Next Article in Journal
Features of the Metabolisms of Cardiac Troponin Molecules—Part 1: The Main Stages of Metabolism, Release Stage
Next Article in Special Issue
Effect of Aqueous Extract of Phenolic Compounds Obtained from Red Wine in Experimental Model of Colitis in Mice
Previous Article in Journal
Multigene Panel Sequencing Reveals Cancer-Specific and Common Somatic Mutations in Colorectal Cancer Patients: An Egyptian Experience
Previous Article in Special Issue
Resveratrol Downregulates Granulocyte-Macrophage Colony-Stimulating Factor-Induced Oncostatin M Production through Blocking of PI3K/Akt/NF-κB Signal Cascade in Neutrophil-like Differentiated HL-60 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Putative Mechanisms Underlying the Beneficial Effects of Polyphenols in Murine Models of Metabolic Disorders in Relation to Gut Microbiota

1
Faculty of Nursing, Shumei University, Yachiyo 276-0003, Japan
2
Department of Advanced Free Radical Science, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2022, 44(3), 1353-1375; https://doi.org/10.3390/cimb44030091
Submission received: 25 February 2022 / Revised: 15 March 2022 / Accepted: 16 March 2022 / Published: 18 March 2022
(This article belongs to the Special Issue Polyphenols as Cellular Metabolic Regulators)

Abstract

:
The beneficial effects of polyphenols on metabolic disorders have been extensively reported. The interaction of these compounds with the gut microbiota has been the focus of recent studies. In this review, we explored the fundamental mechanisms underlying the beneficial effects of polyphenols in relation to the gut microbiota in murine models of metabolic disorders. We analyzed the effects of polyphenols on three murine models of metabolic disorders, namely, models of a high-fat diet (HFD)-induced metabolic disorder, dextran sulfate sodium (DSS)-induced colitis, and a metabolic disorder not associated with HFD or DSS. Regardless of the model, polyphenols ameliorated the effects of metabolic disorders by alleviating intestinal oxidative stress, improving inflammatory status, and improving intestinal barrier function, as well as by modulating gut microbiota, for example, by increasing the abundance of short-chain fatty acid-producing bacteria. Consequently, polyphenols reduce circulating lipopolysaccharide levels, thereby improving inflammatory status and alleviating oxidative imbalance at the lesion sites. In conclusion, polyphenols likely act by regulating intestinal functions, including the gut microbiota, and may be a safe and suitable therapeutic agent for various metabolic disorders.

1. Introduction

Polyphenols, widely distributed in fruits, vegetables, and plant-based beverages, such as tea, coffee, and wine, have health benefits, which have been thought to be due to their antioxidative activity. Polyphenols, alone or as part of mixtures, have been shown to prevent and alleviate oxidative stress-related metabolic disorders due to their intrinsic ability to scavenge free radicals by providing an electron or a hydrogen atom [1,2]. Although polyphenols have low oral bioavailability mainly because of their extensive biotransformation in the intestine and liver, as well as by the gut microbiota [3,4,5], they exert remarkable beneficial effects, which lead to the low bioavailability/high bioactivity paradox.
The human body provides an ecosystem for the habitation of trillions of microbial cells, most of which reside in the gastrointestinal tract; the gut microbiota is most likely associated with metabolic events related to health and disease [6]. The involvement of the gut microbiota in several pathophysiological conditions has been suggested [7], leveraging the advances in genomic techniques, such as 16S and 18S ribosomal RNA sequencing and metagenomic sequencing [8,9].
Besides their effects on oxidative stress-related metabolic disorders, polyphenols substantially interact with the gut microbiota [10,11]. Because an imbalance in the quantity and quality of gut bacteria is associated with several metabolic disorders, the interaction of polyphenols and gut microbiota has been focused upon [1], and in the past decade, several studies have been carried out in this regard. However, the fundamental mechanisms underlying the beneficial effects of polyphenols in relation to the gut microbiota have not yet been fully elucidated. Therefore, this review is aimed at describing the beneficial effects of polyphenols on metabolic disorders in light of their interactions with the gut microbiota.
As data on the effect of polyphenols on human gut microbiota based on human intervention studies are limited, we searched for studies on murine models of metabolic disorders in the PubMed database using the keywords “polyphenol AND gut microbiota AND (rat OR mouse)”. We selected articles from the last decade (2012–2021) discussing the relationship between the heath benefit action of polyphenols and the gut microbiota. Although polyphenols have a variety of compounds, such as flavonoids, phenolic acids, and lignans, we focused on polyphenols in general including plant extracts. Additionally, we excluded the literature discussing the prebiotic action and phytoestrogenic action of polyphenols.

2. Beneficial Effects of Polyphenols on Metabolic Disorders in Relation to the Gut Microbiota in High-Fat Diet (HFD)-Fed Murine Models

HFD-fed mice and rats have been used as in vivo models of chronic metabolic disorders, such as obesity, hyperlipidemia, hyperglycemia, liver injury, and inflammatory dysfunction, and the beneficial effects of polyphenols have been studied using these models in relation to the modulation of the gut microbiota [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34] as summarized in Table 1. In particular, as indicated by a previous study [30], oxidative stress, inflammation, and gut microbial disorders can be induced by long-term HFD. Therefore, an HFD model seems to be suitable for evaluating the putative modes of the action of polyphenols. Of polyphenols, resveratrol, a well-known sirtuin 1 (SIRT1) agonist, has been intensively studied. Resveratrol, alone or in combination with other polyphenolic compounds such as quercetin and sinapic acid or probiotics such as Bifidobacterium longum, alleviates effects of obesity, hyperlipidemia, hyperglycemia, and nonalcoholic fatty liver disease (NAFLD) in HFD-fed mice [17,21,26,28,29,31]. Collectively, the beneficial effects of resveratrol are likely attributable to improved oxidative stress and gut microbial composition. Because the bioavailability of many polyphenols, including resveratrol, is low [35,36], they can be located in the bowel lumen when orally administered. Resveratrol can directly alter the composition of the gut microbiota by increasing the abundance of short-chain fatty acid (SCFA)-producing bacteria, such as Bacteroides and Blautia, and by decreasing the abundance of harmful bacteria, such as Desulfovibrio and Lachnospiraceae_NK4A316_group, as well as improving intestinal oxidative stress by preventing the production of reactive oxygen species (ROS) and improving antioxidant defense mechanisms, for example, by enhancing superoxide dismutase (SOD) and glutathione (GSH) levels [28,29]. The increase in these SCFA-producing bacteria could lead to the anti-obesity effects of resveratrol because these bacteria reportedly correlate negatively with inflammation, insulin resistance, and obesity [37,38]. Indeed, fecal microbiota transplantation (FMT) from resveratrol-treated mice to HFD-fed mice resulted in decreased weight gain and increased insulin sensitivity in the latter [26]. Furthermore, resveratrol could improve the integrity of the gut intestinal barrier through the repair of intestinal mucosal morphology possibly due to improved intestinal redox status, which leads to amelioration of HFD-induced NAFLD [28], because the development of HFD-induced NAFLD is closely associated with a loss of tight junction proteins in the small intestine [39,40].
Aside from resveratrol, other polyphenols and polyphenol-rich extracts and substances also alleviate obesity, hyperlipidemia, liver injury, and inflammatory status secondary to the alteration of the gut microbiota composition [12,13,14,15,16,18,19,20,22,23,24,25,27,30,32,33,34]. Sinapine, a rapeseed polyphenol, ameliorated NAFLD, suppressed intestinal nuclear factor-κB (NF-κB) and tumor necrosis factor-α (TNF-α) expression, and enhanced adipose tissue insulin receptor substrate 1 (IRS-1) expression in HFD-fed mice [22]. Sinapine possibly manifested its effect by modulating the composition of the gut microbiota by decreasing the ratio of Firmicutes to Bacteroidetes and increasing the abundance of probiotics. Phylum-level analyses of Firmicutes and Bacteroidetes have revealed that a reduced population of Bacteroidetes or an increased population of Firmicutes is associated with obesity [41,42,43,44,45]. Among plant-origin polyphenol-rich extracts, the extracts of pomegranate peel, cranberry, cinnamon bark, grape and grape pomace, brown macroalga Lessonia trabeculata, Lonicera caerulea L. berries, and red pepper could attenuate obesity, hyperglycemia, or liver injury by alleviating oxidative stress and modulating the gut microbiota [12,14,15,19,24,25,27,32]. Polyphenol-rich beverages, food, and their ingredients also show beneficial effects on metabolic disorders in relation to the gut microbiota [13,16,18,23,30,33,34]. Tea, a popular beverage consumed worldwide, is known to contain catechins, such as epicatechin, epicatechin-3-gallate, epigallocatechin, and epigallocatechin-3-gallate (EGCG) [46]. Oral administration of tea polyphenols, such as polyphenols from fermented Pu-erh tea and fermented Fu brick tea mainly produced in China, ameliorated obesity and hyperlipidemia by ameliorating effects of inflammation and oxidative stress in the intestine, and improved intestinal barrier function, leading to reduced circulation of lipopolysaccharides (LPS) and modulation of the gut microbiota [16,18,23,34]. The tea polyphenols decreased the abundance of Proteobacteria, a source of LPS, and the Fu brick tea polyphenols increased phylogenetic diversity and decreased the Firmicutes/Bacteroidetes ratio. Pu-erh tea polyphenols reduced circulation of LPS via restoration of gut barrier function and restored HFD-induced gut microbial community structural shift. Chronic consumption of commercially available instant caffeinated coffee also ameliorated obesity and decreased the Firmicutes/Bacteroides ratio [13]. Polyphenol extracts from Shanxi-aged vinegar showed similar effects [30]. Polyphenol-rich whole red grape juice could reduce oxidative stress and inflammatory status by activating the body’s antioxidant system, preventing free radical action, and beneficially modulating the gut microbiota, although it slightly affected the body composition such as body mass, fat mass, and lean mass, and body bone area [33].
The proposed mode of action of polyphenols in HFD-induced metabolic disorders is illustrated in Figure 1. As reported previously, metabolic endotoxemia dysregulates the inflammatory tone mediated by infiltrated macrophages and triggers body weight gain and diabetes [47], and a previous meta-analysis revealed that elevated levels of inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-18, C-reactive protein) and TNF-α and low levels of adiponectin, an antidiabetic and antiatherogenic adipokine, are risk factors for type 2 diabetes [48]. Polyphenols decrease the abundance of bacteria, which are a source of LPS, and restore impaired intestinal tight junctions, possibly by improving the intestinal redox status, leading to lowered circulating LPS levels and alleviated inflammation in the adipose tissue. Subsequently, the levels of circulating adiponectin, an antidiabetic and antiatherogenic adipokine, can be restored, resulting in mitigating insulin resistance. This effect of adiponectin on insulin resistance appears to be mediated, at least in part, by an increase in fatty acid oxidation through activation of AMP-activated protein kinase (AMPK) and also through the peroxisome proliferator-activated receptor (PPAR)-α in the muscles and liver [49,50,51,52,53]. The polyphenols also decrease the Firmicutes/Bacteroidetes ratio, leading to an anti-obesity effect by depressing the increased capacity for energy harvest from the diet associated with obesity [44].

3. Beneficial Effects of Polyphenols on Dextran Sulfate Sodium (DSS)-Induced Colitis in Relation to the Gut Microbiota in Murine Models

Inflammatory bowel disease (IBD), such as Crohn’s disease and ulcerative colitis, is a recurrent and multifaceted inflammatory disorder requiring long-term medication [54,55]. DSS-induced colitis is an animal model of IBD, which has been used to study the beneficial effects of polyphenols in relation to the modulation of the gut microbiota [56,57,58,59,60,61,62,63,64,65] as shown in Table 2. In these studies, the oral intake of single polyphenolic compounds, polyphenol-rich extracts, or polyphenol-rich food substances ameliorated DSS-induced colitis, enhanced colonic barrier integrity, improved oxidative balance and inflammatory status in the blood and/or colon, and modulated the gut microbiota. As for the prophylactic effects, oral pretreatment with bronze tomato extract, quercetin, quercetin monoglycosides, taxifolin, flavanonol, or EGCG prevented the development of DSS-induced colitis, suggesting that these polyphenols can prevent DSS-induced oxidative imbalance and changes in the microbial composition in the colon [58,60,63,64]. A study showed that rectal administration of EGCG tended to exacerbate DSS-induced colitis, indicating that the direct effects of this compound are unlikely to play a primary role in vivo [64]; we presume that biotransformed metabolite(s) of EGCG could be the main driver for its action. In contrast, FMT from EGCG-treated mice to DSS-treated mice (EGCG-FMT) resulted not only in the amelioration of colitis but also in an increased abundance of SCFA-producing bacteria, such as Akkermansia, which showed a positive correlation with antioxidative indices and a negative correlation with inflammatory indices [64]. These results suggest that gut microbiota modulation, especially an increase in SCFA-producing bacteria, and, subsequently, in functional SCFAs, plays a pivotal role in EGCG-treated mice with colitis. The proposed mode of action of polyphenols in DSS-induced colitis is illustrated in Figure 2. The DSS-induced intestinal and systemic oxidative imbalance can be ameliorated by polyphenols, leading to the restoration of the impaired epithelial barrier of the intestine. In addition, polyphenols can increase the number of SCFA-producing bacteria with a subsequent increase in SCFA production, further enhancing the epithelial barrier function.

4. Beneficial Effects of Polyphenols on Metabolic Disorders Not Associated with HFD or DSS in Relation to the Gut Microbiota in Murine Models

The beneficial effects of polyphenols on metabolic disorders not associated with HFD or DSS are summarized in Table 3. Several studies have explored the beneficial effects on liver injury in relation to the gut microbiota. They showed that regardless of the hepatic disorder induced by different factors, such as fructose- or western diet-induced NAFLD, and alcohol-, LPS-, or L-carnitine-induced liver injury, polyphenols could prevent or alleviate liver injuries, ameliorate oxidative stress and inflammatory status, and modulate the composition of the gut microbiota or maintain its normal composition [66,67,68,69,70,71,72]. Polyphenol-treated animals show improved intestinal barrier function and reduced blood LPS levels, with the latter likely contributing to the prevention of necrotic damage to the liver. Four of the six studies on the taxonomic analysis of gut bacteria at the phylum level showed a clear decrease in the Firmicutes/Bacteroidetes ratio [66,67,68,71]. In studies on fructose- and ethanol-induced liver dysfunction [66,68], it was observed that the LPS content and Toll-like receptor 4 (TLR4) expression in the liver were decreased by oral intake of polyphenols. The latter study also showed that the abundance of Bacteroidetes was negatively correlated with parameters of oxidative stress and inflammation and that of Firmicutes was positively correlated; however, the role of the decreased Firmicutes/Bacteroidetes ratio in reduced liver inflammation was not discussed. Based on these results, a proposed mode of action of polyphenols on liver injuries induced by various factors is illustrated in Figure 3. Polyphenols can decrease the abundance of bacteria that are a source of LPS and enhance intestinal barrier function, possibly by improving intestinal redox status and lowering circulating LPS levels, which can, in turn, attenuate inflammation in the liver by suppressing the LPS-TLR4 signaling pathway in sinusoidal Kupffer cells.
Regarding metabolic disorders other than the liver injuries listed in Table 3, beneficial effects of polyphenols were reported on diabetic db/db mice, particulate matter ≤ 2.5 μm (PM2.5)-induced visceral adiposity, cafeteria diet-induced obesity, spontaneous hypertension, doxorubicin (an anti-cancer drug)-induced heart failure, and potassium oxonate-induced hyperuricemia [73,74,75,76,77,78]. Regardless of the experimental conditions and pathological sites, local and/or systemic oxidative stress-induced inflammation was reduced by polyphenol intake, along with altered gut microbiota. Regarding the involvement of gut microbiota in the actions of polyphenols, while in some studies it has been suggested that altered gut microbiota is the primary mechanism underlying the pharmacological actions of polyphenols [74,76], in others, it has been mentioned that further exploration is required to elucidate whether their beneficial effects are mediated by the gut microbiota [74,75,77].

5. Effects of Polyphenols on the Gut Microbiota in Healthy Mice and Rats

The effects of polyphenols on the gut microbiota of healthy animals have been reported [79,80,81,82] and are summarized in Table 4. A study showed that dietary supplementation of polyphenol-rich Jaboticaba (Plinia jaboticaba) peel extract altered the gut microbiota, increasing the abundance of Lactobacillus, Bifidobacterium, and Enterobacteriaceae without disturbing the antioxidant system [79]. Lactobacillus and Bifidobacterium were reported to exert inhibitory actions against harmful bacteria, likely via pH reduction [83]. Three other studies reported that polyphenol-rich dietary plant materials enhanced the hepatic antioxidant capacity and positively modulated the gut microbiota, even in healthy animals. The Firmicutes/Bacteroidetes ratio was significantly reduced by the dietary supplementation of whole golden kiwifruit with peel [80] and by oral gavage of polyphenol-rich Penthorum chinense extract [81]. Long-term oral gavage of anthocyanin-rich Lycium ruthenicum Murray was reported to increase SCFA-producing bacteria and enhance the intestinal barrier function [82]. These studies indicate that beneficial effects on the gut microbiota along with enhanced intestinal barrier function and/or antioxidant capacity could be exerted even in healthy animals.

6. Possible Involvement of Prooxidative Potential of Polyphenols in Intestinal Barrier Function

As described above, improved intestinal barrier function is likely a key player for polyphenols’ ameliorative action on metabolic disorders. It has been questioned if polyphenols directly exert antioxidative action in situ [84,85]. Aside from polyphenols’ direct antioxidative action, they possess prooxidative potential; e.g., antibacterial activity of catechins [86] and cytocidal action of plant polyphenols on cancer cells [87], both of which are exerted by cytotoxic ROS generated by oxidation of phenolic hydroxyl moiety coupled with the reduction of dissolved oxygen. Nuclear factor E2-related factor 2 (Nrf2) assumes the pivotal role in protecting cells, tissues, and organs owing to various genes encoding antioxidant proteins [88,89,90]. During ROS production by polyphenols, cells may activate the Nrf2 pathway independently of the polyphenols’ inherent antioxidant activity. This idea drove us to illustrate on possible involvement of prooxidative potential of polyphenols in intestinal barrier function (Figure 4). ROS generated by prooxidative polyphenols induces mild oxidative stress, which in turn activates Nrf2 followed by induction of antioxidant defense enzymes such as heme oxygenase 1 and NAD(P)H quinine oxidoreductase 1. These antioxidant enzymes could improve intestinal redox status, resulting in potentiated intestinal barrier function that prohibits LPS leakage to blood stream.

7. Future Perspective on Studies on Interaction of Polyphenols and Gut Microbiota

There are several issues that should be elucidated in the future studies. Given polyphenols’ poor absorbability from the digestive tract, their beneficial activity seems to be mediated through interaction with gut microbiota [91]. Accordingly, the number of studies on the interaction of polyphenols’ health beneficial effects and gut microbiota has gradually increased throughout this decade. Although many studies have shown that polyphenols could modulate gut microbiota, most studies failed to show how the polyphenols affected the microbiota on the basis of experimental evidence. In addition, while there have been many studies of polyphenol-rich plant extracts on this matter, there have been relatively a few studies of pure polyphenols. In other words, the possibility that components other than polyphenols could interact with gut microbiota still remains in the effects of plant extracts. Most of the studies performed chemical analyses of the polyphenols on the extracts; one study, for example, determined only the total polyphenol content along with carotenoid and capsinoid content, leaving us with the question of which component was a key player [32]. Next, despite the poor bioavailability of polyphenols, there have been very few reports on the in vivo fate of polyphenols in the literature we cited. To elucidate fundamental mechanisms, information on the absorption, distribution, metabolism, and elimination (ADME) of target polyphenols would be essential. Finally, future studies should also investigate whether polyphenol prooxidant properties are involved in improving intestinal barrier function along with the modulation of gut microbiota via the activation of Nrf2 pathway. Although meaningful findings have been accumulated through the efforts of many researchers, solving the above problem would give a new perspective to the in vivo effects of polyphenols in relation to gut microbiota.
Lastly, we also address the critical reviewing of the literature listed in the tables. Although replace, refine, reduce—the 3 Rs of ethical animal research—are globally accepted, researchers are required to formulate experiments based on enough statistical power to ensure the results of animal experiments, e.g., the message from UK funding agencies is that some experiments use too few animals, a problem that leads to wastage and low-quality results [92]. The National Institutes of Health also sounded a warning that some irreproducible reports using animal models are probably the result of coincidental findings that happen to reach statistical significance, coupled with publication bias [93]. In the tables, the number of animals per group in some papers were five or less, without stating the validity of the sample size [20,58,67,74]. We have to carefully interpret the data in such studies from the point of view of reproducibility.

8. Conclusions

There have been many reports on the beneficial effects of polyphenols on metabolic disorders, and recent studies have focused on their interaction with the gut microbiota. In HFD-fed murine models, polyphenols could ameliorate obesity, hyperlipidemia, and hyperglycemia by the alleviation of oxidative stress and inflammation in the intestine, the improvement of the intestinal barrier function, and the modulation of the gut microbiota, including a reduction in the Firmicutes/Bacteroidetes (F/B) ratio. In murine models of DSS-induced colitis, polyphenols could prevent or ameliorate oxidative imbalance, inflammatory status, and changes in the colonic microbial composition, with an increased abundance of SCFA-producing bacteria, leading to the protection of the intestinal epithelial barrier. In murine models of liver injuries not associated with HFD or DSS, polyphenols could improve the intestinal barrier function and reduced the blood LPS levels, which likely contributes to the prevention of necrotic damage in the liver, along with altered gut microbiota, including a reduction in the Firmicutes/Bacteroidetes ratio. Although some studies with FMT indicate a direct involvement of gut microbiota in the health benefits of polyphenols, further exploration is required in this regard.
Regarding the F/B ratio, it has been focused on by many researchers since the increased ratio was reported to be responsible for an increased capacity for energy harvest from diet [44]. Firmicutes and Bacteroides are the two main phyla of gut microbiota in mammals, playing important roles in maintaining gut microecological homeostasis [94]. Accordingly, it has been reported that alterations in the F/B ratio are associated with a variety of diseases [95,96,97]. However, there has been some inconsistency in the ratio even though similar experimental models were applied. For instance, in ovariectomized mice, one study showed an increased F/B ratio determined by a PCR analysis [98], but another one reported a decreased ratio determined by a 16s rDNA sequencing following DNA amplification by PCR [99]. In this review, five studies determined F/B ratios in Table 2, and among them one study revealed that the ratio increased [65], one study showed almost no change in the ratio [60], and the other three studies showed decreased ratios [56,58,63]. Thus, although the F/B ratio would be a good indicator to reflect gut microecological homeostasis, data should be carefully checked from the point of view of the following: which assay was applied for phylum level analysis, what timing of fecal sampling, how much the ratio changed, and so on.
Finally, the studies investigated thus far are limited to murine models, so that the findings cannot be extrapolated to humans. A review on the bioavailability of phytoestrogens such as isoflavones in murine models [100] noted that data should be carefully interpreted because of the large interspecies variability in the metabolism of phytoestrogens in murine models (e.g., their limited intestinal absorption and rapid excretion, compared to humans). Thus, data from murine models must be interpreted with great caution. In addition, given polyphenols’ poor absorbability in the digestive tract, their activity toward the human host seems to be mediated through interaction with intestinal microbes [101,102]. Considering transformation of dietary polyphenols by gut microbiota, reactions of polyphenols and bacteria are based on the reduction and/or hydrolysis because of anaerobic conditions. A typical well-known example is the bacterial transformation of the soya isoflavone daidzein to equal [103,104,105], which possesses high binding affinity to the estrogen receptor [106]. The O-deglycosylation of flavonoids by gut microbiota was also shown by many studies [102]. A typical example of flavonoid transformation is rutin to quercetin. As demonstrated, flavonoid aglycones, but not their glycosides, may inhibit growth of some intestinal bacteria [107], so that quercetin may have a more inhibitory influence on the intestinal bacteria than rutin. These direct effects of transformed polyphenols on gut microbiota have not been fully discussed in the literature listed in the tables. Since information on ADME of the polyphenols or the extracts is poor, further studies should be conducted in terms of ADME to obtain more information on the interaction between polyphenols and gut microbiota.

Funding

This study was partly supported by JSPS KAKENHI (grant number JP20K09996).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Shirato, Shishido, and Nakamura are members of the Academia–Industry Collaboration Laboratory at Tohoku University Graduate School of Dentistry, which receives funding from Luke Co., Ltd. (Sendai, Japan). This academia–industry collaboration was examined and approved by the Conflict of Interest Management Committee of Tohoku University. Luke Co., Ltd. and the grant funder had no role in the conception of the study design, data collection and interpretation, or in the decision to submit the work for publication.

References

  1. Marranzano, M.; Rosa, R.L.; Malaguarnera, M.; Palmeri, R.; Tessitori, M.; Barbera, A.C. Polyphenols: Plant sources and food industry applications. Curr. Pharm. Des. 2018, 24, 4125–4130. [Google Scholar] [CrossRef] [PubMed]
  2. Rajha, H.N.; Paule, A.; Aragonès, G.; Barbosa, M.; Caddeo, C.; Debs, E.; Dinkova, R.; Eckert, G.P.; Fontana, A.; Gebrayel, P.; et al. Recent advances in research on polyphenols: Effects on microbiota, metabolism, and health. Mol. Nutr. Food Res. 2021, 66, e2100670. [Google Scholar] [CrossRef] [PubMed]
  3. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef] [PubMed]
  4. Bertelli, A.; Biagi, M.; Corsini, M.; Baini, G.; Cappellucci, G.; Miraldi, E. Polyphenols: From theory to practice. Foods 2021, 10, 2595. [Google Scholar] [CrossRef]
  5. Iglesias-Aguirre, C.E.; Cortés-Martín, A.; Ávila-Gálvez, M.; Giménez-Bastida, J.A.; Selma, M.V.; González-Sarrías, A.; Espín, J.C. Main drivers of (poly)phenol effects on human health: Metabolite production and/or gut microbiota-associated metabotypes? Food Funct. 2021, 12, 10324–10355. [Google Scholar] [CrossRef]
  6. Parida, S.; Sharma, D. The microbiome-estrogen connection and breast cancer risk. Cells 2019, 8, 1642. [Google Scholar] [CrossRef] [Green Version]
  7. Yu, Y.; Raka, F.; Adeli, K. The role of the gut microbiota in lipid and lipoprotein metabolism. J. Clin. Med. 2019, 8, 2227. [Google Scholar] [CrossRef] [Green Version]
  8. The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
  9. The Integrative HMP (iHMP) Research Network Consortium. The integrative human microbiome project: Dynamic analysis of microbiome-host omics profiles during periods of human health and disease. Cell Host Microbe 2014, 16, 276–289. [Google Scholar] [CrossRef] [Green Version]
  10. Saint-Georges-Chaumet, Y.; Edeas, M. Microbiota-mitochondria inter-talk: Consequence for microbiota-host interaction. Pathog. Dis. 2016, 74, ftv096. [Google Scholar] [CrossRef] [Green Version]
  11. Zhao, Y.; Jiang, Q. Roles of the polyphenol-gut microbiota interaction in alleviating colitis and preventing colitis-associated colorectal cancer. Adv. Nutr. 2021, 12, 546–565. [Google Scholar] [CrossRef] [PubMed]
  12. Neyrinck, A.M.; Van Hée, V.F.; Bindels, L.B.; De Backer, F.; Cani, P.D.; Delzenne, N.M. Polyphenol-rich extract of pomegranate peel alleviates tissue inflammation and hypercholesterolaemia in high-fat diet-induced obese mice: Potential implication of the gut microbiota. Br. J. Nutr. 2013, 109, 802–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cowan, T.E.; Palmnäs, M.S.; Yang, J.; Bomhof, M.R.; Ardell, K.L.; Reimer, R.A.; Vogel, H.J.; Shearer, J. Chronic coffee consumption in the diet-induced obese rat: Impact on gut microbiota and serum metabolomics. J. Nutr. Biochem. 2014, 25, 489–495. [Google Scholar] [CrossRef]
  14. Anhê, F.F.; Roy, D.; Pilon, G.; Dudonné, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Collins, B.; Hoffman, J.; Martinez, K.; Grace, M.; Lila, M.A.; Cockrell, C.; Nadimpalli, A.; Chang, E.; Chuang, C.C.; Zhong, W.; et al. A polyphenol-rich fraction obtained from table grapes decreases adiposity, insulin resistance and markers of inflammation and impacts gut microbiota in high-fat-fed mice. J. Nutr. Biochem. 2016, 31, 150–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Wang, L.; Zeng, B.; Zhang, X.; Liao, Z.; Gu, L.; Liu, Z.; Zhong, Q.; Wei, H.; Fang, X. The effect of green tea polyphenols on gut microbial diversity and fat deposition in C57BL/6J HFA mice. Food Funct. 2016, 7, 4956–4966. [Google Scholar] [CrossRef]
  17. Zhao, L.; Zhang, Q.; Ma, W.; Tian, F.; Shen, H.; Zhou, M. A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food Funct. 2017, 8, 4644–4656. [Google Scholar] [CrossRef]
  18. Gao, X.; Xie, Q.; Kong, P.; Liu, L.; Sun, S.; Xiong, B.; Huang, B.; Yan, L.; Sheng, J.; Xiang, H. Polyphenol- and caffeine-rich postfermented Pu-erh tea improves diet-induced metabolic syndrome by remodeling intestinal homeostasis in mice. Infect. Immun. 2018, 86, e00601-17. [Google Scholar] [CrossRef] [Green Version]
  19. Van Hul, M.; Geurts, L.; Plovier, H.; Druart, C.; Everard, A.; Ståhlman, M.; Rhimi, M.; Chira, K.; Teissedre, P.L.; Delzenne, N.M.; et al. Reduced obesity, diabetes, and steatosis upon cinnamon and grape pomace are associated with changes in gut microbiota and markers of gut barrier. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E334–E352. [Google Scholar] [CrossRef]
  20. Wu, S.; Hu, R.; Nakano, H.; Chen, K.; Liu, M.; He, X.; Zhang, H.; He, J.; Hou, D.X. Modulation of gut microbiota by Lonicera caerulea L. berry polyphenols in a mouse model of fatty liver induced by high fat diet. Molecules 2018, 23, 3213. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, C.; Deng, Q.; Xu, J.; Wang, X.; Hu, C.; Tang, H.; Huang, F. Sinapic acid and resveratrol alleviate oxidative stress with modulation of gut microbiota in high-fat diet-fed rats. Food Res. Int. 2019, 116, 1202–1211. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Y.; Li, J.; Su, Q.; Liu, Y. Sinapine reduces non-alcoholic fatty liver disease in mice by modulating the composition of the gut microbiota. Food Funct. 2019, 10, 3637–3649. [Google Scholar] [CrossRef] [PubMed]
  23. Ma, H.; Zhang, B.; Hu, Y.; Wang, J.; Liu, J.; Qin, R.; Lv, S.; Wang, S. Correlation analysis of intestinal redox state with the gut microbiota reveals the positive intervention of tea polyphenols on hyperlipidemia in high fat diet fed mice. J. Agric. Food Chem. 2019, 67, 7325–7335. [Google Scholar] [CrossRef] [PubMed]
  24. Yuan, Y.; Zheng, Y.; Zhou, J.; Geng, Y.; Zou, P.; Li, Y.; Zhang, C. Polyphenol-rich extracts from brown macroalgae Lessonia trabeculate attenuate hyperglycemia and modulate gut microbiota in high-fat diet and streptozotocin-induced diabetic rats. J. Agric. Food Chem. 2019, 67, 12472–12480. [Google Scholar] [CrossRef]
  25. Zhao, R.; Long, X.; Yang, J.; Du, L.; Zhang, X.; Li, J.; Hou, C. Pomegranate peel polyphenols reduce chronic low-grade inflammatory responses by modulating gut microbiota and decreasing colonic tissue damage in rats fed a high-fat diet. Food Funct. 2019, 10, 8273–8285. [Google Scholar] [CrossRef]
  26. Wang, P.; Li, D.; Ke, W.; Liang, D.; Hu, X.; Chen, F. Resveratrol-induced gut microbiota reduces obesity in high-fat diet-fed mice. Int. J. Obes. 2020, 44, 213–225. [Google Scholar] [CrossRef]
  27. Li, B.; Cheng, Z.; Sun, X.; Si, X.; Gong, E.; Wang, Y.; Tian, J.; Shu, C.; Ma, F.; Li, D.; et al. Lonicera caerulea L. polyphenols alleviate oxidative stress-induced intestinal environment imbalance and lipopolysaccharide-induced liver injury in HFD-fed rats by regulating the Nrf2/HO-1/NQO1 and MAPK pathways. Mol. Nutr. Food Res. 2020, 64, e1901315. [Google Scholar] [CrossRef]
  28. Wang, P.; Wang, J.; Li, D.; Ke, W.; Chen, F.; Hu, X. Targeting the gut microbiota with resveratrol: A demonstration of novel evidence for the management of hepatic steatosis. J. Nutr. Biochem. 2020, 81, 108363. [Google Scholar] [CrossRef]
  29. Wang, P.; Gao, J.; Ke, W.; Wang, J.; Li, D.; Liu, R.; Jia, Y.; Wang, X.; Chen, X.; Chen, F.; et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic. Biol. Med. 2020, 156, 83–98. [Google Scholar] [CrossRef]
  30. Du, P.; Zhou, J.; Zhang, L.; Zhang, J.; Li, N.; Zhao, C.; Tu, L.; Zheng, Y.; Xia, T.; Luo, J.; et al. GC × GC-MS analysis and hypolipidemic effects of polyphenol extracts from Shanxi-aged vinegar in rats under a high fat diet. Food Funct. 2020, 11, 7468–7480. [Google Scholar] [CrossRef]
  31. Hu, D.; Yang, W.; Mao, P.; Cheng, M. Combined amelioration of prebiotic resveratrol and probiotic Bifidobacteria on obesity and nonalcoholic fatty liver disease. Nutr. Cancer 2021, 73, 652–661. [Google Scholar] [CrossRef] [PubMed]
  32. Sinisgalli, C.; Vezza, T.; Diez-Echave, P.; Ostuni, A.; Faraone, I.; Hidalgo-Garcia, L.; Russo, D.; Armentano, M.F.; Garrido-Mesa, J.; Rodriguez-Cabezas, M.E.; et al. The beneficial effects of red sun-dried Capsicum annuum L. Cv Senise extract with antioxidant properties in experimental obesity are associated with modulation of the intestinal microbiota. Mol. Nutr. Food Res. 2021, 65, e2000812. [Google Scholar] [CrossRef] [PubMed]
  33. Da Fonseca Cardoso, L.M.; de Souza Monnerat, J.A.; de Medeiros Silva, I.W.S.; da Silva Ferreira Fiochi, R.; da Matta Alvarez Pimenta, N.; Mota, B.F.; Dolisnky, M.; do Carmo, F.L.; Barroso, S.G.; da Costa, C.A.S.; et al. Beverages rich in resveratrol and physical activity attenuate metabolic changes induced by high-fat diet. J. Am. Coll. Nutr. 2021, 40, 485–495. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, F.; Li, Y.L.; Zhang, X.; Wang, K.B.; Huang, J.A.; Liu, Z.H.; Zhu, M.Z. Polyphenols from Fu brick tea reduce obesity via modulation of gut microbiota and gut microbiota-related intestinal oxidative stress and barrier function. J. Agric. Food Chem. 2021, 69, 14530–14543. [Google Scholar] [CrossRef]
  35. Morbidelli, L. Polyphenol-based nutraceuticals for the control of angiogenesis: Analysis of the critical issues for human use. Pharmacol. Res. 2016, 111, 384–393. [Google Scholar] [CrossRef]
  36. Carrera-Quintanar, L.; López Roa, R.I.; Quintero-Fabián, S.; Sánchez-Sánchez, M.A.; Vizmanos, B.; Ortuño-Sahagún, D. Phytochemicals that influence gut microbiota as prophylactics and for the treatment of obesity and inflammatory diseases. Mediat. Inflamm. 2018, 2018, 9734845. [Google Scholar] [CrossRef]
  37. Tong, X.; Xu, J.; Lian, F.; Yu, X.; Zhao, Y.; Xu, L.; Zhang, M.; Zhao, X.; Shen, J.; Wu, S.; et al. Structural alteration of gut microbiota during the amelioration of human type 2 diabetes with hyperlipidemia by metformin and a traditional Chinese herbal frmula: A multicenter, randomized, open label clinical trial. mBio 2018, 9, e02392-17. [Google Scholar] [CrossRef] [Green Version]
  38. Wang, J.; Wang, P.; Li, D.; Hu, X.; Chen, F. Beneficial effects of ginger on prevention of obesity through modulation of gut microbiota in mice. Eur. J. Nutr. 2020, 59, 699–718. [Google Scholar] [CrossRef]
  39. Sellmann, C.; Priebs, J.; Landmann, M.; Degen, C.; Engstler, A.J.; Jin, C.J.; Gärttner, S.; Spruss, A.; Huber, O.; Bergheim, I. Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time. J. Nutr. Biochem. 2015, 26, 1183–1192. [Google Scholar] [CrossRef]
  40. Sellmann, C.; Degen, C.; Jin, C.J.; Nier, A.; Engstler, A.J.; Hasan Alkhatib, D.; De Bandt, J.P.; Bergheim, I. Oral arginine supplementation protects female mice from the onset of non-alcoholic steatohepatitis. Amino Acids 2017, 49, 1215–1225. [Google Scholar] [CrossRef] [Green Version]
  41. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Guo, X.; Xia, X.; Tang, R.; Zhou, J.; Zhao, H.; Wang, K. Development of a real-time PCR method for Firmicutes and Bacteroidetes in faeces and its application to quantify intestinal population of obese and lean pigs. Lett. Appl. Microbiol. 2008, 47, 367–373. [Google Scholar] [CrossRef] [PubMed]
  44. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  45. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [Green Version]
  46. Yang, C.S.; Prabhu, S.; Landau, J. Prevention of carcinogenesis by tea polyphenols. Drug Metab. Rev. 2001, 33, 237–253. [Google Scholar] [CrossRef]
  47. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  48. Liu, C.; Feng, X.; Li, Q.; Wang, Y.; Li, Q.; Hua, M. Adiponectin, TNF-α and inflammatory cytokines and risk of type 2 diabetes: A systematic review and meta-analysis. Cytokine 2016, 86, 100–109. [Google Scholar] [CrossRef]
  49. Kersten, S.; Desvergne, B.; Wahli, W. Roles of PPARs in health and disease. Nature 2000, 405, 421–424. [Google Scholar] [CrossRef]
  50. Yamauchi, T.; Kamon, J.; Minokoshi, Y.; Ito, Y.; Waki, H.; Uchida, S.; Yamashita, S.; Noda, M.; Kita, S.; Ueki, K.; et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med. 2002, 8, 1288–1295. [Google Scholar] [CrossRef]
  51. Tomas, E.; Tsao, T.S.; Saha, A.K.; Murrey, H.E.; Zhang, C.C.; Itani, S.I.; Lodish, H.F.; Ruderman, N.B. Enhanced muscle fat oxidation and glucose transport by ACRP30 globular domain: Acetyl-CoA carboxylase inhibition and AMP-activated protein kinase activation. Proc. Natl. Acad. Sci. USA 2002, 99, 16309–16313. [Google Scholar] [CrossRef] [Green Version]
  52. Kahn, B.B.; Alquier, T.; Carling, D.; Hardie, D.G. AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005, 1, 15–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Okada-Iwabu, M.; Yamauchi, T.; Iwabu, M.; Honma, T.; Hamagami, K.; Matsuda, K.; Yamaguchi, M.; Tanabe, H.; Kimura-Someya, T.; Shirouzu, M.; et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 2013, 503, 493–499. [Google Scholar] [CrossRef] [PubMed]
  54. Kostic, A.D.; Xavier, R.J.; Gevers, D. The microbiome in inflammatory bowel disease: Current status and the future ahead. Gastroenterology 2014, 146, 1489–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Danese, S.; Fiocchi, C.; Panés, J. Drug development in IBD: From novel target identification to early clinical trials. Gut 2016, 65, 1233–1239. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, Z.; Wu, X.; Cao, S.; Cromie, M.; Shen, Y.; Feng, Y.; Yang, H.; Li, L. Chlorogenic acid ameliorates experimental colitis by promoting growth of Akkermansia in mice. Nutrients 2017, 9, 677. [Google Scholar] [CrossRef] [Green Version]
  57. Ohno, M.; Nishida, A.; Sugitani, Y.; Nishino, K.; Inatomi, O.; Sugimoto, M.; Kawahara, M.; Andoh, A. Nanoparticle curcumin ameliorates experimental colitis via modulation of gut microbiota and induction of regulatory T cells. PLoS ONE 2017, 12, e0185999. [Google Scholar] [CrossRef] [Green Version]
  58. Scarano, A.; Butelli, E.; De Santis, S.; Cavalcanti, E.; Hill, L.; De Angelis, M.; Giovinazzo, G.; Chieppa, M.; Martin, C.; Santino, A. Combined dietary anthocyanins, flavonols, and stilbenoids alleviate inflammatory bowel disease symptoms in mice. Front. Nutr. 2017, 4, 75. [Google Scholar] [CrossRef] [Green Version]
  59. Wang, K.; Jin, X.; Li, Q.; Sawaya, A.; Le Leu, R.K.; Conlon, M.A.; Wu, L.; Hu, F. Propolis from different geographic origins decreases intestinal inflammation and Bacteroides spp. populations in a model of DSS-induced colitis. Mol. Nutr. Food Res. 2018, 62, e1800080. [Google Scholar] [CrossRef]
  60. Hong, Z.; Piao, M. Effect of quercetin monoglycosides on oxidative stress and gut microbiota diversity in mice with dextran sodium sulphate-induced colitis. BioMed Res. Int. 2018, 2018, 8343052. [Google Scholar] [CrossRef]
  61. Chen, S.; Zhao, H.; Cheng, N.; Cao, W. Rape bee pollen alleviates dextran sulfate sodium (DSS)-induced colitis by neutralizing IL-1β and regulating the gut microbiota in mice. Food Res. Int. 2019, 122, 241–251. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, H.; Cheng, N.; Zhou, W.; Chen, S.; Wang, Q.; Gao, H.; Xue, X.; Wu, L.; Cao, W. Honey polyphenols ameliorate DSS-induced ulcerative colitis via modulating gut microbiota in rats. Mol. Nutr. Food Res. 2019, 63, e1900638. [Google Scholar] [CrossRef] [PubMed]
  63. Hou, J.; Hu, M.; Zhang, L.; Gao, Y.; Ma, L.; Xu, Q. Dietary taxifolin protects against dextran sulfate sodium-induced colitis via NF-κB signaling, enhancing intestinal barrier and modulating gut microbiota. Front. Immunol. 2021, 11, 631809. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, Z.; Huang, S.; Li, T.; Li, N.; Han, D.; Zhang, B.; Xu, Z.Z.; Zhang, S.; Pang, J.; Wang, S.; et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome 2021, 9, 184. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, F.; Tsao, R.; Li, C.; Wang, X.; Zhang, H.; Jiang, L.; Sun, Y.; Xiong, H. Green pea (Pisum sativum L.) hull polyphenol extracts ameliorate DSS-induced colitis through Keap1/Nrf2 pathway and gut microbiota modulation. Foods 2021, 10, 2765. [Google Scholar] [CrossRef] [PubMed]
  66. Li, W.; Yang, H.; Zhao, Q.; Wang, X.; Zhang, J.; Zhao, X. Polyphenol-rich loquat fruit extract prevents fructose-induced nonalcoholic fatty liver disease by modulating glycometabolism, lipometabolism, oxidative stress, inflammation, intestinal barrier, and gut microbiota in mice. J. Agric. Food Chem. 2019, 67, 7726–7737. [Google Scholar] [CrossRef]
  67. Xie, K.; He, X.; Chen, K.; Sakao, K.; Hou, D.X. Ameliorative effects and molecular mechanisms of vine tea on western diet-induced NAFLD. Food Funct. 2020, 11, 5976–5991. [Google Scholar] [CrossRef]
  68. Xia, T.; Duan, W.; Zhang, Z.; Li, S.; Zhao, Y.; Geng, B.; Zheng, Y.; Yu, J.; Wang, M. Polyphenol-rich vinegar extract regulates intestinal microbiota and immunity and prevents alcohol-induced inflammation in mice. Food Res. Int. 2021, 140, 110064. [Google Scholar] [CrossRef]
  69. Zhao, L.; Mehmood, A.; Soliman, M.M.; Iftikhar, A.; Iftikhar, M.; Aboelenin, S.M.; Wang, C. Protective effects of ellagic acid against alcoholic lver disease in mice. Front. Nutr. 2021, 8, 744520. [Google Scholar] [CrossRef]
  70. Kong, Y.; Yan, T.; Tong, Y.; Deng, H.; Tan, C.; Wan, M.; Wang, M.; Meng, X.; Wang, Y. Gut microbiota modulation by polyphenols from Aronia melanocarpa of LPS-induced liver diseases in rats. J. Agric. Food Chem. 2021, 69, 3312–3325. [Google Scholar] [CrossRef]
  71. Zhang, X.; Shi, L.; Chen, R.; Zhao, Y.; Ren, D.; Yang, X. Chlorogenic acid inhibits trimethylamine-N-oxide formation and remodels intestinal microbiota to alleviate liver dysfunction in high L-carnitine feeding mice. Food Funct. 2021, 12, 10500–10511. [Google Scholar] [CrossRef]
  72. Mezhibovsky, E.; Knowles, K.A.; He, Q.; Sui, K.; Tveter, K.M.; Duran, R.M.; Roopchand, D.E. Grape polyphenols attenuate diet-induced obesity and hepatic steatosis in mice in association with reduced butyrate and increased markers of intestinal carbohydrate oxidation. Front. Nutr. 2021, 8, 675267. [Google Scholar] [CrossRef] [PubMed]
  73. Li, X.W.; Chen, H.P.; He, Y.Y.; Chen, W.L.; Chen, J.W.; Gao, L.; Hu, H.Y.; Wang, J. Effects of rich-polyphenols extract of Dendrobium loddigesii on anti-diabetic, anti-inflammatory, anti-oxidant, and gut microbiota modulation in db/db mice. Molecules 2018, 23, 3245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wang, N.; Ma, Y.; Liu, Z.; Liu, L.; Yang, K.; Wei, Y.; Liu, Y.; Chen, X.; Sun, X.; Wen, D. Hydroxytyrosol prevents PM(2.5)-induced adiposity and insulin resistance by restraining oxidative stress related NF-κB pathway and modulation of gut microbiota in a murine model. Free Radic. Biol. Med. 2019, 141, 393–407. [Google Scholar] [CrossRef] [PubMed]
  75. Guirro, M.; Gual-Grau, A.; Gibert-Ramos, A.; Alcaide-Hidalgo, J.M.; Canela, N.; Arola, L.; Mayneris-Perxachs, J. Metabolomics elucidates dose-dependent molecular beneficial effects of hesperidin supplementation in rats fed an obesogenic diet. Antioxidants 2020, 9, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Yao, Y.; Liu, T.; Yin, L.; Man, S.; Ye, S.; Ma, L. Polyphenol-rich extract from Litchi chinensis seeds alleviates hypertension-induced renal damage in rats. J. Agric. Food Chem. 2021, 69, 2138–2148. [Google Scholar] [CrossRef] [PubMed]
  77. Wu, X.; Zhang, N.; Kan, J.; Tang, S.; Sun, R.; Wang, Z.; Chen, M.; Liu, J.; Jin, C. Polyphenols from Arctium lappa L ameliorate doxorubicin-induced heart failure and improve gut microbiota composition in mice. J. Food Biochem. 2021, 46, e13731. [Google Scholar] [CrossRef]
  78. Xu, Y.; Cao, X.; Zhao, H.; Yang, E.; Wang, Y.; Cheng, N.; Cao, W. Impact of Camellia japonica bee pollen polyphenols on hyperuricemia and gut microbiota in potassium oxonate-induced mice. Nutrients 2021, 13, 2665. [Google Scholar] [CrossRef] [PubMed]
  79. Da Silva-Maia, J.K.; Batista, A.G.; Correa, L.C.; Lima, G.C.; Bogusz Junior, S.; Maróstica Junior, M.R. Aqueous extract of berry (Plinia jaboticaba) byproduct modulates gut microbiota and maintains the balance on antioxidant defense system in rats. J. Food Biochem. 2019, 43, e12705. [Google Scholar] [CrossRef]
  80. Alim, A.; Li, T.; Nisar, T.; Ren, D.; Liu, Y.; Yang, X. Consumption of two whole kiwifruit (Actinide chinensis) per day improves lipid homeostasis, fatty acid metabolism and gut microbiota in healthy rats. Int. J. Biol. Macromol. 2020, 156, 186–195. [Google Scholar] [CrossRef]
  81. Yin, J.; Ren, W.; Wei, B.; Huang, H.; Li, M.; Wu, X.; Wang, A.; Xiao, Z.; Shen, J.; Zhao, Y.; et al. Characterization of chemical composition and prebiotic effect of a dietary medicinal plant Penthorum chinense Pursh. Food Chem. 2020, 319, 126568. [Google Scholar] [CrossRef] [PubMed]
  82. Peng, Y.; Yan, Y.; Wan, P.; Dong, W.; Huang, K.; Ran, L.; Mi, J.; Lu, L.; Zeng, X.; Cao, Y. Effects of long-term intake of anthocyanins from Lycium ruthenicum Murray on the organism health and gut microbiota in vivo. Food Res. Int. 2020, 130, 108952. [Google Scholar] [CrossRef] [PubMed]
  83. Likotrafiti, E.; Rhoades, J. Probiotics, prebiotics, synbiotics, and foodborne illness. In Bioactive Foods in Health Promotion: Probiotics, Prebiotics, and Synbiotics; Watson, R.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2015; pp. 469–476. [Google Scholar]
  84. Hollman, P.C.; Cassidy, A.; Comte, B.; Heinonen, M.; Richelle, M.; Richling, E.; Serafini, M.; Scalbert, A.; Sies, H.; Vidry, S. The biological relevance of direct antioxidant effects of polyphenols for cardiovascular health in humans is not established. J. Nutr. 2011, 141, 989s–1009s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Croft, K.D. Dietary polyphenols: Antioxidants or not? Arch. Biochem. Biophys. 2016, 595, 120–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Arakawa, H.; Maeda, M.; Okubo, S.; Shimamura, T. Role of hydrogen peroxide in bactericidal action of catechin. Biol. Pharm. Bull. 2004, 27, 277–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Khan, H.Y.; Zubair, H.; Faisal, M.; Ullah, M.F.; Farhan, M.; Sarkar, F.H.; Ahmad, A.; Hadi, S.M. Plant polyphenol induced cell death in human cancer cells involves mobilization of intracellular copper ions and reactive oxygen species generation: A mechanism for cancer chemopreventive action. Mol. Nutr. Food Res. 2014, 58, 437–446. [Google Scholar] [CrossRef]
  88. Leonard, M.O.; Kieran, N.E.; Howell, K.; Burne, M.J.; Varadarajan, R.; Dhakshinamoorthy, S.; Porter, A.G.; O’Farrelly, C.; Rabb, H.; Taylor, C.T. Reoxygenation-specific activation of the antioxidant transcription factor Nrf2 mediates cytoprotective gene expression in ischemia-reperfusion injury. FASEB J. 2006, 20, 2624–2626. [Google Scholar] [CrossRef]
  89. Shelton, L.M.; Park, B.K.; Copple, I.M. Role of Nrf2 in protection against acute kidney injury. Kidney Int. 2013, 84, 1090–1095. [Google Scholar] [CrossRef]
  90. Smith, R.E.; Tran, K.; Smith, C.C.; McDonald, M.; Shejwalkar, P.; Hara, K. The role of the Nrf2/ARE antioxidant system in preventing cardiovascular diseases. Diseases 2016, 4, 34. [Google Scholar] [CrossRef]
  91. Milutinović, M.; Dimitrijević-Branković, S.; Rajilić-Stojanović, M. Plant Extracts Rich in Polyphenols as Potent Modulators in the Growth of Probiotic and Pathogenic Intestinal Microorganisms. Front. Nutr. 2021, 8, 688843. [Google Scholar] [CrossRef]
  92. Cressey, D. UK funders demand strong statistics for animal studies. Nature 2015, 520, 271–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Collins, F.S.; Tabak, L.A. Policy: NIH plans to enhance reproducibility. Nature 2014, 505, 612–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mahowald, M.A.; Rey, F.E.; Seedorf, H.; Turnbaugh, P.J.; Fulton, R.S.; Wollam, A.; Shah, N.; Wang, C.; Magrini, V.; Wilson, R.K.; et al. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc. Natl. Acad. Sci. USA 2009, 106, 5859–5864. [Google Scholar] [CrossRef] [Green Version]
  95. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  96. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef] [PubMed]
  97. Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef] [Green Version]
  98. Jin, G.; Asou, Y.; Ishiyama, K.; Okawa, A.; Kanno, T.; Niwano, Y. Proanthocyanidin-Rich Grape Seed Extract Modulates Intestinal Microbiota in Ovariectomized Mice. J. Food Sci. 2018, 83, 1149–1152. [Google Scholar] [CrossRef] [PubMed]
  99. Yuan, Y.; Yang, J.; Zhuge, A.; Li, L.; Ni, S. Gut microbiota modulates osteoclast glutathione synthesis and mitochondrial biogenesis in mice subjected to ovariectomy. Cell Prolif. 2022, 55, e13194. [Google Scholar] [CrossRef]
  100. Rowland, I.; Faughnan, M.; Hoey, L.; Wähälä, K.; Williamson, G.; Cassidy, A. Bioavailability of phyto-oestrogens. Br. J. Nutr. 2003, 89 (Suppl. 1), S45–S58. [Google Scholar] [CrossRef]
  101. Duda-Chodak, A.; Tarko, T.; Satora, P.; Sroka, P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: A review. Eur. J. Nutr. 2015, 54, 325–341. [Google Scholar] [CrossRef] [Green Version]
  102. Braune, A.; Blaut, M. Bacterial species involved in the conversion of dietary flavonoids in the human gut. Gut Microbes 2016, 7, 216–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yokoyama, S.; Suzuki, T. Isolation and characterization of a novel equol-producing bacterium from human feces. Biosci. Biotechnol. Biochem. 2008, 72, 2660–2666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Matthies, A.; Blaut, M.; Braune, A. Isolation of a human intestinal bacterium capable of daidzein and genistein conversion. Appl. Environ. Microbiol. 2009, 75, 1740–1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Setchell, K.D.; Clerici, C. Equol: History, chemistry, and formation. J. Nutr. 2010, 140, 1355s–1362s. [Google Scholar] [CrossRef] [Green Version]
  106. Setchell, K.D.; Clerici, C.; Lephart, E.D.; Cole, S.J.; Heenan, C.; Castellani, D.; Wolfe, B.E.; Nechemias-Zimmer, L.; Brown, N.M.; Lund, T.D.; et al. S-equol, a potent ligand for estrogen receptor beta, is the exclusive enantiomeric form of the soy isoflavone metabolite produced by human intestinal bacterial flora. Am. J. Clin. Nutr. 2005, 81, 1072–1079. [Google Scholar] [CrossRef] [Green Version]
  107. Duda-Chodak, A. The inhibitory effect of polyphenols on human gut microbiota. J. Physiol. Pharmacol. 2012, 63, 497–503. [Google Scholar]
Figure 1. The proposed mechanism underlying the beneficial effects of polyphenols on metabolic disorders in high-fat diet (HFD)-fed murine models. LPS: lipopolysaccharide; ROS: reactive oxygen species; F/B ratio: Firmicutes/Bacteroidetes ratio; AMPK: AMP-activated protein kinase; PPAR-α: peroxisome proliferator-activated receptor-α.
Figure 1. The proposed mechanism underlying the beneficial effects of polyphenols on metabolic disorders in high-fat diet (HFD)-fed murine models. LPS: lipopolysaccharide; ROS: reactive oxygen species; F/B ratio: Firmicutes/Bacteroidetes ratio; AMPK: AMP-activated protein kinase; PPAR-α: peroxisome proliferator-activated receptor-α.
Cimb 44 00091 g001
Figure 2. The proposed mechanism underlying the beneficial effects of polyphenols on dextran sulfate sodium (DSS)-induced colitis in murine models. SOD: superoxide dismutase; CAT: catalase; GSH-px: glutathione peroxidase; IL: interleukin; TNF: tumor necrosis factor; SCF: short-chain fatty acid.
Figure 2. The proposed mechanism underlying the beneficial effects of polyphenols on dextran sulfate sodium (DSS)-induced colitis in murine models. SOD: superoxide dismutase; CAT: catalase; GSH-px: glutathione peroxidase; IL: interleukin; TNF: tumor necrosis factor; SCF: short-chain fatty acid.
Cimb 44 00091 g002
Figure 3. The proposed mechanism underlying the beneficial effects of polyphenols on murine liver injuries induced by various factors, except for a high-fat diet. LPS: lipopolysaccharide; ROS: reactive oxygen species; F/B ratio: Firmicutes/Bacteroidetes ratio; TLR: toll-like receptor; NF: nuclear factor; TNF: tumor necrosis factor; IL: interleukin; MCP: monocyte chemotactic protein.
Figure 3. The proposed mechanism underlying the beneficial effects of polyphenols on murine liver injuries induced by various factors, except for a high-fat diet. LPS: lipopolysaccharide; ROS: reactive oxygen species; F/B ratio: Firmicutes/Bacteroidetes ratio; TLR: toll-like receptor; NF: nuclear factor; TNF: tumor necrosis factor; IL: interleukin; MCP: monocyte chemotactic protein.
Cimb 44 00091 g003
Figure 4. Schematic illustration of the possible involvement of prooxidative potential of polyphenols in intestinal barrier function. ROS: reactive oxygen species; Nrf2: nuclear factor E2-related factor 2; HO-1: heme oxygenase 1; NQO-1: NAD(P)H quinine oxidoreductase 1; LPS: lipopolysaccharide.
Figure 4. Schematic illustration of the possible involvement of prooxidative potential of polyphenols in intestinal barrier function. ROS: reactive oxygen species; Nrf2: nuclear factor E2-related factor 2; HO-1: heme oxygenase 1; NQO-1: NAD(P)H quinine oxidoreductase 1; LPS: lipopolysaccharide.
Cimb 44 00091 g004
Table 1. Beneficial effects of polyphenols in metabolic disorders in relation to the gut microbiota in high-fat diet (HFD)-fed murine models.
Table 1. Beneficial effects of polyphenols in metabolic disorders in relation to the gut microbiota in high-fat diet (HFD)-fed murine models.
Reference, Publication Year, Animal Species, Polyphenol(s), and DosageMajor Physiological Issues ImprovedMode of Action
Antioxidative and Anti-Inflammatory ActionGut Microbiota Modulation
[12] 2013
Mice, polyphenol-rich pomegranate peel extract (PPE), p.o. in drinking water containing 0.2% PPE (average consumption of 6 mg/d per mouse) for 4 weeks
Reduced serum cholesterol (total and LDL) levels and alleviated tissue (colon and visceral adipose tissue) inflammationPromoted the growth of gut bacteria, in particular, Bifidobacterium spp.
[13] 2014
Rats, p.o. as instant caffeinated coffee at a concentration of 20 g/L for 8 weeks (HFD was given for 10 weeks)
Reduced weight gain, adiposity, liver triglycerides, and energy intakeDecreased the Firmicutes/Bacteroidetes ratio
[14] 2015
Mice, high fat/high sucrose diet (HFHSD), polyphenol-rich cranberry extract, p.o. at 200 mg/kg/day for 8 weeks
Reduced visceral obesity and improved insulin sensitivityAmeliorated oxidative stress and inflammation in the jejunum and reduced circulating LPSIncreased the relative abundance of Akkermansia spp.
[15] 2016
Mice, extractable polyphenol-rich fraction of table grapes (EP), p.o. with diet containing 1.1 g EP/kg for 16 weeks
Reduced white adipose tissue mass and improved glucose tolerancePartially restored the HFD-mediated reduction in diversity
[16] 2016
Mice, green tea polyphenols (GTP), p.o. with a diet containing 0.05, 0.2, and 0.8% GTP for 8 weeks
Reduced obesity, and improved hepatic steatosisPartially restored the HFD-mediated reduction in diversity
[17] 2017
Rats, a combination of quercetin (Q) and resveratrol (R), p.o. at 30 mg Q + 15 mg R/kg/day for 8 weeks
Reduced obesityAttenuated serum inflammatory markersDecreased the Firmicutes/Bacteroidetes ratio
[18] 2017
Mice, p.o. polyphenol- and caffeine-rich post-fermented Pu-erh tea, p.o. at 750 mg/kg/day for 12 weeks
Improved glucose and lipid metabolism disorderAttenuated expression of inflammation genes in the proximal colon, reduced circulating LPS, and restored gut barrier integrityRestored the HFD-induced gut microbial community structural shift
[19] 2018
Mice, polyphenol-rich cinnamon bark, or grape pomace extract (CBE or PBE), p.o. with a diet containing 0.2% CBE or and 0.8% PBE for 8 weeks
Reduced fat mass gain and adipose tissue inflammation, and ameliorated liver steatosisReduced adipose tissue inflammation, and improved gut barrier functionDecreased abundance of Desulfovibrio and Lactococcus at the genus level
[20] 2018
Mice, Lonicera caerulea L. berry polyphenols (LCBP), p.o. with diet containing 0.5% and 1% LCBP for 45 days
Improved hepatic steatosisAttenuated serum inflammatory markers, and decreased LPS level in serum and liverDecreased the Firmicutes/Bacteroidetes ratio
[21] 2019
Rats, resveratrol (RSV) and sinapic acid (SA), p.o. at 400 mg RSV/kg/day, 200 mg SA/kg/day, or a combination of RSV and SA for 8 weeks
Reduced fasting blood glucose levels and increased HDL-C levels by RSVDecreased ROS and MDA levels in the colon, and increased total antioxidant capacity in the liver by SACombination of RSV and SA: Improved proportion of butyrate producer Blautia and Dorea from the Lachaospiraceae family and inhibited growth of bacterial species associated with diseases and inflammation, such as Bacteroides and Desulfovibrionaceae sp.
[22] 2019
Rats, sinapine (a rapeseed polyphenol), p.o. at 500 mg/kg/day for 12 weeks
Ameliorated NAFLD, reduced body weight and decreased TG and LDL-C levels.Suppressed expression of NF-κB and TNF-α in the intestine and enhanced expression of IRS-1 in the adipose tissueDecreased Firmicutes/Bacteroidetes ratio and increased abundance of probiotics, along with SCFA-mediated upregulation of G protein-coupled receptor 43 (GPR43) to inhibit the expression of inflammatory factors
[23] 2019
Mice, tea polyphenols (TPs) including EGCG, EGC, and ECG, p.o. at 100, 200, and 400 mg/kg/day for 12 weeks
Ameliorated hyperlipidemia, enhanced expression levels of hepatic lipid metabolism genes, and modulated gut microbiotaMaintenance of intestinal redox state by TPsDecreased gut microbiota diversity and relative abundance of Proteobacteria, a source of LPS, possibly due to the antimicrobial activity of TPs
[24] 2019
Rats (treated with HFD + STZ), polyphenol-rich extracts from brown macroalga Lessonia trabeculata containing phlorotannin derivatives, phenolic acid derivatives, and gallocatechin derivatives, p.o. at 200 mg/kg/day for 4 weeks
Lowered fasting blood glucose and insulin levels, as well as better serum lipid profiles and antioxidant stress parametersIncreased response of antioxidant defense systems (e.g., CAT, SOD, and GSH in the liver) to oxidative stressA positive effect on regulating the dysbiosis of the microbial ecology in diabetic rats
[25] 2019
Mice, pomegranate peel polyphenols including gallic acid, punicalagin, and catechin, p.o. at 150 and 300 mg/kg/day for 12 weeks
Alleviated obesity, decreased circulating proinflammatory cytokines, colonic tissue damage, and enhanced protein expression in the colonic tight junctionImproved oxidative damage and inflammation of the intestinal tissues, thereby reversing the reduced levels of tight junction proteinsNormalized the HFD-induced gut microbiota imbalance by increasing the abundance of beneficial bacteria in the colon
[26] 2020
Mice [fecal microbiota transplantation (FMT) to HFD-fed mice], resveratrol (RSV), p.o. at 300 mg/kg/day for 16 weeks followed by transplantation of the RSV-microbiota to HFD-fed mice (HFD-RSVT) to explore the function of the microbiota
HFD-RSVT decreased weight gain and increased insulin sensitivityHFD-RSVT reduced the production of ROS and MDA in the intestineA remarkable alteration in the composition of gut microbiota in mice treated with RSV, for example, enrichment of Bacteroides, Lachnospiraceae_NK4A136_ group, Blautia, Lachnoclostridium, Parabacteroides, and Ruminiclostridium_9, collectively referred to as RSV-microbiota
[27] 2020
Rats, Lonicera caerulea L. polyphenols containing anthocyanins, phenolic acids, and flavonoids, p.o. at 250 mg/kg/day for 8 weeks
Ameliorated intestinal permeability and intestinal inflammation; alleviated LPS-induced liver injuryAmeliorated intestinal oxidative stress damage (through regulation of the Nrf2/HO-1/NQO1 pathway)Increased relative abundance of Bacteroidetes and Tenericutes and decreased relative abundance of Proteobacteria at the phylum level
[28] 2020
Mice (FMT from HFD-fed mice to HFD-fed mice), resveratrol (RSV), p.o. at 300 mg/kg/day for 16 weeks followed by transplantation of the HFDR-microbiota to HFD-fed mice (HFD-RSVT)
Alleviated NAFLD; ameliorated liver oxidative stress by HFD + RSV-microbiota treatmentHFD + RSV-microbiota treatment prevented HFD-induced production of ROS and improved antioxidant defense mechanisms (SOD and GSH levels)The RSV-induced gut microbiota characterized by a decreased abundance of harmful bacteria, including Desulfovibrio, Lachnospiraceae_NK4A316_group, and Alistipes, as well as an increased abundance of SCFA-producing bacteria, such as Allobaculum, Bacteroides, and Blautia
[29] 2020
Mice, resveratrol (RSV), p.o. at 300 mg/kg/day for 16 weeks
Improved obesityA two-part anti-obesity mechanism of RSV through the gut microbiota was proposed:(1) improved composition and function of the gut microbiota as well as the intestinal oxidative state; (2) 3-hydroxyphenylpropionic acid and 4-hydroxyphenylacetic acid (biotransformed from RSV by the gut microbiota), which may be responsible for the beneficial effects of RSV
[30] 2020
Rats, polyphenol extracts from Shanxi-aged vinegar containing at least 41 polyphenols (including 18 phenolic acids), p.o. at 4, 8, and 16 mg/kg/day for 4 weeks
Improved hyperlipidemiaImproved inflammatory stress- and oxidative stress-related indicatorsDecreased the Firmicutes/Bacteroidetes ratio; increased the diversity of microorganisms
[31] 2021
Mice, resveratrol (RSV) with probiotic Bifidobacteria, p.o. at 100 mg RSV/kg/day and probiotic Bifidobacteria for 3 weeks, starting the fifth week of HFD feeding
Coadministration of B. longum and RSV alleviated obesity and NAFLDThe combination of B. longum and RSV exerted an inhibitory effect on inflammatory cytokines and increased the levels of antioxidants, including SOD and GSH, and decreased the levels of MDARSV acted as an excellent prebiotic because most orally administered RSV is located in the bowel lumen
[32] 2021
Mice, Capsicum annuum L. ‘Senise’ extract (CAE) containing polyphenols, lycopene, and capsinoid derivatives, p.o. at 1, 10, and 25 mg/kg/day for 6 weeks
Promoted weight loss and improved plasma markers related to glucose and lipid metabolismReduced the expression of proinflammatory cytokines possibly due to the antioxidant property of CAEDecreased the Firmicutes/Bacteroidetes ratio
[33] 2021
Rats, polyphenol-rich whole red grape juice, p.o. at 10 mL/day + physical training for 60 days
Lowered the concentration of IL-6 and TBARSReduced oxidative stress by activating the body’s antioxidant system, preventing the action of free radicals, and consequently, reducing the expression of inflammatory cytokinesThe juice consumption beneficially modulated the gut microbiota
[34] 2021
Rats, Fu brick tea polyphenols, including EGCG, EGC, and ECG, p.o. at 100 mg/kg for 12 weeks
Improved the intestinal oxidative stress and intestinal barrier function, including intestinal inflammation and the integrity of the intestinal barrierAttenuated HFD-induced gut microbiota dysbiosis, characterized by increased phylogenetic diversity and decreased Firmicutes/Bacteroidetes ratio
p.o., per os; ROS, reactive oxygen species; MDA, malondialdehyde; NAFLD, nonalcoholic fatty liver disease; NF-κB, nuclear factor-kappa B; TG, triglyceride; LDL-C, low-density lipoprotein cholesterol; IRS-1, insulin receptor substrate 1; EGCG, epigallocatechin gallate; EGC, epigallocatechin; ECG, epicatechin gallate; SCFA, short-chain fatty acid; LPS, lipopolysaccharide; STZ, streptozotocin; CAT, catalase; SOD, superoxide dismutase; GSH, glutathione; Nrf2, nuclear factor (erythroid-derived 2)-like 2; HO-1, heme oxygenase 1; NQO1, quinone oxidoreductase 1; FMT, fecal microbiota transplantation; IL-6, interleukin-6; TBARS, thiobarbituric acid-reactive substances; ―, not clearly described.
Table 2. Beneficial effects of polyphenols on dextran sulfate sodium (DSS)-induced colitis in relation to the gut microbiota in murine models.
Table 2. Beneficial effects of polyphenols on dextran sulfate sodium (DSS)-induced colitis in relation to the gut microbiota in murine models.
Reference, Publication Year, Animal Species, Polyphenol(s), and DosageMajor Physiological Issues ImprovedMode of Action
Antioxidative and Anti-Inflammatory ActionGut Microbiota Modulation
[56] 2017
Mice, chlorogenic acid (CA), p.o. with drinking water containing 1 mM CA for 15 days; 2.5% DSS was given during the last 8 days with CA
Ameliorated DSS-induced colitis and improved mucosal damageSuppressed the active NF-κB signaling pathway in the colonDecreased the Firmicutes/Bacteroidetes ratio and increased the relative abundance of Akkermansia
[57] 2017
Mice, nanoparticle curcumin (NC), p.o. with diet containing 0.2% NC for 18 days; 3% DSS was given from day 8 to day 18
Ameliorated DSS-induced colitis and improved mucosal permeabilitySuppressed NF-κB activation in colonic epithelial cellsIncreased the abundance of butyrate-producing bacteria and fecal butyrate levels
[58] 2018
Mice, bronze tomato extract (BTE) rich in anthocyanins, flavonols, and stilbenoids, p.o. with diet containing 1% BTE for 2 weeks; 1% DSS was given from day 14 to day 29
Ameliorated DSS-induced colitisSuppressed LPS-mediated production of pro-inflammatory cytokinesDecreased the Firmicutes/Bacteroidetes ratio
[59] 2018
Rats, polyphenol-rich Chinese propolis (CP) or Brazilian propolis (BP), p.o. at 300 mg/kg/day for 17 days; 3% DSS was given from day 7 for 1 week
Ameliorated DSS-induced colitisAlleviated the intestinal oxidative status, and suppressed inflammatory gene expression in the distal colonReduced populations of Bacteroides spp.
[60] 2018
Mice, quercetin aglycone (Q) or quercetin aglycone with monoglycosides (Q + MQ), p.o. with diet containing 0.21% Q or 0.35% Q + MQ for 7 days; 3% DSS was given from day 8 for 1 week
Ameliorated DSS-induced colitisSuppressed oxidative stress indicated by MPO, GSH, and MDAAmeliorated reduced Firmicutes population and increased Proteobacteria population by DSS
[61] 2019
Mice, rape bee pollen extract rich in polyphenols including kaempferol, sinapic acid, and rosmarinic acid, p.o. at 10.6 and 21.2 g/kg/day for 15 days; 3% DSS was given from day 8 to day 12
Ameliorated DSS-induced colitisAttenuated oxidative stress and downregulated the expression of inflammatory cytokines such as IL-1βReduced the abundance of Allobaculum and Bacteroides, and increased the abundance of Lactobacillus
[62] 2019
Rats, honey polyphenols including caffeic acid, chlorogenic acid, and rutin, p.o. at 10.5 mg/kg twice daily for 7 days; 3% DSS was given from day 0 to day 5
Improved DSS-induced colonic apoptosis and reduced the expression of inflammatory cytokines in the colonImproved the levels of SOD, GSH-Px, NO, and MPO; downregulated the expression of IL-1β, IL-6, TNF-α, and IFN-γ genes, and upregulated the expression of IκB-α geneReduced the population of Bacteroides, Corynebacterium, and Proteus species at the genus level
[63] 2021
Mice, taxifolin, p.o. at 100 mg/kg/day for 7 days; from day 8, mice received 3% DSS for 7 days
Prevented DSS-induced colitisInhibited the secretion of proinflammatory cytokines, and increased the secretion of IL-10, secretory IgA, SOD, and immunoglobulins; increased the expression of intestinal tight junction proteinsRestored the microbiota composition in the colon, including the decrease in the abundance of Bacteroidetes and the Bacteroidetes/Firmicutes ratio at the phylum level
[64] 2021
Mice, Exp. I, epigallocatechin-3-gallate (EGCG), p.o. or rectal administration at 50 mg/kg/day for 3 days starting after supplementation with 2.5% DSS for 7 days;
Exp. II, EGCG, p.o. at 50 mg/kg/day for 3 weeks with 2.5% DSS for the last 6 days;
Exp. III, 2.5% DSS for 7 days followed by FMT for 3 days
Exp. I: oral, but not rectal, EGCG alleviated DSS-induced colitis;
Exp. II: oral EGCG prevented DSS-induced colitis;
Exp. III: FMT alleviated DSS-induced colitis
Oral, but not rectal, EGCG attenuated oxidative stress, and exerted an anti-inflammatory effect along with enhanced integrity of the colonic barrier; oral EGCG pre-supplementation induced beneficial outcomes similar to those achieved with oral EGCG administrationAttenuation of colitis by oral EGCG suggests an intimate involvement of SCFA-producing bacteria, of the genus Akkermansia.
[65] 2021
Mice, green pea (Pisum sativum L.) hull polyphenol extracts containing quercetin and its derivatives, kaempferol trihexanside, and catechin and its derivatives, p.o. at 100 and 600 mg/kg for 14 days; 3% DSS was given for 7 weeks from day 8
Alleviated DSS induced colitisRestored oxidative balance, and regulated inflammatory factors along with repaired colonic functionIncreased the Firmicutes/Bacteroidetes ratio, promoted the growth of Lactobacillaceae and Lachnospiraceae, and improved the level of SCFAs
p.o., per os; IL-1β, interleukin-1β; SOD, superoxide dismutase; GSH-Px, glutathione peroxidase; NO, nitric oxide; IL-6, interleukin-6; TNF-α, tumor necrosis factor-α; IFN-γ, interferon-γ; IκB-α, NF-κB inhibitor-α; IL-10, interleukin-10; IgA, immunoglobulin A; FMT, fecal microbiota transplantation; SCFA, short-chain fatty acids.
Table 3. Beneficial effects of polyphenols on metabolic disorders not associated with a high-fat diet (HFD) or dextran sulfate sodium (DSS) in relation to the gut microbiota in murine models.
Table 3. Beneficial effects of polyphenols on metabolic disorders not associated with a high-fat diet (HFD) or dextran sulfate sodium (DSS) in relation to the gut microbiota in murine models.
Reference, Publication Year, Animal Model of the Disorder, Polyphenol(s), and DosageMajor Physiological Issues ImprovedMode of Action
Antioxidative and Anti-Inflammatory ActionGut Microbiota Modulation
[66] 2019
Mice with fructose-induced NAFLD, loquat fruit extract rich in polyphenols including chlorogenic acid, cryptochlorogenic acid, and oleanolic acid, p.o. at 25 and 50 mg/kg/day for 8 weeks
Prevented fructose-induced NAFLD with mitigation of abnormal body weight and improved lipid metabolismMitigated oxidative stress and inflammation; reduced the endotoxin content and improved fructose-induced breakage of the intestinal barrierMaintained normal Firmicutes/Bacteroidetes ratio
[67] 2020
Mice with western diet-induced NAFLD, vine tea polyphenol extracted from Ampelopsis grossedentata, p.o. in drinking water (0.5, 1, and 2%) for 12 weeks
Decreased the serum levels of cholesterol and triglycerides, and reduced the accumulation of hepatic lipid dropletsActivated Nrf2-mediated expression of hemeoxygenase-1 and quinone oxidoreductase, and reduced hepatic TBARS levels to prevent hepatic oxidative stressIncreased the relative abundance of Akkermansia, and reduced the Firmicutes/Bacteroidetes ratio
[68] 2021
Mice with alcohol-induced liver inflammation, Zhenjiang aromatic vinegar (a traditional fermented food in China) rich in polyphenols including catechin, p-hydroxybenzoic acid, and vanillic acid, p.o. at 200 and 800 mg/kg/day for 30 days; EtOH was given p.o. after 32 h, every day
Protected against alcohol-induced liver injuryInhibited oxidative stress (reduced the levels of ROS, iNOS, MDA, 4-HNE, and 8-OHdG) and LPS-mediated inflammationModulated the composition of the gut microbiota and improved gut immunity and intestinal homeostasis;
decreased the Firmicutes/Bacteroidetes ratio
[69] 2021
Mice with alcoholic liver disease, ellagic acid, p.o. at 50 and 100 mg/kg/day for 4 weeks to mice that received 15% alcohol
Alleviated alcohol-induced liver injuryAlleviated hepatic antioxidant activities (GSH-Px, CAT, MDA, SOD, and GSH), and proinflammatory cytokines levels (IL-6, IL-1β, and TNF-α)Improved the alcohol-induced gut microbiota dysbiosis; restored the relative abundance of microbiota, such as Firmicutes, Verrucomicrobia, Actinobacteria, Bacteroidetes, and Proteobacteria at the phylum level
[70] 2021
Rats with LPS-induced liver disease, Aronia melanocarpa polyphenols containing anthocyanins, flavonols, and hydroxycinnamates, p.o. at 50, 100, and 200 mg/kg/day with LPS (p.o. at 200 μg/kg/day) for 4 weeks
Alleviated the degree of LPS-induced liver diseaseAlleviated LPS-induced oxidative stress in the liver (reduced ROS and increased GSH levels).Modulated the composition of the gut microbiota and improved the intestinal barrier function. At the phylum level, the enrichment of Verrucomicrobia microflora was alleviated and the abundance of Actinobacteria was decreased
[71] 2021
Mice with L-carnitine-induced liver injury, chlorogenic acid, p.o. at 200 and 400 mg/kg/day for 12 weeks with 3% L-carnitine in drinking water
Prevented L-carnitine-induced liver injuryInhibited free radical production and improved the antioxidant defense system;
inhibited the inflammatory reaction (i.e., IL-1, IL-6, TNF-α, and TNF-β levels).
Inhibited the L-carnitine-induced increase in the abundance of Firmicutes and Proteobacteria, and promoted Bacteroidetes at the phylum level
[72] 2021
Mice with obesity and hepatic steatosis induced by a western diet (WD), low in fiber but high in fats and sugars, p.o. as the WD supplemented with 1% grape polyphenols rich in B-type proanthocyanidins
Higher lean mass and lower fat mass, body weight, and hepatic steatosisReduced the intestinal oxidative stressIncreased the abundance of Akkermansia muciniphila, a gut microbe reported to increase energy expenditure
[73] 2018
Diabetic db/db mice, polyphenol-rich extract of Dendrobium loddigesi, p.o. at 50 and 100 mg/kg/day for 8 weeks
Decreased blood glucose, LDL-C, and body weightInhibited oxidative stress (reduced MDA and increased SOD, CAT, and GSH) in liver and kidney, attenuated serum inflammatory markers (IL-6 and TNF-α)Decreased the Firmicutes/Bacteroidetes ratio
[74] 2019
Mice exposed to PM2.5 by instillation, hydroxytyrosol, p.o. at 50 mg/kg/day for 4 weeks
Alleviated PM2.5-induced visceral adiposity and insulin resistanceInhibited PM2.5-induced oxidative stress-mediated activation of NF-κBEnrichment of gut microbiota, and reduction of pathogenic bacteria
[75] 2020
Rats fed an obesogenic cafeteria diet, hesperidin (a citrus polyphenol), p.o. at 40 and 100 mg/kg/day for 8 weeks
Decreased the total cholesterol, LDL-C, and free fatty acids; ameliorated blood pressure and insulin sensitivity, and decreased the markers of arterial stiffness and inflammationMetabolomics revealed an improvement in lower excretion of inflammation- and oxidative stress-related metabolitesExcretion of higher amounts of microbe-derived metabolites, which positively correlated with the Bacteroidaceae family
[76] 2021
Spontaneously hypertensive rats, Litchi chinensis seed extract, rich in polyphenols, including procyanidins, cinnamtannins, and rutin, p.o. at 30 and 60 mg/kg/day for 10 weeks
Reduced blood pressure and alleviated hypertension-induced renal damageAttenuated oxidative stress and inflammationIncreased the relative abundance of Lactobacillus and production of SCFAs in the intestine
[77] 2021
Mice with doxorubicin (DOX)-induced heart failure, purified polyphenols from Arctium lappa L. including arctiin, dicaffeoyl succinoylquinic acid, and luteolin, p.o. at 50 and 150 mg/kg/day for 29 days; on day 22, 24, and 26, DOX was i.p. injected
Reduced heart failure syndrome and reduced serum activities of casein kinase and lactate dehydrogenaseAlleviated serum oxidative stress and reduced serum levels of inflammatory indices (TNF-α and NO).Increased the abundance of Lactobacillaceae, Muribaculaceae, and Ruminococcaceae and decreased the abundance of Proteobacteria, Enterobacteria, and Escherichia_Shigella;
enhanced the abundance of bacteria producing SCFAs
[78] 2021
Mice with potassium oxonate-induced hyperuricemia, Camellia japonica bee pollen extract containing polyphenols including kaempferol, quercetin, and gallic acid, p.o. at 2 and 4 g/kg/day for 3 weeks; starting on the 15th day, mice received potassium oxonate for the following 7 days
Reduced serum uric acid by inhibiting XOD activity and improved renal functionIncreased antioxidant biomarkers, SOD activity, and GSH content, and decreased MDA content in the liverIncreased the abundance of Lactobacillus that has an anti-hyperuricemia effect;
decreased the Firmicutes/Bacteroidetes ratio
p.o., per os; NAFLD, nonalcoholic fatty liver disease; PM2.5, particulate matter (≤2.5 μm); NF-κB, nuclear factor-κB; LDL-C, low-density lipoprotein-cholesterol; Nrf2, nuclear factor (erythroid-derived 2)-like 2; TBARS, thiobarbituric acid reactive substances; ROS, reactive oxygen species; iNOS, inducible nitric oxide synthase; MDA, malondialdehyde; 4-HNE, 4-hydroxy-2-nonenal; 8-OHdG, 8-hydroxy-2′-deoxyguanosine; LPS, lipopolysaccharide; GSH, glutathione; TNF-α, tumor necrosis factor-α; NO, nitric oxide; SCFAs, short-chain fatty acids; XOD, xanthine oxidase; SOD, superoxide dismutase; IL-6, interleukin-6; IL-1β, interleukin-1β; TNF-β, tumor necrosis factor-β.
Table 4. Effects of polyphenols on the gut microbiota in healthy mice and rats.
Table 4. Effects of polyphenols on the gut microbiota in healthy mice and rats.
Reference, Publication Year, Animal Species,
Polyphenol(s), and Dosage
Observations
Antioxidative and Anti-Inflammatory ActionGut Microbiota Modulation
[79] 2019
Rats, Jaboticaba (Plinia jaboticaba) peel extract containing gallic acid, flavonols, and anthocyanins, p.o. in drinking water (5 g/L) for 15 and 49 days
No disturbance of the antioxidant systemIncreased the counts of Lactobacillus, Bifidobacterium, and Enterobacteriaceae
[80] 2020
Rats, freeze-dried whole golden kiwifruit (Actinidia chinensis) containing polyphenols, including quercetin, catechin, and chlorogenic acid, p.o. 4.6 g/kg/day for 28 days
Effectively improved the hepatic lipid profile, lipid peroxidation, long-chain fatty acid profile, and intestinal homeostasisReduced the Firmicutes/Bacteroidetes ratio at the phylum level; increased the abundance of beneficial bacteria (Lactobacillus and Barnesiella) and reduced the abundance of harmful bacteria (Enterococcus, Escherichia, and Staphylococcus)
[81] 2020
Mice, Penthorum chinense extract containing polyphenols, including pinocembrin-7-O-glucoside, thonningianin A, and brevifolin carboxylic acid, p.o. at 0.4 g/kg/day for 5 days
Increased antioxidant capacity (increased plasma SOD and hepatic CAT and GSH-px levels)Increased microbiota diversity, elevated the Bacteroidetes/Firmicutes ratio at the phylum level, and enriched gut health-promoting bacteria
[82] 2020
Mice, anthocyanin-rich Lycium ruthenicum Murray (ACN), p.o. at 200 mg/kg/day for 12 weeks
Increased the antioxidant status in the liver (increased AOC, SOD, CAT, GSH, and GSH-Px levels and decreased the MDA levels); ameliorated the anti-inflammatory status in the colon (reduced the expression of iNOS, COX-2, TNF-α, IL-6, IL-1β, and IFN-γ mRNAs) and increased the intestinal barrier function (increased the expression of ZO-1, Occludin, Claudin-1, and Muc1 mRNAs)Increased the proliferation of Barnesiella, Alistipes, Eisenbergiella, Coprobacter, and Odoribacter;
increased SCFAs in the cecal content and feces.
p.o., per os; SOD, superoxide dismutase; CAT, catalase; GSH-Px, T-AOC, total antioxidant capacity; GSH, glutathione; MDA, malondialdehyde; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; TNF-α, tumor necrosis factor-α; IL-6, interleukin-6; IL-1β, interleukin-1β; IFN-γ, interferon-γ; ZO-1, zonula occludens-1; Muc1, mucin 1; SCFAs, short-chain fatty acids.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Niwano, Y.; Kohzaki, H.; Shirato, M.; Shishido, S.; Nakamura, K. Putative Mechanisms Underlying the Beneficial Effects of Polyphenols in Murine Models of Metabolic Disorders in Relation to Gut Microbiota. Curr. Issues Mol. Biol. 2022, 44, 1353-1375. https://doi.org/10.3390/cimb44030091

AMA Style

Niwano Y, Kohzaki H, Shirato M, Shishido S, Nakamura K. Putative Mechanisms Underlying the Beneficial Effects of Polyphenols in Murine Models of Metabolic Disorders in Relation to Gut Microbiota. Current Issues in Molecular Biology. 2022; 44(3):1353-1375. https://doi.org/10.3390/cimb44030091

Chicago/Turabian Style

Niwano, Yoshimi, Hidetsugu Kohzaki, Midori Shirato, Shunichi Shishido, and Keisuke Nakamura. 2022. "Putative Mechanisms Underlying the Beneficial Effects of Polyphenols in Murine Models of Metabolic Disorders in Relation to Gut Microbiota" Current Issues in Molecular Biology 44, no. 3: 1353-1375. https://doi.org/10.3390/cimb44030091

Article Metrics

Back to TopTop