Next Article in Journal
Comparison of Nutrition and Flavor Characteristics of Five Breeds of Pork in China
Previous Article in Journal
The Bioaccessibility and Antioxidant Activities of Fermented Mango Cultivar Juices after Simulated In Vitro Digestion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease

Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon 24252, Korea
*
Author to whom correspondence should be addressed.
Foods 2022, 11(17), 2703; https://doi.org/10.3390/foods11172703
Submission received: 20 July 2022 / Revised: 19 August 2022 / Accepted: 31 August 2022 / Published: 5 September 2022
(This article belongs to the Section Food Microbiology)

Abstract

:
Diet and lifestyle are crucial factors that influence the susceptibility of humans to nonalcoholic fatty liver disease (NAFLD). Personalized diet patterns chronically affect the composition and activity of microbiota in the human gut; consequently, nutrition-related dysbiosis exacerbates NAFLD via the gut–liver axis. Recent advances in diagnostic technology for gut microbes and microbiota-derived metabolites have led to advances in the diagnosis, treatment, and prognosis of NAFLD. Microbiota-derived metabolites, including tryptophan, short-chain fatty acid, fat, fructose, or bile acid, regulate the pathophysiology of NAFLD. The microbiota metabolize nutrients, and metabolites are closely related to the development of NAFLD. In this review, we discuss the influence of nutrients, gut microbes, their corresponding metabolites, and metabolism in the pathogenesis of NAFLD.

1. Introduction

Genetic background, environmental factors, lifestyle, host physiology, and dietary intake affect human health and the risk of disease throughout life. Of these components, food and nutrients play an essential role in maintaining health [1,2]. Dietary nutrients are absorbed in the intestine and are involved in preserving health through various actions. For example, a high calorie diet may cause metabolic syndrome, and the only approved treatment is exercise and diet control.
Microbiota is a collective term for the microorganisms that live in or on the human body. The microbiome refers to the collection of genomes from all the microorganisms in the environment. In the intestine, nutrients are metabolized by gut microbiota, and the metabolized nutrients and their metabolites move to the liver through the portal vein and are involved in various metabolic processes. This gut–liver axis is involved in the metabolism of nutrients and microbiota in the gut–liver axis play an important role. Trillions of microbiota are present in the intestine and some microbiota may contribute to the pathogenesis of liver diseases [3]. The gut microbiota affect liver disease through various mechanisms, including chronic systemic inflammation, increased gut permeability, the production of short-chain fatty acids, and changes in the metabolism [4]. Dysbiosis is a condition in which microbiome balance is disrupted, resulting in an imbalance in the composition of the microbiota. Gut dysbiosis is related with NAFLD progression by increasing the translocation of microbial metabolites into the liver [5,6].
The human gastrointestinal tract contains many microbiota composed of bacteria, viruses, and fungi. It provides a platform for multiple interactions between the host and the gut microbiome [7]. The gut microbiome forms a complex ecosystem that comprises several biological networks and functional mechanisms that interact with one another. Our knowledge about the crucial roles of the gut microbiome in growth, the immune response, inflammation, and metabolism has increased gradually in the past decade [8,9]. Diversity in diet patterns and food intake are the main drivers of the composition of gut microbial clusters, and a diverse and high-quality diet leads to healthy gut microbiota [10]. An uncontrolled diet may cause dysbiosis, and gut-derived microbial lipopolysaccharides (LPS), a toxic component of the bacterial wall, play an important role in the development of NAFLD [6].
Pathologically, NAFLD includes simple steatosis, nonalcoholic steatohepatitis (NASH), and NASH-related cirrhosis [11]. Total energy intake and nutrients are associated with the development of liver disease, which can be characterized by the proper selection of active nutrients that can play a role in the regulation of the nutritional metabolism [12,13]. Several studies have indicated that an inappropriate diet may lead to a fatty liver [14]. To date, recent research has reported that the composition of gut microbes changes according to nutritional status and that their metabolites are associated with the development of NAFLD. In this study, we intend to discuss the relationship between nutrients and the microbiota-derived metabolites in the pathogenesis of NAFLD.

2. Food and Gut Microbiota

There are many microbiota in our intestines, which are collectively called the “gut microbiota” [15]. The gut microbiota are comprised of all the organisms that are present in the gastrointestinal tract. Bacteria, viruses, fungi, and protozoa make up approximately 100 trillion microbiota in the human gastrointestinal tract [16]. Each microbial species can form colonies from 1012~1014 cells/mL [17]. The composition and function of the gut microbiota can be affected by various factors, including age, gender, genetic factors, lifestyle, medication, and dietary differences [18]. Environmental and genetic factors, as well as changes in the gut microbiota that play a role in the pathogenesis of metabolic disorders, are also implicated. Most gut bacteria exist in a commensal form, including lactobacilli and bifidobacteria. However, there are also Enterococcus and Escherichia coli, which can be harmful under certain conditions such as dysbiosis or in various diseases [19,20].
The gut microbiome contributes to nutrient processing and signaling and produces metabolites with essential functions, such as tryptophan, bile acid, choline, and short-chain fatty acid (SCFA) metabolisms [21]. Gut dysbiosis is associated with various diseases including obesity, insulin resistance, coronary heart disease, gut disease, metabolic syndrome, and infection [22,23].
Recent advances in immunology and microbiology have supported new hypotheses. First, the composition of gut microbiota clusters was significantly affected by diet patterns [24]. Each individual harbors his own distinctive pattern of microbial composition, and changes in the diet lead to changes in the composition (Table 1). Second, it has been found that the typical microbial composition and microbiota-derived products found in various laboratories have shown unexpected effects on immune and inflammatory responses [25].

3. Nonalcoholic Fatty Liver Disease

NAFLD is recognized as the most common disease among chronic liver diseases, and the prevalence of NAFLD has increased recently in the Asia-Pacific region [41]. Diabetes and metabolic syndrome are on the rise worldwide, and as a result, the prevalence of NAFLD is increasing. NAFLD causes NASH, along with other persistent liver diseases, over time, which can subsequently lead to cirrhosis and liver cancer [42,43]. One of the main causes of NAFLD is an inadequate diet, along with an oversupply of calories and an unbalanced intake of fats, grains, fruits, vegetables, protein, and ω-3 fatty acids at the same time [13]. NAFLD is closely related with changes in gut microbiota composition and metabolic activity (Table 2) [44].
In the “multi-hit” theory, various factors, such as dietary intake, environmental factors, insulin resistance, adipose tissue dysfunction, alterations in the gut microbiota, and genetic predisposition, are involved in the pathophysiology of NAFLD [55]. Gut dysbiosis increases the gut permeability of bacteria and increases exposure to harmful substances that increase inflammation and fibrosis in the liver [56]. Lifestyle changes are an effective treatment for NAFLD, leading to weight loss and improving metabolic diseases including heart disease and endocrine disease [57]. Therefore, it is of practical significance to study the relationship between dietary intake-induced liver lipid accumulation and nutrient intake to prevent diseases associated with NAFLD [58].
The gut–liver axis is important for the development, progression, and prognosis of NAFLD. According to the kind of diet and food, gut dysbiosis can occur, and bacteria-derived substances (endotoxin, bacterial DNA, microbe-associated molecular patterns (MAMPs), damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and nematode-associated molecular patterns (NAMPs)) directly affect the liver through the portal vein [6]. In addition, alcohol-secreting strains cause an inflammatory response in the liver and body [59].

4. Nutrients Associated with Microbiota in Nonalcoholic Fatty Liver Disease

Food-derived metabolites and metabolites associated with the gut microbiota are described in Table 3.

4.1. Bile Acid

Bile acids (BAs) are a major component of bile juice and play a crucial role in the absorption of dietary fat and some vitamins because they are bipolar [70]. It is known that bile acids are involved in digestion as surfactants. In many studies in recent decades, bile acids have been reported to act as a wide range of signaling hormones in the human body by binding to various receptors in the cell membrane or nucleus, thus regulating fat metabolism, glucose metabolism, inflammation, and the growth of gut microbes [71,72,73].
BAs are generally divided into primary and secondary BAs. Primary BAs are synthesized directly from cholesterol, such as cholic acid (CA) and chenodeoxycholic acid (CDCA). Most primary BAs are synthesized through the cytochrome p450 enzyme, and among them, cholesterol 7a-hydroxylase (CYP7a1) is a key factor in regulating the synthesis of bile acids [74,75]. Secondary BAs secrete bile into the duodenum during digestion, move along the intestines, and are produced by the metabolism of gut microbes. At this time, deoxycholic acid (DCA) is produced from CA, chenodeoxycholic acid is produced from CACA, and various BAs are present [76,77].
The role of BAs in NAFLD/NASH is increasingly acknowledged [78]. The risk of liver injury is increased by the dysregulated BA metabolism in NAFLD patients [79]. Thus, increased bile acid exposure may be involved in hepatic injury and the pathogenesis of NAFLD and NASH [80].
The complex mechanism of BA action in the development of NAFLD/NASH is controlled primarily by the farnesoid X receptor (FXR), activated by primary BAs, and Takeda G protein-coupled receptors 5 (TGR5), activated by secondary BAs [81]. Since bile acids are cytotoxic, they can cause liver damage and hepatitis, so it is important to maintain a proper amount within the cells. FXR performs various functions related to fat and glucose metabolism and is also involved in the regulation of inflammatory responses. In addition, FXR plays a critical role in maintaining metabolic homeostasis by regulating the synthesis, secretion, and transport of bile acids [82,83,84]. Many recent studies have provided strong evidence that FXR activity inhibits lipopolysaccharide (LPS)-stimulated NF-κB activation and signaling, thereby suppressing hepatic inflammation and fibrosis, and resulting in a hepatoprotective effect [85]. However, one study found that regulating gut microbiota to inhibit gut FXR resulted in BA alteration and increased BA metabolites, further contributing to the amelioration of rat liver steatosis. [86]. Therefore, FXR regulates the gut microbiota and has a wide range of effects in the treatment of NAFLD.

4.2. Short-Chain Fatty Acid

SCFAs are generated through the fermentation of indigestible polysaccharides such as dietary fiber by gut microbes in the large bowel [87]. This fermentation supports the growth of SCFAs and specialized microbiota that produce gases, and the main metabolites of the SCFAs are propionate, acetate, and butyrate [88]. In general, propionate affects hepatic lipogenesis and gluconeogenesis, whereas peripheral acetate functions as a substrate for cholesterol synthesis [89]. Ninety-five percent of SCFAs produced in the cecum and large bowel are rapidly absorbed by colonic cells, and the remaining 5% are excreted in the feces [90]. Additionally, SCFAs modulate the production of several inflammatory cytokines, including TNF-α, IL-6, IL-2, and IL-10 [91].
Butyrate activates gut gluconeogenesis, which has controlling effects on glucose and energy homeostasis [92]. Butyrate is necessary for epithelial cells to consume large amounts of oxygen by beta-oxidation, creating a hypoxic state that maintains oxygen balance in the intestine and prevents dysbiosis in the gut flora [93]. Butyrate also regulates diet-induced insulin resistance in animals. One study reported that sodium butyrate prevents steatohepatitis by modulating immune responses in the gut and liver and reducing microbial imbalance and endotoxemia in high-fat diet-induced mice [94]. As a result, SCFAs can prevent the progression of fatty liver to NASH by promoting hepatic glucagon-like peptide (GLP)-1 receptor expression [68]. Furthermore, increased GLP-2 levels by microbial or subcutaneous GLP-2 administration to mice reduces gut permeability, as well as plasma LPS and cytokine levels, resulting in reduced hepatic oxidative stress and inflammation [69]. Activation of G-protein-coupled receptor-43 revealed the antiinflammatory effect of SCFAs, and colitis was ameliorated by acetate supplementation. This is a finding not found in G-protein-coupled receptor-43 knockout mice [60]. Therefore, SCFAs improve gut permeability and prevent the delivery of harmful microbiota-derived substances and metabolites to the liver.

4.3. Amino Acid and Tryptophan

Amino acids are major building blocks of cellular proteins and precursors of nitrogenous substance synthesis. Polyamines, nitric oxide, catecholamines, creatine, and dopamine are typical amino acids and are essential in systemic homeostasis [95]. Although most dietary proteins are digested and absorbed in the small intestine, significant amounts of proteins and amino acids reach the colon, where various related bacteria degrade them [96,97].
Bacterial metabolites arising from tryptophan, phenylalanine, and tyrosine are known to be involved in the pathogenesis of NAFLD. These amino acids and their metabolites from the gut microbiota have been shown to have a variety of effects on the liver [98]. Tryptophan is an essential amino acid for humans and is found in many protein-based foods, including milk, meat, fruits, and seeds [99].
Dietary tryptophan catabolites include indole, tryptamine, indole ethanol, indole propionic acid, indoleacetic acid, indole lactic acid, skatole, indole aldehyde, and indole acrylic acid, and they may affect host physiology in numerous ways [100]. According to recent evidence, the metabolites of indole are known to be effective in gut and hepatic protection. As such, tryptophan availability is a key factor in controlling protein biosynthesis. This may be one of the important reasons why the immune system uses tryptophan to limit the proliferation of pathogens and malignant cells [101].
Indole is a beneficial signal for maintaining the gut barrier by increasing the expression of antiinflammatory cytokine interleukin (IL)-10 and inhibiting the tumor necrosis factor (TNF)-α-mediated activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and proinflammatory chemokine IL-8 expression [102]. Indoleamine 2,3-dioxygenase (IDO), the most undigested tryptophan, is absorbed and converted to kynurenine through the regulation of a key rate-limiting enzyme expressed in the gastrointestinal tract [103]. A study of IDO found that high-fat diet-induced IDO-/-mice were less infiltrated with inflammatory macrophages and were protected from obesity-related fatty liver and insulin resistance [104]. Therefore, the gut microbiota might regulate IDO activity to affect the development of NAFLD.
Indole compounds strengthen the tight junctions of epithelial cells and relieve the gut’s inflammatory response and damage [105]. There is currently firm evidence linking indoles and improvements in gut and liver health. Indole components induce signal transducer and transcriptional activator 3 (STAT3) phosphorylation by stimulating the secretion of IL-22 through the aryl hydrocarbon receptor (AhR), thus promoting the proliferation of gut epithelial cells and restoring the barrier function [106]. The treatment of hepatic and inflammatory severity in mice on an indole-attenuated high-fat diet was associated with the modulation of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 and the normalization of the gut microbiota [107].
Glutamine is a nonessential amino acid and is considered to be an essential nutrient in severe disease, such as cardiovascular disease, liver disease, and intestinal disease [108,109]. As glutamine is one of the abundant amino acids in the body, it regulates protein synthesis and breakdown and plays an important role in regulating acid–base balance, enhancing immune function, and improving adaptability to stress [110]. Glutamine also appears to play an important role in the gut through glutathione production, which may contribute to the prevention of oxidative damage to the gut [111].

4.4. Choline

Choline is composed of a phospholipid of the cell membrane and is the main nutrient supplied by food and synthesis in the body [112]. Choline is related to biological mechanisms, such as lipid-related pathways and the enterohepatic circulation of bile [112]. Choline-deficient intake is associated with obesity and insulin resistance, and consequently causes NASH through the inhibition of very low-density lipoprotein production [113,114,115,116].
Some gut microbiota may selectively metabolize choline to trimethylamine (TMA). TMA is transformed to trimethylamine-N-oxide (TMAO) by flavin-containing mono-oxygenase in the liver [117,118]. Microbiota-related choline metabolism reduces choline bioactivity and creates a choline deficiency, causing metabolic syndrome [119]. In the previous report, transgenic mice models (knockout of a choline-utilizing C) with modified gut microbiota (choline-metabolizing Escherichia coli group vs. no choline-metabolizing E. coli group). The choline-utilizing group changed the composition of the gut microbiota and metabolites, which induced metabolic disease [119]. In a human trial with 15 subjects, the choline amount in food altered the composition of choline-related gut microbiota, such as Gammaproteobacteria and Erysipelotrichi [120]. NAFLD patients showed increased levels of TMAO in the serum [121]. TMAO changes the glucose metabolism and increases insulin resistance [122]. Furthermore, TMAO is related to inflammation in adipocyte and insulin resistance by activating proinflammatory cytokine C-C motif chemokine 2 [122]. However, the exact mechanism of TMA in the pathogenesis of NAFLD is not fully demonstrated.

4.5. Polyphenol

Polyphenols are naturally occurring bioactive components in fruits and vegetables, and they are the most abundant antioxidants in the human diet [123]. Polyphenols include tea, fruits, vegetables, roots, seeds, cocoa, and wine, which represent a variety of bioactive substances. Approximately 90% to 95% of polyphenols ingested in the diet reach the colon and are then transformed into bioactive products by the gut microbiota [124,125]. In turn, polyphenols metabolized by the gut microbiota improve lipid regulatory bioavailability and act as antioxidants, thus interfering with various metabolic processes that affect cancer cell growth [126,127]. Recent systematic studies demonstrated that polyphenol intake reveals prebiotic properties that control the host’s health by controlling gut dysbiosis and maintaining homeostasis [128,129]. The prebiotic effect of polyphenols is mainly associated with the establishment of probiotics or the inhibition of pathogenic bacteria, resulting in reduced endotoxins, the induction of inflammatory immune responses in the intestine, and other health benefits, such as improved bowel habits and overall well-being [130].
Furthermore, various polyphenols have a protective effect on NAFLD by attenuating oxidative stress, insulin metabolism, and inflammation [131]. Surprisingly, polyphenols showed various effects on lipid metabolism, oxidative stress, inflammation, and insulin resistance, which are the most important pathological processes in the pathogenesis of the liver disease [132]. Additionally, nuts have prebiotic properties due to their fatty acid profile and high contents of protein, fiber, phytosterols, vitamins, and phenols [133]. Phenols show multifaceted effects on chronic liver diseases through a well-known mechanism, indicating a potential for the treatment of liver disease [134].

4.6. ω-3 Polyunsaturated Fatty Acid

ω-3 polyunsaturated fatty acids (PUFAs) are considered as prebiotics, and the consumption of an ω-3-rich diet has been considered beneficial for health. Fish oil contains more than 40 fatty acids, but sources of ω-3 PUFAs include eicosapentaenoic acid (20:5) and alpha-linolenic acid (18:3) in plant oils and docosahexaenoic acid (22:6) in fish oils [135]. Eicosapentaenoic acid and docosahexaenoic acid are found in significant amounts in fish and other seafood, so they are all marine n-3 fatty acids [136]. The ω-6 PUFAs (γ-linolenic acid (18:3), arachidonic acid (20:4), and linoleic acid (18:2)) and ω-3 PUFAs are well known to attenuate inflammation [137]. As a result, an increase in the dietary intake of ω-6/ω-3 PUFAs has been associated with a higher risk of developing diseases such as obesity, arthritis, diabetes, cardiovascular disease, and liver disease [138,139,140]. Previous studies have shown that ω-3 PUFAs might modulate fatty acid metabolism, thus increasing hepatic fatty acid β-oxidation and antiinflammatory effects in NAFLD [141]. Therefore, more attention has been given to the role of ω-3 PUFAs in NAFLD.
ω-3 PUFAs directly affect the composition and diversity of gut microbiota. The unbalanced intake of ω-3/ω-6 PUFAs can lead to a significant increase in the Firmicutes/Bacteroides ratio and dysbiosis [142]. In addition, fish oil exerts an inhibitory effect on various bacteria, and ω-3 PUFAs can have a beneficial effect on the gut microbiota, which reduces the growth of Enterobacteria, increases the growth of Bifidobacteria, and increases metabolic endotoxins. It also potentially inhibits hematuria-associated inflammation [28]. Thus, it is suggested that the gut microbiota might regulate the absorption, bioavailability, and biotransformation of ω-3 PUFAs [143]. Finally, ω-3 PUFAs trigger a healthy chain reaction to increase the amounts of SCFA, serve as farnesoid X receptor (FXR) ligands, and create healthy bile acid reservoirs, thus contributing to liver health maintenance and the inhibition of hepatic inflammation [29,144].

4.7. Others

Among the heterogeneous groups of plant-derived carbohydrates, dietary fiber is not digested by the host and is selectively fermented by the gut microbiota [145]. A diet with legumes, vegetables, grains, and fruit is usually related to fiber intake [146]. The microbiota ferment soluble fiber to produce SCFAs. SCFAs, which are major energy sources for colon cells, are related to the growth of microbiota, the inhibition of pathogenic bacteria, fat reduction, an increase in insulin sensitivity, and systemic inflammation reduction [147]. Fermented foods including vinegar, olives, yogurt, bread, cheese, butters, wine, beer, pickle, sauerkraut, soy sauce, and yogurt are rich in SCFAs
A vegetable diet, especially abundant in indigestible carbohydrates (fiber), is related to a high proportion of Prevotella and Xylanibacter spp., while a Western diet, which generally contains saturated fat and protein from animals, is associated with an increase in Bacteroides spp. and a decrease in general microbiota gene diversity [30]. Insoluble forms, such as cellulose, have a fecal-increasing effect and resist metabolism by the gut microbiota, especially in the unit host. The dietary fiber that microbes can use for carbon and energy includes microbe-accessible carbohydrates [148]. Given that the diet provides both soluble and insoluble fiber, while the most generally used diets contain only insoluble cellulose, an imbalance between fiber sources has a well-established impact on the gut microbiota and metabolism, which has been demonstrated in important experimental results [149].
Today, fat tissues are generally known as physiologic organs that act on the endocrine metabolism. Fatty tissues store excess nutrients in neutral lipids in an overnutrition state, while undernutrition activates lipolysis and fuels lipid decomposition [150]. From a nutritional point of view, a low intake of vegetables, fruits, protein, grains, and ω3-fatty acids, along with an inadequate and excessive intake of saturated fats and calorie oversupply, are major causes of NAFLD [151].
Dietary fructose is strongly associated with the pathogenesis of NAFLD. A large association study found that people who regularly consume sugary drinks, especially those who are obese, show an increased risk of NAFLD [152]. Dietary fructose may cause rapid and deleterious changes in the composition of the gut microbiota, including reduced phylogenetic diversity and low concentrations of the genus Bifidobacterium [153]. Fructose leads to gut bacterial imbalances that increase gut macrophages and reduced tight junction function, which cause translocation and endotoxemia, higher bacterial potential, and increased liver TLR expression [154]. The intake of artificial sweeteners is nowadays also associated with a dysbiosis of the gut microbiota.
Several types of foods have been linked to microbiota. Fermented foods contain a lot of substances beneficial to health and are used in various forms depending on the region. Beans (Cheonggukjang, doenjang, fermented bean curd, miso, natto, soy sauce, stinky tofu, tempeh, and oncom), grains (amazake, beer, bread, choujiu, gamju, injera, kvass, makgeolli, murri, ogi, rejuvelac, sake, sikhye, sourdough, sowans, and rice wine), vegetables (kimchi, mixed pickle, sauerkraut, Indian pickle, gundruk, and tursu), fruit (wine, vinegar, cider, perry, and brandy), honey, dairy, fish (bagoong, faseekh, fish sauce, garum, and ngapi), meat (chorizo, salami, sucuk, pepperoni, nem chua, and som moo), and tea are well-known fermented foods.

5. Conclusions

In recent years, gut microbiota have received great attention in chronic liver disease. The microbiota can enhance energy extraction from the diet, regulate systemic metabolism towards increased fatty acid uptake in adipose tissue, and shift the lipid metabolism from oxidation to de novo production. Therefore, a key role in maintaining gut–liver axis health is attributed to the gut microbiota.
All foods are absorbed in the intestine and used as an energy source by organs, so foods we eat are closely related to the gut microflora. For healthy activity of the gut microbiota, a nutrient-balanced diet and regular meals are necessary. In addition, since heavy drinking, tobacco, drug abuse, lack of exercise, and lack of sleep affect the gut microflora, a healthy diet is necessary along with a healthy life habit.
The liver can be greatly affected by changes in gut microbiota through the portal vein of the liver, and the gut–liver axis is especially important for understanding the pathophysiology of various liver diseases. Gut microbiota-targeted therapy for liver disease is related to metabolites and disease mechanisms, and further research is required for targeted therapy of the gut microbiota (Figure 1). In addition, further studies for new diagnostic methods using specific gut microbiota are needed in the future.

Author Contributions

Conceptualization and drafting, K.T.S.; resources, S.M.W., J.J.J., R.G., H.G., S.P.S., Y.R.C., S.J.Y., J.Y.H., M.K.J., H.J.P., B.H.M. and M.R.C.; writing and editing, J.A.E.; revisions and reviews, K.K.O., D.J.K. and K.T.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Hallym University Research Fund and the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education, Science and Technology (NRF-2020R1I1A3073530 and NRF-2020R1A6A1A03043026).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moco, S.; Martin, F.P.; Rezzi, S. Metabolomics view on gut microbiome modulation by polyphenol-rich foods. J. Proteome Res. 2012, 11, 4781–4790. [Google Scholar] [CrossRef]
  2. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  3. Mokhtari, Z.; Gibson, D.L.; Hekmatdoost, A. Nonalcoholic fatty liver disease, the gut microbiome, and diet. Adv. Nutr. 2017, 8, 240–252. [Google Scholar] [CrossRef]
  4. Schwenger, K.J.; Clermont-Dejean, N.; Allard, J.P. The role of the gut microbiome in chronic liver disease: The clinical evidence revised. JHEP Rep. 2019, 1, 214–226. [Google Scholar] [CrossRef]
  5. Smirnova, E.; Muthiah, M.D.; Narayan, N.; Siddiqui, M.S.; Puri, P.; Luketic, V.A.; Contos, M.J.; Idowu, M.; Chuang, J.C.; Billin, A.N.; et al. Metabolic reprogramming of the intestinal microbiome with functional bile acid changes underlie the development of NAFLD. Hepatology 2022. [Google Scholar] [CrossRef]
  6. Song, Q.; Zhang, X. The Role of Gut-Liver Axis in Gut Microbiome Dysbiosis Associated NAFLD and NAFLD-HCC. Biomedicines 2022, 10, 524. [Google Scholar] [CrossRef]
  7. Yadav, M.; Verma, M.K.; Chauhan, N.S. A review of metabolic potential of human gut microbiome in human nutrition. Arch. Microbiol. 2018, 200, 203–217. [Google Scholar] [CrossRef]
  8. Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef]
  9. Hacquard, S.; Garrido-Oter, R.; González, A.; Spaepen, S.; Ackermann, G.; Lebeis, S.; McHardy, A.C.; Dangl, J.L.; Knight, R.; Ley, R. Microbiota and host nutrition across plant and animal kingdoms. Cell Host Microbe 2015, 17, 603–616. [Google Scholar] [CrossRef]
  10. Frame, L.A.; Costa, E.; Jackson, S.A. Current explorations of nutrition and the gut microbiome: A comprehensive evaluation of the review literature. Nutr. Rev. 2020, 78, 798–812. [Google Scholar] [CrossRef]
  11. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef]
  12. Mouzaki, M.; Allard, J.P. The role of nutrients in the development, progression, and treatment of nonalcoholic fatty liver disease. J. Clin. Gastroenterol. 2012, 46, 457–467. [Google Scholar] [CrossRef]
  13. Stachowska, E.; Ryterska, K.; Maciejewska, D.; Banaszczak, M.; Milkiewicz, P.; Milkiewicz, M.; Gutowska, I.; Ossowski, P.; Kaczorowska, M.; Jamiol-Milc, D.; et al. Nutritional Strategies for the Individualized Treatment of Non-Alcoholic Fatty Liver Disease (NAFLD) Based on the Nutrient-Induced Insulin Output Ratio (NIOR). Int. J. Mol. Sci. 2016, 17, 1192. [Google Scholar] [CrossRef]
  14. Parry, S.A.; Hodson, L. Influence of dietary macronutrients on liver fat accumulation and metabolism. J. Investig. Med. 2017, 65, 1102–1115. [Google Scholar] [CrossRef]
  15. Bruzzese, E.; Volpicelli, M.; Squaglia, M.; Tartaglione, A.; Guarino, A. Impact of prebiotics on human health. Dig. Liver Dis. 2006, 38, S283–S287. [Google Scholar] [CrossRef]
  16. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. Bmj 2018, 361, k2179. [Google Scholar] [CrossRef]
  17. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
  18. Blandino, G.; Inturri, R.; Lazzara, F.; Di Rosa, M.; Malaguarnera, L. Impact of gut microbiota on diabetes mellitus. Diabetes Metab. 2016, 42, 303–315. [Google Scholar] [CrossRef]
  19. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; FitzGerald, M.G.; Fulton, R.S. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207. [Google Scholar]
  20. Meelu, P.; Marin, R.; Clish, C.; Zella, G.; Cox, S.; Yajnik, V.; Nguyen, D.D.; Korzenik, J.R. Impaired innate immune function associated with fecal supernatant from Crohn’s disease patients: Insights into potential pathogenic role of the microbiome. Inflamm. Bowel Dis. 2014, 20, 1139–1146. [Google Scholar] [CrossRef]
  21. Zoetendal, E.G.; Rajilić-Stojanović, M.; de Vos, W.M. High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut 2008, 57, 1605–1615. [Google Scholar] [CrossRef]
  22. Cani, P.D.; Delzenne, N.M.; Amar, J.; Burcelin, R. Role of gut microflora in the development of obesity and insulin resistance following high-fat diet feeding. Pathol. Biol. 2008, 56, 305–309. [Google Scholar] [CrossRef]
  23. Martinez, F.D. The human microbiome. Early life determinant of health outcomes. Ann. Am. Thorac. Soc. 2014, 11, S7–S12. [Google Scholar] [CrossRef]
  24. Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The effect of diet on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 2009, 1, ra14–ra16. [Google Scholar] [CrossRef]
  25. Wen, L.; Ley, R.E.; Volchkov, P.Y.; Stranges, P.B.; Avanesyan, L.; Stonebraker, A.C.; Hu, C.; Wong, F.S.; Szot, G.L.; Bluestone, J.A. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 2008, 455, 1109–1113. [Google Scholar] [CrossRef]
  26. Bäckhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [Google Scholar] [CrossRef]
  27. Friedman, M. Analysis, nutrition, and health benefits of tryptophan. Int. J. Tryptophan Res. 2018, 11, 1178646918802282. [Google Scholar] [CrossRef]
  28. Cao, W.; Wang, C.; Chin, Y.; Chen, X.; Gao, Y.; Yuan, S.; Xue, C.; Wang, Y.; Tang, Q. DHA-phospholipids (DHA-PL) and EPA-phospholipids (EPA-PL) prevent intestinal dysfunction induced by chronic stress. Food Funct. 2019, 10, 277–288. [Google Scholar] [CrossRef]
  29. Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of omega-3 fatty acids on the gut microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef]
  30. Coker, J.K.; Moyne, O.; Rodionov, D.A.; Zengler, K. Carbohydrates great and small, from dietary fiber to sialic acids: How glycans influence the gut microbiome and affect human health. Gut Microbes 2021, 13, 1–18. [Google Scholar] [CrossRef]
  31. Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef] [Green Version]
  32. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef]
  33. Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef]
  34. Brown, K.; DeCoffe, D.; Molcan, E.; Gibson, D.L. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients 2012, 4, 1095–1119. [Google Scholar] [CrossRef]
  35. Cani, P.D.; Delzenne, N.M. Involvement of the gut microbiota in the development of low grade inflammation associated with obesity: Focus on this neglected partner. Acta Gastroenterol Belg 2010, 73, 267–269. [Google Scholar]
  36. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef]
  37. Barrea, L.; Annunziata, G.; Muscogiuri, G.; Di Somma, C.; Laudisio, D.; Maisto, M.; De Alteriis, G.; Tenore, G.C.; Colao, A.; Savastano, S. Trimethylamine-N-oxide (TMAO) as novel potential biomarker of early predictors of metabolic syndrome. Nutrients 2018, 10, 1971. [Google Scholar] [CrossRef]
  38. Zhang, C.; Zhang, M.; Wang, S.; Han, R.; Cao, Y.; Hua, W.; Mao, Y.; Zhang, X.; Pang, X.; Wei, C. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010, 4, 232–241. [Google Scholar] [CrossRef]
  39. Sun, G.; Yin, Z.; Liu, N.; Bian, X.; Yu, R.; Su, X.; Zhang, B.; Wang, Y. Gut microbial metabolite TMAO contributes to renal dysfunction in a mouse model of diet-induced obesity. Biochem. Biophys. Res. Commun. 2017, 493, 964–970. [Google Scholar] [CrossRef]
  40. Chang, C.-J.; Lin, C.-S.; Lu, C.-C.; Martel, J.; Ko, Y.-F.; Ojcius, D.M.; Tseng, S.-F.; Wu, T.-R.; Chen, Y.-Y.M.; Young, J.D. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat. Commun. 2015, 6, 7489. [Google Scholar] [CrossRef]
  41. Terai, S.; Buchanan-Hughes, A.; Ng, A.; Lee, I.-H.; Hasegawa, K. Comorbidities and healthcare costs and resource use of patients with nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) in the Japan medical data vision database. J. Gastroenterol. 2021, 56, 274–284. [Google Scholar] [CrossRef]
  42. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef]
  43. Larsen, O.F.A.; van der Grint, M.; Wiegers, C.; van de Burgwal, L.H.M. The Gut Microbiota: Master of Puppets Connecting the Epidemiology of Infectious, Autoimmune, and Metabolic Disease. Front. Microbiol. 2022, 13, 902106. [Google Scholar] [CrossRef]
  44. Burz, S.D.; Monnoye, M.; Philippe, C.; Farin, W.; Ratziu, V.; Strozzi, F.; Paillarse, J.-M.; Chêne, L.; Blottière, H.M.; Gérard, P. Fecal Microbiota Transplant from Human to Mice Gives Insights into the Role of the Gut Microbiota in Non-Alcoholic Fatty Liver Disease (NAFLD). Microorganisms 2021, 9, 199. [Google Scholar] [CrossRef]
  45. Raman, M.; Ahmed, I.; Gillevet, P.M.; Probert, C.S.; Ratcliffe, N.M.; Smith, S.; Greenwood, R.; Sikaroodi, M.; Lam, V.; Crotty, P. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2013, 11, 868–875. e863. [Google Scholar] [CrossRef]
  46. Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef]
  47. Berlanga, A.; Guiu-Jurado, E.; Porras, J.A.; Auguet, T. Molecular pathways in non-alcoholic fatty liver disease. Clin. Exp. Gastroenterol. 2014, 7, 221. [Google Scholar]
  48. Caussy, C.; Tripathi, A.; Humphrey, G.; Bassirian, S.; Singh, S.; Faulkner, C.; Bettencourt, R.; Rizo, E.; Richards, L.; Xu, Z.Z. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat. Commun. 2019, 10, 1406. [Google Scholar] [CrossRef]
  49. Leong, P.K.; Ko, K.M. Schisandrin B: A double-edged sword in nonalcoholic fatty liver disease. Oxidative medicine and cellular longevity 2016, 2016, 6171658. [Google Scholar] [CrossRef]
  50. Gupta, H.; Youn, G.S.; Shin, M.J.; Suk, K.T. Role of Gut Microbiota in Hepatocarcinogenesis. Microorganisms 2019, 7, 121. [Google Scholar] [CrossRef]
  51. Aravalli, R.N.; Cressman, E.N.; Steer, C.J. Cellular and molecular mechanisms of hepatocellular carcinoma: An update. Arch. Toxicol. 2013, 87, 227–247. [Google Scholar] [CrossRef]
  52. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef]
  53. Zeng, H.; Liu, J.; Jackson, M.I.; Zhao, F.-Q.; Yan, L.; Combs Jr, G.F. Fatty liver accompanies an increase in lactobacillus species in the hind gut of C57BL/6 mice fed a high-fat diet. J. Nutr. 2013, 143, 627–631. [Google Scholar] [CrossRef] [Green Version]
  54. Cano, P.G.; Santacruz, A.; Trejo, F.M.; Sanz, Y. Bifidobacterium CECT 7765 improves metabolic and immunological alterations associated with obesity in high-fat diet-fed mice. Obesity 2013, 21, 2310–2321. [Google Scholar] [CrossRef]
  55. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  56. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
  57. Romero-Gómez, M.; Zelber-Sagi, S.; Trenell, M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017, 67, 829–846. [Google Scholar] [CrossRef]
  58. Cheng, Y.; Zhang, K.; Chen, Y.; Li, Y.; Li, Y.; Fu, K.; Feng, R. Associations between dietary nutrient intakes and hepatic lipid contents in NAFLD patients quantified by 1H-MRS and dual-Echo MRI. Nutrients 2016, 8, 527. [Google Scholar] [CrossRef]
  59. Kolodziejczyk, A.A.; Zheng, D.; Shibolet, O.; Elinav, E. The role of the microbiome in NAFLD and NASH. EMBO Mol. Med. 2019, 11. [Google Scholar] [CrossRef]
  60. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
  61. Daliri, E.B.-M.; Lee, B.H. New perspectives on probiotics in health and disease. Food Sci. Hum. Wellness 2015, 4, 56–65. [Google Scholar] [CrossRef]
  62. Daliri, E.B.-M.; Wei, S.; Oh, D.H.; Lee, B.H. The human microbiome and metabolomics: Current concepts and applications. Crit. Rev. Food Sci. Nutr. 2017, 57, 3565–3576. [Google Scholar] [CrossRef]
  63. de Diego-Cabero, N.; Mereu, A.; Menoyo, D.; Holst, J.J.; Ipharraguerre, I.R. Bile acid mediated effects on gut integrity and performance of early-weaned piglets. BMC Vet. Res. 2015, 11, 111. [Google Scholar] [CrossRef] [Green Version]
  64. Adom, D.; Nie, D. Regulation of autophagy by short chain fatty acids in colon cancer cells. In Autophagy–A Double-Edged Sword–Cell Survival or Death; Intechopen: London, UK, 2013; pp. 235–247. [Google Scholar]
  65. Den Besten, G.; Bleeker, A.; Gerding, A.; van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.-J. Short-chain fatty acids protect against high-fat diet–induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef]
  66. Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef]
  67. Sahuri-Arisoylu, M.; Brody, L.; Parkinson, J.; Parkes, H.; Navaratnam, N.; Miller, A.D.; Thomas, E.; Frost, G.; Bell, J. Reprogramming of hepatic fat accumulation and ‘browning’ of adipose tissue by the short-chain fatty acid acetate. Int. J. Obes. 2016, 40, 955–963. [Google Scholar] [CrossRef]
  68. Zhou, D.; Chen, Y.-W.; Zhao, Z.-H.; Yang, R.-X.; Xin, F.-Z.; Liu, X.-L.; Pan, Q.; Zhou, H.; Fan, J.-G. Sodium butyrate reduces high-fat diet-induced non-alcoholic steatohepatitis through upregulation of hepatic GLP-1R expression. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef]
  69. Cani, P.D.; Possemiers, S.; Van de Wiele, T.; Guiot, Y.; Everard, A.; Rottier, O.; Geurts, L.; Naslain, D.; Neyrinck, A.; Lambert, D.M. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009, 58, 1091–1103. [Google Scholar] [CrossRef]
  70. Joyce, S.A.; Gahan, C.G. Bile acid modifications at the microbe-host interface: Potential for nutraceutical and pharmaceutical interventions in host health. Annu. Rev. Food Sci. Technol. 2016, 7, 313–333. [Google Scholar] [CrossRef]
  71. Fiorucci, S.; Distrutti, E. Bile acid-activated receptors, intestinal microbiota, and the treatment of metabolic disorders. Trends Mol. Med. 2015, 21, 702–714. [Google Scholar] [CrossRef]
  72. Esteller, A. Physiology of bile secretion. World J. Gastroenterol. WJG 2008, 14, 5641. [Google Scholar] [CrossRef]
  73. Zhu, C.; Fuchs, C.D.; Halilbasic, E.; Trauner, M. Bile acids in regulation of inflammation and immunity: Friend or foe. Clin. Exp. Rheumatol. 2016, 34, 25–31. [Google Scholar]
  74. Li, Y.C.; Wang, D.P.; Chiang, J. Regulation of cholesterol 7 alpha-hydroxylase in the liver. Cloning, sequencing, and regulation of cholesterol 7 alpha-hydroxylase mRNA. J. Biol. Chem. 1990, 265, 12012–12019. [Google Scholar] [CrossRef]
  75. Chiang, J.Y. Bile acid regulation of gene expression: Roles of nuclear hormone receptors. Endocr. Rev. 2002, 23, 443–463. [Google Scholar] [CrossRef] [Green Version]
  76. Chiang, J.Y. Bile acids: Regulation of synthesis: Thematic Review Series: Bile Acids. J. Lipid Res. 2009, 50, 1955–1966. [Google Scholar] [CrossRef]
  77. Monte, M.J.; Marin, J.J.; Antelo, A.; Vazquez-Tato, J. Bile acids: Chemistry, physiology, and pathophysiology. World J. Gastroenterol. WJG 2009, 15, 804. [Google Scholar] [CrossRef]
  78. Arab, J.P.; Karpen, S.J.; Dawson, P.A.; Arrese, M.; Trauner, M. Bile acids and nonalcoholic fatty liver disease: Molecular insights and therapeutic perspectives. Hepatology 2017, 65, 350–362. [Google Scholar] [CrossRef]
  79. Mouzaki, M.; Wang, A.Y.; Bandsma, R.; Comelli, E.M.; Arendt, B.M.; Zhang, L.; Fung, S.; Fischer, S.E.; McGilvray, I.G.; Allard, J.P. Bile acids and dysbiosis in non-alcoholic fatty liver disease. PLoS ONE 2016, 11, e0151829. [Google Scholar] [CrossRef]
  80. Ferslew, B.C.; Xie, G.; Johnston, C.K.; Su, M.; Stewart, P.W.; Jia, W.; Brouwer, K.L.; Barritt, A.S. Altered bile acid metabolome in patients with nonalcoholic steatohepatitis. Dig. Dis. Sci. 2015, 60, 3318–3328. [Google Scholar] [CrossRef]
  81. Chávez-Talavera, O.; Tailleux, A.; Lefebvre, P.; Staels, B. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 2017, 152, 1679–1694. e1673. [Google Scholar] [CrossRef]
  82. Houten, S.M.; Watanabe, M.; Auwerx, J. Endocrine functions of bile acids. EMBO J. 2006, 25, 1419–1425. [Google Scholar] [CrossRef]
  83. Nguyen, A.; Bouscarel, B. Bile acids and signal transduction: Role in glucose homeostasis. Cell. Signal. 2008, 20, 2180–2197. [Google Scholar] [CrossRef]
  84. Modica, S.; Murzilli, S.; Salvatore, L.; Schmidt, D.R.; Moschetta, A. Nuclear bile acid receptor FXR protects against intestinal tumorigenesis. Cancer Res. 2008, 68, 9589–9594. [Google Scholar] [CrossRef]
  85. Chiang, J.Y.; Ferrell, J.M. Bile acid metabolism in liver pathobiology. Gene Expr. 2018, 18, 71. [Google Scholar] [CrossRef] [Green Version]
  86. Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.-Z.; Takahashi, S. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Investig. 2015, 125, 386–402. [Google Scholar] [CrossRef]
  87. Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef]
  88. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  89. Schwiertz, A.; Taras, D.; Schäfer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef]
  90. Ruppin, H.; Bar-Meir, S.; Soergel, K.H.; Wood, C.M.; Schmitt Jr, M.G. Absorption of short-chain fatty acids by the colon. Gastroenterology 1980, 78, 1500–1507. [Google Scholar] [CrossRef]
  91. Vinolo, M.A.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef]
  92. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef]
  93. Byndloss, M.X.; Olsan, E.E.; Rivera-Chávez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef]
  94. Da Zhou, Q.P.; Xin, F.-Z.; Zhang, R.-N.; He, C.-X.; Chen, G.-Y.; Liu, C.; Chen, Y.-W.; Fan, J.-G. Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J. Gastroenterol. 2017, 23, 60. [Google Scholar] [CrossRef]
  95. Wu, G.; Meininger, C.J. Regulation of nitric oxide synthesis by dietary factors. Annu. Rev. Nutr. 2002, 22, 61–86. [Google Scholar] [CrossRef]
  96. Evenepoel, P.; Claus, D.; Geypens, B.; Hiele, M.; Geboes, K.; Rutgeerts, P.; Ghoos, Y. Amount and fate of egg protein escaping assimilation in the small intestine of humans. Am. J. Physiol. -Gastrointest. Liver Physiol. 1999, 277, G935–G943. [Google Scholar] [CrossRef]
  97. Smith, E.A.; Macfarlane, G.T. Enumeration of human colonic bacteria producing phenolic and indolic compounds: Effects of pH, carbohydrate availability and retention time on dissimilatory aromatic amino acid metabolism. J. Appl. Bacteriol. 1996, 81, 288–302. [Google Scholar] [CrossRef]
  98. Chen, J.; Vitetta, L. Gut microbiota metabolites in nafld pathogenesis and therapeutic implications. Int. J. Mol. Sci. 2020, 21, 5214. [Google Scholar] [CrossRef]
  99. Herrera, C.P.; Smith, K.; Atkinson, F.; Ruell, P.; Chow, C.M.; O’Connor, H.; Brand-Miller, J. High-glycaemic index and-glycaemic load meals increase the availability of tryptophan in healthy volunteers. Br. J. Nutr. 2011, 105, 1601–1606. [Google Scholar] [CrossRef]
  100. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  101. Gostner, J.M.; Geisler, S.; Stonig, M.; Mair, L.; Sperner-Unterweger, B.; Fuchs, D. Tryptophan metabolism and related pathways in psychoneuroimmunology: The impact of nutrition and lifestyle. Neuropsychobiology 2020, 79, 89–99. [Google Scholar] [CrossRef]
  102. Bansal, T.; Alaniz, R.C.; Wood, T.K.; Jayaraman, A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 228–233. [Google Scholar] [CrossRef]
  103. Cherayil, B.J. Indoleamine 2, 3-dioxygenase in intestinal immunity and inflammation. Inflamm. Bowel Dis. 2009, 15, 1391–1396. [Google Scholar] [CrossRef]
  104. Laurans, L.; Venteclef, N.; Haddad, Y.; Chajadine, M.; Alzaid, F.; Metghalchi, S.; Sovran, B.; Denis, R.G.; Dairou, J.; Cardellini, M. Genetic deficiency of indoleamine 2, 3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat. Med. 2018, 24, 1113–1120. [Google Scholar] [CrossRef]
  105. Ji, Y.; Yin, Y.; Li, Z.; Zhang, W. Gut microbiota-derived components and metabolites in the progression of non-alcoholic fatty liver disease (NAFLD). Nutrients 2019, 11, 1712. [Google Scholar] [CrossRef] [Green Version]
  106. Hou, Q.; Ye, L.; Liu, H.; Huang, L.; Yang, Q.; Turner, J.; Yu, Q. Lactobacillus accelerates ISCs regeneration to protect the integrity of intestinal mucosa through activation of STAT3 signaling pathway induced by LPLs secretion of IL-22. Cell Death Differ. 2018, 25, 1657–1670. [Google Scholar] [CrossRef]
  107. Ma, L.; Li, H.; Hu, J.; Zheng, J.; Zhou, J.; Botchlett, R.; Matthews, D.; Zeng, T.; Chen, L.; Xiao, X. Indole Alleviates Diet-Induced Hepatic Steatosis and Inflammation in a Manner Involving Myeloid Cell 6-Phosphofructo-2-Kinase/Fructose-2, 6-Biphosphatase 3. Hepatology 2020, 72, 1191–1203. [Google Scholar] [CrossRef]
  108. Kessel, A.; Toubi, E.; Pavlotzky, E.; Mogilner, J.; Coran, A.; Lurie, M.; Karry, R.; Sukhotnik, I. Treatment with glutamine is associated with down-regulation of Toll-like receptor-4 and myeloid differentiation factor 88 expression and decrease in intestinal mucosal injury caused by lipopolysaccharide endotoxaemia in a rat. Clin. Exp. Immunol. 2008, 151, 341–347. [Google Scholar] [CrossRef]
  109. Simon, J.; Martinez-Chantar, M.L.; Delgado, T.C. Glutamine, fatty liver disease and aging. Aging 2021, 13, 3165–3166. [Google Scholar] [CrossRef]
  110. Zhang, W.-X.; Zhou, L.-F.; Zhang, L.; Bao, L.; Wang, C.-C.; Meng, H.-Y.; Yin, W. Protective effects of glutamine preconditioning on ischemia-reperfusion injury in rats. Hepatobiliary Pancreat. Dis. Int. 2011, 10, 78–82. [Google Scholar] [CrossRef]
  111. Neu, J.; DeMarco, V.; Li, N. Glutamine: Clinical applications and mechanisms of action. Curr. Opin. Clin. Nutr. Metab. Care 2002, 5, 69–75. [Google Scholar] [CrossRef]
  112. Corbin, K.D.; Zeisel, S.H. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr. Opin. Gastroenterol. 2012, 28, 159–165. [Google Scholar] [CrossRef]
  113. Guerrerio, A.L.; Colvin, R.M.; Schwartz, A.K.; Molleston, J.P.; Murray, K.F.; Diehl, A.; Mohan, P.; Schwimmer, J.B.; Lavine, J.E.; Torbenson, M.S.; et al. Choline intake in a large cohort of patients with nonalcoholic fatty liver disease. Am. J. Clin. Nutr. 2012, 95, 892–900. [Google Scholar] [CrossRef] [PubMed]
  114. Yao, Z.M.; Vance, D.E. Reduction in VLDL, but not HDL, in plasma of rats deficient in choline. Biochem Cell Biol. 1990, 68, 552–558. [Google Scholar] [CrossRef] [PubMed]
  115. Schneider, K.M.; Mohs, A.; Kilic, K.; Candels, L.S.; Elfers, C.; Bennek, E.; Schneider, L.B.; Heymann, F.; Gassler, N.; Penders, J.; et al. Intestinal Microbiota Protects against MCD Diet-Induced Steatohepatitis. Int. J. Mol. Sci. 2019, 20, 308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ye, J.Z.; Li, Y.T.; Wu, W.R.; Shi, D.; Fang, D.Q.; Yang, L.Y.; Bian, X.Y.; Wu, J.J.; Wang, Q.; Jiang, X.W.; et al. Dynamic alterations in the gut microbiota and metabolome during the development of methionine-choline-deficient diet-induced nonalcoholic steatohepatitis. World J. Gastroenterol. 2018, 24, 2468–2481. [Google Scholar] [CrossRef]
  117. Baker, J.R.; Chaykin, S. The biosynthesis of trimethylamine-N-oxide. J. Biol. Chem. 1962, 237, 1309–1313. [Google Scholar] [CrossRef]
  118. Lang, D.H.; Yeung, C.K.; Peter, R.M.; Ibarra, C.; Gasser, R.; Itagaki, K.; Philpot, R.M.; Rettie, A.E. Isoform specificity of trimethylamine N-oxygenation by human flavin-containing monooxygenase (FMO) and P450 enzymes: Selective catalysis by FMO3. Biochem. Pharmacol. 1998, 56, 1005–1012. [Google Scholar] [CrossRef]
  119. Romano, K.A.; Martinez-Del Campo, A.; Kasahara, K.; Chittim, C.L.; Vivas, E.I.; Amador-Noguez, D.; Balskus, E.P.; Rey, F.E. Metabolic, Epigenetic, and Transgenerational Effects of Gut Bacterial Choline Consumption. Cell Host Microbe 2017, 22, 279–290.e277. [Google Scholar] [CrossRef]
  120. Spencer, M.D.; Hamp, T.J.; Reid, R.W.; Fischer, L.M.; Zeisel, S.H.; Fodor, A.A. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011, 140, 976–986. [Google Scholar] [CrossRef]
  121. Chen, Y.M.; Liu, Y.; Zhou, R.F.; Chen, X.L.; Wang, C.; Tan, X.Y.; Wang, L.J.; Zheng, R.D.; Zhang, H.W.; Ling, W.H.; et al. Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci. Rep. 2016, 6, 19076. [Google Scholar] [CrossRef]
  122. Gao, X.; Liu, X.; Xu, J.; Xue, C.; Xue, Y.; Wang, Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J. Biosci. Bioeng. 2014, 118, 476–481. [Google Scholar] [CrossRef] [PubMed]
  123. Shabbir, U.; Rubab, M.; Daliri, E.B.-M.; Chelliah, R.; Javed, A.; Oh, D.-H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  124. Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef] [PubMed]
  125. Ding, S.; Xu, S.; Ma, Y.; Liu, G.; Jang, H.; Fang, J. Modulatory mechanisms of the NLRP3 inflammasomes in diabetes. Biomolecules 2019, 9, 850. [Google Scholar] [CrossRef] [Green Version]
  126. Ma, J.; Zheng, Y.; Tang, W.; Yan, W.; Nie, H.; Fang, J.; Liu, G. Dietary polyphenols in lipid metabolism: A role of gut microbiome. Anim. Nutr. 2020, 6, 404–409. [Google Scholar] [CrossRef]
  127. Bian, Y.; Wei, J.; Zhao, C.; Li, G. Natural polyphenols targeting senescence: A novel prevention and therapy strategy for cancer. Int. J. Mol. Sci. 2020, 21, 684. [Google Scholar] [CrossRef]
  128. Alves-Santos, A.M.; Sugizaki, C.S.A.; Lima, G.C.; Naves, M.M.V. Prebiotic effect of dietary polyphenols: A systematic review. J. Funct. Foods 2020, 74, 104169. [Google Scholar] [CrossRef]
  129. Moorthy, M.; Chaiyakunapruk, N.; Jacob, S.A.; Palanisamy, U.D. Prebiotic potential of polyphenols, its effect on gut microbiota and anthropometric/clinical markers: A systematic review of randomised controlled trials. Trends Food Sci. Technol. 2020, 99, 634–649. [Google Scholar] [CrossRef]
  130. Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104, S1–S63. [Google Scholar] [CrossRef]
  131. Rafiei, H.; Omidian, K.; Bandy, B. Dietary polyphenols protect against oleic acid-induced steatosis in an in vitro model of NAFLD by modulating lipid metabolism and improving mitochondrial function. Nutrients 2019, 11, 541. [Google Scholar] [CrossRef]
  132. Li, A.-N.; Li, S.; Zhang, Y.-J.; Xu, X.-R.; Chen, Y.-M.; Li, H.-B. Resources and biological activities of natural polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef] [PubMed]
  133. Lamuel-Raventos, R.M.; Onge, M.-P.S. Prebiotic nut compounds and human microbiota. Crit. Rev. Food Sci. Nutr. 2017, 57, 3154–3163. [Google Scholar] [CrossRef] [PubMed]
  134. Li, S.; Tan, H.Y.; Wang, N.; Cheung, F.; Hong, M.; Feng, Y. The potential and action mechanism of polyphenols in the treatment of liver diseases. Oxidative Med. Cell. Longev. 2018, 2018, 8394818. [Google Scholar] [CrossRef] [PubMed]
  135. Byelashov, O.A.; Sinclair, A.J.; Kaur, G. Dietary sources, current intakes, and nutritional role of omega-3 docosapentaenoic acid. Lipid Technol. 2015, 27, 79–82. [Google Scholar] [CrossRef] [Green Version]
  136. Calder, P.C. Marine omega-3 fatty acids and inflammatory processes: Effects, mechanisms and clinical relevance. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2015, 1851, 469–484. [Google Scholar] [CrossRef]
  137. Calder, P.C. Omega-3 polyunsaturated fatty acids and inflammatory processes: Nutrition or pharmacology? Br. J. Clin. Pharmacol. 2013, 75, 645–662. [Google Scholar] [CrossRef]
  138. Brown, T.J.; Brainard, J.; Song, F.; Wang, X.; Abdelhamid, A.; Hooper, L. Omega-3, omega-6, and total dietary polyunsaturated fat for prevention and treatment of type 2 diabetes mellitus: Systematic review and meta-analysis of randomised controlled trials. BMJ 2019, 366, l4697. [Google Scholar] [CrossRef]
  139. He, X.-X.; Wu, X.-L.; Chen, R.-P.; Chen, C.; Liu, X.-G.; Wu, B.-J.; Huang, Z.-M. Effectiveness of omega-3 polyunsaturated fatty acids in non-alcoholic fatty liver disease: A meta-analysis of randomized controlled trials. PLoS ONE 2016, 11, e0162368. [Google Scholar] [CrossRef]
  140. Simopoulos, A.P. An increase in the omega-6/omega-3 fatty acid ratio increases the risk for obesity. Nutrients 2016, 8, 128. [Google Scholar] [CrossRef]
  141. de Castro, G.S.; Calder, P.C. Non-alcoholic fatty liver disease and its treatment with n-3 polyunsaturated fatty acids. Clin. Nutr. 2018, 37, 37–55. [Google Scholar] [CrossRef]
  142. Liu, H.-Q.; Qiu, Y.; Mu, Y.; Zhang, X.-J.; Liu, L.; Hou, X.-H.; Zhang, L.; Xu, X.-N.; Ji, A.-L.; Cao, R. A high ratio of dietary n-3/n-6 polyunsaturated fatty acids improves obesity-linked inflammation and insulin resistance through suppressing activation of TLR4 in SD rats. Nutr. Res. 2013, 33, 849–858. [Google Scholar] [CrossRef] [PubMed]
  143. Druart, C.; Bindels, L.B.; Schmaltz, R.; Neyrinck, A.M.; Cani, P.D.; Walter, J.; Ramer-Tait, A.E.; Delzenne, N.M. Ability of the gut microbiota to produce PUFA-derived bacterial metabolites: Proof of concept in germ-free versus conventionalized mice. Mol. Nutr. Food Res. 2015, 59, 1603–1613. [Google Scholar] [CrossRef] [PubMed]
  144. Cieślak, A.; Trottier, J.; Verreault, M.; Milkiewicz, P.; Vohl, M.-C.; Barbier, O. N-3 polyunsaturated fatty acids stimulate bile acid detoxification in human cell models. Can. J. Gastroenterol. Hepatol. 2018, 2018, 6031074. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, Z.; Hyun, J.E.; Thiesen, A.; Park, H.; Hotte, N.; Watanabe, H.; Higashiyama, T.; Madsen, K.L. Sex-Specific Differences in the Gut Microbiome in Response to Dietary Fiber Supplementation in IL-10-Deficient Mice. Nutrients 2020, 12, 2088. [Google Scholar] [CrossRef] [PubMed]
  146. Stephen, A.M.; Champ, M.M.-J.; Cloran, S.J.; Fleith, M.; van Lieshout, L.; Mejborn, H.; Burley, V.J. Dietary fibre in Europe: Current state of knowledge on definitions, sources, recommendations, intakes and relationships to health. Nutr. Res. Rev. 2017, 30, 149–190. [Google Scholar] [CrossRef]
  147. Den Besten, G.; Van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef]
  148. Murga-Garrido, S.M.; Hong, Q.; Cross, T.-W.L.; Hutchison, E.R.; Han, J.; Thomas, S.P.; Vivas, E.I.; Denu, J.; Ceschin, D.G.; Tang, Z.-Z. Gut microbiome variation modulates the effects of dietary fiber on host metabolism. Microbiome 2021, 9, 117. [Google Scholar] [CrossRef]
  149. Kuo, S.-M. The interplay between fiber and the intestinal microbiome in the inflammatory response. Adv. Nutr. 2013, 4, 16–28. [Google Scholar] [CrossRef]
  150. Choe, S.S.; Huh, J.Y.; Hwang, I.J.; Kim, J.I.; Kim, J.B. Adipose tissue remodeling: Its role in energy metabolism and metabolic disorders. Front. Endocrinol. 2016, 7, 30. [Google Scholar] [CrossRef]
  151. Argo, C.K.; Northup, P.G.; Al-Osaimi, A.M.; Caldwell, S.H. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J. Hepatol. 2009, 51, 371–379. [Google Scholar] [CrossRef]
  152. Ma, J.; Fox, C.S.; Jacques, P.F.; Speliotes, E.K.; Hoffmann, U.; Smith, C.E.; Saltzman, E.; McKeown, N.M. Sugar-sweetened beverage, diet soda, and fatty liver disease in the Framingham Heart Study cohorts. J. Hepatol. 2015, 63, 462–469. [Google Scholar] [CrossRef] [PubMed]
  153. Do, M.H.; Lee, E.; Oh, M.-J.; Kim, Y.; Park, H.-Y. High-glucose or-fructose diet cause changes of the gut microbiota and metabolic disorders in mice without body weight change. Nutrients 2018, 10, 761. [Google Scholar] [CrossRef] [PubMed]
  154. Ishimoto, T.; Lanaspa, M.A.; Rivard, C.J.; Roncal-Jimenez, C.A.; Orlicky, D.J.; Cicerchi, C.; McMahan, R.H.; Abdelmalek, M.F.; Rosen, H.R.; Jackman, M.R. High-fat and high-sucrose (western) diet induces steatohepatitis that is dependent on fructokinase. Hepatology 2013, 58, 1632–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Interaction between nutrition and gut microbiome in the progression of chronic liver disease. SCFAs, short-chain fatty acids; GPR43, G-protein-coupled receptor 43; GLP, glucagon-like peptide; LPS, lipopolysaccharide; IL-10, interleukin-10; IL-22, interleukin-22; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TNF, tumor necrosis factor; TMA, trimethylamine; TMAO, trimethylamine-N-oxide; IPA, indole-3-propionic acid; I3A, indole-3-acetic acid; PXR, pregnane X receptor; AhR, aryl hydrocarbon receptor; AMPK, AMP-activated protein kinase; GLP-1, glucagon-like peptide-1; GPR4, G-protein-coupled receptor; FXR, farnesoid X receptor.
Figure 1. Interaction between nutrition and gut microbiome in the progression of chronic liver disease. SCFAs, short-chain fatty acids; GPR43, G-protein-coupled receptor 43; GLP, glucagon-like peptide; LPS, lipopolysaccharide; IL-10, interleukin-10; IL-22, interleukin-22; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TNF, tumor necrosis factor; TMA, trimethylamine; TMAO, trimethylamine-N-oxide; IPA, indole-3-propionic acid; I3A, indole-3-acetic acid; PXR, pregnane X receptor; AhR, aryl hydrocarbon receptor; AMPK, AMP-activated protein kinase; GLP-1, glucagon-like peptide-1; GPR4, G-protein-coupled receptor; FXR, farnesoid X receptor.
Foods 11 02703 g001
Table 1. Changes in gut microbiota and metabolites in different types of diets.
Table 1. Changes in gut microbiota and metabolites in different types of diets.
NutrientMicrobiota ChangesAltered MetabolitesReference
Protein (↑): Bacteroidetes, Lactobacillus(↑): Sulfide, polysaccharide lyases, tryptophan catabolism[26,27]
(↓): Firmicutes, Clostridium
Omega-3 (↑): Bifidobacteria, Lachnospiraceae, Roseburia, Bacteroidetes(↑): SCFAs[28,29]
(↓): Enterobacteria, Faecalibacterium(↓): IL-1β, IL-6, TNF-α
Fiber(↑): Prevotella, Xylanibacter spp., Bifidobacterium, Roseburia, Faecalibacterium(↑): SCFAs[30,31,32]
Low-fiber(↑): Akkermansia, Bacteroides caccae[33]
(↓): Escherichia coli
Fat intake(↑): Firmicutes, Clostridium(↑): LPS, Indoxyl sulfate, p-cresyl sulfate[34,35]
(↓): Lactobacillus, Bacteroidetes, Bifidobacteria spp., Akkermansia
Fructose intake(↑): Clostridium innocuum, Catenibacterium mitsuokai, Enterococcus spp.TMAO[36,37]
High-fat diet(↑): Firmicutes, Proteobacteria(↑): TMAO, LPS[24,38,39,40]
(↓): Bacteroidetes, Bifidobacteria
(↑) increase; (↓) decrease. LPS, lipopolysaccharide; TMAO, trimethylamine N-oxide; SCFA, short-chain fatty acids; PUFA, polyunsaturated fatty acid; IL-1β, interleukin-1 beta; IL-6, interleukin-6; TNF, tumor necrosis factor.
Table 2. The alterations in gut microbiota in the pathogenesis of nonalcoholic fatty liver disease.
Table 2. The alterations in gut microbiota in the pathogenesis of nonalcoholic fatty liver disease.
Liver DiseaseMicrobiota ChangesMajor ImpactsReferences
NAFLD(↑): Proteobacteria, Firmicutes, Lactobacillus, Parabacteroides, Allisonella, C. coccoides
(↓): Oscillibacter, Faecalibacterium, Anaerosporobacter
(↑): FFAs, triglycerides, de novo lipogenesis
(↓): ApoB
[45,46,47]
NAFLD-associated Cirrhosis(↑): Streptococcus, Lactococcus, Enterobacteriaceae
(↓): Bacteroidetes, Bacillus, F. prausnitzii, Prevotella
(↑): PDGF, TGF-β, ECM
[48,49]
NAFLD-associated HCC(↑): E. coli, Actinobacteria
(↓): Parabacteroides, butyrate-producing genera
(↑): HBV, HCV, Wnt/β-catenin, JAK/STAT, Rb, p53, MAPK[50,51]
Model
Methionine choline-deficient diet model, leptin receptor deficiency steatosis model, and the high-fat diet model.(↑): Hepatic steatosis, hepatic inflammation, lipid biosynthesis
(↑): Bacteroidetes, Prevotellaceae, Deferribacteres, Oscillibacter
(↓): Lactobacillus, Bacteroidetes
[52]
High-fat (45% energy) or low-fat (10% energy) diet for 10 weeks.(↑): Body weight (by 34%)
(↓): Hepatic steatosis, hepatic inflammation
(↑): L. gasseri, L.s taiwanensis
[53]
High-fat diet-induced and lean mice (7 weeks): supplemented with B. pseudocatenulatum(↓): Insulin resistance, hepatic fat, serum inflammatory markers, body weight
(↑): Bifidobacteria, Enterobacteriaceae
[54]
(↑) increase; (↓) decrease. NAFLD, nonalcoholic fatty liver disease; HCC, hepatocellular carcinoma; ALD, alcohol-related liver disease; FFAs, free fatty acids; PDGF, platelet-derived growth factor; ECM, extracellular matrix; HBV, hepatitis B virus; HCV, hepatitis C virus; JAK, Janus kinase; STAT, signal transducer and activator of transcription; MAPK, mitogen-activated protein kinase; ROS, reactive oxygen species.
Table 3. Nutrients associated with microbiota in nonalcoholic fatty liver disease.
Table 3. Nutrients associated with microbiota in nonalcoholic fatty liver disease.
NutrientsMicrobiotaFunctionReference
Bile acidBifidobacterium, Lactobacillus, Enterobacter, Clostridium, Bacteroides(↑): Inhibit growth of pathogens, gut-derived hormone secretion, insulin resistance, gut barrier[60,61,62,63]
SCFAsBifidobacterium sp., Roseburia, Clostridium, Faecalibacterium, Coprococcus(↑): Protect from diet-induced obesity,
(↓): Hepatic lipid accumulation
[64,65]
AcetateBacteroidetes(↑): Insulin signaling, hepatic function, activation of GPR43
(↓): Fat accumulation, lipid storage
[66,67]
PropionateBacteroidetes, B. obeum, C. catus, R. inulinivorans, P. copri(↑): Regulation of colonic T reg cell homeostasis,
ButyrateClostridia, F. prausnitzii, Eubacterium, Roseburia, C. catus, A. hadrus(↑): GLP-2, GLP-1R level, antiinflammation, gut–gut barrier function, T reg cell homeostasis, colonic suppression of colonic inflammation, activation of GPR43[60,68,69]
(↑) increase; (↓) decrease. SCFAs, short-chain fatty acids; GPR43, G-protein-coupled receptor 43; GLP, glucagon-like peptide; LPS, lipopolysaccharide.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jeong, M.K.; Min, B.H.; Choi, Y.R.; Hyun, J.Y.; Park, H.J.; Eom, J.A.; Won, S.M.; Jeong, J.J.; Oh, K.K.; Gupta, H.; et al. Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease. Foods 2022, 11, 2703. https://doi.org/10.3390/foods11172703

AMA Style

Jeong MK, Min BH, Choi YR, Hyun JY, Park HJ, Eom JA, Won SM, Jeong JJ, Oh KK, Gupta H, et al. Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease. Foods. 2022; 11(17):2703. https://doi.org/10.3390/foods11172703

Chicago/Turabian Style

Jeong, Min Kyo, Byeong Hyun Min, Ye Rin Choi, Ji Ye Hyun, Hee Jin Park, Jung A Eom, Sung Min Won, Jin Ju Jeong, Ki Kwang Oh, Haripriya Gupta, and et al. 2022. "Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease" Foods 11, no. 17: 2703. https://doi.org/10.3390/foods11172703

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop