clostridium perfringensregulation of jejunal inflammatory signaling pathways TLR/NF-κB and JAK3/STAT6, alleviation of jejunal oxidative stress, inhibition of intestinal barrier damage, prevention of systemic inflammatory responseTang et al., 2022 [293]
ellagic acidmultiple sclerosisattenuation of astrogliosis, astrocyte activation, demyelination, neuroinflammation, and axonal damage via NLRP3 inflammasome and pyroptotic pathwayKiasalari et al., 2021 [295]
ellagic acidcognitive impairments, long-term potentiation deficitssignificant prevention of traumatic brain injury-induced memory impairment and hippocampal long-term potentiation impairmentFarbood et al., 2015 [296]
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

Matsumura, Y.; Kitabatake, M.; Kayano, S.-i.; Ito, T. Dietary Phenolic Compounds: Their Health Benefits and Association with the Gut Microbiota. Antioxidants 2023, 12, 880. https://doi.org/10.3390/antiox12040880

AMA Style

Matsumura Y, Kitabatake M, Kayano S-i, Ito T. Dietary Phenolic Compounds: Their Health Benefits and Association with the Gut Microbiota. Antioxidants. 2023; 12(4):880. https://doi.org/10.3390/antiox12040880

Chicago/Turabian Style

Matsumura, Yoko, Masahiro Kitabatake, Shin-ichi Kayano, and Toshihiro Ito. 2023. "Dietary Phenolic Compounds: Their Health Benefits and Association with the Gut Microbiota" Antioxidants 12, no. 4: 880. https://doi.org/10.3390/antiox12040880

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