Skip to main content
Journal of Advanced Research logoLink to Journal of Advanced Research
. 2023 Jun 2;52:151–170. doi: 10.1016/j.jare.2023.05.011

Gut microbiota: A magical multifunctional target regulated by medicine food homology species

Wei-Fang Zuo 1,1, Qiwen Pang 1,1, Lai-Ping Yao 1, Yang Zhang 1, Cheng Peng 1, Wei Huang 1,, Bo Han 1,
PMCID: PMC10555941  PMID: 37269937

Graphical abstract

graphic file with name ga1.jpg

Keywords: Gut microbiota, Medicine food homology species, Human health, Bioconversion

Highlights

  • Gut microbiota's coevolution with hosts significantly impacts the regulation of human diseases.

  • Medicine food homology species are rich in bio-active substances, which have good medical and nutritional value.

  • Intaking medicine food homology species will affect the composition of gut microbiota and provide metabolites that affect host physiology.

  • The relationship between medicine food homology species–gut microbiota–individual health has been confirmed, but the underlying mechanisms remain far from clear.

Abstract

Background

The relationship between gut microbiota and human health has gradually been recognized. Increasing studies show that the disorder of gut microbiota is related to the occurrence and development of many diseases. Metabolites produced by the gut microbiota are responsible for their extensive regulatory roles. In addition, naturally derived medicine food homology species with low toxicity and high efficiency have been clearly defined owing to their outstanding physiological and pharmacological properties in disease prevention and treatment.

Aim of review

Based on supporting evidence, the current review summarizes the representative work of medicine food homology species targeting the gut microbiota to regulate host pathophysiology and discusses the challenges and prospects in this field. It aims to facilitate the understanding of the relationship among medicine food homology species, gut microbiota, and human health and further stimulate the advancement of more relevant research.

Key scientific concepts of review

As this review reveals, from the initial practical application to more mechanism studies, the relationship among medicine food homology species, gut microbiota, and human health has evolved into an irrefutable interaction. On the one hand, through affecting the population structure, metabolism, and function of gut microbiota, medicine food homology species maintain the homeostasis of the intestinal microenvironment and human health by affecting the population structure, metabolism, and function of gut microbiota. On the other hand, the gut microbiota is also involved in the bioconversion of the active ingredients from medicine food homology species and thus influences their physiological and pharmacological properties.

Introduction

Gut microbiota is a vast and complex ecosystem containing approximately 100 trillion microorganisms and is symbiotic with the host [1]. Research results have indicated that gut microbiota equals the number of human cells and has been dubbed a “forgotten organ” due to its multiple roles in human physiology [2]. These microorganisms bind closely to the intestinal mucosal epithelial cells and form a critical biological barrier to protect the intestinal mucosa from foreign pathogens [3]. Ingested plant polysaccharides and other dietary substances are degraded by the gut microbiota, which enhances the digestion efficiency of the host and simultaneously ensures a steady nutrient supply for the microorganisms. Moreover, extensive interconnections between the physiology of the microbial community and the host have been established due to millions of years of co-evolution, and they have extended beyond the degradation function [4]. The gut microbiota has developed a tactic to release large numbers of metabolites from different chemical categories to exert an extensive role in host organs, regardless of close to or far from the gastrointestinal lumen. These metabolites include but are not limited to, short-chain fatty acids (SCFAs) [5], bile acids (BAs) [6], [7], choline [8], [9], vitamins [10], tryptophan [11], [12], and indole derivatives [13], [14]. Besides, they associate with each other and host cells, thereby further influencing host health and disease states [15], [16], [17], [18]. Increasing pieces of evidence have shown that the imbalance of gut microbiota is related to the incidence of various diseases, including inflammatory bowel disease (IBD) [19], fatty liver [20], [21], obesity [22], [23], diabetes [24], [25], neurological disorders [26], [27], cardiovascular diseases [28], [29], etc. (Fig. 1, right). Although sharing a conserved set of core gut microbiota and core microbiome among individuals at the macro level may be necessary for correct gut function at the macro level, each one has a distinct and highly variable microbiota from an individual point of view [30], [31], [32]. In addition to host factors (such as age and genetics) [33], the composition and function of gut microbiota are also influenced mainly by changing environmental factors (Fig. 1, left), especially diet [34] and medications [35], [36]. Therefore, gut microbiota may be a multifunctional target regulated by easily ingested medicine food homology species.

Fig. 1.

Fig. 1

Relationship between gut microbiota and human. Left: Multiple environmental factors, encompassing diet, antibiotics, pollution, physical inactivity, and disease, may engender perturbations in the gut microbiota. Right: Gut microbiota aberrations have been linked to a myriad of morbidities, spanning neuropsychiatric disorders, cardiovascular afflictions, gastrointestinal ailments, diseases arising from environmental and nutritional factors, endocrinopathies, and more. Figure created via BioRender.com.

The term “Medicinal Food Homology” (MFH) typically refers to species that are traditionally both food and Chinese medicine [37]. It also has various English expressions such as the homology of medicine and food, and the materials for both food and medicine. Globally, there are many concepts similar to MFH, such as functional foods and dietary supplements. Huang Di Nei Jing (475 BCE-221 BCE), the first monograph on medical theory in China, has recorded: “eating on an empty stomach as food and administering to the patient as medication”, which should be the earliest literal definition of the MFH theory. In about 500 CE, Sun Simiao, a medical scientist and pharmacologist at that time, systematically elaborated the theory of diet therapy for the first time and comprehensively expounded the method of combining food and medicine to treat diseases in Prescriptions Worth Thousand Gold for Emergencies [38]. Since modern times, people's health consciousness has gradually shifted from treatment to the combination of prevention and treatment, and medicinal plants with therapeutic effects have been integrated into the diet as dietary supplements, which belong to the category of alternative therapy. Modern pharmacological studies have demonstrated MFH species' preventive and therapeutic effects in various diseases, especially chronic diseases [39], [40], [41]. To ensure the safe and normative use of these species, the National Health Commission of the People's Republic of China has issued specific regulations on the MFH items. As of 2019, nearly a hundred kinds of Chinese medicine have been recognized as MFH species [42] (Fig. 2).

Fig. 2.

Fig. 2

The history of MFH and some representative MFH species discussed in this review. Top: Key historical events in the development of MFH in China. Bottom: According to the taxonomy, these species are divided into nine groups: Liliflorae, Microspermae, Umbelliflorae, Rosales, Polyporales, Tubiflorae, Campanulales, Scitamineae, and others.

Increasing evidence shows a close interaction and relationship between MFH and gut microbiota, which can affect human health and disease processes. This review surveys the number of articles published on the MFH and gut microbiota over the past decade and finds that these fields have experienced vigorous development. However, so far, there are few articles to systematically review and analyze the regulation of MFH on gut microbiota and its influence on human health, which urgently needs replenishment. This review focuses on the research status of the regulation of MFH on gut microbiota and its effects on host health and discusses the interaction between MFH and gut microbiota. The content of the current review is deliberately confined to the critical selection of representative work, and it is not intended to be a comprehensive (encyclopedia) coverage covering all the examples that appear in the literature. According to the pathology catalog, this review is divided into digestive diseases, endocrine diseases, neurological and psychiatric disorders, cardiovascular diseases, environmental and nutritional diseases, etc. Finally, the research direction in this field has been prospected to encourage more researchers to pay attention to the effects of MFH on gut microbiota and human health and to provide new insights for treating clinical diseases.

Medicine food homology species treat diseases by regulating gut microbiota

Digestive system diseases

The digestive system is a functional group of digestive ducts and glands responsible for digestion, absorption, excretion, detoxification, and endocrinology. The interactions between gut microbiota, digestive system, and medicine food homology species are increasingly demonstrated to be essential for maintaining homeostasis and human health (Table 1).

Table 1.

Effects of medicine food homology species on digestive diseases via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Ginseng Radix Et Rhizoma polysaccharide ↓Bacteroidetes; Verrucomicrobia; Proteobacteria
↑Akkermansia
↓inflammatory
↑autophagy; microbiota diversity
SD mice 200 mg/kg IBD [47]
Lycii Fructus AA-2βG ↓Proteobacteria; Parabacteroides; Parasutterella; Clostridiaceae
↑Verrucomicrobia; Prevotellaceae; Helicobacteraceae
↑SCFAs; microbiota diversity C57BL/6 mice
(5 weeks old)
300 mg/kg IBD [48]
Lycii Fructus arabinogalactan ↓Proteobacteria; Enterobacteriaceae
↑Ruminococcaceae; Muribaculaceae; Prevotellaceae; Rikenellaceae
↓inflammatory
↑SCFAs
male C57BL/6J mice
(7 weeks old)
50 mg/kg IBD [49]
Polygonati Odorati Rhizoma extract ↓Desulfobacterota; Campylobacterota
↑Muribaculaceae; Ruminococcus; Alloprevotella
↓H2S
↑SCFAs
male BABL/c mice
(6–8 weeks old)
400 mg/kg IBD [50]
Ganoderma polysaccharide ↓Oscillibacter; Desulfovibrio; Parasutterella; Alistipes
↑Bifidobacterium; Lactobacillus
↓inflammatory; tumorigenesis;
↑SCFAs; intestinal barrier
male C57BL/6 mice
(5 weeks old)
300 mg/kg CRC [66]
Panacis Quinquefolii Radix extract ↓Bacteroidetes; Verrucomicrobia
↑Firmicutes
↓inflammatory; harmful metabolites; tumor multiplicity
↑beneficial metabolites
male A/J mice
(6 weeks old)
30 mg/kg CRC [67]
Jujubae Fructus polysaccharide ↓Firmicutes
↑Bifidobacterium; Bacteroides; Lactobacillus
↓inflammatory
↑SCFAs; Metabolic functions
male C57BL/6 mice
(6–8 weeks old)
1 g/kg CRC [68]
Jujubae Fructus polysaccharide ↓Firmicutes; Cyanobacteria; Spirochaete; Deferribacteres; Mucoromycota
↑Bacteroidetes; Lactobacillaceae; Actinobacteria
↓gut dysbiosis
↑SCFAs
male C57BL/6 mice
(6–8 weeks old)
1 g/kg CRC [69]
Lycii Fructus Lycii Fructus ↓Alloprevotella
↑Akkermansia; Ruminococcaceae_UCG_014
↓inflammation; harmful metabolites
↑SCFAs; antioxidant capacity; beneficial metabolites
female C57BL/6 mice
(4 weeks old)
1% of dry feed weight ALD [72]
Ganoderma extract ↓Clostridium_sensu_stricto_1
↑Ruminiclostridium_9; Prevotellaceae_UCG-001; Oscillibacter; Bilophila; Ruminococcaceae_UCG-009
↓harmful metabolites
↑SCFAs; metabolic function
male Kunming mice
(6–7 weeks old)
100 mg/kg ALD [73]
Ganoderma Ganoderic Acids ↓Helicobacter
↑Lactobacillus; Faecalibaculum; Romboutsia; Bifidobacterium
↓lipid metabolic male Kunming mice
(6 weeks old)
36 mg/kg ALD [74]
Astragali Radix polysaccharides ↓Firmicutes
↑Bacteroidetes; Proteobacteria; Episilonbacteria
↓inflammation
↑intestinal integrity
male SD rats 200 mg/kg NAFLD [86]
Cassiae Semen extract ↓Bilophila; Lactobacillus;
Mucispirillum; Paraprevotella
↑Odoribacter; Rikenella; Desulfovibrio
↓inflammation; lipid accumulation
↑intestinal integrity
C57BL/6 mice 10 mg/kg NAFLD [87]
Rubi Fructus extract ↑Turicibacter; Bifidobacterium ↓inflammation; liver fibrosis male C57BL/6J mice
(8 weeks old)
7.9 g/kg liver fibrosis [93]
Gardeniae Fructus polysaccharides ↓Enterobacteriaceae; Enterococcaceae
↑Bacteroides
↓inflammation; bile acids
↑SCFAs; intestinal integrity
male C57BL/6 mice
(8 weeks old)
400 mg/kg Cholestasis [96]

Inflammatory bowel disease

Inflammatory bowel disease (IBD) is a chronic relapsing disorder typically characterized by nonspecific inflammation and intestinal tissue damage [43]. The intestinal barrier is vital in maintaining intestinal homeostasis and is the primary defense against pathogen invasion [3]. Increasing evidence show that gut microbiota dysbiosis contributes to the initiation, development, and exacerbation of IBD [44]. According to reports, the overactive immune response is closely associated with an increase in lipopolysaccharide (LPS), a substance that makes up the cell walls of Gram-negative bacteria [45]. In addition, the primary intestinal metabolites, SCFAs, enhance the intestinal barrier, aid in nutritional absorption, and inhibit the growth of pathogenic microorganisms [46]. Thus, as an alternative therapy, targeting gut microbiota through the diet may be a new approach for IBD treatment in the future (Fig. 3).

Fig. 3.

Fig. 3

Schematic representation of the proposed mechanism of medicine food homology species on ameliorating IBD and CRC. LPS from Gram-negative bacteria trigger an inflammatory cascade by activating the TLR4/NF-κB signaling pathway and releasing pro-inflammatory factors, thereby inducing an inflammatory response and increasing intestinal permeability. Persistent chronic intestinal inflammation eventually leads to cancer. Medicine food homology species reduced the release of toxic substances and the secretion of pro-inflammatory factors and repaired the intestinal barrier by regulating gut microbiota, thus decreasing the immune response, and playing the role of anti-IBD and CRC. Figure created via BioRender.com.

As a receptor of LPS, activated Toll-like receptor 4 (TLR4) recruits downstream signaling molecules to coordinate the secretion of inflammatory cytokines and oxidative stress response, thereby initiating IBD. Studies have shown that the edible herb Ginseng Radix Et Rhizoma can alleviate IBD. By lowering the abundance of Gram-negative bacteria, such as Bacteroidetes, Verrucomicrobia, Proteobacteria, etc., the concentration of LPS was decreased, resulting in the blocking of TLR4/Nuclear Factor-κB (NF-κB) pathway-associated proteins expression. Meanwhile, autophagy was stimulated to inhibit the activation of NF-κB, contributing to managing the inflammatory response in the intestine, which favorably connected with the restoration of Akkermansia by Ginseng polysaccharide [47]. Lycii Fructus is the most common dietary supplement with various pharmacological activities, such as antioxidant and immune promotion. In colon tissue, it has been shown that treatment with the active ingredient 2-O-β-D-Glucopyranosyl-L-ascorbic acid (AA-2βG) from Lycii Fructus can enhance the content of metabolites SCFAs and increase the mRNA expression level of G protein-coupled receptor 43(GPR43) and GPR41, two SCFAs receptors. Additionally, AA-2βG reduced Parabacteroides, Parasutterella, and Clostridium associated with the development and progression of IBD while increasing Helicobacteraceae, which protects against IBD [48]. Arabinogalactan, another bioactive ingredient from Lycii Fructus, has also been shown to be effective against IBD. It gave some gut bacteria that eat mucin O-glycans essential nutrients, lowering the amount of mucin 2 (MUC2) that some gut microbiota utilized and enhancing intestinal integrity [49]. Gas, a gut microbiota metabolite, can also influence how colitis develops. The administration of Polygonati Odorati Rhizoma extract modulated the gut microbiota in this article to lessen the harmful bacteria (such as Desulfovibrionaceae) linked to the gas generation of H2S, which is poisonous in high quantities. Importantly, their research was the first to establish a link between intestinal gas production and key bacteria associated with ulcerative colitis [50].

In addition, many medicine food homology species can also participate in the treatment of IBD by regulating gut microbiota, including Crataegi Fructus [51], [52], Mel [53], Dioscoreae Rhizoma [54], Chrysanthemi Flos [55], Ziziphi Spinosae Semen [56], Curcumae Longae Rhizoma [57], Astragali Radix [58], [59], Ganoderma [60], etc. These species described above may serve as promising candidates to provide new therapeutic options for IBD.

Colorectal cancer

One of the most prevalent tumors, colorectal cancer (CRC) is responsible for 10% of all cancer diagnoses each year and all cancer-related deaths globally [61], [62]. In addition, CRC risk is higher in patients with long-standing IBD and gut microbiota disorders brought on by unhealthy lifestyle choices [63], [64], [65]. Therefore, it has become increasingly appealing to develop novel, potent, but low-toxicity reagents based on medicine food homology species for the prevention and therapy of CRC.

Ganoderma is one of the most popular and well-known mushrooms of MFH in China. Cancer caused by azoxymethane/dextran sulfate sodium (AOM/DSS) was dramatically reduced by dietary fiber Ganoderma polysaccharide (GLP). According to the findings, GLP consumption increased Firmicutes and decreased Bacteroidetes at the phylum level, which was related to SCFAs production and tumor susceptibility, respectively. In addition, GLP treatment inhibited tumorigenesis by increasing beneficial bacteria Bibifdobacterium and Lactobacillus and decreasing Desulfovibrio, Oscillibacter, and other bacteria positively related to CRC. Significantly, alterations in the levels of SCFAs, GPR43, Zonula Occludens-1 (ZO-1), LPS, and other factors demonstrate that GLP is crucial in preventing inflammation-related CRC carcinogenesis [66]. However, the exact mechanism of how the altered gut microbiota after GLP therapy affects the progression of CRC remains to be investigated. Panacis Quinquefolii Radix is metabolized by the gut microbiota into active components (e.g., compound K, 20S-protopanaxadiol) with anti-tumor potential, which can inhibit the release of cytokines and induce endogenous metabolite changes. Additionally, these components reduced Verrucomicrobia, a Gram-negative bacterium that promotes tumorigenesis [67]. In general, and particularly in China, Jujubae Fructus is regarded as a species with high nutritional value and pharmacological effects. Polysaccharides, as the main component of Jujubae Fructus, have significant gastrointestinal protective effects. It was reported that jujubae polysaccharides (JPs) increased the concentration of total SCFAs and changed the intestinal microbiota units, significantly increasing the abundance of beneficial bacteria, including Bifidobacterium, Bacteroides and Lactobacillus. Additionally, this study also revealed that JPs could alleviate metabolic dysfunction, which was related to specific gut microbiota [68], [69]. These findings suggested that Jujubae Fructus could reduce CRC-associated risks and be a valuable dietary therapeutic for CRC prevention.

Alcoholic liver disease

Alcoholic liver disease (ALD), brought on by prolonged excessive alcohol consumption, is one of the most common and poses a severe threat to human health and social development [70]. Several pathological characteristics of liver lesions, primarily fatty liver, alcoholic hepatitis, and alcoholic cirrhosis can be caused by ALD. In addition, increasing studies have suggested that excess ethanol and its metabolites, such as acetaldehyde, cause disorders of the gut microbiome homeostasis and the disruption of intestinal barrier permeability, leading to perturbations of the “gut-liver axis” and, finally, liver disease [71]. A promising ALD therapy strategy is focusing on the “gut-liver” axis.

In addition to its anti-IBD properties, which were already described, Lycii Fructus may also benefit the alcohol-induced liver injury. Lycii Fructus serves a protective role in the liver by reducing inflammation and enhancing antioxidant capacity, according to recent studies. On the one hand, Lycii Fructus boosted the levels of SCFAs that were positively correlated with Akkermansia and Ruminococcaceae_UCG_014, particularly butyrate, which is thought to be crucial for maintaining the integrity of the intestinal barrier by regulating the expression of tight junction proteins and mucin. Meanwhile, LPS biosynthesis was down-regulated by reducing Proteobacteria, thereby alleviating the subsequent inflammatory response caused by LPS translocation to the liver. On the other hand, glutathione (GSH), an endogenous antioxidant in hepatocytes, has been shown to delay or decrease apoptosis when GSH is blocked. Therefore, the effect of Lycii Fructus on GSH synthesis might be through regulating the growth of Akkermansia, thus improving its antioxidant capacity. Moreover, after L. barbarum intervention, toxic metabolites, such as phenylacetylglutamine, markedly decreased while helpful metabolites for treating ALD, including L-glutamic acid, pyroglutamic acid, vanillic acid, and retinol β-glucuronide were noticeably elevated. In addition to reversing the damage already done, the ability of Lycii Fructus pretreatment to resist the butyric acid drop caused by alcohol intake suggests that it also has disease-preventive effects [72]. Ganoderma could contribute to host health by increasing the abundance of Ruminiclostridium_9, Prevotellaceae_UCG-001, Oscillibacter, Ruminococcaceae_UCG-009, and others. It has been reported that these may be related to functions such as increasing the content of SCFAs, improving lipid metabolism, and inhibiting inflammation. Spearman correlation analysis showed that these bacteria were strongly correlated with a variety of liver metabolites, including caffeic acid [POS_13], erucic acid [POS_39], L-cystine [Neg_12], Ile-Ala [POS_8], etc. [73]. In another report, the active ingredient Ganoderma acids significantly reduced the level of pathogenic bacteria Helicobacter and exerted liver protection by regulating liver lipid metabolism by increasing the Lactobacillus, Faecalibaculum, and Romboutsia [74]. Although the opposite result was shown in Lv's work, the Lactobacillus level was reduced [75], which may be because the excessive abundance of Lactobacillus also disturbs lipid metabolism in the liver [76]. It was reported that the “gut-liver” axis could help Cornel iridoid glycoside (CIG) from Corni Fructus prevent ALD. Notably, the therapeutic efficacy of CIG (200 mg/kg) was superior to that of silymarin, suggesting that it can be an alternative hepatoprotective agent to prevent alcoholic liver injury. But further research is needed to determine the long-term effects of CIG in ALD models. [77]. Studies on medicine food homology species of Hippophae Fructus [78], Puerariae Lobatae Radix [79], Astragali radix, and related processed products [80], [81] have shown a growing role in preventing and alleviating of ALD through the gut microbiota.

Nonalcoholic fatty liver disease

Nonalcoholic fatty liver disease (NAFLD) is characterized by lipid metabolism dysfunction. Its mechanism mainly involves insulin resistance and increased oxidative stress, leading to hepatocyte steatosis and lipid peroxidation [82], [83]. The histological changes of NAFLD are similar to those of ALD, but there is no history of alcoholism in NAFLD patients. Even though pioglitazone, obeticholic acid, and vitamin E have all been tested in clinical trials, no drug has yet received therapeutic approval. [84]. In addition to exercise, dietary intervention is currently the only available therapy [85].

The SCFAs produced by the fermentation of indigestible dietary fiber can provide energy to the host. Studies have shown that SCFAs concentrations are higher in genetically obese or overweight models than in lean animal models, suggesting that microbial energy harvesting is increasing in obese patients. Although polysaccharides extracted from Astragali radix (mAPS) had no significant effect on SCFAs, it significantly reduced the protein expression of GPR41 and GPR43 in the liver and colon, which might be related to the down-regulation of Firmicutes/Bacteroidetes (F/B) ratio under mAPS intervention. These findings suggested that mAPS attenuated the progression of NAFLD partly by regulating host energy balance. In addition, decreased expression of GPR41 and GPR43 was correlated with decreased TLR4-NF-κB-NLRP3 signaling, perhaps exhibiting anti-inflammatory effects [86]. The seeds of Cassia obtusifolia L. and C. tora L. have a long medicinal history in China as a medicinal-edible substance known as Cassiae Semen (CS), with claims that it can ease constipation and have hepatoprotective benefits. In high-fat diet (HFD)-induced liver injury, Tang et al. revealed that CS extracts ameliorated lipid accumulation, intestinal barrier damage, liver damage, and inflammation. Importantly, fecal microbe transplantation (FMT) recapitulated the pharmacological effects of CS, and the efficacy was impaired by antibiotic-induced dysbiosis [87]. In addition, by combining the cholesterol-lowering probiotics DM9054 and 86,066 with anthraquinone from Cassia obtusifolia L., the development of HFD-induced NAFLD was effectively prevented and the effect of co-administration was more pronounced than that of either agent administered alone [88]. Moreover, Amomi Fructus [89], Poria [90], Crataegi Fructus [91], Puerariae Lobatae Radix and its components [92] play a preventive and therapeutic role in NAFLD by regulating gut microbiota and triggering related signals.

Others

Liver disease also includes other subtypes, such as liver fibrosis, cirrhosis, etc. These are often driven by inflammation or environmental factors. Rubi Fructus (RF), the dried fruit of Rubus chingii Hu., is a well-known herb for nourishing the liver and has been documented in many monographs such as “Kai Bao Ben Cao” and “Ben Cao Fa Ming”. Liu et al. revealed that supplementation with RF extract could effectively ameliorate CCl4-induced liver fibrosis while simultaneously modulating the gut microbiota. Correlation analysis indicated that the regulatory effect of RF on intestinal microbiota and its beneficial effects on liver fibrosis were related, among which Bifidobacterium and Turicibacter were the most pertinent bacterial genera, and were closely related to pharmacodynamic parameters, including alanine aminotransferase, aspartate aminotransferase, etc. [93]. Cholestasis can also cause liver damage and progression to liver fibrosis without proper treatment. Gardeniae Fructus, an herb commonly used to treat liver disease [94], is also a constituent in many classic anti-cholestatic prescriptions [95]. For the first time, Ma et al. showed how polysaccharides from Gardeniae Fructus modulated gut microbiota to alleviate cholestatic liver damage. The decreased bile acids levels, the reduced hepatic inflammation mediated by the TLR4/NF-κB pathway, and repaired intestinal barrier were proven to be involved in this process. However, how the polysaccharide from Gardeniae Fructus regulates gut microbiota to reduce bile acid accumulation in the liver requires further investigation [96]. In addition, Portulacae Herba was reported as a supplementary dietary therapeutic strategy to alleviate liver and kidney injury associated with food-source toxic metal Cd exposure by increasing the beneficial bacteria Akkermansia and Faecalibaculum [97].

Environmental and nutritional diseases

Nutritional diseases

Obesity is an over-nourishing disease defined as the accumulation of body fat and has become a serious global health problem [98]. Given the negative impact of obesity on individual health and the economy, it is urgent to explore new ways to prevent and treat obesity. Here, several medicine food homology species exert their anti-obesity activity by regulating gut microbiota and related signals (Table 2).

Table 2.

Effects of medicine food homology species on environmental and nutritional diseases via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Dendrobii Officinalis Caulis extract ↓Bilophila
↑Muribaculum; Akkermansia; Bifidobacterium
↓inflammatory; oxidant stress;
↑intestinal barrier; glucose homeostasis; SCFAs
male C57BL/6 mice
(6 weeks old)
1 g/kg Obesity [99]
Ganoderma extract ↓the ratio of F/B; Proteobacteria; Escherichia ↓inflammatory
↑intestinal barrier
male C57BL/6N mice
(8 weeks old)
8 % WEGL Obesity [100]
Citri Reticulatae Pericarpium Polymethoxyflavone ↓Proteobacteria; F/B; Acetatifactor; Bilophila
↑Akkermansia; Allobaculum
↓BAs metabolism
↑SCFAs;
male C57BL/6J mice
(6 weeks old)
0.5% extract Obesity [101]
Lycii Fructus polysaccharide ↓Proteobacteria
↑Lactobacillus
↓inflammatory
↑SCFAs
male C57BL/6 mice 50 mg/kg Obesity [102]
Glycyrrhizae Radix Et Rhizoma total flavonoids ↑Lactobacillus; Muribaculum; Roseburia; Anaerotruncus; ↓inflammatory
↑SCFAs
male C57BL/6 mice
(6 weeks old)
135 mg/kg ADR [103]
Poria polysaccharides ↓Proteobacteria; Cyanobacteria; Ruminococcaceae; Helicobacteraceae
↑Erysipelotrichaceae; Prevotellaceae
↓inflammatory
↑intestinal barrier
male C57BL/6 mice
(6–8 weeks old)
7.6 mg/kg ADR [104]
Amomi Fructus volatile oil
bornyl acetate
↓Escherichia; Bacteroides; Helicobacter
↑Lactobacillus; Bifidobacterium;
↓inflammatory
↑intestinal barrier
male SD rats
(8 weeks old)
32 mg/kg
8 mg/kg
ADR [105]
Codonopsis Radix polysaccharides ↓Escherichia coli
↑Lactobacillus
↑immunity SPF BALB/c mice
(8 weeks old)
200 mg/kg ADR [106]
Lycii Fructus AA-2βG ↓Proteobacteria; Deferribacteraceae
↑Muribaculaceae
↑immunity; amino acid metabolism and biosynthesis BALB/c mice
(5 weeks old)
100 mg/kg ADR [109]
Ginseng Radix Et Rhizoma Ginsenoside Rg1 ↑B. vulgatus ↓tryptophan metabolism; 5-HT; 5-HTR1B; 5-HTR2A female BALB/c mice
(6 weeks old)
200 mg/kg ADR [111]

Dendrobii Officinalis is an edible medicinal plant and several studies revealed that D. officinale and its components exert the therapeutic effects on metabolic syndrome. By targeting gut microbiota and host metabolism, D. Officinale dietary fiber (DODF) effectively alleviated obesity-related symptoms in obese mice. The gut microbiota of obese mice was restructured as indicated by increased Akkermansia, Bifidobacterium, and Muribaculum and decreased Bilophila. In addition, metabolic phenotypes were changed following DODF administration, as seen by upregulation of energy metabolism, an increase in acetate and taurine, and a reduction in serum low-density/very low-density lipoproteins (LDL/VLDL). These indicated that DODF might be utilized as a prebiotic for managing obesity (Fig. 4) [99]. Furthermore, water extract of Ganoderma mycelium (WEG) has been shown to effectively reduce body weight, inflammation, and insulin resistance in HFD mice. Combined with the fecal transplants experiment, WEG not only reversed the gut dysbiosis, including the drop in the F/B ratio and the level of endotoxin-producing Proteobacteria, but it also maintained the intestinal barrier integrity and reduced metabolic endotoxemia. Further experiments suggested that high molecular weight polysaccharides (4300 kDa) isolated from WEG exhibited similar anti-obesity and microbiota-modulating actions, illuminating the underlying material foundation [100]. Dysregulation of BAs may lead to obesity. Citri Reticulatae Pericarpium extracts dose-dependently alleviated HFD-induced growth of Acetatifactor and Bilophila, bacteria involved in BAs metabolism [101]. In addition, Polysaccharides from Lycii Fructus (LBPs) could be used as prebiotics to ameliorate obesity by altering the genus-specific gut microbiota (Proteobacteria and Lactobacillus) and SCFAs metabolism [102].

Fig. 4.

Fig. 4

The protective effects of DODF in obesity. DODF remodeled the metabolic phenotype of the host by reshaping gut microbiota, thus enhancing insulin signaling pathway and liver glycogen synthesis and alleviating systemic inflammatory responses and oxidative stress. Figure created via BioRender.com.

Adverse drug reactions

Therapeutic drug injury, also known as adverse drug reactions, is the effect of drugs that have nothing to do with the therapeutic effect and are detrimental to the body when the drug treats the disease. Consequently, we have explored some medicine food homology species which can alleviate the damage caused by these drugs (Table 2).

Intestinal inflammation is the most common adverse drug reaction. Although irinotecan (CPT-11) is an effective chemotherapeutic agent for solid tumors, severe gastrointestinal toxicity significantly limits its anticancer effects. Duan et al. reported total flavonoids of Glycyrrhiza uralensis (TFGU) could correct the whole intestinal microbial imbalance and fecal metabolic disorder in CPT-11-induced colitis mice. It mainly call-backed SCFAs-producing bacteria, including Muribaculum, Roseburia, Anaerotruncus, and Lactobacillus, and corrected Bacteroides and Clostridium, which were closely related to β-glucuronidase. In addition, TFGU could also increase the abundance of the Lactobacillus and butyrate-producing bacteria Roseburia, both of which are conducive to balancing the intestinal environment in colitis [103]. Poria extract showed an excellent protective effect on intestinal injury caused by cisplatin. It could mainly alleviate the imbalance of intestinal microflora induced by cisplatin, which significantly reduced pathogens such as Helicobacteraceae and promoted the growth of probiotics such as Erysipelotrichaceae and Prevotellaceae [104]. The chemotherapeutic 5-fluorouracil (5-FU) can produce common toxic side effects, such as intestinal mucositis. The volatile oil from Amomi Fructus and its active component, bornyl acetate, have been proven to effectively reduce the number of pathogenic bacteria (including Escherichia, Bacteroides, Helicobacter, etc., increase probiotics (including Lactobacillus, Bifidobacterium) and alleviate intestinal mucositis caused by 5-FU [105].

Cyclophosphamide (CTX) is a common cancer chemotherapeutic drug in clinics that can cause immunosuppression and intestinal mucosal damage when used in large doses. Zou et al. found that polysaccharides from Codonopsis Radix (CRP) could regulate the imbalance and inhibit the growth of Escherichia coli by promoting the growth of Lactobacillus. CRP has a significant protective effect on immunosuppression, especially on mucosal immune damage caused by CTX, and is a potential source of innate immune regulation. A subsequent study on the changes in immune indexes further confirmed that the intestinal immune system might be the active target site of polysaccharides isolated from Codonopsis Radix [106], [107]. In the immunosuppressive model of C57BL/6 mice induced by CTX, Huang et al. found the co-treatment with Panacis Quinquefolii Radix polysaccharide and ginsenoside (AGP_AGG) could alleviate the side effects of CTX by regulating intestinal flora and related metabolism. Research showed that the increase in the beneficial bacteria Lactobacillus, Bifidobacterium, Parasutterella, Lachnospiraceae, etc., were positively correlated with the curative effect of AGP_AGG [108]. AA-2βG also showed regulatory and immunomodulatory effects on intestinal and colonic microflora in mice in similar models. Especially in the microflora of the mouse colon and small intestine, the relative abundance of Muribaculaceae increased significantly, although the interaction between AA-2βG and mouse lipids needs further investigation [109]. Recently, Sun et al. found that the active fraction from Polygonati Rhizoma (PSE30 and PSE75) could enhance the immunity by improving intestinal microorganisms and activating macrophages in the immunosuppressive mouse model constructed by CTX. The preliminary study showed that PSE75 could call back the CTX-induced increased abundance of Bacteroidales, Lachnospiraceae, Lachnospiraceae, and Lachnospiraceae and increase the relative abundance of Lactobacillus, Desulfovibrio, and Enterorhabdus, indicating that PSE75 is expected to be an excellent immune booster [110].

Drug abuse is an undesirable behavior that violates medical use and social norms and can easily cause physical, emotional, mental, or sensory damage. Morphine dependence is a persistent brain disease caused by the opioid morphine, which causes significant physical and mental damage to abusers. Ginsenoside Rg1, an herbal-based dietary supplement derived from Ginseng Radix Et Rhizoma, was shown to be helpful in protection against morphine dependence. Significantly increased B. vulgatus after ginsenoside Rg1 administration inhibited tryptophan metabolism and reduced serotonin (5-HT) and 5-hydroxytryptamine receptor 1B (5-HTR1B)/5-hydroxytryptamine receptor 2A (5-HTR2A) levels, thus inhibiting excitatory nerve conduction and alleviating morphine dependence. This seminal evidence suggested that ginsenoside Rg1 intervention in gut microbiome-tryptophan metabolism-serotonin signaling in morphine disease may be a novel strategy for treating drug dependence [111].

Neurological and psychiatric disorders

Neurological and psychiatric disorders are a widespread group of chronic diseases that cause severe mental and physical health problems for patients. Many studies have highlighted the vital role of gut microbiota in the occurrence and development of these diseases. Biological products produced by gut microbiota act as messengers through the gut-brain axis [112]. Thus, many medicine food homology species have been shown to alleviate neurological and psychiatric disorders by targeting gut microbiota to regulate the gut-brain axis (Table 3).

Table 3.

Effects of medicine food homology species on neurological and psychiatric disorders via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Poria extract ↓Cyanobacteria; Proteobacteria; Roseburia; Bacteroides; Deferribacteraceae, Enterobacteriales
↑Lactobacillus
↓Aβ plaques; secondary BAs; BMAA; inflammatory male APP/PS1 mice 1.2 g/kg AD [116]
Astragali
Radix
flavonoids ↑Roseburia; Lactobacillus; Allobaculum ↓AGEs; Aβ protein
↑SCFAs
male C57BL/6J mice
(10 weeks old)
50 mg/kg AD [117]
Gastrodiae Rhizoma extract ↓Rikenelleos-RC9
↑Bacilli; Lactobacillus johnsonii; Lactobacillus murinus; Lactobacillus reuteri.
↓P-TauThr231 levels SPF mice
(8 weeks old)
300 mg/kg AD [118]
Astragali
Radix
polysaccharide ↓Osscillospira; Mucispirillum
↑Lactobacillus; Sutterella
↓metabolic disorders
↑SCFAs
male C57BL/6J mice 600 mg/kg AD [120]
Panacis Quinquefolii Radix Panacis Quinquefolii Radix ↑Akkermansia muciniphila; Lactobacillus ↑SCFAs Human
(18–40 years old)
200 mg/day AD [119]
Sojae Semen Praeparatum Semen Sojae Praeparatum ↓Bacteroides
↑Ruminococcaceae_UCG-008
↓SCFAs;
↑5-HT; GABA; BDNF; NE
male SD rats 0.97 g/kg Depression [127]
Cistanches Herba extract ↓Bacteroides
↑Ruminococcus
↓SCFAs
↑5-HT; GABA; BDNF; NE
male SD rats 400 mg/kg Depression [128]
Poria triterpenoids extracts ↓ [Prevotella; Allobaculum; Ochrobactrum ↓palmitoylcarnitine; enoxolone
↑taurocholic acid; citric acid; creatine
male SD rats 15 g/kg Depression [129]
Poria polysaccharide ↓Blautial
↑Ruminococcus; Prevotella;
↓inflammatory male Wistar rats
(8 weeks old)
100 mg/kg Anxiety [138]

Neurological disorders

As the most common neurodegenerative disease, Alzheimer's disease (AD) has a complex pathogenesis. Extracellular deposition of amyloid beta protein (Aβ) and intracellular neurofibrillary tangles of tau protein are typical pathological markers [113], [114]. In addition, in pathological conditions, various factors, such as most secondary BAs and endotoxins, can promote the generation of reactive oxygen species and induce neuroinflammation, thereby causing brain damage [115]. The later stages of AD are accompanied by increasingly severe cognitive impairment and no effective therapeutic drugs. The gut microbiota is known as “the second brain “. There is mounting evidence that the microbiota-gut-brain axis has a role in regulating the progression of AD (Fig. 5c), such as butyrate, a metabolite of gut microbes, can cross the blood–brain barrier (BBB) to reduce Aβ aggregation. Therefore, “drugging the microbiome “may be a new strategy for neurological disease treatment.

Fig. 5.

Fig. 5

Scheme illustrating the role of Poria in AD treatment. (a) Regulation of gut-brain axis with Poria intervention. (b) In the dysregulation of gut microbiota, the increase of harmful bacteria and pro-inflammatory factors in the gut leads to increased intestinal and blood–brain barrier permeability. The ensuing peripheral immune response over activates the microglia and weakens their ability to clear damaged neurons and proteins. In addition, harmful secretions (such as BMAA) are produced by the corresponding bacteria and accelerate the formation of Aβ plaques. All these adverse factors were reversed with the Poria intervention. (c) The homeostatic gut interacts with the brain through neural, metabolic, endocrine, and immune pathways. Figure created via BioRender.com.

Due to its unique advantages, traditional Chinese herbal medicine has shown significant effects on various diseases with complex mechanisms, especially in the field of neuroprotection. Poria, a conventional herbal and dietary supplement, dramatically reduced the abundance of Roseburia and Bacteroides, which can convert primary BAs into secondary BAs and thus help to alleviate AD-related pathologies by restoring BA homeostasis. Meanwhile, Cyanobacteria, which produces toxic amino acids (β-N-methylamino-L-alanine, BMAA) that misfold Aβ protein, has also been significantly inhibited in reducing Aβ plaques. Deferribacteraceae, Enterobacteriales, and Proteobacteria produce endotoxins and induce neuroinflammation, resulting in excessive activation of microglia and astrocytes and thus leading to a decrease in Aβ proteins clearance. All these harmful bacteria were reduced after the Poria intervention. Moreover, the abundance of Lactobacillus, a probiotic that produces the neurotransmitter γ-aminobutyric acid (GABA), was increased (Fig. 5b) [116]. In another study, decreased levels of advanced glycosylation end products (AGEs) in serum also contributed to retard deposition of Aβ protein in the brain, as the increased abundance of Roseburia and Lactobacillus by flavonoids from Astragali Radix [117]. Notably, Gastrodiae Rhizoma, a medicine food homology species, improved the cognitive impairment in AD mice by reducing the intraneuronal accumulation of hyperphosphorylated TauThr231 in a dose-dependent manner. Increased abundance of Lactobacillus johnsonii, Lactobacillus murinus, Lactobacillus reuteri, etc., and reversed Rikenelleos-RC9 might be a possible mechanistic connection between gut microbiota dysregulation and P-TauThr231 levels in AD progression [118]. SCFAs, especially butyrate, were significantly increased after intervention with Panacis Quinquefolii Radix and Astragali Radix, which linked with the observed increase in Allobaculum, Akkermansia mucinphila and Lactobacillus, etc. [119], [120].

Psychiatric disorders

Depression is a pervasive and incapacitating mental disorder that has become one of the most concerning diseases [121]. The pathogenesis of depression is extremely complex. Decreased levels of monoamines in the brain's nervous system are among the most accepted hypotheses, such as GABA, norepinephrine (NE), and serotonin (5-HT) [122], [123]. Brain-derived neurotrophic factor (BDNF) is a key neuronal substrate for regulating depressive-like behavior, and levels of BDNF in the hippocampus and serum are downregulated by severe psychological stress and mood disorders [124], [125], [126].

Sojae Semen Praeparatum (SSP), a traditional edible herb, showed an antidepressant effect in a chronic unpredictable mild stress (CUMS)-induced mice model. Specifically, this species enhanced the population of the Ruminococcaceae_UCG-008 to slow the depression-related drop in GABA and BDNF. Meanwhile, 5-HT, BDNF, and NE levels were noticeably rising while Bacteroides was reducing [127]. Contrarily, Cistanches Herba demonstrated the exact opposite regulatory impact of SSP on Bacteroides and Ruminococcus while also being able to raise 5-HT levels in the hippocampal region in the same paradigm as described above [128]. Qin et al. also observed that triterpenoids extract from Poria could increase intestinal microbial diversity in CUMS rats and exert antidepressant effects by reducing the abundance of Prevotella, Allobaculum and Ochrobactrum to regulate pathways such as inflammation, energy metabolism, and immunity [129]. In addition, growing preclinical evidence points to the gut-brain axis through the vagus nerve as a critical role in the bidirectional communication between gut microbiota and the brain [130], [131]. For instance, subdiaphragmatic vagotomy stopped the manifestation of depressive-like behavior in rodents [132], [133], [134], [135], [136]. It's apparent that the MFH species are highly correlated with the vagal pathway in the aspect of combating psychiatric disorders, but the underlying mechanism awaits further investigation.

Anxiety is another psychiatric disorder that often accompanies depression. The core symptoms of the disease are excessive nervousness and worry, accompanied by symptoms such as insomnia, which seriously affect the quality of patients’ life [137]. Poria has been widely used to treat restlessness as an ancient medicine and modern functional food. Ye et al. demonstrated that Poria exerted anxiolytic effects by reducing Blautial and increasing Ruminococcus and Prevotella, which were associated with decreased inflammatory response in a rat model of anxiety induced by chronic sleep deprivation [138]. In addition, Lilii Bulbus and Ziziphi Spinosae Semen are traditional Chinese herbs used for treating insomnia for thousands of years. Modern studies revealed that these species ameliorated the disturbance of gut microbiota caused by insomnia and significantly regulated the gut microbiota-related metabolites, thus showing therapeutic effects through the microbe-gut-brain axis [139], [140].

Cardiovascular diseases

Atherosclerosis (AS) is a risk factor for cardiovascular diseases, which has become the leading cause of death worldwide [141]. Various factors can predispose to AS, particularly dyslipidemia. Natural species have unique advantages in regulating these indicators (Table 4). In the current study, the active ingredient, ginsenoside Rc (GRc), alleviates AS by modifying the gut microbiota and fecal metabolites. At the genus levels, the relative abundances of the beneficial bacteria Muribaculaceae, Lactobacillus, Ileibacterium, and Bifidobacterium increased significantly under the GRc intervention. In contrast, the abundance of the harmful bacteria Faecalibaculum, Oscillibacter, and Eubacterium_coprostanoligenes_group decreased. These changes were closely related to a variety of metabolites involved in the metabolism of taurine and purine, ATP binding cassette (ABC) transporters, the biosynthesis of primary BAs and arginine, the tricarboxylic acid (TCA) cycle, and other pathways [142]. Circulating trimethylamine N-Oxide (TMAO), derived from the oxidation of trimellitic anhydride (TMA) by liver-secreted flavin monooxygenase 3 (FMO3), can be used as a predictive signal for the early stages of AS, which can influence cholesterol metabolism and oxidative stress. Ginkgolide B (GB) is a naturally occurring chemical derived from Ginkgo Semen that has been identified as a therapy for AS. The mRNA and protein expression of FMO3 were dramatically suppressed by GB therapy, leading to a decrease in TMA and TMAO concentrations, which were linked to the altered gut microbiota in HFD mice. Increased Bacteroides and decreased Helicobacter might contribute to the anti­atherosclerotic effects of GB [143]. 7α-hydroxylase (CYP7A1) is a specific rate-limiting enzyme. Polyphenol extract and essential oil from Tsaoko Fructus altered the abundance of Allobaculum, Desulfovibrio, and Ruminococcus_2, the gut microbiota associated with CYP7A1, and upregulated the expression of CYP7A1 at both the mRNA and protein levels, thus exhibiting cholesterol-lowering effect by converting cholesterol into BAs and excreting them out [144]. Additionally, intervention with Ganoderma and its active ingredient could also ameliorate dyslipidemia by selectively promoting the growth of specific benign bacteria (Prevotella and SCFAs producers) [145], [146].

Table 4.

Effects of medicine food homology species on cardiovascular diseases via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Ginseng Radix Et Rhizoma ginsenoside Rc ↓Faecalibaculum; Oscillibacter; Eubacterium_coprostanoligenes_group
↑Muribaculaceae; Lactobacillus; Ileibacterium; Bifidobacterium
↓uric acid; adenine; fumaric acid; malic acid; citric acid; cholesterol
↑L-glutamine; ABC
Transporter; BAs; FXR; TGR
male C57/BL6 ApoE−/−mice
(7 weeks old)
40 mg/kg AS [142]
Ginkgo Semen Ginkgolide B ↓Helicobacter
↑Bacteroides
↓TMA; TMAO; cholesterol
↑FMO3
male C57/BL6 ApoE−/− mice
(8 weeks old)
30 mg/kg AS [143]
Tsaoko Fructus Polyphenol extract/
essential oil
↓Allobaculum, Desulfovibrio
↑Ruminococcus_2
↓cholesterol
↑CYP7A1
male Golden Syrian hamsters
(8 weeks old)
1000 mg/kg 200 mg/kg AS [144]
Ganoderma extract ↓Turicibacter; Clostridium XVIII
↑Prevotella; Alloprevotella; Ruminococcus
↓blood lipid levels
↑SCFAs
male Wistar rats 150 mg/kg AS [145]
Ganoderma Ganoderic acid A ↑Alistipes; Bacteroides ↓blood lipid levels
↑SCFAs
male SPF Kunming mice
(6 weeks old)
75 mg/kg AS [146]

Endocrine diseases

Diabetes is a metabolic disorder in which hyperglycemia results from inadequate insulin production and/or action. According to 2021 data from the International Diabetes Federation, it makes diabetes prevention and treatment are a global priority, with 537 million people living with diabetes [147]. Growing evidence suggests that the gut microbiota is crucial in controlling host physiology and metabolism, and its dysbiosis is associated with the progression of diabetes [148], [149]. Therefore, targeting the gut microbiota and its metabolites has become a required field in diabetes prevention and treatment (Table 5).

Table 5.

Effects of medicine food homology species on endocrine diseases via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Lycii Fructus polysaccharides ↓Allobaculum; Dubosiella; Romboutsia
↑Bacteroides; Ruminococcaceae_UCG-014; Mucispirillum; Intestinimonas; Ruminococcaceae_UCG-009
↓inflammatory;
↑SCFAs; insulin
male SPF C57BL/6 mice
(6 weeks old)
200 mg/kg Diabetes [150]
Lycii Fructus polysaccharides ↑Allobaculum ↓inflammatory
↑intestinal barrier
male C57BL/6 mice
(5 weeks old)
Caco-2 cells
200 mg/kg
200 μg/kg
Diabetes [154]
Portulacae Herba extract ↓Firmicutes ↓BCAAs male C57BL/6J mice
(3 weeks old)
200 mg/kg Diabetes [158]
Siraitia Fructus mogrosides ↓Lachnospiraceae_UCG-004; Desulfovibrio; Escherichia Shigella
↑Elusimicrobium,
↓blood glucose; insulin resistance; 1β-hydroxycholic acid
↑SCFAs
male SD rats 20 mg/kg Diabetes [155]
Mori Folium extract ↓Romboutsia; Oscillatoriales_cyanobacterium
↑Alloprevotella; Parabacteroides; Muribaculaceae
↓inflammatory; blood glucose; insulin resistance Male C57BJ/6J mice
(8 weeks old)
250 mg/kg Diabetes [156]

LBPs altered the gut microbiota composition in diabetic mice, specifically increasing SCFAs-producing bacteria Ruminococcaceae_UCG-014 and Intestinimonas. Then, the increased SCFAs promoted insulin secretion and improved insulin sensitivity by directly activating the GPCRs (GPR41 and GPR43) receptor to increase the secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) by intestinal L cells [150]. In addition, increased intestinal permeability and chronic inflammation due to intestinal barrier dysfunction are thought to be responsible for impaired islet β cell function and insulin signaling [151], [152]. SCFAs exert a protective effect on the intestinal barrier by promoting epithelial cell proliferation, increasing MUC2 gene expression, and promoting tight junction assembly [153]. Here, SCFAs were elevated, and LPS was decreased by altering the specific gut microbiota after LBPs intervention, thereby repairing the intestinal barrier, alleviating the inflammatory response, and relieving blood glucose levels in diabetic mice [150], [154]. The disturbance of gut microbiota in diabetic mice was restored by Siraitia Fructus (SF) intervention. The relative abundance of Elasimicrobium negatively correlated with fasting glucose and insulin resistance was significantly increased, while Lachnospiraceae_UCG-004 positively correlated with these factors was reduced. Additionally, the inhibition effect of SF and its component on opportunistic pathogens Desulfovibrio and Escherichia Shigella might contribute to diabetes treatment [155]. Mori Folium also alleviated diabetes by mediating the host–microbial metabolic axis. The bacteria Alloprevotella, Parabacteroides, Muribaculaceae, Romboutsia, and Oscillatoriales_cyanobacterium associated with lactic acid and other metabolites were all reversed under Mori Folium intervention [156]. Serum-branched-chain amino acids (BCAAs) levels have been reported to be associated with insulin resistance [157]. Portulacae Herba extract intervention reduced the biosynthesis of BCAAs, which was mainly associated with the phylum Firmicutes, thereby reducing the level of serum BCAAs and alleviating the progression of diabetes [158].

Others

Medicine food homology species targeting the gut microbiota regulate various diseases (Table 6). For example, chronic nonbacterial prostatitis (CNP) is a common urological disease with a high incidence and low cure rate. Previous studies have demonstrated that Poria polysaccharides (PPs) effectively relieved CNP by regulating the gut microbiota [159], [160]. Parabacteroides, Fusicatenibacter, and Parasutterella, as well as their metabolites haloperidol glucoside acid and 7-ketodeoxycholic acid, were significantly enriched by PPs in colonic epithelium, altering the expression of several critical genes in colon epithelium. Meanwhile, the ratio of dihydrotestosterone to estradiol (DTH/E2) was regulated, prostate inflammation was eventually suppressed, and CNP was reduced. Among them, haloperidol glucuronide and 7-ketodeoxycholic acid could be the signaling molecules of the “gut-prostate” axis to alleviate CNP [161]. These results provide new guidelines for treating CNP using natural-derived species to target the gut microbiota.

Table 6.

Effects of medicine food homology species on other diseases via gut microbiota.

Species Category of Active Ingredient Microbiota Findings Mechanism Animals Dose Disease Ref.
Poria polysaccharides ↑Parabacteroides; Fusicatenibacter; Parasutterella ↓inflammatory; Cyp1a1; Hsd17b7
↑Pla2g2f; Alox15; DTH/E2
male SD rats
(7–8 weeks old)
250 mg/kg Chronic nonbacterial prostatitis [161]
Astragali Radix polysaccharides ↓Bifidobacterium_pseudolongum
↑Lactobacillus_johnsonii
↓MDSC
↑immunity; CD8 T cells; L-glutamate; creatine
male C57BL/6 mice
(5–6 weeks old)
200 mg/kg Malignant melanoma [162]
Poria extract ↓Desulfovibrio; Mucispirillum
↑Lactobacillus; Bifidobacterium
↓inflammatory; putrescine
↑intestinal barrier
female (BALB/c mice
(4 weeks old)
700 mg/kg Breast cancer [163]
Eucommiae Folium extract ↑Lactobacillus bulgaricus ↑SCFAs; gut microbiota diversity male SAMP6 mice
(16 weeks old)
3.0 g/kg osteoporosis [164]
Poria poricoic acid A ↓Escherichia_Shigella; Blautia; Enterorhabdus; Parasutterella ↓inflammatory; polyamine metabolites; glycine-conjugated compounds; male SD rats 10 mg/kg chronic kidney disease [165]

Malignant melanoma is an invasive skin cancer characterized by high mortality and poor prognosis. Myeloid-derived suppressor cells (MDSCs) can interfere with antitumor activity by impairing the tumor-killing effect of CD8 T cells. Administration of Astragali Radix polysaccharides (APs) significantly decreased the number of MDSCs and the expression of the MDSCs-related molecule while increasing the number of CD8 T cells. It was shown that the ability of MDSCs to inhibit CD8 T cells from killing tumor cells was weakened. Subsequent FMT and antibiotic (ABX) interference experiments demonstrated that this function was achieved by regulating the gut microbiota. L-glutamate and creatine, which were negatively correlated with Bifidobacterium_pseudolongum and positively correlated with Lactobacillus_johnsonii, were significantly upregulated under APs intervention. All these data revealed the therapeutic capacity of APs to inhibit tumor growth, which was related to the degenerative immunosuppressive efficiency of MDSCs and depended on the remodeling and metabolic function of the gut microbiota [162].

Medicine food homology species also show profound and extensive effects targeting the gut microbiota. Poria improved dysbacteriosis in breast cancer mice by boosting the beneficial bacteria Lactobacillus and Bifidobacterium and decreasing the sulfate-reducing bacteria Desulfovibrio and the inflammatory-associated bacteria Mucispirillum, etc. [163]. In vivo studies in the senescence-accelerated mouse model proved that Eucommiae Folium extract supplementation promoted the growth of Lactobacillus bulgaricus, increased the concentration of SCFAs in feces and serum, and exhibited anti-osteoporosis activity [164]. As a new dietary and therapeutic strategy, Poria supplementation could delay the development of chronic kidney disease by regulating the gut microbiota and its metabolites [165].

Bioconversion of active constituents by gut microbiota

The gut microbiota is not only regulated by ingested substances but also has the tremendous capability to convert the active ingredients of these species into diverse xenobiotics. The metabolization of the azo drug Prontosil into sulfonamide with significant antimicrobial activity by azo reductase is a classic example of microbiome-driven activation of precursor drugs [166]. Oral administration is the dominant use in medicine food homology species, which inevitably leads to direct contact and interaction between the gut microbiota and these species. The essence of the conversion of these natural-derived active components by gut microbiota is chemical reactions that occur when enzymes produced by gut microbiota modify the structure of these exogenous substrates. The modified structures show altered physicochemical and biological properties that enhance the therapeutic efficacy and increase the biological diversity of ingested natural products [167], [168]. Compared with the modification of these natural products by chemical synthesis, the biotransformation of gut microbiota has unique advantages: 1) Most of the reactions driven by the gut microbiota secreted catalytic enzymes are challenging to complete by chemical synthesis; 2) Enzymatic reactions are highly selective, especially without the need to protect the groups that do not participate in the conversion; 3) The reaction conditions of biotransformation are relatively mild, which can maximize the protection of the active ingredients from the damage of external conditions, such as high temperature and pressure [169], [170], [171]. The biotransformation of gut microbiota mainly includes demethylation, deprenylation, deglycosylation, dihydroxylation, and acetylation, and some representative examples are listed below (Fig. 6).

Fig. 6.

Fig. 6

Modification of bioactive ingredients from medicine food homology species by gut microbiota.

Demethylation and deprenylation

Furanocoumarins are bioactive ingredients in foods and herbs and exhibit various pharmacological activities, including anti-inflammatory, anti-cancer, neuroprotection, and so on [172]. However, oral administration of furanocoumarins may also be phototoxic. Furanocoumarin ingestion and subsequent exposure to ultraviolet have been reported to cause photosensitive dermatitis [173]. In addition, furanocoumarins inhibit the drug-metabolizing enzyme CYP3A in the liver, which prolongs the drug's duration of action and leads to serious adverse effects [174]. Angelicae Dahuricae Radix contains a variety of furanocoumarins, including xanthotoxin, bergapten, imperatorin, and isoimperatorin, etc. In the current study, the Co O-methyltransferase expressed by the gut microbe Blautia sp. MRG-PMF1 lysed the methyl aryl ether and converted xanthotoxin and bergapten to the products xanthotoxol and bergaptol, respectively. Importantly, the prenyl aryl ether group was cleavaged in the same way. Deprenylation of imperatorin and isoimperatorin by gut microbial conversion also resulted in the products described above [175]. These demethylated and deprenylated phenolic metabolites with better solubility can be excreted rapidly with urine and thus serve as its detoxification mechanism [176] (Fig. 6a).

Deglycosylation

Many complex natural products are usually in glycosylated forms, such as ginsenosides. After oral administration of Ginseng, the major ginsenosides are metabolized to deglycosylated products by the gut microbiota before absorption into the bloodstream. The biotransformation of Ginsenoside Rb1 with Bifidobacterium led to the deglycosylated metabolite Rd. And compound K with anticancer and antidiabetic activities was also obtained after subsequent continuous deglycosylation [177]. In addition, deglycosylated metabolites possess potent pharmacological activity compared to basic ginsenosides. For example, the 20(S)-protopanaxatriol (PPT) obtained from the deglycosylation of Rg1 has a stronger anti-fatigue activity than the original saponin [178]. Calycosin-7-O-β-D-glucoside (CG) has been investigated intensively as the major isoflavonoids in Astragali Radix and has been reported to possess extensive pharmacological properties. It was reported that aglycone calycosin was obtained from the deglycosylation of CG by Bacteroides sp.13 isolated from human gut bacteria [179]. Importantly, it has a more vital antiviral ability than the corresponding glycoside [180] and can protect PC12 cells from glutamate-induced injury, while CG cannot [181]. These results indicate that the deglycosylation of natural products with gut microbiota is critical to finding new therapeutic molecules with more potent and diverse pharmacological activities (Fig. 6b).

Dehydroxylation and acetylation

Dehydroxylation is also one of the main metabolic ways of natural products by gut microbiota. For example, Formononetin is a well-known phytoestrogen used to prevent and treat breast, prostate, and colon cancers and osteoporosis [182], [183]. It was reported that Bacteroides sp.58 from human intestinal bacteria could dehydroxylate calycosin and convert it into Formononetin. In addition, the acetylation of calycosin was accomplished by strain Clostridium sp.21–2 [179] (Fig. 6c).

Conclusion and future perspectives

To sum up, as a “forgotten organ”, the gut microbiota plays a crucial role in regulating human health. Targeting the gut microbiota by medicine food homology species to affect the host favorably is the dawn of disease prevention and treatment. This review summarizes the interaction between MFH species and gut microbiota and its effects on human health from a macro point of view. The changes in the composition and metabolites of gut microbiota and the conversion of natural active components after MFH species intervention have proved these interactions. According to the results of literature research, species from Rosales are the most abundant in regulating the involvement of gut flora in host pathophysiology (Fig. 7a). In addition, the statistical results also revealed that saccharides are the most widely studied and widely used (Fig. 7b). These results provide a direction for further searching for MFH species and active ingredients.

Fig. 7.

Fig. 7

Distribution of (a) medicine food homology species and (b) bioactive ingredients from medicine food homology species.

As this review shows, from the initial practical application to more mechanism studies, the relationship between MFH–gut microbiota–human health has evolved into an irrefutable interaction. But obviously, there are still a lot of challenges:

  • (I)

    The category of MFH species needs to be further clarified. This may require screening and research performed by various countries and regions in combination with the actual local situation, as each region has different dietary sources and habits that must be taken into account, and drastic changes in diet can upset the body's balance and even lead to more complex health problems. In addition, medical institutions or the government should constantly update the standards and catalogs of MFH since they are a continuous development process.

  • (II)

    Most existing studies only establish the association among MFH, gut microbiota, and disease phenotype, but it is not causal. Future research should further clarify the causal relationship among MFH, gut microbiota, and therapeutic effect, especially at the cellular and molecular level, which is helpful for the transformation from experimental research to clinical application.

  • (III)

    Although it has been proven that the regulation of gut microbiota is an effective method for disease treatment. How to define healthy gut microbiota, quickly detect and analyze, and select appropriate MFH for individuals are worthy of in-depth study because there are significant differences in individuals' gut microbiota. Besides, eating habits, living environment, and other factors are individual independent variables. Therefore, a new method of observation and analysis may be proposed to achieve detailed communication and more insights into diet, gut microbiome population, and their relationship with host health, thus obtaining personalized healthy diet guidance and disease treatment for human beings [184].

  • (IV)

    Nowadays, the discoveries related to the role of MFH, gut microbiota, and human disease state are exploding. Efforts should be made to integrate, process, and analyze this massive data to transform it into useful and applicable knowledge [185]. This may require new methods such as omics analysis [186], [187], [188], [189] and machine learning [190], [191]. These methods are expected to accurately predict the disease state and find appropriate treatment means by using more samples across the population and time.

  • (V)

    Explaining the advantages of the MFH theory in managing gut microbiota requires an advanced theory and a broader perspective, both of which are made possible by the rapid development of modern means. This requires continuous cooperation in various areas, such as traditional Chinese medicine, neurology, microbiology, and life sciences, to develop comprehensive and relevant approaches to identify the mechanisms of action that are still based on observations. At the same time, more efforts should be made to translate these findings into improved human health [192], [193], [194], [195], [196].

Looking ahead, the knowledge and experience accumulated by studying the effects of MFH on gut microbiota will open a new field to prevent and treat human diseases. It is hoped that the treatment of complex diseases and the regulation of gut microbiota can find more “overlap” in MFH, and more clinicians will regulate the microbiota through MFH and use it as an alternative for disease prevention and diagnosis.

Compliance with ethics requirements

This article does not contain any studies with human or animal subjects.

CRediT authorship contribution statement

Wei-Fang Zuo: Writing – original draft, Writing – review & editing, Visualization. Qiwen Pang: Writing – original draft, Writing – review & editing, Visualization. Lai-Ping Yao: Writing – review & editing, Formal analysis. Yang Zhang: Formal analysis, Visualization. Cheng Peng: Funding acquisition. Wei Huang: Conceptualization, Funding acquisition. Bo Han: Conceptualization, Supervision, Funding acquisition.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

We are grateful for financial support from the National Natural Science Foundation of China (Grant Nos. 82073998 and 82073997), Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (Grant Nos. ZYYCXTD-D-202209).

Biographies

graphic file with name fx1.jpg

Wei-Fang Zuo was born in Sichuan Province, China, in 1997. She obtained her Master's degree from Chengdu University of Traditional Chinese Medicine in 2022. She is now pursuing her Ph.D. at the same university under the supervision of Prof Bo Han and Prof Wei Huang. Currently, her main research interests are synthesizing and modifying active compounds in traditional Chinese medicine and elucidating the pharmacological mechanisms and metabolic processes of bioactive substances.

graphic file with name fx2.jpg

Qiwen Pang was born in 1996 in Sichuan Province, China. In 2019, he received his Bachelor's degree in Pharmaceutical Engineering from Jiangxi University of Science and Technology, College of Pharmacy. Now, he is a postgraduate student in the School of Pharmacy at Chengdu University of Traditional Chinese Medicine. His research interests are structural modification, activity screening, and mechanism investigation of active ingredients of traditional Chinese medicine and natural products.

graphic file with name fx3.jpg

Laiping Yao received her Bachelor's degree in Traditional Chinese Medicine from the School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, College of Science and Technology in 2022. Now, she is pursuing her Master's degree at the College of Pharmacy, Chengdu University of Traditional Chinese Medicine. Her main research interests are the discovery and structural modification of bioactive components in traditional Chinese medicine.

graphic file with name fx4.jpg

Yang Zhang was born in 2001 in Chongqing Municipality, China. He enrolled at Chengdu University of Traditional Chinese Medicine in 2019. Now, he is pursuing his Bachelor's degree in Chinese Pharmacy at the College of Pharmacy, Chengdu University of TCM.

graphic file with name fx5.jpg

Cheng Peng was born in 1964 in Sichuan, China. He graduated from Chengdu University of Traditional Chinese Medicine with a degree in Chinese medicine and received his Ph.D. in 1996. He joined the Chengdu University of Traditional Chinese Medicine in 1986 and became a professor in 1999. He is currently the Vice President of the Chengdu University of Chinese Medicine and the Director of the State Key Laboratory of Southwest Specialty Chinese Medicine Resources. His research interests are based on the discovery of active ingredients in traditional Chinese medicine and natural products, the investigation of the relationship between toxicity and therapeutic efficacy, and the study of the mechanism of action and biotransformation of bioactive molecules in the intestinal microbiota. He has received many awards, including the National Science and Technology Progress Award, and is a member of the Chinese Pharmacopoeia Commission.

graphic file with name fx6.jpg

Wei Huang was born in Chengdu, China in 1980. She received her B.S. and M.S. degrees from West China College of Pharmacy, Sichuan University, in 2003 and 2006, respectively, and her Ph.D. degree in Chinese medicine from Chengdu University of Traditional Chinese Medicine in 2010. Currently, she is a professor at the College of Pharmacy, Chengdu University of Chinese Medicine. Her main research interests are chemical modification, structure-activity relationships, and potential molecular mechanisms of active ingredients in traditional Chinese medicine and natural products.

graphic file with name fx7.jpg

Bo Han received his Ph.D. degree from the West China School of Pharmacy, Sichuan University, in 2010. In 2017–2018, he was a visiting scientist at RNA Institute, State University of New York, USA. Since 2015, he has worked as a Professor at the State Key Laboratory of Southwestern Chinese Medicine Resources at Chengdu University of TCM. His interest is in the design, synthesis and modification of lead compounds based on biologically active compounds discovered from traditional medicinal plants and the study of their pharmacological efficacy and mechanism of action.

Contributor Information

Wei Huang, Email: huangwei@cdutcm.edu.cn.

Bo Han, Email: hanbo@cdutcm.edu.cn.

References

  • 1.Bäckhed F., Ley R.E., Sonnenburg J.L., Peterson D.A., Gordon J.I. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915–1920. doi: 10.1126/science.1104816. [DOI] [PubMed] [Google Scholar]
  • 2.Sender R., Fuchs S., Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337–340. doi: 10.1016/j.cell.2016.01.013. [DOI] [PubMed] [Google Scholar]
  • 3.Citi S. Intestinal barriers protect against disease. Science. 2018;359(6380):1097–1098. doi: 10.1126/science.aat0835. [DOI] [PubMed] [Google Scholar]
  • 4.Hooper L.V., Littman D.R., Macpherson A.J. Interactions between the microbiota and the immune system. Science. 2012;336(6086):1268–1273. doi: 10.1126/science.1223490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Sanna S., van Zuydam N.R., Mahajan A., Kurilshikov A., Vich Vila A., Võsa U., et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet. 2019;51(4):600–605. doi: 10.1038/s41588-019-0350-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ma C., Han M., Heinrich B., Fu Q., Zhang Q., Sandhu M., et al. Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360(6391) doi: 10.1126/science.aan5931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cheng H., Liu J., Zhang D., Tan Y., Feng W., Peng C. Gut microbiota, bile acids, and nature compounds. Phytother Res. 2022;36(8):3102–3119. doi: 10.1002/ptr.7517. [DOI] [PubMed] [Google Scholar]
  • 8.Wang Z., Klipfell E., Bennett B.J., Koeth R., Levison B.S., DuGar B., et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi: 10.1038/nature09922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tang W.H.W., Wang Z., Levison B.S., Koeth R.A., Britt E.B., Fu X., et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–1584. doi: 10.1056/NEJMoa1109400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhang T., Sun P., Geng Q., Fan H., Gong Y., Hu Y., et al. Disrupted spermatogenesis in a metabolic syndrome model: the role of vitamin A metabolism in the gut–testis axis. Gut. 2022;71(1):78. doi: 10.1136/gutjnl-2020-323347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Qi Q., Li J., Yu B., Moon J.-Y., Chai J.C., Merino J., et al. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut. 2022;71(6):1095. doi: 10.1136/gutjnl-2021-324053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Agus A., Planchais J., Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–724. doi: 10.1016/j.chom.2018.05.003. [DOI] [PubMed] [Google Scholar]
  • 13.Devlin A.S., Marcobal A., Dodd D., Nayfach S., Plummer N., Meyer T., et al. Modulation of a circulating uremic solute via rational genetic manipulation of the gut microbiota. Cell Host Microbe. 2016;20(6):709–715. doi: 10.1016/j.chom.2016.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Dodd D., Spitzer M.H., Van Treuren W., Merrill B.D., Hryckowian A.J., Higginbottom S.K., et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652. doi: 10.1038/nature24661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Flint H.J. Gut microbial metabolites in health and disease. Gut Microbes. 2016;7(3):187–188. doi: 10.1080/19490976.2016.1182295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wilmes P., Martin-Gallausiaux C., Ostaszewski M., Aho V.T.E., Novikova P.V., Laczny C.C., et al. The gut microbiome molecular complex in human health and disease. Cell Host Microbe. 2022;30(9):1201–1206. doi: 10.1016/j.chom.2022.08.016. [DOI] [PubMed] [Google Scholar]
  • 17.Zitvogel L., Daillère R., Roberti M.P., Routy B., Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15(8):465–478. doi: 10.1038/nrmicro.2017.44. [DOI] [PubMed] [Google Scholar]
  • 18.Nicholson J.K., Holmes E., Kinross J., Burcelin R., Gibson G., Jia W., et al. Host-Gut Microbiota Metabolic Interactions. Science. 2012;336(6086):1262–1267. doi: 10.1126/science.1223813. [DOI] [PubMed] [Google Scholar]
  • 19.Clemente J.C., Manasson J., Scher J.U. The role of the gut microbiome in systemic inflammatory disease. BMJ (Int Ed) 2018;360 doi: 10.1136/bmj.j5145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mokhtari Z., Gibson D.L., Hekmatdoost A. Nonalcoholic Fatty Liver Disease, the Gut Microbiome, and Diet. Adv Nutr. 2017;8(2):240–252. doi: 10.3945/an.116.013151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sharpton S.R., Ajmera V., Loomba R. Emerging Role of the Gut Microbiome in Nonalcoholic Fatty Liver Disease: From Composition to Function. Clin Gastroenterol Hepatol. 2019;17(2):296–306. doi: 10.1016/j.cgh.2018.08.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Greenhill C. Gut microbiome and serum metabolome changes. Nat Rev Endocrinol. 2017;13(9):501. doi: 10.1038/nrendo.2017.89. [DOI] [PubMed] [Google Scholar]
  • 23.Turnbaugh P.J., Hamady M., Yatsunenko T., Cantarel B.L., Duncan A., Ley R.E., et al. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480–484. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Vatanen T., Franzosa E.A., Schwager R., Tripathi S., Arthur T.D., Vehik K., et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 2018;562(7728):589–594. doi: 10.1038/s41586-018-0620-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Qin J., Li Y., Cai Z., Li S., Zhu J., Zhang F., et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60. doi: 10.1038/nature11450. [DOI] [PubMed] [Google Scholar]
  • 26.Cryan J.F., O'Riordan K.J., Sandhu K., Peterson V., Dinan T.G. The gut microbiome in neurological disorders. Lancet Neurol. 2020;19(2):179–194. doi: 10.1016/S1474-4422(19)30356-4. [DOI] [PubMed] [Google Scholar]
  • 27.Sorboni Shokufeh G., Moghaddam Hanieh S., Jafarzadeh-Esfehani R., Soleimanpour S. A Comprehensive Review on the Role of the Gut Microbiome in Human Neurological Disorders. Clin Microbiol Rev. 2022;35(1):e00338–e420. doi: 10.1128/CMR.00338-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tang W.H.W., Li D.Y., Hazen S.L. Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol. 2019;16(3):137–154. doi: 10.1038/s41569-018-0108-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tang W.H.W., Hazen S.L. The gut microbiome and its role in cardiovascular diseases. Circulation. 2017;135(11):1008–1010. doi: 10.1161/CIRCULATIONAHA.116.024251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ding T., Schloss P.D. Dynamics and associations of microbial community types across the human body. Nature. 2014;509(7500):357–360. doi: 10.1038/nature13178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Qin J., Li R., Raes J., Arumugam M., Burgdorf K.S., Manichanh C., et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Guthrie L, Spencer SP, Perelman D, Van Treuren W, Han S, Yu FB, et al. Impact of a 7-day homogeneous diet on interpersonal variation in human gut microbiomes and metabolomes. Cell Host Microbe. 2022;30 doi: 10.1016/j.chom.2022.05.003. 863–74.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.de la Cuesta-Zuluaga J, Kelley Scott T, Chen Y, Escobar Juan S. Mueller Noel T, Ley Ruth E, et al. Age- and Sex-Dependent Patterns of Gut Microbial Diversity in Human Adults. mSystems. 2019;4(4) doi: 10.1128/msystems.00261-19. e00261–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.David L.A., Maurice C.F., Carmody R.N., Gootenberg D.B., Button J.E., Wolfe B.E., et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vich Vila A., Collij V., Sanna S., Sinha T., Imhann F., Bourgonje A.R., et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat Commun. 2020;11(1):362. doi: 10.1038/s41467-019-14177-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Feng W., Ao H., Peng C., Yan D. Gut microbiota, a new frontier to understand traditional Chinese medicines. Pharmacol Res. 2019;142:176–191. doi: 10.1016/j.phrs.2019.02.024. [DOI] [PubMed] [Google Scholar]
  • 37.Hou Y., Jiang J.-G. Origin and concept of medicine food homology and its application in modern functional foods. Food Funct. 2013;4(12):1727–1741. doi: 10.1039/C3FO60295H. [DOI] [PubMed] [Google Scholar]
  • 38.Feng S., Huang L.Q., Guo J., Chen M. History and development of “One Root of Medicine and Food”. Chin Bull Sci. 2015;27(08):1061–1069. doi: 10.13376/j.cbls/2015146. [DOI] [Google Scholar]
  • 39.Ma A., Zou F., Zhang R., Zhao X. The effects and underlying mechanisms of medicine and food homologous flowers on the prevention and treatment of related diseases. J Food Biochem. 2022 doi: 10.1111/jfbc.14430. [DOI] [PubMed] [Google Scholar]
  • 40.Song D.-x., Jiang J.-g. Hypolipidemic components from medicine food homology species used in China: pharmacological and health effects. Arch Med Res. 2017;48(7):569–581. doi: 10.1016/j.arcmed.2018.01.004. [DOI] [PubMed] [Google Scholar]
  • 41.Ji M.-Y., Bo A., Yang M., Xu J.-F., Jiang L.-L., Zhou B.-C., et al. The pharmacological effects and health benefits of Platycodon grandiflorus—a medicine food homology species. Foods. 2020 doi: 10.3390/foods9020142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Guo P., Zhang B., Zhao J., Wang C., Wang Z., Liu A., et al. Medicine-food herbs against Alzheimer’s disease: a review of their traditional functional features, substance basis, clinical practices and mechanisms of action. Molecules. 2022 doi: 10.3390/molecules27030901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cosnes J, Gower–Rousseau C, Seksik P. Cortot A Epidemiology and natural history of inflammatory bowel diseases. Gastroenterology. 2011;140 doi: 10.1053/j.gastro.2011.01.055. 1785–94.e4. [DOI] [PubMed] [Google Scholar]
  • 44.Zuo T., Ng S.C. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front Microbiol. 2018:9. doi: 10.3389/fmicb.2018.02247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhao L., Xie Q., Evivie S.E., Yue Y., Yang H., Lv X., et al. Bifidobacterium longum subsp. longum K5 alleviates inflammatory response and prevents intestinal barrier injury induced by LPS in vitro based on comparative genomics. J Funct Foods. 2022;92 doi: 10.1016/j.jff.2022.105030. [DOI] [Google Scholar]
  • 46.Parada Venegas D., De la Fuente M.K., Landskron G., González M.J., Quera R., Dijkstra G., et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019:10. doi: 10.3389/fimmu.2019.00277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang D., Shao S., Zhang Y., Zhao D., Wang M. Insight into polysaccharides from Panax ginseng C. A. Meyer in improving intestinal inflammation: modulating intestinal microbiota and autophagy. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.683911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Huang K., Dong W., Liu W., Yan Y., Wan P., Peng Y., et al. 2-O-beta-d-glucopyranosyl-l-ascorbic acid, an ascorbic acid derivative isolated from the fruits of Lycium barbarum L., modulates gut microbiota and palliates colitis in dextran sodium sulfate-induced colitis in mice. J Agric Food Chem. 2019;67(41):11408–11419. doi: 10.1021/acs.jafc.9b04411. [DOI] [PubMed] [Google Scholar]
  • 49.Cao C., Zhu B., Liu Z., Wang X., Ai C., Gong G., et al. An arabinogalactan from Lycium barbarum attenuates DSS-induced chronic colitis in C57BL/6J mice associated with the modulation of intestinal barrier function and gut microbiota. Food Funct. 2021;12(20):9829–9843. doi: 10.1039/d1fo01200b. [DOI] [PubMed] [Google Scholar]
  • 50.Ye X., Pi X., Zheng W., Cen Y., Ni J., Xu L., et al. The Methanol Extract of Polygonatum odoratum Ameliorates Colitis by Improving Intestinal Short-Chain Fatty Acids and Gas Production to Regulate Microbiota Dysbiosis in Mice. Front Nutr. 2022;9 doi: 10.3389/fnut.2022.899421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Guo C., Wang Y., Zhang S., Zhang X., Du Z., Li M., et al. Crataegus pinnatifida polysaccharide alleviates colitis via modulation of gut microbiota and SCFAs metabolism. Int J Biol Macromol. 2021;181:357–368. doi: 10.1016/j.ijbiomac.2021.03.137. [DOI] [PubMed] [Google Scholar]
  • 52.Guo C., Zhang S., Wang Y., Li M., Ding K. Isolation and structure characterization of a polysaccharide from Crataegus pinnatifida and its bioactivity on gut microbiota. Int J Biol Macromol. 2020;154:82–91. doi: 10.1016/j.ijbiomac.2020.03.058. [DOI] [PubMed] [Google Scholar]
  • 53.Zhao H., Cheng N., Zhou W., Chen S., Wang Q., Gao H., et al. Honey Polyphenols Ameliorate DSS-Induced Ulcerative Colitis via Modulating Gut Microbiota in Rats. Mol Nutr Food Res. 2019;63 doi: 10.1002/mnfr.201900638. [DOI] [PubMed] [Google Scholar]
  • 54.Li P., Xiao N., Zeng L., Xiao J., Huang J., Xu Y., et al. Structural characteristics of a mannoglucan isolated from Chinese yam and its treatment effects against gut microbiota dysbiosis and DSS-induced colitis in mice. Carbohydr Polym. 2020;250 doi: 10.1016/j.carbpol.2020.116958. [DOI] [PubMed] [Google Scholar]
  • 55.Ren D.D., Li S.S., Lin H.M., Xia Y.S., Li Z.M., Bo P.P., et al. Panax quinquefolius Polysaccharides Ameliorate Antibiotic-Associated Diarrhoea Induced by Lincomycin Hydrochloride in Rats via the MAPK Signaling Pathways. J Immunol Res. 2022;2022:4126273. doi: 10.1155/2022/4126273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liu Y., Zhao X., Lin T., Wang Q., Zhang Y., Xie J. Molecular mechanisms of polysaccharides from Ziziphus jujuba Mill var. spinosa seeds regulating the bioavailability of spinosin and preventing colitis. Int J Biol Macromol. 2020;163:1393–1402. doi: 10.1016/j.ijbiomac.2020.07.229. [DOI] [PubMed] [Google Scholar]
  • 57.Li C., Zhang W., Wu X., Cai Q., Tan Z., Hong Z., et al. Aromatic-turmerone ameliorates DSS-induced ulcerative colitis via modulating gut microbiota in mice. Inflammopharmacology. 2022;30(4):1283–1294. doi: 10.1007/s10787-022-01007-w. [DOI] [PubMed] [Google Scholar]
  • 58.Wu J., Li C., Bai L., Wu J., Bo R., Ye M., et al. Structural differences of polysaccharides from Astragalus before and after honey processing and their effects on colitis mice. Int J Biol Macromol. 2021;182:815–824. doi: 10.1016/j.ijbiomac.2021.04.055. [DOI] [PubMed] [Google Scholar]
  • 59.Ruan J.-Q., Li S., Li Y.-P., Wu W.-J., Lee S.-M.-Y., Yan R. The Presystemic Interplay between Gut Microbiota and Orally Administered Calycosin-7-O-β-d-Glucoside. Drug Metab Dispos. 2015;43(10):1601. doi: 10.1124/dmd.115.065094. [DOI] [PubMed] [Google Scholar]
  • 60.Xie J., Liu Y., Chen B., Zhang G., Ou S., Luo J., et al. <em>Ganoderma lucidum</em> polysaccharide improves rat DSS-induced colitis by altering cecal microbiota and gene expression of colonic epithelial cells. Food Nutr Res. 2019;63 doi: 10.29219/fnr.v63.1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
  • 62.Dekker E., Tanis P.J., Vleugels J.L.A., Kasi P.M., Wallace M.B. Colorectal cancer. Lancet. 2019;394(10207):1467–1480. doi: 10.1016/S0140-6736(19)32319-0. [DOI] [PubMed] [Google Scholar]
  • 63.Jess T., Rungoe C., Peyrin-Biroulet L. Risk of colorectal cancer in patients with ulcerative colitis: a meta-analysis of population-based cohort studies. Clin Gastroenterol Hepatol. 2012;10(6):639–645. doi: 10.1016/j.cgh.2012.01.010. [DOI] [PubMed] [Google Scholar]
  • 64.Nakatsu G., Li X., Zhou H., Sheng J., Wong S.H., Wu W.K.K., et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun. 2015;6(1):8727. doi: 10.1038/ncomms9727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wang L., Peng F., Peng C., Du J.-R. Gut microbiota in tumor microenvironment: a critical regulator in cancer initiation and development as potential targets for Chinese medicine. Am J Chin Med. 2021;49(03):609–626. doi: 10.1142/S0192415X21500270. [DOI] [PubMed] [Google Scholar]
  • 66.Guo C., Guo D., Fang L., Sang T., Wu J., Guo C., et al. Ganoderma lucidum polysaccharide modulates gut microbiota and immune cell function to inhibit inflammation and tumorigenesis in colon. Carbohydr Polym. 2021;267 doi: 10.1016/j.carbpol.2021.118231. [DOI] [PubMed] [Google Scholar]
  • 67.Wang C.Z., Yu C., Wen X.D., Chen L., Zhang C.F., Calway T., et al. American ginseng attenuates colitis-associated colon carcinogenesis in mice: impact on gut microbiota and metabolomics. Cancer Prev Res (Phila) 2016;9(10):803–811. doi: 10.1158/1940-6207.CAPR-15-0372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ji X., Hou C., Zhang X., Han L., Yin S., Peng Q., et al. Microbiome-metabolomic analysis of the impact of Zizyphus jujuba cv. Muzao polysaccharides consumption on colorectal cancer mice fecal microbiota and metabolites. Int J Biol Macromol. 2019;131:1067–1076. doi: 10.1016/j.ijbiomac.2019.03.175. [DOI] [PubMed] [Google Scholar]
  • 69.Ji X., Hou C., Gao Y., Xue Y., Yan Y., Guo X. Metagenomic analysis of gut microbiota modulatory effects of jujube (Ziziphus jujuba Mill.) polysaccharides in a colorectal cancer mouse model. Food Funct. 2020;11(1):163–173. doi: 10.1039/c9fo02171j. [DOI] [PubMed] [Google Scholar]
  • 70.Seitz H.K., Bataller R., Cortez-Pinto H., Gao B., Gual A., Lackner C., et al. Alcoholic liver disease. Nat Rev Dis Primers. 2018;4(1):16. doi: 10.1038/s41572-018-0014-7. [DOI] [PubMed] [Google Scholar]
  • 71.Szabo G. Gut-liver axis in alcoholic liver disease. Gastroenterology. 2015;148(1):30–36. doi: 10.1053/j.gastro.2014.10.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Guo L., Guan Q., Duan W., Ren Y., Zhang X.J., Xu H.Y., et al. Dietary goji shapes the gut microbiota to prevent the liver injury induced by acute alcohol intake. Front Nutr. 2022;9 doi: 10.3389/fnut.2022.929776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Guo W.L., Cao Y.J., You S.Z., Wu Q., Zhang F., Han J.Z., et al. Ganoderic acids-rich ethanol extract from Ganoderma lucidum protects against alcoholic liver injury and modulates intestinal microbiota in mice with excessive alcohol intake. Curr Res Food Sci. 2022;5:515–530. doi: 10.1016/j.crfs.2022.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cao Y.J., Huang Z.R., You S.Z., Guo W.L., Zhang F., Liu B., et al. The protective effects of ganoderic acids from Ganoderma lucidum fruiting body on alcoholic liver injury and intestinal microflora disturbance in mice with excessive alcohol intake. Foods. 2022;11(7) doi: 10.3390/foods11070949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lv X.C., Wu Q., Cao Y.J., Lin Y.C., Guo W.L., Rao P.F., et al. Ganoderic acid A from Ganoderma lucidum protects against alcoholic liver injury through ameliorating the lipid metabolism and modulating the intestinal microbial composition. Food Funct. 2022;13(10):5820–5837. doi: 10.1039/d1fo03219d. [DOI] [PubMed] [Google Scholar]
  • 76.Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2013;11 doi: 10.1016/j.cgh.2013.02.015. 868–75.e3. [DOI] [PubMed] [Google Scholar]
  • 77.Han X., Liu J., Bai Y., Hang A., Lu T., Mao C. An iridoid glycoside from Cornus officinalis balances intestinal microbiome disorder and alleviates alcohol-induced liver injury. J Funct Foods. 2021;82 doi: 10.1016/j.jff.2021.104488. [DOI] [Google Scholar]
  • 78.Zhao H, Kong L, Shao M, Liu J, Sun C, Li C, et al. Protective effect of flavonoids extract of Hippophae rhamnoides L. on alcoholic fatty liver disease through regulating intestinal flora and inhibiting TAK1/p38MAPK/p65NF-kappaB pathway. J Ethnopharmacol. 2022;292:115225. doi: 10.1016/j.jep.2022.115225. [DOI] [PubMed] [Google Scholar]
  • 79.Zhao W., Peng D., Li W., Chen S., Liu B., Huang P., et al. Probiotic-fermented Pueraria lobata (Willd.) Ohwi alleviates alcoholic liver injury by enhancing antioxidant defense and modulating gut microbiota. J Sci Food Agric. 2022;102(15):6877–6888. doi: 10.1002/jsfa.12049. [DOI] [PubMed] [Google Scholar]
  • 80.Zhou J., Zhang N., Zhao L., Wu W., Zhang L., Zhou F., et al. Astragalus polysaccharides and saponins alleviate liver injury and regulate gut microbiota in alcohol liver disease mice. Foods. 2021;10(11) doi: 10.3390/foods10112688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhou J., Zhang N., Zhao L., Mohamed Soliman M., Wu W., Li J., et al. Protective effects of honey-processed astragalus on liver injury and gut microbiota in mice induced by chronic alcohol intake. J Food Qual. 2022;2022 doi: 10.1155/2022/5333691. [DOI] [Google Scholar]
  • 82.Wang X.J., Malhi H. Nonalcoholic fatty liver disease. Ann Intern Med. 2018;169(9):ITC65–ITC80. doi: 10.7326/AITC201811060. [DOI] [PubMed] [Google Scholar]
  • 83.Pierantonelli I., Svegliati-Baroni G. Nonalcoholic fatty liver disease: basic pathogenetic mechanisms in the progression from NAFLD to NASH. Transplantation. 2019;103(1):e1–e13. doi: 10.1097/TP.0000000000002480. [DOI] [PubMed] [Google Scholar]
  • 84.Gottlieb A., Mosthael W., Sowa J.P., Canbay A. Nonalcoholic-fatty-liver-disease and nonalcoholic steatohepatitis: successful development of pharmacological treatment will depend on translational research. Digestion. 2019;100(2):79–85. doi: 10.1159/000493259. [DOI] [PubMed] [Google Scholar]
  • 85.El-Agroudy N.N., Kurzbach A., Rodionov R.N., O’Sullivan J., Roden M., Birkenfeld A.L., et al. Are lifestyle therapies effective for NAFLD treatment? Trends Endocrinol Metab. 2019;30(10):701–709. doi: 10.1016/j.tem.2019.07.013. [DOI] [PubMed] [Google Scholar]
  • 86.Zhong M., Yan Y., Yuan H., Xu G., Cai F., et al. Astragalus mongholicus polysaccharides ameliorate hepatic lipid accumulation and inflammation as well as modulate gut microbiota in NAFLD rats. Food Funct. 2022;13(13):7287–7301. doi: 10.1039/d2fo01009g. [DOI] [PubMed] [Google Scholar]
  • 87.Luo H., Wu H., Wang L., Xiao S., Lu Y., Liu C., et al. Hepatoprotective effects of Cassiae Semen on mice with non-alcoholic fatty liver disease based on gut microbiota. Commun Biol. 2021;4(1):1357. doi: 10.1038/s42003-021-02883-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mei L., Tang Y., Li M., Yang P., Liu Z., Yuan J., et al. Co-administration of cholesterol-lowering probiotics and anthraquinone from Cassia obtusifolia L.ameliorate non-alcoholic fatty liver. PLoS ONE. 2015;10 doi: 10.1371/journal.pone.0138078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lu S., Zhang T., Gu W., Yang X., Lu J., Zhao R., et al. Volatile oil of Amomum villosum inhibits nonalcoholic fatty liver disease via the gut-liver axis. Biomed Res Int. 2018;2018 doi: 10.1155/2018/3589874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Tan Y.Y., Yue S.R., Lu A.P., Zhang L., Ji G., Liu B.C., et al. The improvement of nonalcoholic steatohepatitis by Poria cocos polysaccharides associated with gut microbiota and NF-kappaB/CCL3/CCR1 axis. Phytomedicine. 2022;103 doi: 10.1016/j.phymed.2022.154208. [DOI] [PubMed] [Google Scholar]
  • 91.Han X., Zhao W., Zhou Q., Chen H., Yuan J., Xiaofu Z., et al. Procyanidins from hawthorn (Crataegus pinnatifida) alleviate lipid metabolism disorder via inhibiting insulin resistance and oxidative stress, normalizing the gut microbiota structure and intestinal barrier, and further suppressing hepatic inflammation and lipid accumulation. Food Funct. 2022;13(14):7901–7917. doi: 10.1039/d2fo00836j. [DOI] [PubMed] [Google Scholar]
  • 92.Yang Y., Li M., Wang Q., Huang H., Zhao Y., Du F., et al. Pueraria lobata starch regulates gut microbiota and alleviates high-fat high-cholesterol diet induced non-alcoholic fatty liver disease in mice. Food Res Int. 2022;157 doi: 10.1016/j.foodres.2022.111401. [DOI] [PubMed] [Google Scholar]
  • 93.Wu J., Zhang D., Zhu B., Wang S., Xu Y., Zhang C., et al. Rubus chingii Hu. unripe fruits extract ameliorates carbon tetrachloride-induced liver fibrosis and improves the associated gut microbiota imbalance. Chin Med. 2022;17(1):56. doi: 10.1186/s13020-022-00607-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chen L., Li M., Yang Z., Tao W., Wang P., Tian X., et al. Gardenia jasminoides Ellis: ethnopharmacology, phytochemistry, and pharmacological and industrial applications of an important traditional Chinese medicine. J Ethnopharmacol. 2020;257 doi: 10.1016/j.jep.2020.112829. [DOI] [PubMed] [Google Scholar]
  • 95.Chen Z., Lin T., Liao X., Li Z., Lin R., Qi X., et al. Network pharmacology based research into the effect and mechanism of Yinchenhao Decoction against Cholangiocarcinoma. Chin Med. 2021;16(1):13. doi: 10.1186/s13020-021-00423-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fang S., Wang T., Li Y., Xue H., Zou J., Cai J., et al. Gardenia jasminoides Ellis polysaccharide ameliorates cholestatic liver injury by alleviating gut microbiota dysbiosis and inhibiting the TLR4/NF-kappaB signaling pathway. Int J Biol Macromol. 2022;205:23–36. doi: 10.1016/j.ijbiomac.2022.02.056. [DOI] [PubMed] [Google Scholar]
  • 97.Tian X, Ding Y, Kong Y, Wang G, Wang S, Cheng D. Purslane (Portulacae oleracea L.) attenuates cadmium-induced hepatorenal and colonic damage in mice: Role of chelation, antioxidant and intestinal microecological regulation. Phytomedicine. 2021;92:153716. doi: 10.1016/j.phymed.2021.153716. [DOI] [PubMed] [Google Scholar]
  • 98.Chooi Y.C., Ding C., Magkos F. The epidemiology of obesity. Metabolism. 2019;92:6–10. doi: 10.1016/j.metabol.2018.09.005. [DOI] [PubMed] [Google Scholar]
  • 99.Zheng H., Ji H., Fan K., Xu H., Huang Y., Zheng Y., et al. Targeting gut microbiota and host metabolism with dendrobium officinale dietary fiber to prevent obesity and improve glucose homeostasis in diet-induced obese mice. Mol Nutr Food Res. 2022;66:2100772. doi: 10.1002/mnfr.202100772. [DOI] [PubMed] [Google Scholar]
  • 100.Chang C.J., Lin C.S., Lu C.C., Martel J., Ko Y.F., Ojcius D.M., et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat Commun. 2015;6:7489. doi: 10.1038/ncomms8489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zhang M., Zhu J., Zhang X., Zhao D.G., Ma Y.Y., Li D., et al. Aged citrus peel (chenpi) extract causes dynamic alteration of colonic microbiota in high-fat diet induced obese mice. Food Funct. 2020;11(3):2667–2678. doi: 10.1039/c9fo02907a. [DOI] [PubMed] [Google Scholar]
  • 102.Yang Y., Chang Y., Wu Y., Liu H., Liu Q., Kang Z., et al. A homogeneous polysaccharide from Lycium barbarum: structural characterizations, anti-obesity effects and impacts on gut microbiota. Int J Biol Macromol. 2021;183:2074–2087. doi: 10.1016/j.ijbiomac.2021.05.209. [DOI] [PubMed] [Google Scholar]
  • 103.Yue S.J., Qin Y.F., Kang A., Tao H.J., Zhou G.S., Chen Y.Y., et al. Total flavonoids of Glycyrrhiza uralensis alleviates irinotecan-induced colitis via modification of gut microbiota and fecal metabolism. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.628358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zou Y.T., Zhou J., Wu C.Y., Zhang W., Shen H., Xu J.D., et al. Protective effects of Poria cocos and its components against cisplatin-induced intestinal injury. J Ethnopharmacol. 2021;269 doi: 10.1016/j.jep.2020.113722. [DOI] [PubMed] [Google Scholar]
  • 105.Zhang T., Lu S.H., Bi Q., Liang L., Wang Y.F., Yang X.X., et al. Volatile oil from amomi fructus attenuates 5-fluorouracil-induced intestinal mucositis. Front Pharmacol. 2017;8:786. doi: 10.3389/fphar.2017.00786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Fu Y.P., Feng B., Zhu Z.K., Feng X., Chen S.F., Li L.X., et al. The polysaccharides from Codonopsis pilosula modulates the immunity and intestinal microbiota of cyclophosphamide-treated immunosuppressed mice. Molecules. 2018;23(7) doi: 10.3390/molecules23071801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zou Y.F., Zhang Y.Y., Fu Y.P., Inngjerdingen K.T., Paulsen B.S., Feng B., et al. A polysaccharide isolated from Codonopsis pilosula with immunomodulation effects both in vitro and in vivo. Molecules. 2019;24(20) doi: 10.3390/molecules24203632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zhou R., He D., Xie J., Zhou Q., Zeng H., Li H., et al. The synergistic effects of polysaccharides and ginsenosides from American ginseng (Panax quinquefolius L.) ameliorating cyclophosphamide-induced intestinal immune disorders and gut barrier dysfunctions based on microbiome-metabolomics analysis. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.665901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Huang K., Yan Y., Chen D., Zhao Y., Dong W., Zeng X., et al. Ascorbic acid derivative 2-O-beta-d-glucopyranosyl-l-ascorbic acid from the fruit of Lycium barbarum modulates microbiota in the small intestine and colon and exerts an immunomodulatory effect on cyclophosphamide-treated BALB/c mice. J Agric Food Chem. 2020;68(40):11128–11143. doi: 10.1021/acs.jafc.0c04253. [DOI] [PubMed] [Google Scholar]
  • 110.He Y., Huang L., Jiang P., Xu G., Sun T. Immunological regulation of the active fraction from Polygonatum sibiricum F. Delaroche based on improvement of intestinal microflora and activation of RAW264.7 cells. J Ethnopharmacol. 2022;293 doi: 10.1016/j.jep.2022.115240. [DOI] [PubMed] [Google Scholar]
  • 111.Chen Z., Lin Y., Zhou Q., Xiao S., Li C., Lin R., et al. Ginsenoside Rg1 mitigates morphine dependence via regulation of gut microbiota, tryptophan metabolism, and serotonergic system function. Biomed Pharmacother. 2022;150 doi: 10.1016/j.biopha.2022.112935. [DOI] [PubMed] [Google Scholar]
  • 112.Mitrea L., Nemeş S.-A., Szabo K., Teleky B.-E., Vodnar D.-C. Guts imbalance imbalances the brain: a review of gut microbiota association with neurological and psychiatric disorders. Front Med. 2022:9. doi: 10.3389/fmed.2022.813204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang H., Wei W., Zhao M., Ma L., Jiang X., Pei H., et al. Interaction between Aβ and Tau in the pathogenesis of Alzheimer's disease. Int J Biol Sci. 2021;17(9):2181–2192. doi: 10.7150/ijbs.57078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ittner A., Ittner L.M. Dendritic Tau in Alzheimer’s disease. Neuron. 2018;99(1):13–27. doi: 10.1016/j.neuron.2018.06.003. [DOI] [PubMed] [Google Scholar]
  • 115.MahmoudianDehkordi S., Arnold M., Nho K., Ahmad S., Jia W., Xie G., et al. Altered bile acid profile associates with cognitive impairment in Alzheimer's disease—an emerging role for gut microbiome. Alzheimers Dement. 2019;15(1):76–92. doi: 10.1016/j.jalz.2018.07.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sun Y., Liu Z., Pi Z., Song F., Wu J., Liu S. Poria cocos could ameliorate cognitive dysfunction in APP/PS1 mice by restoring imbalance of Abeta production and clearance and gut microbiota dysbiosis. Phytother Res. 2021 doi: 10.1002/ptr.7014. [DOI] [PubMed] [Google Scholar]
  • 117.Li X., Zhao T., Gu J., Wang Z., Lin J., Wang R., et al. Intake of flavonoids from Astragalus membranaceus ameliorated brain impairment in diabetic mice via modulating brain-gut axis. Chin Med. 2022;17(1):22. doi: 10.1186/s13020-022-00578-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhao W., Wang J., Latta M., Wang C., Liu Y., Ma W., et al. Rhizoma gastrodiae water extract modulates the gut microbiota and pathological changes of P-Tau(Thr231) to protect against cognitive impairment in mice. Front Pharmacol. 2022;13 doi: 10.3389/fphar.2022.903659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bell L., Whyte A., Duysburgh C., Marzorati M., Van den Abbeele P., Le Cozannet R., et al. A randomized, placebo-controlled trial investigating the acute and chronic benefits of American Ginseng (Cereboost(R)) on mood and cognition in healthy young adults, including in vitro investigation of gut microbiota changes as a possible mechanism of action. Eur J Nutr. 2022;61(1):413–428. doi: 10.1007/s00394-021-02654-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Liu Y., Liu W., Li J., Tang S., Wang M., Huang W., et al. A polysaccharide extracted from Astragalus membranaceus residue improves cognitive dysfunction by altering gut microbiota in diabetic mice. Carbohydr Polym. 2019;205:500–512. doi: 10.1016/j.carbpol.2018.10.041. [DOI] [PubMed] [Google Scholar]
  • 121.McCarron R.M., Shapiro B., Rawles J., Depression L.J. Ann Intern Med. 2021;174(5):ITC65–ITC80. doi: 10.7326/AITC202105180. [DOI] [PubMed] [Google Scholar]
  • 122.Shao X., Zhu G. Associations among monoamine neurotransmitter pathways, personality traits, and major depressive disorder. Front Psychiatry. 2020:11. doi: 10.3389/fpsyt.2020.00381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Uzbekov M., Maximova N. Biochemical bases of monoamine and hormonal interactions in pathogenesis of anxious depression: a hypothesis. Eur Psychiatry. 2015;30:542. doi: 10.1016/S0924-9338(15)30426-0. [DOI] [PubMed] [Google Scholar]
  • 124.Bath K.G., Schilit A., Lee F.S. Stress effects on BDNF expression: effects of age, sex, and form of stress. Neuroscience. 2013;239:149–156. doi: 10.1016/j.neuroscience.2013.01.074. [DOI] [PubMed] [Google Scholar]
  • 125.Yang Y., Hu Z., Du X., Davies H., Huo X., Fang M. miR-16 and fluoxetine both reverse autophagic and apoptotic change in chronic unpredictable mild stress model rats. Front Neurosci. 2017:11. doi: 10.3389/fnins.2017.00428. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 126.Lu B., Martinowich K. Cell biology of BDNF and its relevance to schizophrenia. Novartis Found Symp. 2008;289:119–135. doi: 10.1002/9780470751251.ch10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chen Y., Xiao N., Chen Y., Chen X., Zhong C., Cheng Y., et al. Semen Sojae Praeparatum alters depression-like behaviors in chronic unpredictable mild stress rats via intestinal microbiota. Food Res Int. 2021;150(Pt B) doi: 10.1016/j.foodres.2021.110808. [DOI] [PubMed] [Google Scholar]
  • 128.Li Y., Peng Y., Ma P., Yang H., Xiong H., Wang M., et al. Antidepressant-like effects of Cistanche tubulosa extract on chronic unpredictable stress rats through restoration of gut microbiota homeostasis. Front Pharmacol. 2018;9:967. doi: 10.3389/fphar.2018.00967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gao X., Feng Y., Xue H., Meng M., Qin X. Antidepressant-like effect of triterpenoids extracts from Poria cocos on the CUMS rats by 16S rRNA gene sequencing and LC–MS metabolomics. J Liq Chromatogr Relat Technol. 2020;43(13–14):494–507. doi: 10.1080/10826076.2020.1737107. [DOI] [Google Scholar]
  • 130.Chang L., Wei Y., Hashimoto K. Brain–gut–microbiota axis in depression: a historical overview and future directions. Brain Res Bull. 2022;182:44–56. doi: 10.1016/j.brainresbull.2022.02.004. [DOI] [PubMed] [Google Scholar]
  • 131.Bonaz B., Bazin T., Pellissier S. The Vagus nerve at the interface of the microbiota-gut-brain axis. Front Neurosci. 2018;12:42. doi: 10.3389/fnins.2018.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Pu Y., Tan Y., Qu Y., Chang L., Wang S., Wei Y., et al. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain Behav Immun. 2021;94:318–326. doi: 10.1016/j.bbi.2020.12.032. [DOI] [PubMed] [Google Scholar]
  • 133.Wang S., Ishima T., Zhang J., Qu Y., Chang L., Pu Y., et al. Ingestion of Lactobacillus intestinalis and Lactobacillus reuteri causes depression- and anhedonia-like phenotypes in antibiotic-treated mice via the vagus nerve. J Neuroinflammation. 2020;17(1):241. doi: 10.1186/s12974-020-01916-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Zhang J., Ma L., Chang L., Pu Y., Qu Y., Hashimoto K. A key role of the subdiaphragmatic vagus nerve in the depression-like phenotype and abnormal composition of gut microbiota in mice after lipopolysaccharide administration. Transl Psychiatry. 2020;10(1):186. doi: 10.1038/s41398-020-00878-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wang S., Ishima T., Qu Y., Shan J., Chang L., Wei Y., et al. Ingestion of Faecalibaculum rodentium causes depression-like phenotypes in resilient Ephx2 knock-out mice: a role of brain–gut–microbiota axis via the subdiaphragmatic vagus nerve. J Affective Disord. 2021;292:565–573. doi: 10.1016/j.jad.2021.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yang Y., Eguchi A., Wan X., Chang L., Wang X., Qu Y., et al. A role of gut–microbiota–brain axis via subdiaphragmatic vagus nerve in depression-like phenotypes in Chrna7 knock-out mice. Prog Neuro-Psychopharmacol Biol Psychiatry. 2023;120 doi: 10.1016/j.pnpbp.2022.110652. [DOI] [PubMed] [Google Scholar]
  • 137.Craske M.G., Stein M.B. Anxiety Lancet. 2016;388(10063):3048–3059. doi: 10.1016/S0140-6736(16)30381-6. [DOI] [PubMed] [Google Scholar]
  • 138.Zhang D.D., Li H.J., Zhang H.R., Ye X.C. Poria cocos water-soluble polysaccharide modulates anxiety-like behavior induced by sleep deprivation by regulating the gut dysbiosis, metabolic disorders and TNF-alpha/NF-kappaB signaling pathway. Food Funct. 2022;13(12):6648–6664. doi: 10.1039/d2fo00811d. [DOI] [PubMed] [Google Scholar]
  • 139.Si Y., Chen X., Guo T., Wei W., Wang L., Zhang F., et al. Comprehensive 16S rDNA sequencing and LC-MS/MS-based metabolomics to investigate intestinal flora and metabolic profiles of the serum, hypothalamus and hippocampus in pchlorophenylalanine-induced insomnia rats treated with Lilium brownie. Neurochem Res. 2022;47(3):574–589. doi: 10.1007/s11064-021-03466-z. [DOI] [PubMed] [Google Scholar]
  • 140.Hua Y, Guo S, Xie H, Zhu Y, Yan H, Tao WW, et al. Ziziphus jujuba Mill. var. spinosa (Bunge) Hu ex H. F. Chou seed ameliorates insomnia in rats by regulating metabolomics and intestinal flora composition. Front Pharmacol. 2021;12:653767. doi: 10.3389/fphar.2021.653767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kobiyama K., Ley K. Atherosclerosis. Circ Res. 2018;123(10):1118–1120. doi: 10.1161/CIRCRESAHA.118.313816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Xie B., Zu X., Wang Z., Xu X., Liu G., Liu R. Ginsenoside Rc ameliorated atherosclerosis via regulating gut microbiota and fecal metabolites. Front Pharmacol. 2022;13 doi: 10.3389/fphar.2022.990476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lv Z., Shan X., Tu Q., Wang J., Chen J., Yang Y. Ginkgolide B treatment regulated intestinal flora to improve high-fat diet induced atherosclerosis in ApoE(-/-) mice. Biomed Pharmacother. 2021;134 doi: 10.1016/j.biopha.2020.111100. [DOI] [PubMed] [Google Scholar]
  • 144.Liu L., Zhao Y., Ming J., Chen J., Zhao G., Chen Z.Y., et al. Polyphenol extract and essential oil of Amomum tsao-ko equally alleviate hypercholesterolemia and modulate gut microbiota. Food Funct. 2021;12(23):12008–12021. doi: 10.1039/d1fo03082e. [DOI] [PubMed] [Google Scholar]
  • 145.Hu R., Guo W., Huang Z., Li L., Liu B., Lv X. Extracts of Ganoderma lucidum attenuate lipid metabolism and modulate gut microbiota in high-fat diet fed rats. J Funct Foods. 2018;46:403–412. doi: 10.1016/j.jff.2018.05.020. [DOI] [Google Scholar]
  • 146.Guo W.L., Guo J.B., Liu B.Y., Lu J.Q., Chen M., Liu B., et al. Ganoderic acid A from Ganoderma lucidum ameliorates lipid metabolism and alters gut microbiota composition in hyperlipidemic mice fed a high-fat diet. Food Funct. 2020;11(8):6818–6833. doi: 10.1039/d0fo00436g. [DOI] [PubMed] [Google Scholar]
  • 147.Sun H., Saeedi P., Karuranga S., Pinkepank M., Ogurtsova K., Duncan B.B., et al. IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183 doi: 10.1016/j.diabres.2021.109119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Iatcu C.O., Steen A., Covasa M. Gut microbiota and complications of type-2 diabetes. Nutrients. 2022 doi: 10.3390/nu14010166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Yang G., Wei J., Liu P., Zhang Q., Tian Y., Hou G., et al. Role of the gut microbiota in type 2 diabetes and related diseases. Metabolism. 2021;117 doi: 10.1016/j.metabol.2021.154712. [DOI] [PubMed] [Google Scholar]
  • 150.Ma Q., Zhai R., Xie X., Chen T., Zhang Z., Liu H., et al. Hypoglycemic effects of Lycium barbarum polysaccharide in type 2 diabetes mellitus mice via modulating gut microbiota. Front Nutr. 2022;9 doi: 10.3389/fnut.2022.916271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Tilg H., Zmora N., Adolph T.E., Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020;20(1):40–54. doi: 10.1038/s41577-019-0198-4. [DOI] [PubMed] [Google Scholar]
  • 152.Verdu E.F., Danska J.S. Common ground: shared risk factors for type 1 diabetes and celiac disease. Nat Immunol. 2018;19(7):685–695. doi: 10.1038/s41590-018-0130-2. [DOI] [PubMed] [Google Scholar]
  • 153.Zhou W., Chen G., Chen D., Ye H., Zeng X. The antidiabetic effect and potential mechanisms of natural polysaccharides based on the regulation of gut microbiota. J Funct Foods. 2020;75 doi: 10.1016/j.jff.2020.104222. [DOI] [Google Scholar]
  • 154.Zhou W, Yang T, Xu W, Huang Y, Ran L, Yan Y, et al. The polysaccharides from the fruits of Lycium barbarum L. confer anti-diabetic effect by regulating gut microbiota and intestinal barrier. Carbohydr Polym. 2022;291:119626. doi: 10.1016/j.carbpol.2022.119626. [DOI] [PubMed] [Google Scholar]
  • 155.Zhang Y., Peng Y., Zhao L., Zhou G., Li X. Regulating the gut microbiota and SCFAs in the faeces of T2DM rats should be one of antidiabetic mechanisms of mogrosides in the fruits of Siraitia grosvenorii. J Ethnopharmacol. 2021;274 doi: 10.1016/j.jep.2021.114033. [DOI] [PubMed] [Google Scholar]
  • 156.Du Y., Li D.X., Lu D.Y., Zhang R., Zheng X.X., Xu B.J., et al. Morus alba L. water extract changes gut microbiota and fecal metabolome in mice induced by high-fat and high-sucrose diet plus low-dose streptozotocin. Phytother Res. 2022;36(3):1241–1257. doi: 10.1002/ptr.7343. [DOI] [PubMed] [Google Scholar]
  • 157.Jin Q., Ma R.C. Metabolomics in diabetes and diabetic complications: insights from epidemiological studies. Cell. 2021 doi: 10.3390/cells10112832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Bao M, Hou K, Xin C, Zeng D, Cheng C, Zhao H, et al. Portulaca oleracea L. Extract alleviated type 2 diabetes via modulating the gut microbiota and serum branched-chain amino acid metabolism. Mol Nutr Food Res. 2022;66:2101030. doi: 10.1002/mnfr.202101030. [DOI] [PubMed] [Google Scholar]
  • 159.Liu J., Liu L., Zhang G., Peng X. Poria cocos polysaccharides attenuate chronic nonbacterial prostatitis by targeting the gut microbiota: comparative study of Poria cocos polysaccharides and finasteride in treating chronic prostatitis. Int J Biol Macromol. 2021;189:346–355. doi: 10.1016/j.ijbiomac.2021.08.139. [DOI] [PubMed] [Google Scholar]
  • 160.Liu J., Yu J., Peng X. Poria cocos polysaccharides alleviates chronic nonbacterial prostatitis by preventing oxidative stress, regulating hormone production, modifying gut microbiota, and remodeling the DNA methylome. J Agric Food Chem. 2020;68(45):12661–12670. doi: 10.1021/acs.jafc.0c05943. [DOI] [PubMed] [Google Scholar]
  • 161.Yu J., Hu Q., Liu J., Luo J., Liu L., Peng X. Metabolites of gut microbiota fermenting Poria cocos polysaccharide alleviates chronic nonbacterial prostatitis in rats. Int J Biol Macromol. 2022;209(Pt B):1593–1604. doi: 10.1016/j.ijbiomac.2022.04.029. [DOI] [PubMed] [Google Scholar]
  • 162.Ding G., Gong Q., Ma J., Liu X., Wang Y., Cheng X. Immunosuppressive activity is attenuated by Astragalus polysaccharides through remodeling the gut microenvironment in melanoma mice. Cancer Sci. 2021;112(10):4050–4063. doi: 10.1111/cas.15078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Jiang Y., Fan L. The effect of Poria cocos ethanol extract on the intestinal barrier function and intestinal microbiota in mice with breast cancer. J Ethnopharmacol. 2021;266 doi: 10.1016/j.jep.2020.113456. [DOI] [PubMed] [Google Scholar]
  • 164.Zhao X., Wang Y., Nie Z., Han L., Zhong X., Yan X., et al. Eucommia ulmoides leaf extract alters gut microbiota composition, enhances short-chain fatty acids production, and ameliorates osteoporosis in the senescence-accelerated mouse P6 (SAMP6) model. Food Sci Nutr. 2020;8(9):4897–4906. doi: 10.1002/fsn3.1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Feng Y.L., Cao G., Chen D.Q., Vaziri N.D., Chen L., Zhang J., et al. Microbiome-metabolomics reveals gut microbiota associated with glycine-conjugated metabolites and polyamine metabolism in chronic kidney disease. Cell Mol Life Sci. 2019;76(24):4961–4978. doi: 10.1007/s00018-019-03155-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wilson I.D., Nicholson J.K. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl Res. 2017;179:204–222. doi: 10.1016/j.trsl.2016.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Koppel N., Maini Rekdal V., Balskus E.P. Chemical transformation of xenobiotics by the human gut microbiota. Science. 2017;356(6344) doi: 10.1126/science.aag2770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Dikeocha I.J., Al-Kabsi A.M., Miftahussurur M., Alshawsh M.A. Pharmacomicrobiomics: influence of gut microbiota on drug and xenobiotic metabolism. FASEB J. 2022;36(6) doi: 10.1096/fj.202101986r. [DOI] [PubMed] [Google Scholar]
  • 169.Shankar N.B. Developments in taxol production through endophytic fungal biotechnology: a review. Orient Pharm Exp Med. 2019;19(1):1–13. doi: 10.1007/s13596-018-0352-8. [DOI] [Google Scholar]
  • 170.Fei-Liang Z., Rui M., Mingliang J., Wei-Wei D., Jun J., Songquan W., et al. Cloning and characterization of ginsenoside-hydrolyzing β-glucosidase from lactobacillus brevis that transforms ginsenosides Rb1 and F2 into ginsenoside Rd and compound K. J Microbiol Biotechnol. 2016;26(10):1661–1667. doi: 10.4014/jmb.1605.05052. [DOI] [PubMed] [Google Scholar]
  • 171.Yan C., Hao C., Jin W., Dong W.-W., Quan L.-H. Biotransformation of Ginsenoside Rb1 to Ginsenoside F2 by recombinant β-glucosidase from rat intestinal Enterococcus gallinarum. Biotechnol Bioprocess Eng. 2021;26(6):968–975. doi: 10.1007/s12257-021-0008-2. [DOI] [Google Scholar]
  • 172.Bruni R., Barreca D., Protti M., Brighenti V., Righetti L., Anceschi L., et al. Botanical sources, chemistry, analysis, and biological activity of furanocoumarins of pharmaceutical interest. Molecules. 2019 doi: 10.3390/molecules24112163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Wagner A.M., Wu J.J., Hansen R.C., Nigg H.N., Beiere R.C. Bullous phytophotodermatitis associated with high natural concentrations of furanocoumarins in limes. Dermatitis®. 2002;13(1) doi: 10.1053/ajcd.2002.29948. [DOI] [PubMed] [Google Scholar]
  • 174.Yamasaki K., Iohara D., Oyama Y., Nishizaki N., Kawazu S., Nishi K., et al. Processing grapefruit juice with γ-cyclodextrin attenuates its inhibitory effect on cytochrome P450 3A activity. J Pharm Pharmacol. 2020;72(3):356–363. doi: 10.1111/jphp.13212. [DOI] [PubMed] [Google Scholar]
  • 175.Tan S.R.S., Eser B.E., Han J. Gut metabolism of furanocoumarins: proposed function of Co O-methyltransferase. ACS Omega. 2020;5(47):30696–30703. doi: 10.1021/acsomega.0c04879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Ngo L., Tran P., Ham S.-H., Cho J.-H., Cho H.-Y., Lee Y.-B. Simultaneous determination of imperatorin and its metabolite xanthotoxol in rat plasma and urine by LC–MS/MS and its application to pharmacokinetic studies. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1044–1045:30–38. doi: 10.1016/j.jchromb.2016.12.037. [DOI] [PubMed] [Google Scholar]
  • 177.Dong W.W., Xuan F.L., Zhong F.L., Jiang J., Wu S., Li D., et al. Comparative analysis of the rats' gut microbiota composition in animals with different ginsenosides metabolizing activity. J Agric Food Chem. 2017;65(2):327–337. doi: 10.1021/acs.jafc.6b04848. [DOI] [PubMed] [Google Scholar]
  • 178.Oh H.A., Kim D.-E., Choi H.J., Kim N.J., Kim D.-H. Anti-fatigue effects of 20(S)-protopanaxadiol and 20(S)-protopanaxatriol in mice. Biol Pharm Bull. 2015;38(9):1415–1419. doi: 10.1248/bpb.b15-00230. [DOI] [PubMed] [Google Scholar]
  • 179.Zhang W., Jiang S., Qian D., Shang E.X., Duan J.A. Analysis of interaction property of calycosin-7-O-beta-D-glucoside with human gut microbiota. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;963:16–23. doi: 10.1016/j.jchromb.2014.05.015. [DOI] [PubMed] [Google Scholar]
  • 180.Chen L., Li Z., Tang Y., Cui X., Luo R., Guo S., et al. Isolation, identification and antiviral activities of metabolites of calycosin-7-O-β-d-glucopyranoside. J Pharm Biomed Anal. 2011;56(2):382–389. doi: 10.1016/j.jpba.2011.05.033. [DOI] [PubMed] [Google Scholar]
  • 181.Yu D., Duan Y., Bao Y., Wei C., An L. Isoflavonoids from Astragalus mongholicus protect PC12 cells from toxicity induced by l-glutamate. J Ethnopharmacol. 2005;98(1):89–94. doi: 10.1016/j.jep.2004.12.027. [DOI] [PubMed] [Google Scholar]
  • 182.Ong S.K., Shanmugam M.K., Fan L., Fraser S.E., Arfuso F., Ahn K.S., et al. Focus on formononetin: anticancer potential and molecular targets. Cancers (Basel) 2019 doi: 10.3390/cancers11050611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Ha H., Lee H.Y., Lee J.-H., Jung D., Choi J., Song K.-Y., et al. Formononetin prevents ovariectomy-induced bone loss in rats. Arch Pharm Res. 2010;33(4):625–632. doi: 10.1007/s12272-010-0418-8. [DOI] [PubMed] [Google Scholar]
  • 184.Wang D.D., Hu F.B. Precision nutrition for prevention and management of type 2 diabetes. Lancet Diabetes Endocrinol. 2018;6(5):416–426. doi: 10.1016/S2213-8587(18)30037-8. [DOI] [PubMed] [Google Scholar]
  • 185.Poyet M., Groussin M., Gibbons S.M., Avila-Pacheco J., Jiang X., Kearney S.M., et al. A library of human gut bacterial isolates paired with longitudinal multiomics data enables mechanistic microbiome research. Nat Med. 2019;25(9):1442–1452. doi: 10.1038/s41591-019-0559-3. [DOI] [PubMed] [Google Scholar]
  • 186.Jiao W., Sang Y., Wang X., Wang S. Metabonomics and the gut microbiome analysis of the effect of 6-shogaol on improving obesity. Food Chem. 2023;404 doi: 10.1016/j.foodchem.2022.134734. [DOI] [PubMed] [Google Scholar]
  • 187.Bjerrum JT, Rantalainen M, Wang Y, Olsen J, Nielsen OH. 1031 Integration of transcriptomics and metabonomics: improving diagnostics and phenotyping in ulcerative colitis. Gastroenterology. 2013;144(5, Supplement 1):S-193. 10.1016/s0016-5085(13)60678-2 [DOI] [PMC free article] [PubMed]
  • 188.Menni C., Zierer J., Valdes A.M., Spector T.D. Mixing omics: combining genetics and metabolomics to study rheumatic diseases. Nat Rev Rheumatol. 2017;13(3):174–181. doi: 10.1038/nrrheum.2017.5. [DOI] [PubMed] [Google Scholar]
  • 189.Lloyd-Price J., Arze C., Ananthakrishnan A.N., Schirmer M., Avila-Pacheco J., Poon T.W., et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–662. doi: 10.1038/s41586-019-1237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Cammarota G., Ianiro G., Ahern A., Carbone C., Temko A., Claesson M.J., et al. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat Rev Gastroenterol Hepatol. 2020;17(10):635–648. doi: 10.1038/s41575-020-0327-3. [DOI] [PubMed] [Google Scholar]
  • 191.Seyed Tabib N.S., Madgwick M., Sudhakar P., Verstockt B., Korcsmaros T., Vermeire S. Big data in IBD: big progress for clinical practice. Gut. 2020;69(8):1520. doi: 10.1136/gutjnl-2019-320065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Armet A.M., Deehan E.C., O’Sullivan A.F., Mota J.F., Field C.J., Prado C.M., et al. Rethinking healthy eating in light of the gut microbiome. Cell Host Microbe. 2022;30(6):764–785. doi: 10.1016/j.chom.2022.04.016. [DOI] [PubMed] [Google Scholar]
  • 193.Klimenko N.S., Odintsova V.E., Revel-Muroz A., Tyakht A.V. The hallmarks of dietary intervention-resilient gut microbiome. NPJ Biofilms Microbiomes. 2022;8(1):77. doi: 10.1038/s41522-022-00342-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Procházková N, Falony G, Dragsted LO, Licht TR, Raes J, Roager HM. Advancing human gut microbiota research by considering gut transit time. Gut. 2022:gutjnl-2022-328166. 10.1136/gutjnl-2022-328166 [DOI] [PMC free article] [PubMed]
  • 195.Wardman J.F., Bains R.K., Rahfeld P., Withers S.G. Carbohydrate-active enzymes (CAZymes) in the gut microbiome. Nat Rev Microbiol. 2022;20(9):542–556. doi: 10.1038/s41579-022-00712-1. [DOI] [PubMed] [Google Scholar]
  • 196.Liu X., Tang S., Zhong H., Tong X., Jie Z., Ding Q., et al. A genome-wide association study for gut metagenome in Chinese adults illuminates complex diseases. Cell Discov. 2021;7(1):9. doi: 10.1038/s41421-020-00239-w. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Advanced Research are provided here courtesy of Elsevier

RESOURCES