ABSTRACT
The fruits and vegetables we consume as part of our diet are rich in bioactive metabolites that can prevent and ameliorate cardiometabolic diseases, cancers, and neurological conditions. Polyphenols are a major metabolite family that has been intensively investigated in this context. However, for these compounds to exert their optimal bioactivity, they rely on the enzymatic capacity of an individual’s gut microbiota. Indeed, for most polyphenols, the human host is restricted to more basic metabolism such as deglycosylation and hepatic conjugation. In this review, we discuss the mechanisms by which gut bacteria metabolize the core scaffold of polyphenol substrates, and how their conversion into bioactive small molecules impacts host health.
KEYWORDS: Gut microbiome, polyphenols, flavonoids, monophenolic acids, urolithins, metabolic pathways
1. Introduction
Polyphenols are an abundant family of secondary metabolites produced by plants, and they are mainly studied for their various health benefits1. The polyphenol family of compounds is very large and structurally diverse and includes flavonoids, tannins and lignans, stilbenes, and phenolic acids. Among these different polyphenol classes, flavonoids have received most of the attention to date. Due to the presence of aromatic chromophores in their structure, flavonoids are the pigments that are responsible for the distinctive colors of many fruits, vegetables, and flowers. For plants, they play important roles in pollination and reproduction, as well as in protection from pathogens and UV radiation.2,3 The basic structure of flavonoids consists of two aromatic rings, designated A and B, which are connected by a central heterocyclic pyrene C-ring.4 Depending on the degree of oxidation and saturation of the C-ring, as well as the position and number of hydroxyl and other functional groups on the A- and B-rings, flavonoids are further classified into different subgroups, including flavones, flavanols, flavanones, and isoflavones (Figure 1).1
Figure 1.

Flavonoids are classified according to the oxidation state of their central C-ring into six major classes: flavones, flavonols, flavanones, isoflavones, anthocyanidins, and chalcones.
Flavonoids are widely consumed in the human diet through colorful fruits and vegetables, red wine, chocolate, and tea, accounting for an average daily intake of 200–600 mg in adults. It is estimated that there are over 4,000 dietary flavonoids, of which several dozen are investigated for their potential positive effects in preventing and managing various diseases, including cardiovascular and metabolic diseases,5–7 cancer,8–10 and Alzheimer’s disease.11,12 Flavonoids act in a variety of ways, including serving as antioxidant,13,14 anti-inflammatory,15,16 antimutagenic,8,17–19 antiatherogenic,20,21 antidiabetic,22–24 and antithrombotic compounds.25–27 It is worth noting that most of these epidemiological studies involving a dietary intervention are not designed to specifically test the health benefits of individual compounds and might be confounded by additional variables such as a higher dietary fiber content. Therefore, additional controlled intervention studies will be required help elucidate the health benefits of individual dietary compounds, and the potential role of the gut microbiota in their bioactivity.
Despite their various attributed health benefits, the bioavailability of polyphenols in humans is low, as the majority of ingested polyphenols are not absorbed in the small intestine.28 Host metabolism of flavonoids and other polyphenols is limited to select deglycosylations, as well as hepatic phase I conjugation with glucuronic acid, sulfate, or glutathione.29,30 These conjugations render the metabolites more hydrophilic and facilitate their urinary excretion from the body or their enterohepatic circulation to the intestine, where they can be further metabolized by the gut bacteria. To date, no mammalian enzymes have been identified that can break down the core flavonoid three-ring structure.
The bulk of ingested polyphenols reaches the colon relatively unmodified, where they present as a substrate for catabolism by certain gut microbiota.31 The metabolism of polyphenols by the gut microbiota involves a series of enzymatic reactions, including hydrolysis, reduction, and dihydroxylation,32–35 which result in the formation of various metabolites such as monophenolic acids, dihydroflavonols, and urolithins.36–38 These metabolites can be absorbed by the colonic epithelium and reach the systemic circulation, where they can exert their biological activities. Our group recently demonstrated that bacterial flavonoid catabolite 4-hydroxyphenylacetic acid was sufficient to revert diet-induced hepatic steatosis in mice.7
The metabolism of polyphenols by the gut microbiota is a complex process that is influenced by various factors, including the structure and oxidation state of the molecule itself, as well as the composition of the gut microbiota, and their enzymatic repertoire.39 Gut bacterial enzymes are essential for the metabolism and processing of a wide range of dietary components that are not digestible by human enzymes.40 In this review, we focus on the different gut microbiota and their enzymatic activities involved in polyphenol catabolism characterized to date (Table 1). Whereas several microbes and enzymes have been identified, we’ve likely only scratched the surface in this field, leaving many pathways to be discovered.
Table 1.
Gut microbial enzymes involved in polyphenol metabolism.
| Substrate | Enzyme | Products | Corresponding gut bacteria |
|---|---|---|---|
| Deglycosylation | |||
| O-linked flavonoid glycosides | Glycosidase | Flavonoid aglycone | Bifidobacteriaceae, Lactobacillaceae, and Lachnospiraceae41–47 |
| C-linked flavonoid glycosides | Glycosidase | Flavonoid aglycone | Lachnospiraceae, Enterococcaceae,Streptococcaceae.44,45,48,49 |
| Flavonoid glycosides | β-glucosidase | Flavonoid aglycone | Lactobacillus crispatus, Companilactobacillus paralimentarius, Lacticaseibacillus paracasei, Lactiplantibacillus plantarum Levilactobacillus hammesii Furfurilactobacillus milii50 |
| Flavones | |||
| Flavone | Flavone reductases | Dihydroflavone | F. plautii ATCC 49,53135 Clostridium difficile DSM 630 and Clostridium perfringens ATCC 13,12435 |
| Dihydroflavone | Chalcone isomerase | Chalcone | E. ramulus DSM 16,296, F. plautii51 |
| Chalcone | Enoate reductase | Dihydrochalcone | E. ramulus32, F. plautii ATCC 49,53135 |
| Dihydrochalcone | Phloretin hydrolase | Phloroglucinol +3-(4-hydroxyphenyl)propionic acid | E. ramulus,32F. plautii ATCC 49,53135 |
| Flavonol | |||
| Flavonol | Flavone reductases | Dihydroflavonol | E. ramulus,32F. plautii ATCC 49,53135 |
| Dihydroflavonol | Chalcone isomerase | Hydroxyaurone | E. ramulus,32F. plautii ATCC 49,53135 |
| Hydroxyaurone | Phloroglucinol +4-Hydroxyphenylacetic acid | E. ramulus,32F. plautii ATCC 49,53135 | |
| Quercetin | |||
| Quercetin | Quercetinases | Protocatechuic acid + 2,4,6-Trihydroxybenzoic acid | Bacillus subtilis52–54 |
| Isoflavones | |||
| Daidzin | Daidzein reductase | (R)-Dihydrodaidzein + (S)- Dihydrodaidzein |
Eggerthella sp. strain YY7918 (99% identity), and Slackia (only 43% identity).55,56
Lactococcus 20-92 Lactobacillus sp. Niu-O16, Eggerthella sp. Julong 732, Lactococcus garvieae 20–92, Slackia isoflavoniconvertens, and Adlercreutzia equolifaciens57–60 |
| (R)-Dihydrodaidzein | Dihydrodaidzein racemase | (S)- Dihydrodaidzein | Lactobacillus sp. Niu-O16, Eggerthella sp. Julong 732, Lactococcus garvieae 20–92, Slackia isoflavoniconvertens, and Adlercreutzia equolifaciens57–60 |
| (S)- Dihydrodaidzein | Dihydrodaidzein reductase | (3S, 4 R)-tetrahydrodaidzein | Lactobacillus sp. Niu-O16, Eggerthella sp. Julong 732, Lactococcus garvieae 20–92, Slackia isoflavoniconvertens, and Adlercreutzia equolifaciens57–60 |
| (3S, 4 R)-tetrahydrodaidzein | Tetrahydrodiadzein reductase | (S)-equol |
Slackia isoflavoniconvertens and Slackia sp. strain NATTS Lactococcus sp.Eggerthella sp. strain YY791856 Lactobacillus sp. Niu-O16, Eggerthella sp. Julong 732, Lactococcus garvieae 20–92, Slackia isoflavoniconvertens, and Adlercreutzia equolifaciens57–60 |
| Hydroxycinnamic acids | |||
| Chlorogenic acids | Hydroxycinnamic acid esterase | hydroxycinnamic acid + Quinic acid | L. plantarum,61L.johnsonii,62Lactobacillus acidophilus63, B. longum64 and B. animalis .65 |
| Hydroxycinnamic acid | Hydroxycinnamate reductase | Phenylpropionic acid | L. plantarum .66 L. fermentum L. plantarum enzyme .67 L. rossiae L. plantarum reductase ,67 L. kunkeei ,68 and Weissella cibaria .67 |
| Hydroxycinnamic acids | Phenolic acid decarboxylase | vinyl derivatives | Lactobacillus spp69 |
| Vinyl derivatives | vinyl phenol reductase | Ethyl derivatives | Lactobacillus plantarum WCFS170 |
| Urolithins | |||
| Urolithin M5 | 10-dehydroxylase | Urolithin D | Entrocloster spp71 |
| Urolithin M5 | 9-dehydroxylase | Urolithin E | Entrocloster spp71 |
| Urolithin M5 | 4-dehydroylase | Urolithin 6 | Ellagibacter spp38 |
| Urolithin M6 | 10-dehydroxylase | Urolithin C | Gordonibacter urolithinfaciens (DSM 27213T)36 |
| Urolithin M6 | 8-dehydroxylation | Isourolithin A | Ellagibacter38 |
| Urolithin D | 4-dehydroylase | Urolithin C | Gordonibacter urolithinfaciens (DSM 27213T)36 |
| Urolithin D | 4-dehydroylase | Urolithin G | Entrocloster spp71 |
| Urolithin C | 8-dehydroxylation | Isourolithin A | Ellagibacter38 |
| Urolithin C | 4-dehydroylase | Urolithin A | E. bolteae CEBAS S4A9 and DSM 15670T, E. asparagiformis DSM 15981T, and E. citroniae DSM 19261T |
| Urolithin M7 | 10-dehydroylase | urolithin A | E. clostridioformis DSM 933T |
| Isourolithin A | 9-dehydroxylation | Urolithin B | Ellagibacter38,72 |
2. Deglycosylation: a key first step in flavonoid metabolism
Flavonoids occur predominantly in plants as either O- or C-glycosides as this enhances their solubility. O-glycosylated flavonoids contain mono- or disaccharides attached to the hydroxyl groups on the backbone A or C rings, while C-glycosides involve the attachment of a sugar molecule to the carbon atom of the A-ring.73 Deglycosylation of flavonoids by the host or intestinal microbiota is critical for their absorption and further phase I hepatic metabolism. On the host side, the O-deglycosylation process can be facilitated by enzymes such as lactase-phlorizin hydrolase and β-glucosidase expressed in the intestinal epithelium,74,75 whereas flavonoid C-glycosides are refractory to host enzymes (Figure 2). Hydrolysis of flavonoid O-glycosides by the gut microbiota is well documented across several genera of gut microbes, including Bifidobacteriaceae, Lactobacillaceae, and Lachnospiraceae.41–47 Cleavage of C-glycosides is far less common and relatively understudied but has been described for specific species of Lachnospiraceae, Enterococcaceae, and Streptococcaceae.44,45,48,49 Food-fermenting lactobacilli encode a wealth of enzymes specifically capable of phytochemical β-glucoside hydrolysis, including phospho-β-glucosidases (for example encoded by Lactobacillus crispatus, Companilactobacillus paralimentarius, and Lactiplantibacillus plantarum), and 3β-glucosidases (for example from Levilactobacillus hammesii, and Furfurilactobacillus milii).50
Figure 2.

Deglycosylation of C-linked flavonoid glycosides is exclusively catalyzed by the small intestinal microbiota, whereas cleavage of O-linked sugars can also be performed in the stomach and small intestine by host enzymes such as lactase-phlorizin hydrolase and β-glucosidase.
Since both the host and the gut microbiota are capable of deglycosylation, there is some controversy regarding the relative role of microbial enzymes and the need for deglycosylation to ensure flavonoid absorption. Some studies report direct uptake of glycosylated flavonoids into the bloodstream, facilitated by glucose transporters in the intestinal lumen. For example, quercetin glucosides were shown to be transported through sodium-glucose co-transporters (SGLTs) and GLUT2 within 30 minutes after oral ingestion, and the amount of absorbed quercetin was enhanced when conjugated to glucose.76–79 In a separate study, intestinal transport of anthocyanidins like delphinidin 3-rutinoside, cyanidin 3-rutinoside, and cyanidin 3-glucoside, also resulted in increased plasma concentrations a few hours after ingestion.80 Interestingly, this systemic uptake does not seem to work analogous for glycosylated isoflavones, which rely on the gut microbiome for metabolism.81,82
Overall, the deglycosylation of flavonoids in the gut involves both human and bacterial enzymes, and it represents an important first step for their further metabolism, as well as has implications for their bioavailability and associated health effects.
3. Enzymatic conversion of the flavonoid core
Gut microbial catabolism of the core structure of flavanols, flavanonols, flavones, and flavanones is catalyzed by similar enzymatic processes, yet can yield different products depending on the bacteria involved and their enzymatic repertoire. An additional source of catabolite variation is caused by differences in B-ring decorations of the parent flavonoid. Flavones, such as apigenin and luteolin, are converted to 3-(3-hydroxyphenyl)propionic acid and 3,4-dihydroxyphenylacetic acid by different gut bacteria, including Eubacterium ramulus, Blautia glucerasea, and Eggerthella lenta.83,84 Flavanones, such as hesperetin and naringenin, are converted to 3-(3-hydroxyphenyl)propionic acid and phloroglucinol by strains of Bacteroides thetaiotaomicron and Flavonifractor plautii (previously classified as Clostridium orbiscindens).85,86 The catabolism of flavones into monophenolic acids involves a four-step metabolic pathway, initiated by C-ring reduction and subsequent cleavage, followed by further reduction of the resulting α,β-unsaturated ketone moiety, and finally, a hydrolysis reaction that cleaves the molecule.
The enzymes involved in these reactions and their corresponding genes have been identified in studies with E. ramulus and F. plautii35,47 and are discussed in more detail below (Table 2).
Table 2.
Uniprot identifiers for functionally characterized polyphenol metabolizing enzymes.
3.1. Reduction of the central C-ring by flavone reductase
Flavone reductases catalyze the first step in the catabolic pathway of flavone and flavonol aglycones, using a flavin mononucleotide (FMN) cofactor molecule. While catalyzing the first step in the pathway, this novel class of ene-reductases enzymes was only recently functionally characterized in F. plautii ATCC 49,531.35 The activity of the F. plautii flavone reductase (FLR) was demonstrated in an in vitro assay using apigenin as a substrate and involves the transfer of a hydride ion from the FMNH2 cofactor to the C2=C3 double bond in the flavone’s C-ring. This reduction converts apigenin to its dihydroflavone form, naringenin (Figure 3).
Figure 3.

The catabolism of flavones into monophenolic acids involves a four-step metabolic pathway. Flavone reductase (FLR) initiates by reducing the C-ring, followed by chalcone isomerase (chi)-mediated cleavage, and subsequent enoate reductase (EnoR)-mediated further reduction of the resulting α,β-unsaturated ketone moiety. Finally, phloretin hydrolase (PHY) catalyzes a second hydrolysis reaction that cleaves the molecule.
The crystal structure of the FLR holoenzyme in complex with apigenin, a flavone substrate, revealed the location of the active site and the residues involved in catalysis.35 A reaction mechanism was proposed and supported by circular dichroism spectrum analysis. FLR consists of a compact N-terminal compact α-β-α domain and an extended C-terminal domain. FLR forms a homodimer and binds the FMN cofactor at the dimer interface, which is formed involving parts of both the N-terminal and C-terminal domains and burying about a third of the enzyme’s surface area. While functionally similar to Old-Yellow-Enzymes (OYEs), FLR differs in two notable ways.32,87 Firstly, FLR lacks a suitable residue that forms direct hydrogen bonds to the C4-carbonyl group of apigenin for substrate activation. Instead, a hydrogen-bond relay is formed through the hydroxyl of Thr279 via an H2O molecule, and the hydride from FMNH- is transferred to apigenin’s β-carbon. Secondly, FLR does not contain a direct proton donor to reduce the α-carbon of apigenin, which contrasts with OYEs that utilize a tyrosine residue as a general acid. Since the crystal structure revealed that the apigenin α-carbon is exposed to the bulk solvent, the required proton for the formation of the naringenin product likely comes from a water molecule.35
Interestingly, this reduction catalyzed by FLR is reversible, yet the reduction of flavone/flavonol, is favored over the oxidation of dihydroflavones. The substrate promiscuity of FLR was demonstrated in the reduction of other flavone and flavonol substrates, including quercetin, kaempferol, myricetin, chrysin, luteolin, and diosmetin. Interestingly, the F. plautii FLR is unable to reduce flavones that contain methoxy groups, such as tangeretin and nobiletin. FLR-like genes were identified in members of the human gut microbiome, as well as in pathogenic gut bacteria such as Clostridium difficile DSM 630 and Clostridium perfringens ATCC 13,124.35 Because of their unique fold and function, flavone reductases were proposed to form a novel enzyme class separate from previously identified reductases or flavone synthases.
3.2. Chalcone isomerase opens the central C-ring
The conversion of dihydroflavone to chalcone is the second step in the flavonone catabolic pathway, and the chalcone isomerase (CHI) catalyzing this reaction has been extensively studied in E. ramulus DSM 16,296 (exemplified by the conversion of naringenin to naringenin chalcone in Figure 3).88 Purified CHI was next demonstrated to participate in the catabolism of various flavonols and flavanonols, including taxifolin and dihydrokaempferol. The E. ramulus CHI crystal structure has a hexameric organization, consisting of three dimer subunits.32 Enzymatic conversion efficiencies for various flavanone substrates indicated that a B-ring containing a para hydroxyl group is crucial for CHI activity. Molecular docking of naringenin to the active site confirmed that this hydroxyl forms hydrogen bonds with Asp79 and Gln101, ensuring proper substrate orientation. The substrate specificity of CHI is relatively relaxed, allowing flavanones with B-rings that contain hydroxyl or methoxy groups at the meta position. His33 is a critical amino acid located in the active site of CHI, which is postulated to play a crucial role in the acid-base catalysis-mediated opening of the heterocyclic C-ring. The proton transfer from the C-3 carbon of the flavonoid intermediate to His33 generates a negative charge, which facilitates ring opening. The key catalytic role of His33 was confirmed by site-specific mutations, which resulted in loss of CHI activity.51,89 It is worth noting that both the A- and B-rings are vital for the formation and stability of the CHI enzyme-substrate complex. CHI homologs have since been identified across various other genera of flavonoid-catabolizing bacteria, including F. plautii.51
3.3. Reduction of the chalcone double bond by enoate reductase
The third step in the flavone/flavonol catabolic pathway is the reduction of the C=C bond, which was introduced as a result of the CHI reaction. This reduction converts the chalcones to their respective dihydrochalcones, and is catalyzed by the enzyme enoate reductase32,89,90(Figure 3). Similar to FLR -which initiates the flavone catabolic pathway-, enoate reductase also belongs to the enzyme class of ene-reductases. An important distinction between the two types of enzymes is that enoate reductase uses NADH as a reducing agent, whereas FLR is incapable of using NAD(P)H.90 The E. ramulus enoate reductase crystal structure revealed that it has a multidomain structure, consisting of a barrel domain and an oxygen-sensitive domain with an iron-sulfur cluster.32 The activity of this enzyme was demonstrated by converting naringenin chalcone to phloretin. To date, E. ramulus and F. plautii are the only documented gut bacteria that can produce the dihydrochalcone phloretin.
3.4. Final cleavage of the dihydrochalcone by phloretin hydrolase
The final step in flavone/flavanone catabolism is catalyzed by phloretin hydrolase (PHY). Anecdotally, the E. ramulus phy gene was the first gene to be discovered in the bacterial flavonoid degradation pathway. Phy is transcribed from an 822 base-pair open reading frame and forms a homodimer in solution, consisting of two 30 kDa subunits.32 PHY hydrolyzes the C-C bond at the A-ring of phloretin, yielding phloroglucinol and the monophenolic acid 3-(4-hydroxyphenyl)propionic acid (Figure 3).91 This C-C bond cleavage occurs through a reverse Claisen mechanism and is facilitated by a 6-membered transition state.92 Tryptophan, which is part of the substrate binding site, plays a vital role in catalysis.91 The phloretin hydrolase enzyme in F. plautii ATCC 49,531 shares 46.92% amino acid sequence similarity with the PHY enzyme in E. ramulus DSM 16,296.35
4. An alternative pathway for flavonoid catabolism using bacterial dioxygenases
An alternative, oxygen-dependent pathway for microbial flavonoid catabolism was discovered in a series of in vitro degradation studies of the flavonol quercetin by the Bacillus subtilis enzyme YxaG. Gut microbial catabolism of the quercetin typically yields protocatechuic acid (3,4-dihydroxybenzoic acid) and 3,4-dihydroxyphenylacetic acid as primary metabolites. The enzymatic release of protocatechuic acid from quercetin can be facilitated by YxaG-like bacterial quercetin dioxygenases, also known as quercetinases.52–54 Quercetinase uses molecular oxygen to catalyze the simultaneous cleavage of two carbon-carbon bonds in the heterocyclic C-ring, resulting in the formation of an ester that links the two aromatic A- and B-rings the release of carbon monoxide. The enzymatic mechanism requires a divalent cation to chelate the 4-keto/2,3-enol moiety of quercetin, thereby promoting the 3,4-diketo tautomer. First, the hydrogen atom at position 2 is removed, and the resulting carbon 2-centered radical next reacts with superoxide to form a five-membered endo-peroxide. The endo-peroxide is finally rearranged through the conversion of the 3-keto into an acylium ion, leading to the formation of a 2-protocatechuoyl-phloroglucinol carboxylic acid ester, and the release of carbon monoxide. The ester can subsequently be hydrolyzed into protocatechuic acid and 2,4,6-trihydroxybenzoic acid, while the latter can be further decarboxylated to form the gut microbial metabolite phloroglucinol52,92,93 (Figure 4). Further investigations into gut microbial dioxygenase-dependent metabolism of quercetin and other flavonols will elucidate if these oxygen-dependent reactions are relevant in the anaerobic intestinal environment and whether they are perhaps localized to the small intestine or close to the mucosal surface, as well as identify potential enzymes involved in the latter hydrolysis and decarboxylation steps of this pathway.
Figure 4.

An alternative pathway for opening the flavonoid C-ring involves the action of bacterial dioxygenases, like quercetinase, which require molecular oxygen for their activity.
5. Enzymatic conversion of isoflavones
Isoflavones represent a remarkable structural and functional subclass of polyphenols because several have been characterized as phytoestrogens because they are chemically and structurally similar to 17β-estradiol.94–96 Due to this similarity, isoflavones are investigated for their modulatory effect in certain hormone-dependent diseases, such as breast cancer, colon cancer, and cardiometabolic diseases.97–99
Daidzein is one of the most intensively investigated isoflavones and is present primarily in soybeans. It is primarily found in its glycosylated form, daidzin.100–102 Like other flavonoids, isoflavones are poorly absorbed by the small intestine due to their bulky structure.103–105 This provides ample substrate availability for the gut microbiota, which have an important role in their metabolism and bioactivity. Different types of gut microbial products of daidzein include (S)-equol and O-desmethylangolensin (O-DMA).57,104–106 The human host does not produce enzymes capable of metabolizing daidzein, and depending on the gut microbial enzymatic repertoire, individuals are either (S)-equol producers or non-producers.107 An estimated 30–50% of the total human population harbors (S-)-equol producing bacteria in their gut microbiome, while approximately 40–50% of people’s microbiota are estimated to be capable of O-DMA production.97,108
Only a few bacterial strains have been characterized to metabolize daidzein to (S)-equol, including Lactobacillus sp. Niu-O16, Eggerthella sp. Julong 732, Lactococcus garvieae 20–92, Slackia isoflavoniconvertens, and Adlercreutzia equolifaciens .57–60 In addition, certain bacterial strains are incapable of fully converting daidzein to (S)-equol, likely due to the absence of one or two enzymes in the pathway. It is of note that these partial converters may work together with different strains in the gut microbiota that complement their capabilities and can still result in an (S)-equol producer individual.59,60
A characterized pathway for the metabolism of daidzein to (S)-equol involves four subsequent enzymatic steps.48,104,109 Daidzein is made available to (S)-equol producing microbes after deglycosylation of daidzin, via β-glucosidase enzymes that could be produced by either host intestinal epithelial cells, or the gut microbiota.110,111 A portion of this aglycone can get absorbed by the basolateral membrane of the intestine, and become conjugated in the liver.112 Some conjugated daidzein can, in turn, reenter the blood circulation, while a large proportion is re-introduced in the small intestine via enterohepatic circulation, and can be further metabolized by the gut microbiota.113
5.1. Daidzein reductase initiates the conversion of non-glycosylated isoflavones
The first step in the gut bacterial daidzein conversion pathway is catalyzed by the oxidoreductase called daidzein reductase (DZR) and yields dihydrodaidzein (Figure 5).34 This enzyme was initially identified in Lactococcus 20–92, and the dzr gene that encodes it has been shown to be upregulated by the presence of daidzein.114 Functional homologs were later identified in (S)-equol producers Eggerthella sp. strain YY7918 (99% identity), and Slackia (only 43% identity).55,56
Figure 5.

Gut microbial conversion of isoflavonoids to phytoestrogens (S)-equol and O-desmethylangolensin (O-DMA) involves a series of reduction steps and is more efficient if a racemase is present in the microbiota. DZR = daidzein reductase; DDRC = dihydrodaidzein racemase; DDR = dihydrodaidzein reductase; TDR = tetrahydrodaidzein reductase.
Daidzein reductase has a cysteine-rich [4Fe-4S] cluster domain, which assists in the reduction of the C2-C3 double bond to yield mainly the ®-enantiomer of dihydrodaidzein.33,115 Three cofactor binding motifs for NAD(P)H, FAD, and FMN were identified, yet daidzein reductase mainly uses NADPH as a cofactor resulting in its classification as a NADP(H)-dependent daidzein reductase.34
5.2. Dihydrodaidzein racemase provides an (S)-enantiomer supply for the subsequent step
Due to an asymmetric carbon atom at the C-3 position, dihydrodaidzein has two enantiomers, (R)-dihydrodaidzein and (S)-dihydrodaidzein.116 The next step in the conversion pathway involves dihydrodaidzein racemase (DDRC), which is capable of dihydrodaidzein conversation into its mirror-image isomer, thereby producing a racemic mixture.109 This reaction catalyzed by dihydrodaidzein racemase involves an enolization and epimerization (Figure 5).115 The gene ddrc encoding for this enzyme is found in all of the characterized bacterial strains capable of the conversion of daidzein to (S)-equol, except Slackia sp. NATTS .117 Dihydrodaidzein racemase was the last enzyme to be functionally characterized, thereby completing the mapping of enzymes onto the conversion pathway of daidzein to (S)-equol.115 Since the production of (S)-equol is a stereospecific process, dihydrodaidzein racemase, in essence, ensures the supply of (S)-enantiomer substrate from the total dihydrodaidzein pool.
5.3. Dihydrodaidzein reductase is key for stereospecific product formation
Dihydrodaidzein reductase (DDR) catalyzes the third step in the daidzein conversion pathway, the reduction of dihydrodaidzein to tetrahydrodaidzein (Figure 5). Dihydrodaidzein reductase operates primarily by involving the transfer of electrons from a cofactor molecule, such as NADH or NADPH, to its (S)-dihydrodaidzein substrate, reducing the ketone group on position C-4 to an alcohol.114 This reduction results in the stereospecific production of (3S,4 R)- tetrahydrodiadzein.117 Dihydrodaidzein reductase belongs to the short-chain dehydrogenase/reductase (SDR) superfamily, which is characterized by a conserved NAD(P)H-binding domain involved in electron transfer during oxidation-reduction reactions.55,56,114,118
A subset of individuals harbor gut microbes capable of performing the first enzymatic reactions in the pathway (DZR and DDRC), but lack DDR and hence cannot produce (S)-equol. For these people, the intermediate metabolite (S)-dihydrodaidzein can be transformed into O-desmethylangolensin (O-DMA) via cleavage of the C-ring. Individuals with an (S)-equol-producing microbiota could, in addition, produce O-DMA, but analysis of urinary samples suggests that the conversion pathway to (S)-equol is predominant.119,120
5.4. Tetrahydrodaidzein reductase produces (S)-equol
The final step in the daidzein conversion pathway is the reduction of tetrahydrodaidzein to (S)-equol, catalyzed by the glycyl radical enzyme tetrahydrodaidzein reductase (TDR, Figure 5).114 TDR from Slackia isoflavoniconvertens and Slackia sp. strain NATTS contains a characteristic RVXG glycyl radical motif, which is not completely conserved in the TDR enzymes of (S)-equol producers Lactococcus sp. strain 20–92 and Eggerthella sp. strain YY7918.56 Glycyl radical enzymes only function in the absence of oxygen and catalyze the abstraction of substrate hydrogen atoms in a mechanism involving a cation radical intermediate.33,121 First, TDR is proposed to remove a hydrogen atom from tetrahydrodaidzein’s C3 position, forming a stable benzylic radical. Subsequently, the benzylic radical mediates the loss of the hydroxyl group located at the C4 position, leading to the formation of a delocalized radical cation. This radical cation is next reduced by the addition of a hydride donor at the C4 position. Finally, a hydrogen atom is added back to the C3 position, producing (S)-equol.33,56,122,123
A study of the TDR from Eggerthella sp. strain YY7918 indicated that the enzyme might not function as a reductase, but rather as a novel type of dismutase. The enzyme is proposed to function in the absence of cofactors such as NAD(P)H to transform tetrahydrodaidzein into (S)-equol and produce dihydrodaidzein as a by-product via a disproportionation reaction.55 Further investigations will shed more light on the different proposed TDR mechanisms in Slackia and Eggerthella.
While the key enzymes in the bacterial pathway for conversion of daidzein to (S)-equol have been identified, several questions remain unresolved. In particular, the role of dihydrodaidzein racemase (DDRC) has received little attention thus far.55 Previous studies have demonstrated that the presence of the ddrc gene in combination with the three reductase genes (dzr, ddr, and tdr) results in higher (S)-equol production levels compared to when the ddrc racemase gene is absent.115,124 However, since lower levels of (S)-equol production are still observed with only the three reductases present,34 this suggests that the racemase is not essential yet makes the production process more efficient. It is tempting to speculate that this increase in efficiency is due to the increased supply of (S)-dihydrodaidzein for further stereospecific reduction by DDR, thereby depleting the total pool of (RS)-dihydrodaidzein. In the absence of the racemase, both dihydrodaidzein enantiomers are formed and only the (S)-dihydrodaidzein half of the pool can be metabolized further, resulting in lower yields.
6. Enzymatic conversion of hydroxycinnamic acids
Hydroxycinnamic acids are a widely distributed family of nonflavonoid phenolic acids which are characterized by their double bond-containing carboxylic acid side chain linked to the benzene ring (Figure 6). Hydroxycinnamic acids have a variety of beneficial effects on host health, attributed to their antioxidant, antimicrobial, and anti-inflammatory functions and their plasma concentrations are correlated with a reduced risk of cardiovascular diseases, diabetes, hypertension, and colorectal cancer.125–127
Figure 6.

Hydroxycinnamic acids occur as simple free fatty acids or in an esterified form. Examples of simple hydroxycinnamic acids include caffeic acid, ferulic acid, sinapic acid, and coumaric acid. The family of hydroxycinnamic acids that are esterified to quinic acid is named chlorogenic acids.
Hydroxycinnamic acids are most abundantly present in berries, potatoes, coffee, tea, and wine128 and they occur in free acid or conjugated forms. Examples of non-conjugated hydroxycinnamic acids include caffeic acid (3,4-dihydroxycinnamic acid), ferulic acid (4-hydroxy-3-methoxycinnamic acid), sinapic acid (4-hydroxy-3,5-dimethoxycinnamic acid), and coumaric acid (4-hydroxycinnamic acid). Hydroxycinnamic acids can also occur in an esterified form, bound to sugars like arabinose or galactose, or in case of the chlorogenic acids bound to quinic acid, yielding 5-O-caffeoylquinic acid, 5-O-feruloylquinic acid, and 5-O-coumaroylquinic acid (Figure 6).129
Hydroxycinnamic acids can be absorbed through passive diffusion and the monocarboxylic acid transporter system, or it can be absorbed post-deconjugation. After absorption, hydroxycinnamic acids can be sulfated, methylated, or glucuronidated by phase II enzymes in the liver or small intestine. Alternately, the fraction of non-absorbed hydroxycinnamic acids can be metabolized by the gut microbiome.130,131
6.1. Hydrolysis of hydroxycinnamic acid esters by feruloyl esterase (FAE)
Several lactic acid bacteria, including members of the lactobacilli and bifidobacteria, produce hydroxycinnamic esterases, also known as feruloyl esterases (FAEs; Figure 7). The hydrolysis reaction catalyzed by these esterases cleaves chlorogenic acids, yielding quinic acid and hydroxycinnamic acid products, or sugar residues and hydroxycinnamic acid. Hydroxycinnamic esterases have been characterized in L. plantarum ,61 L. johnsonii ,62 Lactobacillus acidophilus 63, B. longum ,64 and B. animalis .65
Figure 7.

Gut microbial conversion of different chlorogenic acids to corresponding hydroxycinnamic acid and quinic acid. FAE = feruloyl esterase.
6.2. Reduction of hydroxycinnamic acids to phenylpropionic acid by hydroxycinnamate reductase
Hydroxycinnamate reductase (HcrB, also known as phenolic acid reductase), is a heterodimeric NADH-dependent reductase that reduces the double bonds in hydroxycinnamic acids to form phenylpropionic acids while re-oxidizing the reduced NADH co-factor to NAD+ .132 Hence, caffeic acid is reduced to dihydrocaffeic acid, coumaric acid to phloretic acid, and ferulic acids to dihydroferulic acids.67
Similar to FAE enzymes, HcrB is also present in lactic acid bacteria, and has been characterized in L. plantarum .66 The L. fermentum hydroxycinnamic acid reductase is encoded by the hcrF gene and shares 63% amino acid homology with HcrB from L. plantarum.67 The L. rossiae hydroxycinnamate reductase, Par1, shares an even lower sequence identity of ~ 25% compared to the functional homologues found in L. plantarum reductase ,67 L. kunkeei ,68 and Weissella cibaria .67
6.3. Decarboxylation of hydroxycinnamic acids to vinyl phenol derivatives via phenolic acid decarboxylase (PAD)
Decarboxylation of hydroxycinnamic acids is an important mechanism to alleviate their potential antimicrobial effects. This reaction is catalyzed by enzymes of the phenolic acid decarboxylase (PAD) family, members of which are well-characterized and upregulated in the presence of caffeic acid. Decarboxylation results in the formation of vinyl derivatives that exert less cytotoxic stress on the lactobacillus cell wall. The decarboxylation of caffeic acid yields vinyl catechol, while the decarboxylation of ferulic acid and coumaric acid result in vinyl guaiacol and vinyl phenol respectively69 (Figure 8). Several Lactobacillus species are capable of hydroxycinnamic acid decarboxylation, yet select strains are devoid of a pad homolog, including Lactobacillus delbrueckii, and Lacticaseibacillus casei BL23.132
Figure 8.

Further metabolism of hydroxycinnamic acids to their corresponding phenolic metabolites by the gut bacteria. PAD = phenolic acid decarboxylase; VprA = vinyl phenol reductase; HcrB = hydroxycinnamate reductase.
6.4. Further reduction of vinyl phenols to their ethyl derivatives by vinyl phenol reductase
The L. plantarum WCFS1 vinyl phenol reductase, VprA, is capable of reducing vinyl catechol to 4-ethyl catechol, vinyl guaiacol to 4-ethyl guaiacol, and vinyl phenol to 4-ethyl phenol.70 Interestingly, the phylogenetic distribution of the vprA gene appears to be limited to lactobacilli, indicating that this last conversion step might be more specialized.70,132 The vinyl phenol reductase reaction has a prime use in the food industry, where it is used to modulate the release of volatile molecules, thereby improving the aroma and sensory characteristics of fruit and vegetable fermentations.133 Ethyl phenols produced by Lactobacillus spp. in the human gut microbiome have demonstrated beneficial effects on brain activity, anxiety, and colon cancer.134,135
7. Enzymatic conversion of ellagitannins and ellagic acid
In the past decade, urolithins have gained significant attention due to their profound anti-obesity and antitumor properties. It is of note that urolithin A has been approved by the Food and Drug Administration as a Generally Recognized as Safe oral supplement, and clinical trials have been performed to study its potential benefits on muscle strength in middle-aged adults.39,136–141 Urolithins are a family of dibenzopyran-6-one metabolites that are formed from the microbial metabolism of the non-flavonoid phenolics ellagitannin and ellagic acid, which are present in pomegranates, raspberries, strawberries, and nuts. The microbial involvement in urolithin metabolism was demonstrated using germ-free rats, which lack gut microbiota. These animals did not produce urolithin metabolites after being fed an ellagitannin and ellagic acid-rich diet, highlighting the essential role of gut microbial metabolism.36,136,138,142–144
Ellagitannins, a class of polyphenols, structurally consist of one or more hexahydroxydiphenic acid esters bound to a single glucose. However, some ellagitannins appear to as a monomer in structure, whereas others appear more dimeric. Due to these variabilities within their structure, ellagitannins cannot be absorbed in the small intestine without first undergoing extensive metabolism to smaller compounds, which have improved bioavailability.145
Host enzymes in the jejunum hydrolyze the glucose moiety, after which hexahydroxydiphenic acid spontaneously lactonizes to form ellagic acid (Figure 9). Ellagic acid itself can also occur in the diet since it is present in berries and nuts. A small fraction of ellagic acid is absorbed in the upper small intestine, which explains the presence of dimethyl ellagic acid-glucuronide in bile, portal plasma, and urine.136 The majority of the intestinal ellagic acid pool can provide a substrate for further metabolism by the microbiota. Metabolism of ellagic acid starts in the small intestine, where a series of transformations is mediated by gut microbial catechol dehydroxylases. The initial dehydroxylation in the catabolic pathway is the result of a decarboxylation reaction. One of the lactone moieties undergoes hydrolysis, leading to the formation of a free carboxylic acid and keto-enol tautomerization of the p-hydroxy group. The resulting carboxylic acid is then decarboxylated and subsequently, the p-hydroxy group leaves as water.
Figure 9.

Ellagitannins (such as pedunculagin) are hydrolyzed by host enzymes in the jejunum. The resulting hexahydroxydiphenic acid products undergo spontaneous lactonization to ellagic acid, which is the main substrate for urolithin metabolism by the gut microbiota.
Further enzymatic removal of hydroxyl groups results in the production of a variety of urolithin intermediates, such as urolithin D and urolithin C.136 These intermediates are further metabolized to generate urolithin A, isourolithin A, and urolithin B, which are detected in high concentrations in plasma, urine, and other tissues.41,146,147 Urolithin A is the most potent of these metabolites, and the only one detectable in feces due to its rapid absorption and subsequent glucuronidation or sulfation in the liver. To date, a total of 25 urolithins have been identified.36
The metabolism of ellagic acid into urolithins is dependent upon specific gut bacteria, whose presence drastically varies among individuals (Figure 10).148 This variability in gut microbial enzymatic capacity gives rise to differences in both the types and amounts of urolithins. A study of urinary urolithin excretion revealed that, after ingestion of walnut and pomegranate extracts, urolithin A is predominantly present and in its glucuronidated form.138 A subset of participants failed to produce urolithin A and instead produced isourolithin A and urolithin B. A noticeable fraction of the study did not produce any detectable urolithins, most likely indicating the absence of an appropriate urolithin-metabolizing microbiome.139 These corroborating observations led to the classification of healthy individuals into three phenotypes, or “metabotypes” based on their ability to produce and excrete different urolithin variants. Individuals of phenotype A produce only urolithin A, whereas phenotype B produce urolithin A, isourolithin A, and urolithin B. In individuals with “phenotype 0”, no production of bioactive urolithin is detected due to the lack of any gut microbiota to fully metabolize ellagic acid.136,140 Interestingly, individuals with chronic diseases characterized by dysbiosis, such as obesity, diabetes, and colon cancer, were mostly phenotype B. A similar trend was observed in the elderly population.138
Figure 10.

Ellagic acid catabolism capacity depends on an individual’s microbiota enzymatic repertoire, and can be classified into three main ‘phenotypes’. Individuals with phenotype 0 do not produce bioactive urolithin metabolites (shaded red). The majority of the population has phenotype a and can produce urolithin A, as well as various intermediates (yellow). The main metabolic pathway in phenotype B individuals culminates in production of urolithin B due to the presence of Ellagibacter spp. with 8-dehydroxylase capability (blue). Individuals belonging to phenotype a or B that harbor bacteria with 3-dehydroxylases can produce R-urolithins in small quantities (green).
Further in-depth investigations revealed the metabolic pathways involved in ellagic acid breakdown for the different phenotypes.140 Phenotype A, which represents the majority of the population at around 70%, is characterized by gut microbiota that can readily catabolize ellagic acid by hydrolyzing a lactone ring and catalyzing three consecutive dehydroxylation reactions to yield urolithin A as the ultimate product.136 On the other hand, phenotype B, which accounts for approximately 20% of the population, utilizes a similar pathway as phenotype A but with an additional dihydroxylation step leading to the production of urolithin B. In contrast, phenotype 0, which is relatively rare in Chinese and Spanish populations at 10% but more prevalent at 60% in the United States, is characterized by a gut microbiota that can only initiate the metabolic pathway of ellagic acid by opening one of the lactone rings to produce the urolithin M5 precursor.149
Several bacterial species have now been isolated and characterized from humans, which contribute to the ellagic acid catabolic pathway of ellagic acid in humans. Notable converters Gordonibacter urolithinfaciens DSM 27213T and Gordonibacter pamelaeae DSM 19378T belong to the Eggerthellaceae family and are capable of producing intermediate urolithin metabolites. Gordonibacter urolithinfaciens (DSM 27213T) can dehydroxylate urolithin D and urolithin M6, yielding urolithin C as the final metabolite through its 4-dehydroylase and 10-dehydroxylase activities, respectively.36 However, Gordonibacter lacks the ability to produce urolithin A from urolithin C, or urolithin B from urolithin A, or any other urolithin conversion involving dehydroxylation at the 9-position.36,136 Despite this observation, a positive correlation was identified between the presence of urolithin A in feces and the presence of Gordonibacter urolithinfaciens, as well as a negative correlation between this bacterium and isourolithin A and urolithin B in feces.140,150
The closely related species Ellagibacter 38 is capable of producing isourolithin from ellagic acid by performing a distinct a 8-dehydroxylation of urolithins M6 and urolithin C (Figure 10). While this species is a key member of phenotype B gut microbiota, it is incapable of performing this dehydroxylation to produce urolithin B. Additionally, Ellagibacter can be present in phenotype A, indicating the requirement for another bacterial species to convert isourolithin A into urolithin B.38,72 Several new urolithin metabolites, including urolithin M6R, urolithin M7R, urolithin CR, and urolithin AR, have been detected in small quantities in the feces of individuals that harbor bacteria with a 3-dehydroxylase. Since previously identified urolithins all maintained their 3-hydroxyl group these R-urolithins represent a new arm of the ellagic acid metabolic pathway and their bioactive role remains to be determined.143
A recent study identified complementary bacterial species with 9-dehydroxylase and 10-dehydroxylase activity, which are required for the final steps of converting urolithin C into urolithin A and isourolithin A into urolithin B. Four different Enterocloster strains (E. bolteae CEBAS S4A9 and DSM 15670T, E. asparagiformis DSM 15981T, and E. citroniae DSM 19261T) were identified that can convert urolithin C to urolithin A. In addition, E. clostridioformis DSM 933T was able to yield urolithin A via converting urolithin M6 to urolithin M7. However, none of these species are capable of metabolizing ellagic acid, for which they depend on a different member of the microbiota such as Gordonibacter. These Enterocloster species can also produce the novel metabolite, 3,4,8-trihydroxy-urolithin, also known as urolithin G.71
Co-culturing experiments investigated the discrepancies in urolithin metabolism between those with phenotype A versus B. Co-culturing G. urolithinfaciens DSM 27213T with E. bolteae CEBAS S4A9 strains resembled the urolithin profile to that of individuals with phenotype A, whereas co-culturing E. isourolithinifaciens with E. bolteae CEBAS S4A9 strains resembled the urolithin metabolic profile of individuals with the phenotype B.71 Other Enterocloster species which were tested exhibited similar results, except for Enterocloster citroniae DSM 19261T.
8. Health benefits of gut bacterial polyphenol-derived metabolites
Gut microbial bioconversion of dietary polyphenols into bioactive metabolites might well account for a significant share of the beneficial health effects attributed to their parent compounds and dietary sources. There is a growing interest in characterizing the responsible enzymes from the microbiota to better understand their role in disease progression and leverage their potential therapeutic properties.
Gut microbial flavonoid degradation products have received a large part of the attention and in vitro as well as select in vivo studies have highlighted health benefits for several of these metabolites. The common flavonoid metabolite phloroglucinol has been investigated as a medication for the reduction of digestive, biliary, and urinary spasms.151 In this capacity, phloroglucinol is promising for relieving the symptoms of irritable bowel syndrome,152,153 and protecting the gastric mucosa from ethanol-induced injury.154 Phloroglucinol might also be promising for treatment for nonalcoholic fatty liver disease by lowering inflammation, fat accumulation, and apoptosis in liver cells.155
Several monophenolic acid products of gut microbial flavonoid catabolism have beneficial effects on host health, either localized to the intestine or impacting distal targets. Phloretic acid (3-(4-hydroxyphenyl)propionic acid) reduced inflammation in Dextran Sulfate Sodium (DSS)-induced colitis in mice, and decreased the development of atherosclerosis by preventing plasma lipid accumulation and inhibiting foam cell formation.156,157 Protocatechuic acid (PCA) has a protective effect on distal organs attributed to its antioxidant activity, its ability to scavenge free radicals and reactive oxygen species,158 chelate metals,159 and induce the enzymatic activity of glutathione and superoxide dismutase.5,160 PCA has pro-apoptotic activity in breast, cervix, liver, prostate, bone, stomach and lung cancer,161–164 as well as prevents accumulation of mutations by blocking carcinogens from binding to DNA.165,166 In addition, PCA has antidiabetic effects, stimulating the insulin signaling pathway in human adipocytes, which leads to a reduction in plasma glucose levels.22,167 PCA also reduces diabetes-associated complications, including dyslipidemia, fatty liver disease, and retinopathy.5,168–170 PCA has an anti-inflammatory effect,164,171,172 and can act as a neuroprotective agent in animal models of neurodegenerative diseases.173–175 Additional research will be required to uncover the molecular mechanisms and pathways impacted by PCA across this plethora of conditions. Nonetheless, the versatile beneficial effects of PCA make it a good therapeutic option for diseases that are characterized by oxidative stress, including metabolic diseases. 3,4-Dihydroxyphenylacetic acid, 3-hydroxyphenylacetic acid, and 4-hydroxyphenylpropionic acid exhibit antiproliferative activity for prostate and colon cancer,176,177 in addition to their antioxidant activity.177,178 Furthermore, 3-hydroxyphenylacetic acid reduces alcohol-induced liver damage,179 acts as a vasodilator to decrease blood pressure,180 and reduces dysregulated spermatogenesis of old mice.181 4-Hydroxyphenylacetic acid can reverse obesity-induced hepatic steatosis, and this effect is associated with the activation of AMP-activated protein kinase α (AMPKα).10 4-Hydroxyphenylacetic acid also improves inflammation and lipid accumulation in alcohol-induced liver disease,182 protects the liver against acetaminophen-induced liver injury,183 and attenuates sepsis-induced acute kidney injury.184
Among the various polyphenolic gut microbial metabolites, the bioactivity of the isoflavonoid-derived metabolite (S)-equol and the urolithin compound family have been most intensely investigated. (S)-equol helps improve postmenopausal symptoms, such as hot flashes, osteoporosis.185–190 It can also decrease the risk of hormone-associated cancers, including breast cancer191,192 and prostate cancer.193–195 (S)-equol can also help in the amelioration of cardiometabolic diseases, by reducing liver steatosis,196 improving arterial stiffness and blood pressure,197,198 and increasing lipid metabolism in an obese population.199 Alleviation of several aging-associated conditions has been attributed to (S)-equol, including prevention of cognitive impairment in dementia and Alzheimer patients,200–202 as well as reducing inflammation and bone erosion-induced rheumatoid arthritis in mice.203 Urolithins are investigated for their capability to reduce inflammation in the small intestine and colon204–206 as well as their antioxidant and anticancer properties.145 Urolithin B is mainly of interest for its anti-cancer properties206–212 acting on cell cycle arrest, aromatase inhibition, and promotion of autophagy as well as senescence.213 In addition to potential anti-cancer activity, urolithin B can inhibit cardiomyocyte apoptosis in ischemic heart disease,214 and reduce inflammation and oxidative stress in neurodegenerative diseases.147,215 The closely related compound urolithin A has a variety of complementary bioactivities, including triggering neurogenesis resulting in improved cognition in mice216 and protection against ischemic neuronal injury.217–219 Urolithin A also ameliorates cardiometabolic disease145 and induces adipocyte browning, improving lipid metabolism.204 Finally, urolithin D has potential protective effects in cancer, as it inhibits ephrin type-A receptor 2 (EPHA2) phosphorylation in a cell-based assay.220
9. Conclusion and outlook
The discovery of polyphenol-metabolizing bacteria and their diverse metabolic pathways has shed light on the health-promoting effects of different classes of polyphenols and their bioactive metabolites, emphasizing the crucial role of gut microbiota in the transformation of a healthy diet to more bioavailable, and easily absorbed compounds in the small intestine. In this review, we described each enzymatic step in the metabolic pathway of different gut microbiota members that are necessary for the breakdown of flavonoids, ellagic acid, and ellagitannins to monophenolic acids, phloroglucinol, dihydroflavonols, and urolithins. Further investigations are needed to fully understand the mechanisms behind this process, but overall, the study of polyphenols metabolism and the identification of gut bacteria involved in that mechanism is a promising avenue for the development of new nutraceuticals and therapies for various diseases, as well as a way the development of probiotic interventions.
Funding Statement
This study was supported by a Research Grant from the Prevent Cancer Foundation [PCF2019-JC], an American Cancer Society Institutional Research Grant [IRG-16-186-21] and a Jump Start Award [CA043703] from the Case Comprehensive Cancer Center, and seed funding from the Cleveland Clinic Foundation (JC). JC is additionally supported by a National Institutes of Health grant [R01 AI153173], and a research grant from the Harold and Leila Mathers Foundation [HLMCJC1022].
Disclosure statement
No potential conflict of interest was reported by the author(s).
Author contributions
Writing of the original draft: SA. Figures: SA, JC. Writing, review and interpretation: SA, JC.
References
- 1.Panche AN, Diwan AD, Chandra SR.. Flavonoids: an overview. J Nutr Sci. 2016;5:e47. doi: 10.1017/jns.2016.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Agati G, Azzarello E, Pollastri S, Tattini M. Flavonoids as antioxidants in plants: location and functional significance. Plant Sci. 2012;196:67–28. doi: 10.1016/j.plantsci.2012.07.014. [DOI] [PubMed] [Google Scholar]
- 3.Samanta A, Das G, Das S. Roles of flavonoids in plants. Int J Pharm Sci Technol. 2011;6:12–35. [Google Scholar]
- 4.Iwashina T. Flavonoid properties of five families newly incorporated into the order caryophyllales (review). Bull Natl Museum Nat Sci Ser B, Botany. 2013;39:25–51. [Google Scholar]
- 5.Abdelmageed ME, Shehatou GSG, Suddek GM, Salem HA. Protocatechuic acid improves hepatic insulin resistance and restores vascular oxidative status in type-2 diabetic rats. Environ Toxicol Pharmacol. 2021;83:103577. doi: 10.1016/j.etap.2020.103577. [DOI] [PubMed] [Google Scholar]
- 6.Corcoran MP, McKay DL, Blumberg JB. Flavonoid basics: chemistry, sources, mechanisms of action, and safety. J Nutr Gerontol Geriatr. 2012;31(3):176–189. doi: 10.1080/21551197.2012.698219. [DOI] [PubMed] [Google Scholar]
- 7.Osborn LJ, Schultz K, Massey W, DeLucia B, Choucair I, Varadharajan V, Banerjee R, Fung K, Horak AJ, Orabi D, et al. A gut microbial metabolite of dietary polyphenols reverses obesity-driven hepatic steatosis. Proc Natl Acad Sci USA. 2022;119(48):e2202934119. doi: 10.1073/pnas.2202934119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Abotaleb M, Samuel S, Varghese E, Varghese S, Kubatka P, Liskova A, Büsselberg D. Flavonoids in cancer and apoptosis. Cancers. 2019;11(1):28. doi: 10.3390/cancers11010028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Appleby PN, Crowe FL, Bradbury KE, Travis RC, Key TJ. Mortality in vegetarians and comparable nonvegetarians in the United Kingdom. Am J Clin Nutr. 2016;103(1):218–230. doi: 10.3945/ajcn.115.119461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Osborn LJ, Schultz K, Massey W, DeLucia B, Choucair I, Varadharajan V, Banerjee R, Fung K, Horak AJ III, Orabi D, et al. A gut microbial metabolite of dietary polyphenols reverses obesity-driven hepatic steatosis. 2021; 2021.09.16.460661. Preprint at. 10.1101/2021.09.16.460661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bakhtiari M, Panahi Y, Ameli J, Darvishi B. Protective effects of flavonoids against alzheimer’s disease-related neural dysfunctions. Biomed & Pharmacother. 2017;93:218–229. doi: 10.1016/j.biopha.2017.06.010. [DOI] [PubMed] [Google Scholar]
- 12.Baptista FI, Henriques AG, Silva AMS, Wiltfang J, da Cruz e Silva OAB. Flavonoids as therapeutic compounds targeting key proteins involved in Alzheimer’s disease. ACS Chem Neurosci. 2014;5(2):83–92. doi: 10.1021/cn400213r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bratu MM, Doroftei E, Negreanu-Pirjol T, Hostina C, Porta S. 2012. Determination of Antioxidant Activity and Toxicity of Sambucus nigra Fruit Extract Using Alternative Methods. Food Technol. Biotechnol. 50 (2): 177–182. ftb.com.hr.
- 14.Jovanovic SV, Steenken S, Tosic M, Marjanovic B, Simic MG. Flavonoids as antioxidants. ACS Publ. 2002; doi: 10.1021/ja00090a032. [DOI] [Google Scholar]
- 15.García-Lafuente A, Guillamón E, Villares A, Rostagno MA, Martínez JA. Flavonoids as anti-inflammatory agents: implications in cancer and cardiovascular disease. Inflamm Res. 2009;58(9):537–552. doi: 10.1007/s00011-009-0037-3. [DOI] [PubMed] [Google Scholar]
- 16.Serafini M, Peluso I, Raguzzini A. 2010. Flavonoids as anti-inflammatory agents. Proc. Nutr. Soc.69:273–278. doi: 10.1017/S002966511000162X. [DOI] [PubMed] [Google Scholar]
- 17.Loarca-Piña G, Neri M, Figueroa JDD, Castaño-Tostado E, Ramos-Gómez M, Reynoso R, Mendoza S. Chemical characterization, antioxidant and antimutagenic evaluations of pigmented corn. J Food Sci Technol. 2019;56(7):3177–3184. doi: 10.1007/s13197-019-03671-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Matsumoto T, Nishikawa T, Furukawa A, Itano S, Tamura Y, Hasei T, Watanabe T. Antimutagenic effects of polymethoxy flavonoids of citrus unshiu. Nat Prod Commun. 2017;12(1):1934578X1701200108. doi: 10.1177/1934578X1701200108. [DOI] [PubMed] [Google Scholar]
- 19.Miyazawa M, Hisama M. Antimutagenic activity of flavonoids from chrysanthemum morifolium. Biosci, Biotechnol, Biochem. 2003;67(10):2091–2099. doi: 10.1271/bbb.67.2091. [DOI] [PubMed] [Google Scholar]
- 20.Basu A, Das AS, Majumder M, Mukhopadhyay R. Antiatherogenic roles of dietary flavonoids chrysin, quercetin, and Luteolin. J Cardiovasc Pharmacol. 2016;68(1):89–96. doi: 10.1097/FJC.0000000000000380. [DOI] [PubMed] [Google Scholar]
- 21.Mulvihill EE, Huff MW. Antiatherogenic properties of flavonoids: implications for cardiovascular health. Can J Cardiol. 2010;26:17A–21A. doi: 10.1016/S0828-282X(10)71056-4. [DOI] [PubMed] [Google Scholar]
- 22.Gray A, Abdel-Wahab Y, Flatt P. Insulin-like and insulin-releasing actions of the traditional antidiabetic plant Sambucus nigra (elder). J Nutr. 2000;130(1):15–20. doi: 10.1093/jn/130.1.15. [DOI] [PubMed] [Google Scholar]
- 23.Ren N, Kim E, Li B, Pan H, Tong T, Yang CS, Tu Y. Flavonoids alleviating insulin resistance through inhibition of inflammatory signaling. J Agric Food Chem. 2019;67(19):5361–5373. doi: 10.1021/acs.jafc.8b05348. [DOI] [PubMed] [Google Scholar]
- 24.Song Y, Manson JE, Buring JE, Sesso HD, Liu S. Associations of dietary flavonoids with risk of type 2 diabetes, and markers of insulin resistance and systemic inflammation in women: a prospective study and cross-sectional analysis. J Am Coll Nutr. 2005;24(5):376–384. doi: 10.1080/07315724.2005.10719488. [DOI] [PubMed] [Google Scholar]
- 25.Bojić M, Maleš Ž, Antolić A, Babić I, Tomičić M. Antithrombotic activity of flavonoids and polyphenols rich plant species. Acta Pharmaceutica. 2019;69(4):483–495. doi: 10.2478/acph-2019-0050. [DOI] [PubMed] [Google Scholar]
- 26.Han N, Gu Y, Ye C, Cao Y, Liu Z, Yin J. Antithrombotic activity of fractions and components obtained from raspberry leaves (Rubus chingii). Food Chem. 2012;132(1):181–185. doi: 10.1016/j.foodchem.2011.10.051. [DOI] [PubMed] [Google Scholar]
- 27.Ruihua G, Dong D, Yiting Z, Yiwen F, Wenhui W. Flavonnoids with antithrombotic activities: a review. Med Res. 2017;1(1):12. doi: 10.21127/yaoyimr20170006. [DOI] [Google Scholar]
- 28.Chen L, Cao H, Huang Q, Xiao J, Teng H. Absorption, metabolism and bioavailability of flavonoids: a review. Crit Rev Food Sci Nutr. 2022;62(28):7730–7742. doi: 10.1080/10408398.2021.1917508. [DOI] [PubMed] [Google Scholar]
- 29.Williamson G, Kay CD, Crozier A. The bioavailability, transport, and bioactivity of dietary flavonoids: a review from a historical perspective. Compr Rev Food Sci Food Saf. 2018;17(5):1054–1112. doi: 10.1111/1541-4337.12351. [DOI] [PubMed] [Google Scholar]
- 30.Spencer JPE. Metabolism of tea flavonoids in the gastrointestinal tract. J Nutr. 2003;133(10):3255S–3261S. doi: 10.1093/jn/133.10.3255S. [DOI] [PubMed] [Google Scholar]
- 31.Gaya P, Medina M, Sánchez-Jiménez A, Landete JM. Phytoestrogen metabolism by adult human gut microbiota. Molecules. 2016;21(8):1034. doi: 10.3390/molecules21081034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gall M, Thomsen M, Peters C, Pavlidis IV, Jonczyk P, Grünert PP, Beutel S, Scheper T, Gross E, Backes M, et al. Enzymatic conversion of flavonoids using bacterial chalcone isomerase and Enoate Reductase. Angew Chem Int Ed. 2014;53(5):1439–1442. doi: 10.1002/anie.201306952. [DOI] [PubMed] [Google Scholar]
- 33.Kim M, Marsh ENG, Kim S-U, Han J. Conversion of (3S,4R)-tetrahydrodaidzein to (3S)-equol by THD reductase: proposed mechanism involving a radical intermediate. Biochemistry. 2010;49(26):5582–5587. doi: 10.1021/bi100465y. [DOI] [PubMed] [Google Scholar]
- 34.Shimada Y, Yasuda S, Takahashi M, Hayashi T, Miyazawa N, Sato I, Abiru Y, Uchiyama S, Hishigaki H. Cloning and expression of a novel NADP(h)-dependent Daidzein Reductase, an enzyme involved in the metabolism of Daidzein, from equol-producing Lactococcus strain 20-92. Appl Environ Microbiol. 2010;76(17):5892–5901. doi: 10.1128/AEM.01101-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yang G, Hong S, Yang P, Sun Y, Wang Y, Zhang P, Jiang W, Gu Y. Discovery of an ene-reductase for initiating flavone and flavonol catabolism in gut bacteria. Nat Commun. 2021;12(1):790. doi: 10.1038/s41467-021-20974-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.García-Villalba R, Giménez‐Bastida JA, Cortés‐Martín A, Ávila‐Gálvez MÁ, Tomás‐Barberán FA, Selma MV, Espín JC, González‐Sarrías A. Urolithins: a comprehensive update on their metabolism, bioactivity, and associated gut microbiota. Mol Nutr & Food Res. 2022;66(21):2101019. doi: 10.1002/mnfr.202101019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ávila-Gálvez MÁ, García‐Villalba R, Martínez‐Díaz F, Ocaña‐Castillo B, Monedero‐Saiz T, Torrecillas‐Sánchez A, Abellán B, González‐Sarrías A, Espín JC. Metabolic profiling of dietary polyphenols and Methylxanthines in normal and malignant mammary tissues from breast cancer patients. Mol Nutr Food Res. 2019;63(9):e1801239. doi: 10.1002/mnfr.201801239. [DOI] [PubMed] [Google Scholar]
- 38.Selma MV, Beltrán D, Luna MC, Romo-Vaquero M, García-Villalba R, Mira A, Espín JC, Tomás-Barberán FA. Isolation of human intestinal bacteria capable of producing the bioactive metabolite isourolithin a from ellagic acid. Front Microbiol. 2017;8. doi: 10.3389/fmicb.2017.01521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Singh RK, Chang H-W, Yan D, Lee KM, Ucmak D, Wong K, Abrouk M, Farahnik B, Nakamura M, Zhu TH, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15(1):73. doi: 10.1186/s12967-017-1175-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Flint HJ, Duncan SH, Scott KP, Louis P. Links between diet, gut microbiota composition and gut metabolism. Proc Nutr Soc. 2015;74(1):13–22. doi: 10.1017/S0029665114001463. [DOI] [PubMed] [Google Scholar]
- 41.Ávila M, Hidalgo M, Sánchez-Moreno C, Pelaez C, Requena T, Pascual-Teresa SD. Bioconversion of anthocyanin glycosides by bifidobacteria and Lactobacillus. Food Res Int. 2009;42(10):1453. doi: 10.1016/j.foodres.2009.07.026. [DOI] [Google Scholar]
- 42.Miyake Y, Yamamoto K, Osawa T. Metabolism of antioxidant in lemon fruit (Citrus limon BURM. f.) by human intestinal bacteria. J Agric Food Chem. 1997;45(10):3738–3742. doi: 10.1021/jf970403r. [DOI] [Google Scholar]
- 43.Bang S-H, Hyun Y-J, Shim J, Hong S-W, Kim D-H. Metabolism of Rutin and Poncirin by human intestinal microbiota and cloning of their metabolizing α-L-Rhamnosidase from bifidobacterium dentium. J Microbiol Biotechnol. 2015;25(1):18–25. doi: 10.4014/jmb.1404.04060. [DOI] [PubMed] [Google Scholar]
- 44.Braune A, Blaut M. Intestinal bacterium eubacterium cellulosolvens deglycosylates flavonoid C- and O-Glucosides. Appl Environ Microbiol. 2012;78(22):8151–8153. doi: 10.1128/AEM.02115-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Braune A, Blaut M. Deglycosylation of puerarin and other aromatic C-glucosides by a newly isolated human intestinal bacterium. Environ Microbiol. 2011;13(2):482–494. doi: 10.1111/j.1462-2920.2010.02352.x. [DOI] [PubMed] [Google Scholar]
- 46.Schneider H, Schwiertz A, Collins MD, Blaut M. Anaerobic transformation of quercetin-3-glucoside by bacteria from the human intestinal tract. Arch Microbiol. 1999;171(2):81–91. doi: 10.1007/s002030050682. [DOI] [PubMed] [Google Scholar]
- 47.Schneider H, Blaut M. Anaerobic degradation of flavonoids by eubacterium ramulus. Arch Microbiol. 2000;173(1):71–75. doi: 10.1007/s002030050010. [DOI] [PubMed] [Google Scholar]
- 48.Kim M, Lee J, Han J. Deglycosylation of isoflavone C-glycosides by newly isolated human intestinal bacteria. J Sci Food Agric. 2015;95(9):1925–1931. doi: 10.1002/jsfa.6900. [DOI] [PubMed] [Google Scholar]
- 49.Lou Y, Zheng J, Hu H, Lee J, Zeng S. Application of ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry to identify curcumin metabolites produced by human intestinal bacteria. J Chromatogr B Analyt Technol Biomed Life Sci. 2015;985:38–47. doi: 10.1016/j.jchromb.2015.01.014. [DOI] [PubMed] [Google Scholar]
- 50.Dymarska M, Widenmann A, Low KE, Abbott DW, Guan L, Gänzle MG. Conversion of phytochemicals by Lactobacilli: (phospho)-β-glucosidases are specific for glucosylated phytochemicals rather than disaccharides. J Agric Food Chem. 2024;72(10):5428–5438. doi: 10.1021/acs.jafc.3c08535. [DOI] [PubMed] [Google Scholar]
- 51.Braune A, Engst W, Elsinghorst PW, Furtmann N, Bajorath J, Gütschow M, Blaut M. Chalcone isomerase from Eubacterium ramulus catalyzes the ring contraction of Flavanonols. J Bacteriol. 2016;198(21):2965–2974. doi: 10.1128/JB.00490-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Schaab MR, Barney BM, Francisco WA. Kinetic and spectroscopic studies on the quercetin 2,3-dioxygenase from Bacillus subtilis. Biochemistry. 2006;45(3):1009–1016. doi: 10.1021/bi051571c. [DOI] [PubMed] [Google Scholar]
- 53.Bowater L, Fairhurst SA, Just VJ, Bornemann S. Bacillus subtilis YxaG is a novel Fe-containing quercetin 2,3-dioxygenase. FEBS Lett. 2004;557(1–3):45–48. doi: 10.1016/S0014-5793(03)01439-X. [DOI] [PubMed] [Google Scholar]
- 54.Barney BM, Schaab MR, LoBrutto R, Francisco WA. Evidence for a new metal in a known active site: purification and characterization of an iron-containing quercetin 2,3-dioxygenase from Bacillus subtilis. Protein Expr Purif. 2004;35(1):131–141. doi: 10.1016/j.pep.2004.01.005. [DOI] [PubMed] [Google Scholar]
- 55.Kawada Y, Yokoyama S, Yanase E, Niwa T, Suzuki T. The production of S-equol from daidzein is associated with a cluster of three genes in Eggerthella sp. YY7918. Biosci of Microbiota, Food Health. 2016;35(3):113–121. doi: 10.12938/bmfh.2015-023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schröder C, Matthies A, Engst W, Blaut M, Braune A. Identification and expression of genes involved in the conversion of daidzein and genistein by the equol-forming bacterium slackia isoflavoniconvertens. Appl Environ Microbiol. 2013;79(11):3494–3502. doi: 10.1128/AEM.03693-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Maruo T, Sakamoto M, Ito C, Toda T, Benno Y. Adlercreutzia equolifaciens gen. nov. sp. nov. an equol-producing bacterium isolated from human faeces, and emended description of the genus eggerthella. Int J Systematic Evol Microbiol. 2008;58(5):1221–1227. doi: 10.1099/ijs.0.65404-0. [DOI] [PubMed] [Google Scholar]
- 58.Yokoyama S, Suzuki T. Isolation and characterization of a novel equol-producing bacterium from human feces. Biosci, Biotechnol, Biochem. 2008;72(10):2660–2666. doi: 10.1271/bbb.80329. [DOI] [PubMed] [Google Scholar]
- 59.Tamura M, Tsushida T, Shinohara K. Isolation of an isoflavone-metabolizing, clostridium-like bacterium, strain TM-40, from human faeces. Anaerobe. 2007;13(1):32–35. doi: 10.1016/j.anaerobe.2006.10.001. [DOI] [PubMed] [Google Scholar]
- 60.Wang X-L, Kim H-J, Kang S-I, Kim S-I, Hur H-G. Production of phytoestrogen S-equol from daidzein in mixed culture of two anaerobic bacteria. Arch Microbiol. 2007;187(2):155–160. doi: 10.1007/s00203-006-0183-8. [DOI] [PubMed] [Google Scholar]
- 61.Esteban-Torres M, Reverón I, Mancheño JM, de Las Rivas B, Muñoz R. Characterization of a feruloyl esterase from Lactobacillus plantarum. Appl Environ Microbiol. 2013;79(17):5130–5136. doi: 10.1128/AEM.01523-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lai KK, Lorca GL, Gonzalez CF. Biochemical properties of two cinnamoyl esterases purified from a Lactobacillus johnsonii strain isolated from stool samples of diabetes-resistant rats. Appl Environ Microbiol. 2009;75(15):5018–5024. doi: 10.1128/AEM.02837-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang X, Geng X, Egashira Y, Sanada H. Purification and characterization of a feruloyl esterase from the intestinal bacterium Lactobacillus acidophilus. Appl Environ Microbiol. 2004;70(4):2367–2372. doi: 10.1128/AEM.70.4.2367-2372.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kelly SM, O’Callaghan J, Kinsella M, van Sinderen D. Characterisation of a hydroxycinnamic acid esterase from the Bifidobacterium longum subsp. longum taxon. Front Microbiol. 2018;9:2690. doi: 10.3389/fmicb.2018.02690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Raimondi S, Anighoro A, Quartieri A, Amaretti A, Tomás‐Barberán FA, Rastelli G, Rossi M. Role of bifidobacteria in the hydrolysis of chlorogenic acid. Microbiologyopen. 2015;4(1):41–52. doi: 10.1002/mbo3.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Santamaría L, Reverón I, López de Felipe F, de Las Rivas B, Muñoz R, Dudley EG. Unravelling the reduction pathway as an alternative metabolic route to hydroxycinnamate decarboxylation in Lactobacillus plantarum. Appl Environ Microbiol. 2018;84(15):e01123–18. doi: 10.1128/AEM.01123-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Filannino P, Gobbetti M, De Angelis M, Di Cagno R, Elkins CA. Hydroxycinnamic acids used as external acceptors of electrons: an energetic advantage for strictly heterofermentative lactic acid bacteria. Appl Environ Microbiol. 2014;80(24):7574–7582. doi: 10.1128/AEM.02413-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Filannino P, Di Cagno R, Addante R, Pontonio E, Gobbetti M, Elkins CA. Metabolism of fructophilic lactic acid bacteria isolated from the Apis mellifera L. Bee gut: phenolic acids as external electron acceptors. Appl Environ Microbiol. 2016;82(23):6899–6911. doi: 10.1128/AEM.02194-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.López de Felipe F. Revised aspects into the molecular bases of hydroxycinnamic acid metabolism in Lactobacilli. Antioxidants. 2023;12(6):1294. doi: 10.3390/antiox12061294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Santamaría L, Reverón I, de Felipe FL, de las Rivas B, Muñoz R, Dudley EG. Ethylphenol formation by Lactobacillus plantarum: identification of the enzyme involved in the reduction of vinylphenols. Appl Environ Microbiol. 2018;84(17):e01064–18. doi: 10.1128/AEM.01064-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Iglesias-Aguirre CE, García-Villalba R, Beltrán D, Frutos-Lisón MD, Espín JC, Tomás-Barberán FA, Selma MV. Gut bacteria involved in ellagic acid metabolism to yield human urolithin metabotypes revealed. J Agric Food Chem. 2023;71(9):4029–4035. doi: 10.1021/acs.jafc.2c08889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Żary-Sikorska E, Fotschki B, Jurgoński A, Kosmala M, Milala J, Kołodziejczyk K, Majewski M, Ognik K, Juśkiewicz J. Protective effects of a strawberry ellagitannin-rich extract against pro-oxidative and pro-inflammatory dysfunctions induced by a high-fat diet in a rat model. Molecules. 2020;25(24):5874. doi: 10.3390/molecules25245874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Guven H, Arici A, Simsek O. Flavonoids in our foods: a short review. J Basic Clin Health Sci. 2019;3:96–106. doi: 10.30621/jbachs.2019.555. [DOI] [Google Scholar]
- 74.Németh K, Plumb GW, Berrin J-G, Juge N, Jacob R, Naim HY, Williamson G, Swallow DM, Kroon PA. Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur J Nutr. 2003;42(1):29–42. doi: 10.1007/s00394-003-0397-3. [DOI] [PubMed] [Google Scholar]
- 75.Day AJ, Cañada FJ, Díaz JC, Kroon PA, Mclauchlan R, Faulds CB, Plumb GW, Morgan MRA, Williamson G. Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. FEBS Lett. 2000;468(2–3):166–170. doi: 10.1016/S0014-5793(00)01211-4. [DOI] [PubMed] [Google Scholar]
- 76.Hollman P, de Vries J, van Leeuwen S, Mengelers M, Katan M. Absorption of dietary quercetin glycosides and quercetin in healthy ileostomy volunteers. Am J Clin Nutr. 1995;62(6):1276–1282. doi: 10.1093/ajcn/62.6.1276. [DOI] [PubMed] [Google Scholar]
- 77.Gee JM, DuPont MS, Day AJ, Plumb GW, Williamson G, Johnson IT. Intestinal transport of quercetin glycosides in rats involves both deglycosylation and interaction with the hexose transport pathway. J Nutr. 2000;130(11):2765–2771. doi: 10.1093/jn/130.11.2765. [DOI] [PubMed] [Google Scholar]
- 78.Hollman PC, van Trijp JMP, Buysman MNCP, Vd Gaag MS, Mengelers MJB, de Vries JHM, Katan MB. Relative bioavailability of the antioxidant flavonoid quercetin from various foods in man. FEBS Lett. 1997;418(1–2):152–156. doi: 10.1016/S0014-5793(97)01367-7. [DOI] [PubMed] [Google Scholar]
- 79.Li S, Liu J, Li Z, Wang L, Gao W, Zhang Z, Guo C. Sodium-dependent glucose transporter 1 and glucose transporter 2 mediate intestinal transport of quercetrin in Caco-2 cells. Food Nutr Res. 2020;64(0). doi: 10.29219/fnr.v64.3745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Matsumoto H, Inaba H, Kishi M, Tominaga S, Hirayama M, Tsuda T. Orally administered delphinidin 3-rutinoside and cyanidin 3-rutinoside are directly absorbed in rats and humans and appear in the blood as the intact forms. J Agric Food Chem. 2001;49(3):1546–1551. doi: 10.1021/jf001246q. [DOI] [PubMed] [Google Scholar]
- 81.Xu X, Harris KS, Wang HJ, Murphy PA, Hendrich S. Bioavailability of soybean isoflavones depends upon gut microflora in women. J Nutr. 1995;125(9):2307–2315. doi: 10.1093/jn/125.9.2307. [DOI] [PubMed] [Google Scholar]
- 82.van Duynhoven J, Vaughan EE, Jacobs, DM, Kemperman RA, van Velzen EJ, Gross G, Roger LC, Possemiers S, Smilde AK, Doré J, et al. 2011. Metabolic fate of polyphenols in the human superorganism. Proc. Natl. Acad. Sci. 108: 4531–4538. doi: 10.1073/pnas.1000098107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hidalgo M, Oruna-Concha MJ, Kolida S, Walton GE, Kallithraka S, Spencer JPE, Gibson GR, de Pascual-Teresa S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J Agric Food Chem. 2012;60(15):3882–3890. doi: 10.1021/jf3002153. [DOI] [PubMed] [Google Scholar]
- 84.Murota K, Nakamura Y, Uehara M. Flavonoid metabolism: the interaction of metabolites and gut microbiota. Biosci, Biotechnol, Biochem. 2018;82(4):600–610. doi: 10.1080/09168451.2018.1444467. [DOI] [PubMed] [Google Scholar]
- 85.Crespy V, Morand C, Besson C, Cotelle N, Vézin H, Demigné C, Rémésy C. The splanchnic metabolism of flavonoids highly differed according to the nature of the compound. Am J Physiol-Gastrointestinal Liver Physiol. 2003;284(6):G980–G988. doi: 10.1152/ajpgi.00223.2002. [DOI] [PubMed] [Google Scholar]
- 86.Selma MV, Espín JC, Tomás-Barberán FA. Interaction between phenolics and gut microbiota: role in human health. J Agric Food Chem. 2009;57(15):6485–6501. doi: 10.1021/jf902107d. [DOI] [PubMed] [Google Scholar]
- 87.Shi Q, Wang H, Liu J, Li S, Guo J, Li H, Jia X, Huo H, Zheng Z, You S, et al. Old yellow enzymes: structures and structure-guided engineering for stereocomplementary bioreduction. Appl Microbiol Biotechnol. 2020;104(19):8155–8170. doi: 10.1007/s00253-020-10845-z. [DOI] [PubMed] [Google Scholar]
- 88.Herles C, Braune A, Blaut M. First bacterial chalcone isomerase isolated from Eubacterium ramulus. Arch Microbiol. 2004;181(6):428–434. doi: 10.1007/s00203-004-0676-2. [DOI] [PubMed] [Google Scholar]
- 89.Thomsen M, Tuukkanen A, Dickerhoff J, Palm GJ, Kratzat H, Svergun DI, Weisz K, Bornscheuer UT, Hinrichs W. Structure and catalytic mechanism of the evolutionarily unique bacterial chalcone isomerase. Acta Cryst D. 2015;71(4):907–917. doi: 10.1107/S1399004715001935. [DOI] [PubMed] [Google Scholar]
- 90.Toogood HS, Scrutton NS. Discovery, characterization, engineering, and applications of ene-reductases for industrial biocatalysis. ACS Catal. 2018;8(4):3532–3549. doi: 10.1021/acscatal.8b00624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Schoefer L, Braune A, Blaut M. Cloning and expression of a phloretin hydrolase gene from eubacterium ramulus and characterization of the recombinant enzyme. Appl Environ Microbiol. 2004;70(10):6131–6137. doi: 10.1128/AEM.70.10.6131-6137.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Stevens JF, Maier CS. The chemistry of gut microbial metabolism of polyphenols. Phytochem Rev. 2016;15(3):425–444. doi: 10.1007/s11101-016-9459-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ulbrich K, Reichardt N, Braune A, Kroh LW, Blaut M, Rohn S. The microbial degradation of onion flavonol glucosides and their roasting products by the human gut bacteria eubacterium ramulus and flavonifractor plautii. Food Res Int. 2015;67:349–355. doi: 10.1016/j.foodres.2014.11.051. [DOI] [Google Scholar]
- 94.Kenneth DS. Phytoestrogens: the biochemistry, physiology, and implications for human health of soy isoflavones12. Am J Clin Nutr. 1998;68(6):S1333–S1346. doi: 10.1093/ajcn/68.6.1333S. [DOI] [PubMed] [Google Scholar]
- 95.Yuan J-P, Wang J-H, Liu X. Metabolism of dietary soy isoflavones to equol by human intestinal microflora – implications for health. Mol Nutr Food Res. 2007;51(7):765–781. doi: 10.1002/mnfr.200600262. [DOI] [PubMed] [Google Scholar]
- 96.Patisaul HB, Jefferson W. The pros and cons of phytoestrogens. Front Neuroendocrinol. 2010;31(4):400–419. doi: 10.1016/j.yfrne.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Atkinson C, Frankenfeld CL, Lampe JW. Gut bacterial metabolism of the soy isoflavone daidzein: exploring the relevance to human health. Exp Biol Med (Maywood). 2005;230(3):155–170. doi: 10.1177/153537020523000302. [DOI] [PubMed] [Google Scholar]
- 98.Aso T, et al. A natural S-Equol supplement alleviates hot flushes and other menopausal symptoms in equol nonproducing postmenopausal Japanese women. J Women’s Health. 2012;21:92–100. doi: 10.1089/jwh.2011.2753. [DOI] [PubMed] [Google Scholar]
- 99.Setchell KDR, Brown NM, Lydeking-Olsen E. The clinical importance of the metabolite equol—a clue to the effectiveness of soy and its isoflavones. J Nutr. 2002;132(12):3577–3584. doi: 10.1093/jn/132.12.3577. [DOI] [PubMed] [Google Scholar]
- 100.Munro IC, Harwood M, Hlywka JJ, Stephen AM, Doull J, Flamm WG, Adlercreutz H. Soy isoflavones: a safety review. Nutr Rev. 2003;61(1):1–33. doi: 10.1301/nr.2003.janr.1-33. [DOI] [PubMed] [Google Scholar]
- 101.Rowland I, Wiseman H, Sanders T, Adlercreutz H, Bowey E. Metabolism of oestrogens and phytoestrogens: role of the gut microflora. Biochemical Soc Trans. 1999;27(2):304–308. doi: 10.1042/bst0270304. [DOI] [PubMed] [Google Scholar]
- 102.Xu L, Du B, Xu B. A systematic, comparative study on the beneficial health components and antioxidant activities of commercially fermented soy products marketed in China. Food Chem. 2015;174:202–213. doi: 10.1016/j.foodchem.2014.11.014. [DOI] [PubMed] [Google Scholar]
- 103.Hur H-G, Lay JO Jr., Beger RD, Freeman JP, Rafii F. Isolation of human intestinal bacteria metabolizing the natural isoflavone glycosides daidzin and genistin. Arch Microbiol. 2000;174(6):422–428. doi: 10.1007/s002030000222. [DOI] [PubMed] [Google Scholar]
- 104.Raimondi S, Roncaglia L, De Lucia M, Amaretti A, Leonardi A, Pagnoni UM, Rossi M. Bioconversion of soy isoflavones daidzin and daidzein by Bifidobacterium strains. Appl Microbiol Biotechnol. 2009;81(5):943–950. doi: 10.1007/s00253-008-1719-4. [DOI] [PubMed] [Google Scholar]
- 105.Richelle M, Pridmore-Merten S, Bodenstab S, Enslen M, Offord EA. Hydrolysis of isoflavone glycosides to aglycones by β-glycosidase does not alter plasma and urine isoflavone pharmacokinetics in postmenopausal women. J Nutr. 2002;132(9):2587–2592. doi: 10.1093/jn/132.9.2587. [DOI] [PubMed] [Google Scholar]
- 106.Mayo B, Vázquez L, Flórez AB. Equol: a bacterial metabolite from the Daidzein isoflavone and its presumed beneficial health effects. Nutrients. 2019;11(9):2231. doi: 10.3390/nu11092231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Rafii F. The role of colonic bacteria in the metabolism of the natural isoflavone daidzin to Equol. Metabolites. 2015;5(1):56–73. doi: 10.3390/metabo5010056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Guo K, Zhang B, Chen C, Uchiyama S, Ueno T, Chen Y, Su Y. Daidzein-metabolising phenotypes in relation to serum lipids and uric acid in adults in Guangzhou, China. Br J Nutr. 2010;104(1):118–124. doi: 10.1017/S0007114510000279. [DOI] [PubMed] [Google Scholar]
- 109.Wang X-L, Hur H-G, Lee JH, Kim KT, Kim S-I. Enantioselective synthesis of S-equol from dihydrodaidzein by a newly isolated anaerobic human intestinal bacterium. Appl Environ Microbiol. 2005;71(1):214–219. doi: 10.1128/AEM.71.1.214-219.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Day AJ, DuPont MS, Ridley S, Rhodes M, Rhodes MJC, Morgan MRA, Williamson G. Deglycosylation of flavonoid and isoflavonoid glycosides by human small intestine and liver β-glucosidase activity. FEBS Lett. 1998;436(1):71–75. doi: 10.1016/S0014-5793(98)01101-6. [DOI] [PubMed] [Google Scholar]
- 111.Setchell KDR, Clerici C. Equol: history, chemistry, and formation. J Nutr. 2010;140(7):1355S–1362S. doi: 10.3945/jn.109.119776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gu L, House SE, Prior RL, Fang N, Ronis MJJ, Clarkson TB, Wilson ME, Badger TM. Metabolic phenotype of isoflavones differ among female rats, pigs, monkeys, and women. J Nutr. 2006;136(5):1215–1221. doi: 10.1093/jn/136.5.1215. [DOI] [PubMed] [Google Scholar]
- 113.Harris RM, et al. Phytoestrogens are potent inhibitors of estrogen sulfation: implications for breast cancer risk and treatment. J Clin Endocrinol & Metab. 2004;89:1779–1787. doi: 10.1210/jc.2003-031631. [DOI] [PubMed] [Google Scholar]
- 114.Shimada Y, Takahashi M, Miyazawa N, Ohtani T, Abiru Y, Uchiyama S, Hishigaki H. Identification of two novel reductases involved in equol biosynthesis in Lactococcus strain 20–92. MIP. 2011;21(3–4):160–172. doi: 10.1159/000335049. [DOI] [PubMed] [Google Scholar]
- 115.Shimada Y, Takahashi M, Miyazawa N, Abiru Y, Uchiyama S, Hishigaki H. Identification of a novel dihydrodaidzein racemase essential for biosynthesis of equol from daidzein in Lactococcus sp. Strain 20-92. Appl Environ Microbiol. 2012;78(14):4902–4907. doi: 10.1128/AEM.00410-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wang X-L, Shin K-H, Hur H-G, Kim S-I. Enhanced biosynthesis of dihydrodaidzein and dihydrogenistein by a newly isolated bovine rumen anaerobic bacterium. J Biotechnol. 2005;115(3):261–269. doi: 10.1016/j.jbiotec.2004.08.014. [DOI] [PubMed] [Google Scholar]
- 117.Kim M, Kim S-I, Han J, Wang X-L, Song D-G, Kim S-U. Stereospecific biotransformation of dihydrodaidzein into (3S)-equol by the human intestinal bacterium eggerthella strain julong 732. Appl Environ Microbiol. 2009;75(10):3062–3068. doi: 10.1128/AEM.02058-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Persson B, Kallberg Y, Bray JE, Bruford E, Dellaporta SL, Favia AD, Duarte RG, Jörnvall H, Kavanagh KL, Kedishvili N, et al. The SDR (short-chain dehydrogenase/reductase and related enzymes) nomenclature initiative. Chemico-Biol Interact. 2009;178(1–3):94–98. doi: 10.1016/j.cbi.2008.10.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wang Q, Spenkelink B, Boonpawa R, Rietjens IMCM. Use of physiologically based pharmacokinetic modeling to predict human gut microbial conversion of daidzein to S-Equol. J Agric Food Chem. 2022;70(1):343–352. doi: 10.1021/acs.jafc.1c03950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Vitale DC, Piazza C, Melilli B, Drago F, Salomone S. Isoflavones: estrogenic activity, biological effect and bioavailability. Eur J Drug Metab Pharmacokinet. 2013;38(1):15–25. doi: 10.1007/s13318-012-0112-y. [DOI] [PubMed] [Google Scholar]
- 121.Frey PA, Hegeman AD, Ruzicka FJ. The radical SAM superfamily. Crit Rev Biochem Mol Biol. 2008;43(1):63–88. doi: 10.1080/10409230701829169. [DOI] [PubMed] [Google Scholar]
- 122.Shisler KA, Broderick JB. Glycyl radical activating enzymes: structure, mechanism, and substrate interactions. Arch Biochem Biophys. 2014;546:64–71. doi: 10.1016/j.abb.2014.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Buckel W, Golding BT. Radical enzymes in anaerobes. Annu Rev Microbiol. 2006;60(1):27–49. doi: 10.1146/annurev.micro.60.080805.142216. [DOI] [PubMed] [Google Scholar]
- 124.Ruiz de la Bastida A, Peirotén Á, Langa S, Arqués JL, Landete JM. Heterologous production of equol by lactic acid bacteria strains in culture medium and food. Int J Food Microbiol. 2021;360:109328. doi: 10.1016/j.ijfoodmicro.2021.109328. [DOI] [PubMed] [Google Scholar]
- 125.Coman V, Vodnar DC. Hydroxycinnamic acids and human health: recent advances. J Sci Food Agric. 2020;100(2):483–499. doi: 10.1002/jsfa.10010. [DOI] [PubMed] [Google Scholar]
- 126.Chao C, Mong M, Chan K, Yin M. Anti‐glycative and anti‐inflammatory effects of caffeic acid and ellagic acid in kidney of diabetic mice. Mol Nutr Food Res. 2010;54(3):388–395. doi: 10.1002/mnfr.200900087. [DOI] [PubMed] [Google Scholar]
- 127.Janicke B, Hegardt C, Krogh M, Önning G, Åkesson B, Cirenajwis HM, Oredsson SM. The antiproliferative effect of dietary fiber phenolic compounds ferulic acid and p-coumaric acid on the cell cycle of Caco-2 cells. Nutr Cancer. 2011;63(4):611–622. doi: 10.1080/01635581.2011.538486. [DOI] [PubMed] [Google Scholar]
- 128.El-Seedi HR, Taher EA, Sheikh BY, Anjum S, Saeed A, AlAjmi MF, Moustafa MS, Al-Mousawi SM, Farag MA, Hegazy ME et al. 2018. Chapter 8 - hydroxycinnamic acids: natural sources, biosynthesis, possible biological activities, and roles in islamic medicine. Stud. Nat. Prod. Chem. 55: 269–292. 10.1016/B978-0-444-64068-0.00008-5 [DOI] [Google Scholar]
- 129.Clifford MN, Jaganath IB, Ludwig IA, Crozier A. Chlorogenic acids and the acyl-quinic acids: discovery, biosynthesis, bioavailability and bioactivity. Nat Prod Rep. 2017;34(12):1391–1421. doi: 10.1039/C7NP00030H. [DOI] [PubMed] [Google Scholar]
- 130.Di Pede G, Mena P, Bresciani L, Achour M, Lamuela-Raventós RM, Estruch R, Landberg R, Kulling SE, Wishart D, Rodriguez-Mateos A, et al. A systematic review and comprehensive evaluation of human intervention studies to unravel the bioavailability of hydroxycinnamic acids. Antioxid Redox Signal. 2024;40(7–9):510–541. doi: 10.1089/ars.2023.0254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Gómez-Juaristi M, Martínez-López S, Sarria B, Bravo L, Mateos R. Bioavailability of hydroxycinnamates in an instant green/roasted coffee blend in humans. Identification of novel colonic metabolites. Food Funct. 2018;9(1):331–343. doi: 10.1039/C7FO01553D. [DOI] [PubMed] [Google Scholar]
- 132.Gaur G, Oh J-H, Filannino P, Gobbetti M, van Pijkeren J-P, Gänzle MG. Genetic determinants of hydroxycinnamic acid metabolism in heterofermentative lactobacilli. Appl Environ Microbiol. 2020;86(5):e02461–19. doi: 10.1128/AEM.02461-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tlais AZA, Lemos Junior WJF, Filannino P, Campanaro S, Gobbetti M, Di Cagno R. How microbiome composition correlates with biochemical changes during sauerkraut fermentation: a focus on neglected bacterial players and functionalities. Microbiol Spectr. 2022;10(4):e00168–22. doi: 10.1128/spectrum.00168-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Needham BD, Funabashi M, Adame MD, Wang Z, Boktor JC, Haney J, Wu W-L, Rabut C, Ladinsky MS, Hwang S-J, et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature. 2022;602(7898):647–653. doi: 10.1038/s41586-022-04396-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kuji M, Itoh N, Ohba Y, Yamada K, Hashimoto K. Inhibitory effect of 4-ethylcatechol on β-glucuronidase activity. Food Sci Technol Res. 2021;27(5):797–806. doi: 10.3136/fstr.27.797. [DOI] [Google Scholar]
- 136.Tomás-Barberán FA, García-Villalba R, González-Sarrías A, Selma MV, Espín JC. Ellagic acid metabolism by human gut microbiota: consistent observation of three urolithin phenotypes in intervention trials, independent of food source, age, and health status. J Agric Food Chem. 2014;62(28):6535–6538. doi: 10.1021/jf5024615. [DOI] [PubMed] [Google Scholar]
- 137.Xian W, Yang S, Deng Y, Yang Y, Chen C, Li W, Yang R. Distribution of urolithins metabotypes in healthy Chinese youth: difference in gut microbiota and predicted metabolic pathways. J Agric Food Chem. 2021;69(44):13055–13065. doi: 10.1021/acs.jafc.1c04849. [DOI] [PubMed] [Google Scholar]
- 138.Cortés-Martín A, García-Villalba R, González-Sarrías A, Romo-Vaquero M, Loria-Kohen V, Ramírez-de-Molina A, Tomás-Barberán FA, Selma MV, Espín JC. The gut microbiota urolithin metabotypes revisited: the human metabolism of ellagic acid is mainly determined by aging. Food Funct. 2018;9(8):4100–4106. doi: 10.1039/C8FO00956B. [DOI] [PubMed] [Google Scholar]
- 139.García-Villalba R, Vissenaekens H, Pitart J, Romo-Vaquero M, Espín JC, Grootaert C, Selma MV, Raes K, Smagghe G, Possemiers S, et al. Gastrointestinal simulation model TWIN-SHIME shows differences between human urolithin-metabotypes in gut microbiota composition, pomegranate polyphenol metabolism, and transport along the intestinal tract. J Agric Food Chem. 2017;65(27):5480–5493. doi: 10.1021/acs.jafc.7b02049. [DOI] [PubMed] [Google Scholar]
- 140.Romo-Vaquero M, García-Villalba R, González-Sarrías A, Beltrán D, Tomás-Barberán FA, Espín, JC, Selma MV. Interindividual variability in the human metabolism of ellagic acid: contribution of gordonibacter to urolithin production. J Funct Foods. 2015;17:785–791. doi: 10.1016/j.jff.2015.06.040. [DOI] [Google Scholar]
- 141.Singh A, D’Amico D, Andreux PA, Dunngalvin G, Kern T, Blanco-Bose W, Auwerx J, Aebischer P, Rinsch C. Direct supplementation with urolithin a overcomes limitations of dietary exposure and gut microbiome variability in healthy adults to achieve consistent levels across the population. Eur J Clin Nutr. 2022;76(2):297–308. doi: 10.1038/s41430-021-00950-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Aune D, Chan DSM, Vieira AR, Rosenblatt DAN, Vieira R, Greenwood DC, Norat T. Fruits, vegetables and breast cancer risk: a systematic review and meta-analysis of prospective studies. Breast Cancer Res Treat. 2012;134(2):479–493. doi: 10.1007/s10549-012-2118-1. [DOI] [PubMed] [Google Scholar]
- 143.Garcia D, Hellberg K, Chaix A, Wallace M, Herzig S, Badur MG, Lin T, Shokhirev MN, Pinto AFM, Ross DS, et al. Genetic liver-specific AMPK activation protects against diet-induced obesity and NAFLD. Cell Rep. 2019;26(1):192–208.e6. doi: 10.1016/j.celrep.2018.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Doyle B, Griffiths LA. The metabolism of ellagic acid in the rat. Xenobiotica. 1980;10(4):247–256. doi: 10.3109/00498258009033752. [DOI] [PubMed] [Google Scholar]
- 145.García-Villalba R, Giménez‐Bastida JA, Ávila‐Gálvez MA, Tomás‐Barberán FA, Espín JC, González‐Sarrías A. Ellagitannins and their gut microbiota-derived metabolites: urolithins. In: Dietary polyphenols. John Wiley & Sons, Ltd; 2020. p. 319–364. doi: 10.1002/9781119563754.ch9. [DOI] [Google Scholar]
- 146.Nuñez-Sánchez MA, García-Villalba R, Monedero-Saiz T, García-Talavera NV, Gómez-Sánchez MB, Sánchez-Álvarez C, García-Albert AM, Rodríguez-Gil FJ, Ruiz-Marín M, Pastor-Quirante FA, et al. Targeted metabolic profiling of pomegranate polyphenols and urolithins in plasma, urine and colon tissues from colorectal cancer patients. Mol Nutr Food Res. 2014;58(6):1199–1211. doi: 10.1002/mnfr.201300931. [DOI] [PubMed] [Google Scholar]
- 147.González-Sarrías A, Giménez‐Bastida JA, García‐Conesa MT, Gómez‐Sánchez MB, García‐Talavera NV, Gil‐Izquierdo A, Sánchez‐Álvarez C, Fontana‐Compiano LO, Morga‐Egea JP, Pastor‐Quirante FA, et al. Occurrence of urolithins, gut microbiota ellagic acid metabolites and proliferation markers expression response in the human prostate gland upon consumption of walnuts and pomegranate juice. Mol Nutr Food Res. 2010;54(3):311–322. doi: 10.1002/mnfr.200900152. [DOI] [PubMed] [Google Scholar]
- 148.Djedjibegovic J, Marjanovic A, Panieri E, Saso L. Ellagic acid-derived urolithins as modulators of oxidative stress. Oxid Med Cell Longev. 2020;2020:1–15. doi: 10.1155/2020/5194508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.González-Sarrías A, García‐Villalba R, Romo‐Vaquero M, Alasalvar C, Örem A, Zafrilla P, Tomás‐Barberán FA, Selma MV, Espín JC. Clustering according to urolithin metabotype explains the interindividual variability in the improvement of cardiovascular risk biomarkers in overweight-obese individuals consuming pomegranate: a randomized clinical trial. Mol Nutr Food Res. 2017;61(5):1600830. doi: 10.1002/mnfr.201600830. [DOI] [PubMed] [Google Scholar]
- 150.Milala J, Kosmala M, Karlińska E, Juśkiewicz J, Zduńczyk Z, Fotschki B. Ellagitannins from strawberries with different degrees of polymerization showed different metabolism through gastrointestinal tract of rats. J Agric Food Chem. 2017;65(49):10738–10748. doi: 10.1021/acs.jafc.7b04120. [DOI] [PubMed] [Google Scholar]
- 151.Corvino A, Magli E, Minale M, Autelitano A, Valente V, Pierantoni GM. Phloroglucinol-derived medications are effective in reducing pain and spasms of urinary and biliary tracts: results of phase 3 multicentre, open-label, randomized, comparative studies of clinical effectiveness and safety. Adv Ther. 2023;40(2):619–640. doi: 10.1007/s12325-022-02347-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Shin SY, Cha BK, Kim W-S, Park JY, Kim JW, Choi CH. The effect of phloroglucinol in patients with diarrhea-predominant irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Neurogastroenterol Motil. 2020;26(1):117–127. doi: 10.5056/jnm19160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Jafri W, Yakoob J, Hussain S, Jafri N, Islam M. Phloroglucinol in irritable bowel syndrome. J Pak Med Assoc. 2006;56:5–8. doi: 10.14309/00000434-200509001-00910. [DOI] [PubMed] [Google Scholar]
- 154.Li N-S, Luo X-J, Zhang Y-S, He L, Liu Y-Z, Peng J. Phloroglucinol protects gastric mucosa against ethanol-induced injury through regulating myeloperoxidase and catalase activities. Fundam Clin Pharmacol. 2011;25(4):462–468. doi: 10.1111/j.1472-8206.2010.00877.x. [DOI] [PubMed] [Google Scholar]
- 155.Drygalski K, Siewko K, Chomentowski A, Odrzygóźdź C, Zalewska A, Krętowski A, Maciejczyk M. Phloroglucinol strengthens the antioxidant barrier and reduces oxidative/nitrosative stress in nonalcoholic fatty liver disease (NAFLD). Oxid Med Cell Longev. 2021;2021(1):8872702. doi: 10.1155/2021/8872702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zhang Y-Y, Li X-L, Li T-Y, Li M-Y, Huang R-M, Li W, Yang R-L. 3-(4-Hydroxyphenyl)propionic acid, a major microbial metabolite of procyanidin A2, shows similar suppression of macrophage foam cell formation as its parent molecule. RSC Adv. 2018;8(12):6242–6250. doi: 10.1039/C7RA13729J. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lee M-K, Park YB, Moon S-S, Bok SH, Kim D-J, Ha T-Y, Jeong T-S, Jeong K-S, Choi M-S. Hypocholesterolemic and antioxidant properties of 3-(4-hydroxyl)propanoic acid derivatives in high-cholesterol fed rats. Chem Biol Interact. 2007;170(1):9–19. doi: 10.1016/j.cbi.2007.06.037. [DOI] [PubMed] [Google Scholar]
- 158.Graton ME, Ferreira BHSH, Troiano JA, Potje SR, Vale GT, Nakamune ACMS, Tirapelli CR, Miller FJ, Ximenes VF, Antoniali C, et al. Comparative study between apocynin and protocatechuic acid regarding antioxidant capacity and vascular effects. Front Physiol. 2022;13:1047916. doi: 10.3389/fphys.2022.1047916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Li X, Wang X, Chen D, Chen S. Antioxidant activity and mechanism of protocatechuic acid in vitro. Funct Foods Health Dis. 2011;1(7):232–244. doi: 10.31989/ffhd.v1i7.127. [DOI] [Google Scholar]
- 160.Varì R, D’Archivio M, Filesi C, Carotenuto S, Scazzocchio B, Santangelo C, Giovannini C, Masella R. Protocatechuic acid induces antioxidant/detoxifying enzyme expression through jnk-mediated Nrf2 activation in murine macrophages. J Nutr Biochem. 2011;22(5):409–417. doi: 10.1016/j.jnutbio.2010.03.008. [DOI] [PubMed] [Google Scholar]
- 161.Yin M-C, Lin C-C, Wu H-C, Tsao S-M, Hsu C-K. Apoptotic effects of protocatechuic acid in human breast, lung, liver, cervix, and prostate cancer cells: potential mechanisms of action. J Agric Food Chem. 2009;57(14):6468–6473. doi: 10.1021/jf9004466. [DOI] [PubMed] [Google Scholar]
- 162.Yip ECH, Chan ASL, Pang H, Tam YK, Wong YH. Protocatechuic acid induces cell death in HepG2 hepatocellular carcinoma cells through a c-Jun N-terminal kinase-dependent mechanism. Cell Biol Toxicol. 2006;22(4):293–302. doi: 10.1007/s10565-006-0082-4. [DOI] [PubMed] [Google Scholar]
- 163.Wu Y-X, et al. Protocatechuic acid inhibits osteoclast differentiation and stimulates apoptosis in mature osteoclasts. Biomed & Pharmacother. 2016;82:399–405. doi: 10.1016/j.biopha.2016.05.008. [DOI] [PubMed] [Google Scholar]
- 164.Tsao S, Hsia T, Yin M. Protocatechuic acid inhibits lung cancer cells by modulating FAK, MAPK, and nf-κB pathways. Nutr Cancer. 2014;66(8):1331–1341. doi: 10.1080/01635581.2014.956259. [DOI] [PubMed] [Google Scholar]
- 165.Tanaka T, Tanaka T, Tanaka M. Potential cancer chemopreventive activity of protocatechuic acid. J Exp & Clin Med. 2011;3(1):27–33. doi: 10.1016/j.jecm.2010.12.005. [DOI] [Google Scholar]
- 166.Cadena-Iñiguez J, Santiago-Osorio E, Sánchez-Flores N, Salazar-Aguilar S, Soto-Hernández RM, Riviello-Flores MDLL, Macías-Zaragoza VM, Aguiñiga-Sánchez I. The cancer-protective potential of protocatechuic acid: a narrative review. Molecules. 2024;29(7):1439. doi: 10.3390/molecules29071439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Scazzocchio B, Varì R, Filesi C, Del Gaudio I, D’Archivio M, Santangelo C, Iacovelli A, Galvano F, Pluchinotta FR, Giovannini C, et al. Protocatechuic acid activates key components of insulin signaling pathway mimicking insulin activity. Mol Nutr Food Res. 2015;59(8):1472–1481. doi: 10.1002/mnfr.201400816. [DOI] [PubMed] [Google Scholar]
- 168.El-Sonbaty YA, Suddek GM, Megahed N, Gameil NM. Protocatechuic acid exhibits hepatoprotective, vasculoprotective, antioxidant and insulin-like effects in dexamethasone-induced insulin-resistant rats. Biochimie. 2019;167:119–134. doi: 10.1016/j.biochi.2019.09.011. [DOI] [PubMed] [Google Scholar]
- 169.Wan L, Li J, Chen W-B, Wu G-Q. Beneficial effects of protocatechuic acid on diabetic retinopathy in streptozocin-induced diabetic rats. Int J Ophthalmol. 2023;16(6):855–862. doi: 10.18240/ijo.2023.06.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Tan J, Hu R, Gong J, Fang C, Li Y, Liu M, He Z, Hou D-X, Zhang H, He J, et al. Protection against metabolic associated fatty liver disease by protocatechuic acid. Gut Microbes. 2023;15(1):2238959. doi: 10.1080/19490976.2023.2238959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Kakkar S, Bais S. A review on protocatechuic acid and its pharmacological potential. ISRN Pharmacol. 2014;2014:952943. doi: 10.1155/2014/952943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Zheng J, Li Q, He L, Weng H, Su D, Liu X, Ling W, Wang D. Protocatechuic acid inhibits vulnerable atherosclerotic lesion progression in older apoe-/- mice. J Nutr. 2020;150(5):1167–1177. doi: 10.1093/jn/nxaa017. [DOI] [PubMed] [Google Scholar]
- 173.Koza LA, Winter AN, Holsopple J, Baybayon-Grandgeorge AN, Pena C, Olson JR, Mazzarino RC, Patterson D, Linseman DA. Protocatechuic acid extends survival, improves motor function, diminishes gliosis, and sustains neuromuscular junctions in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Nutrients. 2020;12(6):1824. doi: 10.3390/nu12061824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Winter AN, Ross EK, Wilkins HM, Stankiewicz TR, Wallace T, Miller K, Linseman DA. An anthocyanin-enriched extract from strawberries delays disease onset and extends survival in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Nutr Neurosci. 2018;21(6):414–426. doi: 10.1080/1028415X.2017.1297023. [DOI] [PubMed] [Google Scholar]
- 175.Thapa R, Goyal A, Gupta G, Bhat AA, Singh SK, Subramaniyan V, Sharma S, Prasher P, Jakhmola V, Singh SK, et al. Recent developments in the role of protocatechuic acid in neurodegenerative disorders. Excli J. 2023;22:595–599. doi: 10.17179/excli2023-5940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Gao K, Xu A, Krul C, Venema K, Liu Y, Niu Y, Lu J, Bensoussan L, Seeram NP, Heber D, et al. Of the major phenolic acids formed during human microbial fermentation of tea, citrus, and soy flavonoid supplements, only 3,4-dihydroxyphenylacetic acid has antiproliferative activity. J Nutr. 2006;136:52–57. doi: 10.1093/jn/136.1.52. [DOI] [PubMed] [Google Scholar]
- 177.Amić A, Marković Z, Marković JMD, Jeremić S, Lučić B, Amić D. Free radical scavenging and COX-2 inhibition by simple colon metabolites of polyphenols: a theoretical approach. Comput Biol Chem. 2016;65:45–53. doi: 10.1016/j.compbiolchem.2016.09.013. [DOI] [PubMed] [Google Scholar]
- 178.Jaganath IB, Mullen W, Lean MEJ, Edwards CA, Crozier A. In vitro catabolism of rutin by human fecal bacteria and the antioxidant capacity of its catabolites. Free Radic Biol Med. 2009;47(8):1180–1189. doi: 10.1016/j.freeradbiomed.2009.07.031. [DOI] [PubMed] [Google Scholar]
- 179.Liu Y, Myojin T, Li K, Kurita A, Seto M, Motoyama A, Liu X, Satoh A, Munemasa S, Murata Y, et al. A major intestinal catabolite of quercetin glycosides, 3-hydroxyphenylacetic acid, protects the hepatocytes from the acetaldehyde-induced cytotoxicity through the enhancement of the total aldehyde dehydrogenase activity. Int J Mol Sci. 2022;23(3):1762. doi: 10.3390/ijms23031762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Dias P, Pourová J, Vopršalová M, Nejmanová I, Mladěnka P. 3-hydroxyphenylacetic acid: a blood pressure-reducing flavonoid metabolite. Nutrients. 2022;14(2):328. doi: 10.3390/nu14020328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Jin Z, Yang Y, Cao Y, Wen Q, Xi Y, Cheng J, Zhao Q, Weng J, Hong K, Jiang H, et al. The gut metabolite 3-hydroxyphenylacetic acid rejuvenates spermatogenic dysfunction in aged mice through GPX4-mediated ferroptosis. Microbiome. 2023;11(1):212. doi: 10.1186/s40168-023-01659-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhang T, Bao L, Zhao Q, Wu ZE, Dai M, Rao Q, Li F. Metabolomics reveals gut microbiota contribute to PPARα deficiency-induced alcoholic liver injury. J Proteome Res. 2023;22(7):2327–2338. doi: 10.1021/acs.jproteome.3c00093. [DOI] [PubMed] [Google Scholar]
- 183.Zhao H, Jiang Z, Chang X, Xue H, Yahefu W, Zhang X. 4-hydroxyphenylacetic acid prevents acute APAP-Induced liver injury by increasing phase II and Antioxidant enzymes in mice. Front Pharmacol. 2018;9:653. doi: 10.3389/fphar.2018.00653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.An S, Yao Y, Wu J, Hu H, Wu J, Sun M, Li J, Zhang Y, Li L, Qiu W, et al. Gut-derived 4-hydroxyphenylacetic acid attenuates sepsis-induced acute kidney injury by upregulating ARC to inhibit necroptosis. Biochim Biophys Acta Mol Basis Dis. 2024;1870(1):166876. doi: 10.1016/j.bbadis.2023.166876. [DOI] [PubMed] [Google Scholar]
- 185.Jackson RL, Greiwe JS, Schwen RJ. Emerging evidence of the health benefits of S-equol, an estrogen receptor β agonist. Nutr Rev. 2011;69(8):432–448. doi: 10.1111/j.1753-4887.2011.00400.x. [DOI] [PubMed] [Google Scholar]
- 186.He F-J, Chen J-Q. Consumption of soybean, soy foods, soy isoflavones and breast cancer incidence: differences between Chinese women and women in Western countries and possible mechanisms. Food Sci Hum Wellness. 2013;2(3–4):146–161. doi: 10.1016/j.fshw.2013.08.002. [DOI] [Google Scholar]
- 187.Ni X, Wu B, Li S, Zhu W, Xu Z, Zhang G, Cui H, Bai Q, Wang J. Equol exerts a protective effect on postmenopausal osteoporosis by upregulating OPG/RANKL pathway. Phytomedicine. 2023;108:154509. doi: 10.1016/j.phymed.2022.154509. [DOI] [PubMed] [Google Scholar]
- 188.Ishiwata N, Melby MK, Mizuno S, Watanabe S. New equol supplement for relieving menopausal symptoms: randomized, placebo-controlled trial of Japanese women. Menopause. 2009;16(1):141–148. doi: 10.1097/gme.0b013e31818379fa. [DOI] [PubMed] [Google Scholar]
- 189.Pawlowski JW, Martin BR, McCabe GP, McCabe L, Jackson GS, Peacock M, Barnes S, Weaver CM. Impact of equol-producing capacity and soy-isoflavone profiles of supplements on bone calcium retention in postmenopausal women: a randomized crossover trial. Am J Clin Nutr. 2015;102(3):695–703. doi: 10.3945/ajcn.114.093906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Yoshikata R, Myint KZY, Ohta H. Effects of equol supplement on bone and cardiovascular parameters in middle-aged Japanese women: a prospective observational study. J Altern Complement Med. 2018;24(7):701–708. doi: 10.1089/acm.2018.0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Verheus M, van Gils CH, Keinan-Boker L, Grace PB, Bingham SA, Peeters PHM. Plasma phytoestrogens and subsequent breast cancer risk. J Clin Oncol. 2007;25(6):648–655. doi: 10.1200/JCO.2006.06.0244. [DOI] [PubMed] [Google Scholar]
- 192.Fuhrman BJ, Teter BE, Barba M, Byrne C, Cavalleri A, Grant BJ, Horvath PJ, Morelli D, Venturelli E, Muti PC, et al. Equol status modifies the association of soy intake and mammographic density in a sample of postmenopausal women. Cancer Epidemiol Biomarker Prev. 2008;17(1):33–42. doi: 10.1158/1055-9965.EPI-07-0193. [DOI] [PubMed] [Google Scholar]
- 193.Mahmoud AM, Yang W, Bosland MC. Soy isoflavones and prostate cancer: a review of molecular mechanisms. J Steroid Biochem Mol Biol. 2014;140:116–132. doi: 10.1016/j.jsbmb.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Akaza H, Miyanaga N, Takashima N, Naito S, Hirao Y, Tsukamoto T, Fujioka T, Mori M, Kim WJ, Song JM, et al. Comparisons of percent equol producers between prostate cancer patients and controls: case-controlled studies of isoflavones in Japanese, Korean and American residents. Jpn J Clin Oncol. 2004;34(2):86–89. doi: 10.1093/jjco/hyh015. [DOI] [PubMed] [Google Scholar]
- 195.Applegate CC, Rowles JL, Ranard KM, Jeon S, Erdman JW. Soy consumption and the risk of prostate cancer: an updated systematic review and meta-analysis. Nutrients. 2018;10(1):40. doi: 10.3390/nu10010040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Luo Z, Yang L, He J, Han Y, Rong X, Luo D, Guo J. Equol, a metabolite of gut microbiota, attenuates liver steatosis via the SIRT1/HNF4α pathway in NAFLD mice. J Funct Foods. 2023;107:105640. doi: 10.1016/j.jff.2023.105640. [DOI] [Google Scholar]
- 197.Nestel P, Fujii A, Zhang L. An isoflavone metabolite reduces arterial stiffness and blood pressure in overweight men and postmenopausal women. Atherosclerosis. 2007;192(1):184–189. doi: 10.1016/j.atherosclerosis.2006.04.033. [DOI] [PubMed] [Google Scholar]
- 198.Törmälä R, Appt S, Clarkson TB, Groop P-H, Rönnback M, Ylikorkala O, Mikkola TS. Equol production capability is associated with favorable vascular function in postmenopausal women using tibolone; no effect with soy supplementation. Atherosclerosis. 2008;198(1):174–178. doi: 10.1016/j.atherosclerosis.2007.09.010. [DOI] [PubMed] [Google Scholar]
- 199.Usui T, Tochiya M, Sasaki Y, Muranaka K, Yamakage H, Himeno A, Shimatsu A, Inaguma A, Ueno T, Uchiyama S, et al. Effects of natural S-equol supplements on overweight or obesity and metabolic syndrome in the Japanese, based on sex and equol status. Clin Endocrinol (Oxf). 2013;78(3):365–372. doi: 10.1111/j.1365-2265.2012.04400.x. [DOI] [PubMed] [Google Scholar]
- 200.Lee Y-B, Lee HJ, Sohn HS. Soy isoflavones and cognitive function. J Nutr Biochem. 2005;16(11):641–649. doi: 10.1016/j.jnutbio.2005.06.010. [DOI] [PubMed] [Google Scholar]
- 201.Sekikawa A, Wharton W, Butts B, Veliky CV, Garfein J, Li J, Goon S, Fort A, Li M, Hughes TM, et al. Potential protective mechanisms of S-equol, a metabolite of soy isoflavone by the gut microbiome, on cognitive decline and dementia. Int J Mol Sci. 2022;23(19):11921. doi: 10.3390/ijms231911921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lu Y, An Y, Lv C, Ma W, Xi Y, Xiao R. Dietary soybean isoflavones in Alzheimer’s disease prevention. Asia Pac J Clin Nutr. 2020;27:946–954. [DOI] [PubMed] [Google Scholar]
- 203.Lin I-C, Yamashita S, Murata M, Kumazoe M, Tachibana H. Equol suppresses inflammatory response and bone erosion due to rheumatoid arthritis in mice. J Nutr Biochem. 2016;32:101–106. doi: 10.1016/j.jnutbio.2016.02.012. [DOI] [PubMed] [Google Scholar]
- 204.Zhang M, Cui S, Mao B, Zhang Q, Zhao J, Zhang H, Tang X, Chen W. Ellagic acid and intestinal microflora metabolite urolithin A: a review on its sources, metabolic distribution, health benefits, and biotransformation. Crit Rev Food Sci Nutr. 2023;63(24):6900–6922. doi: 10.1080/10408398.2022.2036693. [DOI] [PubMed] [Google Scholar]
- 205.Piwowarski JP, Kiss AK, Granica S, Moeslinger T. Urolithins, gut microbiota-derived metabolites of ellagitannins, inhibit lps-induced inflammation in RAW 264.7 murine macrophages. Mol Nutr Food Res. 2015;59(11):2168–2177. doi: 10.1002/mnfr.201500264. [DOI] [PubMed] [Google Scholar]
- 206.Giménez-Bastida JA, Larrosa M, González-Sarrías A, Tomás-Barberán F, Espín JC, García-Conesa M-T. Intestinal ellagitannin metabolites ameliorate cytokine-induced inflammation and associated molecular markers in human colon fibroblasts. J Agric Food Chem. 2012;60(36):8866–8876. doi: 10.1021/jf300290f. [DOI] [PubMed] [Google Scholar]
- 207.González-Sarrías A, Espín J-C, Tomás-Barberán FA, García-Conesa M-T. Gene expression, cell cycle arrest and MAPK signalling regulation in Caco-2 cells exposed to ellagic acid and its metabolites, urolithins. Mol Nutr Food Res. 2009;53(6):686–698. doi: 10.1002/mnfr.200800150. [DOI] [PubMed] [Google Scholar]
- 208.González-Sarrías A, Giménez-Bastida JA, Núñez-Sánchez MÁ, Larrosa M, García-Conesa MT, Tomás-Barberán FA, Espín JC. Phase-ii metabolism limits the antiproliferative activity of urolithins in human colon cancer cells. Eur J Nutr. 2014;53(3):853–864. doi: 10.1007/s00394-013-0589-4. [DOI] [PubMed] [Google Scholar]
- 209.González-Sarrías A, Larrosa M, Tomás-Barberán FA, Dolara P, Espín JC. Nf-κB-dependent anti-inflammatory activity of urolithins, gut microbiota ellagic acid-derived metabolites, in human colonic fibroblasts. Br J Nutr. 2010;104(4):503–512. doi: 10.1017/S0007114510000826. [DOI] [PubMed] [Google Scholar]
- 210.González-Sarrías A, Núñez‐Sánchez MÁ, Tomé‐Carneiro J, Tomás‐Barberán FA, García‐Conesa MT, Espín JC. Comprehensive characterization of the effects of ellagic acid and urolithins on colorectal cancer and key-associated molecular hallmarks: MicroRNA cell specific induction of CDKN1A (p21) as a common mechanism involved. Mol Nutr Food Res. 2016;60(4):701–716. doi: 10.1002/mnfr.201500780. [DOI] [PubMed] [Google Scholar]
- 211.Cerdá B, Tomás-Barberán FA, Espín JC. Metabolism of antioxidant and chemopreventive ellagitannins from strawberries, raspberries, walnuts, and oak-aged wine in humans: identification of biomarkers and individual variability. J Agric Food Chem. 2005;53(2):227–235. doi: 10.1021/jf049144d. [DOI] [PubMed] [Google Scholar]
- 212.Lv M-Y, Shi C-J, Pan F-F, Shao J, Feng L, Chen G, Ou C, Zhang J-F, Fu W-M. Urolithin B suppresses tumor growth in hepatocellular carcinoma through inducing the inactivation of Wnt/β-catenin signaling. J Cell Biochem. 2019;120(10):17273–17282. doi: 10.1002/jcb.28989. [DOI] [PubMed] [Google Scholar]
- 213.Al-Harbi SA, Abdulrahman AO, Zamzami MA, Khan MI. Urolithins: the gut based polyphenol metabolites of Ellagitannins in cancer prevention, a review. Front Nutr. 2021;8:647582. doi: 10.3389/fnut.2021.647582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Huang X, Gao H, Jiang X, Zheng Z. Urolithin B, a gut microbiota metabolite, reduced susceptibility to myocardial arrhythmic predisposition after hypoxia. Dis Markers. 2022;2022:6517266. doi: 10.1155/2022/6517266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Lee G, Park J-S, Lee E-J, Ahn J-H, Kim H-S. Anti-inflammatory and antioxidant mechanisms of urolithin B in activated microglia. Phytomedicine. 2019;55:50–57. doi: 10.1016/j.phymed.2018.06.032. [DOI] [PubMed] [Google Scholar]
- 216.Gong Z, Huang J, Xu B, Ou Z, Zhang L, Lin X, Ye X, Kong X, Long D, Sun X, et al. Urolithin a attenuates memory impairment and neuroinflammation in APP/PS1 mice. J Neuroinflammation. 2019;16(1):62. doi: 10.1186/s12974-019-1450-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Ahsan A, Zheng Y-R, Wu X-L, Tang W-D, Liu M-R, Ma S-J, Jiang L, Hu W-W, Zhang X-N, Chen Z, et al. Urolithin A-activated autophagy but not mitophagy protects against ischemic neuronal injury by inhibiting ER stress in vitro and in vivo. CNS Neurosci Ther. 2019;25(9):976–986. doi: 10.1111/cns.13136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Chen P, Chen F, Lei J, Li Q, Zhou B. Activation of the miR-34a-mediated SIRT1/mTOR signaling pathway by urolithin a attenuates D-Galactose-induced brain aging in mice. Neurotherapeutics. 2019;16(4):1269–1282. doi: 10.1007/s13311-019-00753-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Lim JR, Kim SY, Yoon JH, Cho JH, Lee S-J, et al. Urolithin a suppresses high glucose-induced neuronal amyloidogenesis by modulating TGM2-dependent er-mitochondria contacts and calcium homeostasis. Cell Death Differ. 2021;28(1):184–202. doi: 10.1038/s41418-020-0593-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Giorgio C, Mena P, Del Rio D, Brighenti F, Barocelli E, Hassan‐Mohamed I, Callegari D, Lodola A, Tognolini M. The ellagitannin colonic metabolite urolithin D selectively inhibits EphA2 phosphorylation in prostate cancer cells. Mol Nutr Food Res. 2015;59(11):2155–2167. doi: 10.1002/mnfr.201500470. [DOI] [PubMed] [Google Scholar]
