Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disorder in the world, yet the pathogenesis of the disease is not well elucidated. Due to the close anatomic and functional association between the intestinal lumen and the liver through the portal system, it is speculated that the gut microbiome may play a pivotal role in the pathogenesis of NAFLD. Furthermore, diet, which can modulate the gut microbiome and several metabolic pathways involved in NAFLD development, shows a potential tripartite relation between the gut, diet, and the liver. In this review, we summarize the current evidence that supports the association between NAFLD, the gut microbiome, and the role of diet.
Keywords: nonalcoholic fatty liver disease, gut microbiota, diet, NAFLD, NASH, probiotics
Introduction
Nonalcoholic fatty liver disease (NAFLD)6 is the most common cause of chronic liver diseases in the world (1). NAFLD is defined as a liver pathologic spectrum, which is initiated by steatosis that may progress to nonalcoholic steatohepatitis (NASH). NASH manifestations beyond steatosis include necro-inflammation and fibrosis (2). The disease can progress to cirrhosis and eventual hepatocellular carcinoma (3). The reported prevalence of NAFLD is 30% in Western countries (1, 4) and 12–24% in Asia (5), which shows the global importance of understanding the disease pathology. In addition to genetic background, environmental factors such as diet and the intestinal microbiome should be considered critical factors in the development of NAFLD (6, 7).
The gut microbiome, the trillions of microbes living in the gut, may contribute to the pathogenesis of liver diseases (8, 9). Increasing evidence in recent decades shows that the gut microbiome plays a significant role in metabolism, health, and disease of the host (10), which suggests that the gut microbiome is a metabolic organ in the host. Considering that the anatomy of the liver facilitates a close interaction between the liver and gut microbiome, it is possible that gut microbial metabolites and circulating byproducts influence liver function. Indeed, it is known that slow blood flow and fenestrated endothelium in the sinusoids lead to the interaction between substances derived from the gut and hepatocytes, parenchymal cells, and hepatic immune cells (11). Similarly, dietary antigens are known to affect liver function, and because dietary components are among the strongest factors that predict the ecosystem that makes up the gut microbiome (12, 13), both diet and the microbiome need to be considered in the pathogenesis of NAFLD. In the present review, we summarize the current evidence that supports the association between NAFLD, the gut microbiome, and dietary factors.
Current Status of Knowledge
NAFLD and the gut microbiome.
The gut harbors ∼1013–1014 bacteria, which is predicted to encode genes >100 times as in the human genome (14). Despite the wide microbial diversity in humans, only 4 main phyla dominate: Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria. Approximately 90% of the microbes come from the Firmicutes and Bacteroidetes phyla (14) and each has unique functions. Major contributors of Firmicutes are as follows: Lachnospiraceae, which are composed mostly of microbes from the genera Blautia, Clostridium, Coprococcus, Eubacterium, Roseburia, and Ruminococcaceae, which are composed mostly of the genera Faecalibacterium, Oscillospira, and Ruminococcus. Major contributors of the Bacteroidetes phylum are Bacteroides, Parabacteroides, Porphyromonas, Prevotellaceae (Prevotella), and Rikenellaceae (Alistipes). The most prominent Actinobacteria are Bifidobacteriaceae (Bifidobacterium), and the most abundant Proteobacteria are the Enterobacteriaceae (Escherichia) (15, 16). Although the functions of most Firmicutes are not yet clear, there is some evidence that indicates that some members of this phylum are among the butyrate-producing bacteria that increase energy extraction from the diet (17–19). On the other hand, members of the phylum Bacteroidetes participate in carbohydrate metabolism and accomplish this by expressing enzymes similar to glycosyl transferases, glycoside hydrolases, and polysaccharide lyases (20). Understanding the functions associated with the microbial community is important, because alterations in the intestinal microbiota have been associated with host diseases, including obesity, type 2 diabetes, and NAFLD (8, 21, 22). In patients with NAFLD, the microbiome’s abundance (23) and community structure is altered (also called dysbiosis) (15, 24). The importance of the gut microbiome in the prevention and treatment of NAFLD is highlighted by the fact that fecal transplantation from mice with NAFLD into wild-type mice caused NAFLD in mice that received the microbiota (25). In addition, the alteration of microbiota can change the disease severity (26–30). Overall, these results suggest that the microbiome is important in NAFLD.
Several lines of evidence have shown that NAFLD can be affected by the gut microbiome through various proposed mechanisms. The contribution of increased intestinal permeability (20), overgrowth of bacteria (15), and elevated serum LPS (31, 32) has been shown in patients with NAFLD. Overgrowth of bacteria, in particular gram-negative bacteria, increases the production of hepatotoxic products such as LPS (23). The disruption of the intestinal integrity increases intestinal permeability, which leads to bacterial translocation, and bacterial endotoxins penetrate into the portal vein, which increases the risk of NAFLD development through the activation of hepatic inflammatory cells (23, 33). Bacterial endotoxins are recognized by toll-like receptors (TLRs) on hepatocytes, which recognize several components of microbes and initiate immunologic responses (32).When bacterial LPS signals through TLR4, signaling ultimately activates NF-κB and proinflammatory cytokines (32, 34). Moreover, dysbiosis is associated with reduced synthesis and secretion of fasting-induced adipocyte factor [(FIAF) also known as angiopoietin-like 4] in enterocytes, which results in increased activity of lipoprotein lipase (LPL) and the accumulation of TGs in the liver (35, 36).
Dysbiosis and inflammasome deficiency may be important in the development of hepatic steatosis and inflammation (37, 38). Inflammasomes are cytoplasmic multiprotein complexes that sense endogenous or exogenous stress- or damage-associated molecular patterns (39, 40). Upon sensing the relevant signal, they assemble, typically together with the adaptor protein, apoptosis-associated speck-like protein (ASC), into a multiprotein complex that governs caspase-1 activation and subsequent cleavage of effector proinflammatory cytokines, including pro–IL-1β and pro–IL-18, which ultimately leads to apoptosis (38). In inflammasome deficiency, large amounts of bacterial endotoxins enter the portal vein, and the liver then has to process high amounts of endotoxins, which leads to aberrant and damaging inflammatory responses that promote progression to NASH (37). Another mechanism by which the gut microbiota contributes to the development of NAFLD may be through increasing the number of ethanol-producing bacteria (e.g., Escherichia coli) (15). These bacteria produce alcohol, which could participate in the disruption of gut permeability, the generation of ROS, and consequently, liver inflammation (41, 42). However, the specific mechanisms of this hypothesis require further investigation.
The characterization of the gut microbial composition and comparing it between patients and healthy individuals is regarded as one way of identifying a healthy microbial ecosystem (43). Indeed, a few studies have examined dysbiosis in NAFLD (Table 1). Patients with NASH harbor a lower abundance of Faecalibacterium and Firmicutes (Clostridiales family, Anaerosporobacter) but a higher abundance of Parabacteroides and Allisonella in their fecal microbiome (45). Importantly, it has been reported that improved intrahepatic TG content is related to a lower abundance of Firmicutes and a higher abundance of Bacteroidetes (45). In support of this, obese individuals with NAFLD have increased members of the phylum Firmicutes [Lachnospiraceae (Dorea, Robinsoniella, and Roseburia)] (46). Yet, children and adolescents with NASH do not show similar patterns and instead have increased Bacteriodetes [Prevotellaceae (Prevotella, Porphyromonas)] and Proteobacteria [Enterobacteriaceae (Escherichia), Alcaligenaceae] and a decrease in Firmicutes [Lachnospiraceae (Blautia, Coproccus, Eubacterium, Roseburia), Ruminococcaceae (Faecalibacterium, Oscillospira, Ruminococcus)] and Actinobacteria [Bifidobacteriaceae (Bifidobacterium)] in their fecal microbiomes (15). In support of these studies, pediatric patients with NAFLD showed an increase in Bradyrhizobium, Anaerococcus, Peptoniphilus, Propionibacterium acnes, Dorea, and Ruminococcus and a decrease in Oscillospira and Rikenellaceae (44). Although it is clear that there may be some discrepancies in what defines dysbiosis in liver disease, the frequency of disease also occurs in association with obesity and is considered a manifestation of metabolic syndrome. Thus, the dysbiosis may be related to these metabolic disturbances, considering that several studies suggested that increased Firmicutes and reduced Bacteroidetes may be a cause of obesity (17, 47). However, the reduction in Bacteroidetes is not simply a cause of obesity in patients with NASH, considering that Bacteroidetes abundance is reduced in these patients even after adjusting for BMI and fat intake (24).
TABLE 1.
Intestinal microbiota composition in patients with NAFLD1
Study, year (ref) | Subjects | Method | Change in microbiota |
Del Chierico et al., 2016 (44) | Pediatric NAFLD, NASH, or obesity (n = 61); healthy subjects (n = 54) | 16S rRNA pyrosequencing | NAFLD vs. healthy controls: |
↑Bradyrhizobium, Anaerococcus, Peptoniphilus, Propionibacterium acnes, Dorea, Ruminococcus | |||
↓Oscillospira and Rikenellaceae | |||
Mouzaki et al., 2013 (24) | NASH (n = 22); SS (n = 11); healthy controls (n = 17) | qPCR | NASH vs. both SS and healthy controls: |
↓Percentage of Bacteroidetes (Bacteroidetes to total bacteria counts) | |||
NASH vs. SS: | |||
↑Clostridium coccoides | |||
Wong et al., 2013 (45) | NASH (n = 16); healthy controls (n = 22) | 16S rRNA pyrosequencing | NASH vs. healthy controls: |
↓Faecalibacterium, ↓Anaerosporobacter | |||
↑Parabacteroides, ↑Allisonella | |||
Improvement in intrahepatic TG content was associated with a reduction in the abundance of Firmicutes (R2 = 0.4820, P = 0.0028) and an increase in Bacteroidetes (R2 = 0.4366, P = 0.0053) | |||
Raman et al., 2013 (46) | Obese NAFLD patients (n = 30); healthy controls (n = 30) | 16S rRNA pyrosequencing | Obese NAFLD vs. healthy controls: |
↑Lactobacillus species | |||
↑Firmicutes (Lachnospiraceae; genera: Dorea, Robinsoniella, and Roseburia) | |||
↓One member of phylum Firmicutes (Ruminococcaceae; genus, Oscillibacter) | |||
↓A member of phylum Bacteroidetes (Porphyromonadaceae) | |||
Zhu et al., 2013 (15) | NASH children (n = 22); obese children (n = 25); healthy children (n = 16) | 16S rRNA pyrosequencing | NASH vs. healthy controls: |
↑Proteobacteria [Enterobacteriaceae (Escherichia), Alcaligenaceae] | |||
↑Bacteroidetes [Prevotellaceae (Prevotella), Porphyromonas], ↓Rikenellaceae (Alistipes) | |||
↓Actinobacteria [Bifidobacteriaceae (Bifidobacterium)] | |||
↓Firmicutes [Lachnospiraceae (Blautia, Coproccus, Eubacterium, Roseburia), Ruminococcaceae (Faecalibacterium, Oscillospira, Ruminococcus)], ↑Peptoniphilus | |||
Wigg et al., 2001 (23) | Healthy controls (n = 23) | C-d-xylose and lactulose breath test | Small intestinal bacterial overgrowth was present in 50% of patients with nonalcoholic steatosis and in 22% of control subjects (P = 0.048) |
NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; ref, reference; rRNA, ribosomal RNA; SS, simple steatosis; ↑, increased; ↓, decreased.
Overall, the evidence suggests that the gut microbiome may have an important role in NAFLD pathology, but the studies have not identified a particular microbe involved due to the heterogeneous results. Similar to other microbiome studies, discrepancies can be due to variations in the study designs. Some of the studies, such as the study conducted by Zhu et al. (15), used patients with no history of antibiotics, probiotics, proton pump inhibitors, and histamine receptor antagonists within 3 mo before examining the fecal microbiota; however, others did not consider all of these precautions. In addition, the collection and processing of fecal samples have been shown to produce large variances and inaccuracies in the interpretation of the taxa present in the microbiota (48), which may be a contributing factor to the conflicting data found in NAFLD microbiome studies. Even so, all of these studies have evaluated the association, and well-designed studies are needed to unravel any causal relation between the gut microbes and NAFLD.
Diet and the gut microbiome.
Dietary factors are strong predictors of the gut microbiota composition (49–51). In fact, it has been projected that dietary factors play a more important role in shaping the gut microbiota composition than do genetic factors (52). To understand the role of nutrition, the gut microbiome, and NAFLD, we summarized the experimental studies that evaluated this potential relation (Table 2).
TABLE 2.
Studies evaluating the effects of dietary factors on the gut microbiota in animal models of NAFLD1
Study, year (ref) | Animal model | Treatment | Method(s) | Main results |
Saha and Reimer, 2014 (53) | Wistar rats | HF or HP diet from 3 to 15 wk of age; a high-fat, high-sucrose diet from 15 to 21 wk; the respective HF or HP diets from 21 to 25 wk | qPCR | HF vs. control group: |
↑Total bacteria, ↑Bifidobacteria, and ↑Bacteroides:Prevotella | ||||
HF vs. HP: | ||||
↓Firmicutes, ↓Firmicutes:Bacteroidetes, ↓hepatic cholesterol content | ||||
Negative correlation between liver weight and Bacteroides:Prevotella (r = −0.415, P = 0.044) | ||||
Bomhof et al., 2014 (54) | Sprague-Dawley rats | Initiate with a high-fat, high-sucrose diet for 8 wk and then prebiotic OFSs vs. the probiotic BB-12 for 8 wk | qPCR | Prebiotic oligofructose vs control: |
↓Energy intake, ↓ weight gain, ↓fat mass, ↑PYY, ↑Bifidobacteria, ↑Lactobacilli | ||||
Improved glycemia and ↓insulin concentrations, ↓liver TGs in OFSs and BB-12 | ||||
↑GLP-1 in OFSs | ||||
↑GLP-2 in probiotic BB-12 | ||||
No differences in plasma LPS, TNF-α, IL-6, IL-1β | ||||
Ritze et al., 2014 (55) | C57BL/6 mice | High-fructose diet with LGG vs. high-fructose diet over 8 wk | qPCR | High-fructose diet with LGG vs. high-fructose diet: |
↓ALT, ↓fat, ↓accumulation in liver, ↓ChREBP, ↓ACC1, ↓FAS, ↓TNF-α, ↓IL-1β, ↑occludin, ↓LPS, ↑total bacterial numbers | ||||
Zeng et al., 2013 (56) | C57BL/6 mice | HFD vs. LFD for 10 wk | Sequencing 16S rRNA | HFD vs. LFD: |
↑Hepatic lipid accumulation, ↑inflammatory cell infiltration, ↑leptin, ↑TNF-α | ||||
↑Lactobacillus gasseri and/or ↑Lactobacillus taiwanensis | ||||
Positive correlation of L. gasseri and/or L. taiwanensis DNA and lipid droplets in liver | ||||
Park et al., 2013 (57) | C57BL/6J mice | HFD + probiotic (Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032) vs. HFD + placebo for 10 wk. | Sequencing 16S rRNA | HFD + probiotic vs. HFD + placebo: |
↓ALT, ↑FA oxidation–related genes, ↓proinflammatory genes (TNFa, IL6, IL1b), ↓gut microbiota diversity, ↑Bifidobacterium Pseudolongum | ||||
Pachikian et al., 2013 (58) | C57BL/6J mice | n–3 PUFA–depleted diet for 3 mo supplemented with FOSs during the last 10 d of treatment | DGGE, qPCR | n-3 PUFA-depleted diet supplemented with FOSs vs. n-3 PUFA-depleted diet: |
↑Bifidobacterium spp. ↓Roseburia spp., ↓SREBP2, ↑PPAR-α, ↓LDL, ↓HDL, ↑GLP-1 | ||||
Cano et al., 2013 (59) | C57BL/6 mice | HFD supplemented with Bifidobacterium pseudocatenulatum CECT 7765 vs. HFD for 7 wk | qPCR | HFD supplemented with B. pseudocatenulatum CECT 7765 vs. HFD: |
↓Serum cholesterol, ↓serum TGs, ↓serum glucose, ↓insulin resistance, ↓hepatocytes with grade 3 steatosis, ↓fat absorption, ↓leptin, ↓IL-6, ↓MCP-1, ↑IL-4, ↑IL-10, ↑Bifidobacteria, ↓Enterobacteriaceae, ↓body weight gain, ↓food intake | ||||
Axling et al., 2012 (60) | C57BL/6J mice | HFD with or without a supplement GT and supplemented with Lp or the combination of both (Lp + GT) for 22 wk | Sequencing 16S rRNA, qPCR | Lp + GT vs. control: |
↑Lactobacillus, ↑diversity of bacteria, ↓liver weights, ↓liver size, ↓TGs, ↓ALT | ||||
Akkermansia correlated negatively with body fat content (r = −0.43, P = 0.04), plasma insulin (r = −0.47, P = 0.03), and liver TGs (r = −0.44, P = 0.03). | ||||
Neyrinck et al., 2011 (61) | C57BL/6J mice | HFD supplemented with arabinoxylans from wheat vs. HFD for 4 wk | DGGE, qPCR | HFD supplemented with arabinoxylans from wheat vs. HFD: |
↑Bifidobacteria, ↓adipocyte size, ↓serum and hepatic cholesterol accumulation and insulin resistance, ↑tight junction proteins (ZO-1 and occludin), ↓inflammatory markers (IL-6 and MCP-1) | ||||
Cani et al., 2009 (62) | C57BL/6mice | Prebiotic (oligofructose) vs. nonprebiotic carbohydrates (microcrystalline cellulose) for 4 wk | DGGE, qPCR analysis | Prebiotic vs. nonprebiotic: |
↑Total bacteria count, ↑Lactobacillus spp., ↑Bifidobacterium spp., ↑Clostridium coccoides–Eubacterium rectale cluster, ↓LPS, ↑ZO-1 and occludin, ↓PAI-1, ↓NADPHox, ↓iNOS, ↓TLR4, ↓TNF-α, ↑GLP-2, ↓hepatic expression of inflammatory and oxidative stress markers | ||||
Cani et al., 2007 (33) | C57BL/6J mice | High-fat and carbohydrate-free diet vs. normal-diet group for 4 wk | FISH | High-fat and carbohydrate-free diet vs. normal-diet group: |
↑Liver weight, ↑Liver TGs, ↑TNF-α, ↑IL-1, ↑IL-6, ↑LPS, ↓Bifidobacterium spp., E. rectale/C. coccoides |
ACC1, acetyl-CoA carboxylase 1; ALT, alanin transferase; BB-12, Bifidobacterium animalis subsp. lactis BB-12; ChREBP, transcription factor carbohydrate-responsive element-binding protein; DGGE, denaturing gradient gel electrophoresis; FAS, fatty acid synthase; FISH, Fuorescent in situ hybridization; FOS, fructo-oligosaccharide; GLP, glucagon-like peptide; GT, green tea powder; HF, high prebiotic fiber; HFD, high-fat diet; HP, high protein; iNOS, inducible nitric oxide synthase; LFD, low-fat diet; LGG, Lactobacillus rhamnosus GG; Lp, Lactobacillus plantarum DSM 15313; MCP-1, monocyte chemoattractant protein-1; NADPHox, NAD(P)H oxidase; NAFLD, nonalcoholic fatty liver disease; OFS, oligofructosaccharide; PAI-1, plasminogen activator inhibitor 1; PYY, peptide YY; ref, reference; rRNA, ribosomal RNA; SREBP2, sterol regulatory element binding protein 2; TLR4, Toll-like receptor 4; TNFa, tumor necrosis factor α (gene); ZO-1, zonula occludens-1; ↑, increased; ↓, decreased.
Energy homeostasis and the gut microbiota.
An imbalance between energy intake and expenditure is regarded as the main cause of increased hepatic lipogenesis. Increased lipogenesis and reduced FA oxidation result in increased susceptibility to hepatic steatosis and the subsequent consequences (63). Several studies indicated that an increase in the relative abundance of the members of Firmicutes, and especially of some Clostridium clusters, is involved in harvesting energy from the diet (17, 18, 64), and this contributes to the development of excessive weight gain and hepatic lipogenesis.
With the use of germ-free animal models, it has been shown that the gut microbiota increases energy harvested from dietary polysaccharides and augments key enzymes in hepatic de novo FA biosynthesis, including acetyl-CoA carboxylase (ACC) and FA synthase (FAS), and consequently, hepatic TG accumulation. On the other hand, the gut microbiota drives the host to increase LPL activity and hepatic production of TGs by the inhibition of FIAF (an LPL inhibitor) in intestinal epithelium cells (35). In addition, germ-free mice showed increased AMP-activated protein kinase (AMPK) activity, which is an intracellular energy sensor in the liver and skeletal muscle, leading to increased FA oxidation and insulin sensitivity and reduced glycogen concentrations. This would protect germ-free mice against diet-induced obesity. In normal mice with normal gut microbiota, inactive AMPK is associated with decreased FA oxidation in skeletal muscle (36). In addition, gut satiety hormones are affected by the gut microbiome through the gut-brain axis (65). Therefore, the gut microbiome influences energy intake in addition to energy expenditure. Any changes in the gut microbial taxa can increase gastrointestinal satiety hormones, hence influencing energy hemostasis (66–68).
Another key function of the intestinal bacteria is to hydrolyze polysaccharides and oligosaccharides that are not completely catabolized by the host enzymes. Although much is still to be learned about which microbes synthesize and/or metabolize the SCFAs and the roles of these potent metabolites, saccharolytic fermentations are major players in metabolic disorders (69). A wide range of bacterial groups are able to produce acetate, whereas the production of propionate and butyrate seems to be more specific. Akkermansia municiphila has been known as a major propionate-producing bacterium (70) and Eubacterium hallii, Eubacterium rectale, Roseburia inulinivorans, Faecalibacterium prausnitzii, Clostridium lavalense, Bacteroides uniformis, and Ruminococcus bromii appear to be responsible for most of butyrate production (70–72). Butyrate and propionate at low amounts exert multiple advantageous effects on the host, including the prevention of colonic carcinogenesis, inflammation, and oxidative stress; improvement in intestinal barrier function; and stimulation of satiety and lipid oxidation in hepatocytes (73, 74). Acetate may be more important for modulating insulin sensitivity and metabolic disease (75) and the development of diabetes (76). However, more studies are required to understand which microbes produce each SCFA and the relation this has on liver health and disease.
SCFAs are ligands for G-protein–coupled receptor (GPR) 43 expressed on intestinal epithelial cells. The stimulation of GPR43 by SCFAs is necessary during immune responses (77). GPR41 is another SCFA receptor. Binding to GPR41 is associated with upregulation of peptide YY (PYY) and glucagon-like peptide 1 [(GLP1), anorectic hormones] that have effects on appetite control; therefore, SCFAs are linked to food intake by activating these receptors (78, 79). Current evidence indicates that SCFAs have paradoxical effects on hepatic health. SCFAs have been identified as rich sources of energy for the host and act as intestinotrophic agents to promote intestinal absorption of nutrients. On the other hand, through their receptor GPR43 and other pathways, they reduce gut permeability and invasion of bacterial products through the portal vein and liver, and thus protect the liver from the ensuing damaging inflammation, which then results in downregulation of insulin signaling in adipose tissue, thereby decreasing fat accumulation (80). Thus, it seems that SCFAs are beneficial for hepatic health if the body is receiving a balanced diet, whereas they exaggerate steatosis when extra calories are consumed. However, more mechanistic studies are required to understand the role of each of the SCFAs on NAFLD.
Macronutrient intakes and the gut microbiome in NAFLD.
Several studies have shown that high-fat diets (HFDs) participate in the development of dysbiosis through diet-induced reductions in Bacteroidetes, Lactobacilli, and Bifidobacteria and increases in Firmicutes (81, 82). Thus, an HFD may contribute to the development of NAFLD at least partially through dysbiosis, which may increase intestinal permeability. Furthermore, HFD exposure impairs choline metabolism via dysbiosis. Choline is converted to methylamines (dimethylamine, trimethylamine, and trimethylamine-N-oxide) by intestinal microbes with consumption of an HFD. It is associated with a reduction in plasma phosphatidylcholine and increases in urinary methylamines. Indeed, changes in the gut microbiota mimic the effect of a choline-deficient diet, which leads to accumulation of TGs in hepatocytes and reduced VLDL secretion (83). Maintenance of mice fed an HFD for 4 wk resulted in a significant increase in Bifidobacterium spp. and the E. rectale/Clostridium coccoides group, while decreasing the C. coccoides group and Bifidobacteria E. rectale. Moreover, fat consumption can produce a wide amount of chylomicrons, which translocate LPS toward other organs, especially the liver (84), which can bind to TLR4 on hepatic immune cells and initiate the inflammatory process. It has been shown that plasma inflammatory cytokines are positively correlated with plasma LPS concentrations and negatively correlated with intestinal Bifidobacteria count (85). An HFD not only changes the gut microbiota but also plays a role in the development of steatohepatitis in mice (56). It has been shown that Lactobacillus gasseri and Lactobacillus taiwanensis increase after consumption of an HFD. These bacteria are bile acid–resistant and probably contribute to bile acid deconjugation and reduction in fat absorption. There was a positive significant association between L. gasseri and/or L. taiwanensis DNA and lipid droplets in the liver (R2 = 0.3, P = 0.02). Increased fat intake simultaneously increases bile acid secretion and bile acid–resistant bacteria. Increased Lactobacillus in HFD may affect lipid metabolism through bile acids (86, 87).
A high-protein diet also affects the gut microbiome, favoring a potentially pathogenic and proinflammatory microbiota profile with increased ammonia, phenols, and hydrogen sulfide concentrations, which induce mucosal inflammation and alter the enteric nervous system and intestinal motility (88). In addition, the dietary source of proteins plays a critical role in shaping the gut microbiota and their ability to produce secondary metabolites, so that meat protein–fed rats showed a higher relative abundance of the beneficial genus Lactobacillus but lower amounts of SCFAs and SCFA-producing bacteria, including Fusobacterium, Bacteroides, and Prevotella spp. compared with the soy protein–fed group (89). Overall, studies evaluating the effects of different dietary patterns on the human gut microbiome are limited; further studies are needed to elucidate more details in this area.
Probiotics and the gut microbiome in NAFLD.
Current studies hypothesize the use of probiotics for modulating dysbiosis in many diseases, including NAFLD. Probiotics are beneficial microorganisms when ingested in specified dosages. Several studies showed that gut microbiota manipulation by probiotic supplements is associated with reduced liver damage, decreased concentrations of LPS, as well as improved aminotransferase concentrations (90–93). A recent review summarized the studies that show that probiotic supplementation in animal models and human studies improve inflammatory status and clinical manifestations in NAFLD (94). Manipulating the gut microbiota with probiotics may improve dysbiosis associated with an HFD. In support of this, consumption of an HFD upregulates the expression of genes involved in hepatic lipid biosynthesis, and supplementation with Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 upregulates the expression of genes involved in hepatic FA oxidation (57). Another study showed that increasing tissue n–3 PUFAs in the gut alters the gut bacterial composition, which is associated with a decrease in LPS and gut permeability and eventually results in metabolic endotoxemia and inflammation (95). Overall, although there are some exciting possibilities with regard to the use of probiotics as a treatment for NAFLD, experimental studies are still required to test the mechanisms of action and clinical trials are still required to test their efficacy.
Prebiotics and intestinal microbiota in NAFLD.
In addition to probiotics being potentially protective in NAFLD, accumulating data also suggest that there are beneficial effects of prebiotics in NAFLD. A prebiotic is defined as “a selectively fermented ingredient that results in specific changes, in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefit(s) upon host health” (96). Prebiotics are naturally found in foods such as onions, asparagus, garlic, and leeks and resistant starch (RS) sources such as legumes, potatoes, and cereals. Prebiotics could modulate the intestinal microbiota, because they would increase beneficial bacteria for the host, in particular the indigenous Bifidobacteria and Lactobacilli, and inhibit bacterial activities harmful to host health (97). Furthermore, in the large intestine, inulin-type fructans and RS are completely catabolized by the microbiota into SCFAs, bacterial biomass, and organic acids such as lactic acid and gases (carbon dioxide, hydrogen, methane).
Prebiotics have beneficial effects on the gut microbiota, serum lipids, and the prevention of hepatic steatosis (61, 62, 98, 99). In fact, the addition of RS to rodent feed pellets increases Bacteriodetes, Actinobacteria (Bifidobacteria spp.), and Verrucomicrobia (A. muciniphilia) while decreasing Firmicutes (100–102). Also, in humans, the administration of RS has been shown to induce phylum-level changes, selectively increasing numbers of Actinobacteria and Bacteroidetes and reducing Firmicutes; at the species level, an increase in Bifidobacterium adolescentis, E. rectale, Roseburia spp., and R. bromii have been reported (103–107). These various bacteria differ significantly in the production of metabolic byproducts and potential interactions with the host. It has been well established that Bifidobacteria spp., a source of lactate and acetate and low amounts of ethanol and formate, are broadly used as probiotics and have health-promoting properties because of their potential modulating effects on the immune system, metabolism, and elimination of pathogenic bacteria of the host (108). Members of E. rectale, Roseburia, and R. bromii are main producers of butyrate in the colon (71, 107, 109). It has been shown that increasing butyrate-producing bacteria and butyrate production is closely linked with prevention of the progression from steatosis to hepatocarcinogenesis and improving hepatic inflammatory and oxidative stress indexes and gut integrity (74, 110, 111). This would appear to indicate that there is a mutualistic relation between the gut microbiota and the host; dietary ingredients such as RS could induce special species of bacteria that produce metabolic products such as butyrate. Butyrate could act as a signaling molecule in the host that participates in various signaling pathways (110); thus, products of the gut microbiota could explain part of the role of nutritional factors in the gut liver axis and their relation with NAFLD.
It has been reported that prebiotic supplementation can modulate the effects of HFDs. Prebiotic consumption leads to increased Bifidobacteria and total bacteria Bacteroides:Prevotella spp. and decreased Firmicutes (53). Cano et al. (59) reported that Bifidobacterium pseudocatenulatum consumption improved glucose tolerance and inflammatory status while decreasing serum LPS and hepatic steatosis in HFD-fed mice. Another study showed that prebiotic oligofructose in combination with probiotic Bifidobacterium animalis subsp. lactis BB-12 decreased the ratio of Firmicutes to Bacteroidetes and improved glycemia. This decrease in the Firmicutes-to-Bacteroidetes ratio is hypothesized to decrease obesity-inducing effects of the microbiome (54). In addition, oligofructose decreases energy intake, weight gain, and fat mass (54). In this study, prebiotics and probiotics had no effect on increased LPS and inflammatory factors induced by high-fat and high-sucrose diets (54). A significant increase in the abundance of Bifidobacterium spp. and Lactobacillus spp. by the administration of inulin-type fructo-oligosaccharides (FOSs) has been documented (112). Increasing total bacteria counts of Lactobacillus spp. and Bifidobacterium spp. by prebiotics improved intestinal barrier function and hepatic inflammation and oxidation. Furthermore, stimulating the proliferation of these bacteria was associated with a reduction in intestinal endotoxin and modulated gut mucosal function (113, 114). Lactobacillus spp. inhibit proinflammatory responses that result from endotoxins by suppressing NF-κB (115).
Inulin-type fructans or FOSs are also able to modify groups of bacteria other than Bifidobacteria or Lactobacilli. Inulin-containing diets result in an increase in bacteria belonging to C. coccoides and E. rectale cluster in animal models (116). In addition, inulin can stimulate the metabolic activity of Roseburia spp. and E. rectale genera of the Firmicutes phylum (117, 118). Roseburia spp. and E. rectale belong to the butyrate-producing bacteria; therefore, they may mediate some of the advantageous properties of prebiotics. Furthermore, the consumption of inulin, galacto-oligosaccharides, and raffinose stimulates F. prausnitzii (119–121). F. prausnitzii, which belong to the butyrate-producing bacteria, could be crucial to gut barrier function and could modulate systemic inflammation (common to NAFLD) (79, 122). All of these proposed actions may play a role in the beneficial effects of prebiotics in NAFLD prevention and/or treatment. However, well-designed clinical trials are required to support the efficacy of prebiotics during NAFLD.
Choline and other nutrients and the gut microbiome in NAFLD.
Choline deficiency is related to NAFLD pathogenesis. Although choline is synthesized in the body, it is considered to be an essential nutrient and dietary intake is required to fulfill its roles in the body (123). Choline plays a role in brain function, acetylcholine and neurotransmitter synthesis, cell signaling, lipid transport, and lipid metabolism (124, 125). Several studies have shown that choline is critical for the development of hepatic steatosis due to its contribution to lipid metabolism (125–127). When hepatic de novo lipogenesis or TG synthesis from diacylglycerol is greater than the lipid secretion from the liver and/or fat oxidation, hepatic steatosis occurs. Choline is one of the methyl group donors in the body that contributes to hepatic synthesis of phosphatidylcholine (125). Phosphatidylcholine is required for VLDL synthesis and secretion. Reduced phosphatidylcholine contributes to TG accumulation in the liver and NAFLD (128). Choline-deficient diets disturb the mitochondrial function, resulting in increased hepatic lipid accumulation that contributes to NAFLD pathogenesis (129). Gut bacteria can convert choline to trimethylamine and reduce choline bioavailability, mimicking choline deficiency side effects (130). A reduction in choline bioavailability is associated with increased reactive oxygen species resulting from FFA oxidation, more hepatic lipid accumulation, and reduced hepatic VLDL excretion, all of which contribute to the development of hepatic steatosis (125, 126). Choline metabolism is mainly disrupted by 3 main bacterial phyla in the gut microbiota: Proteobacteria, Firmicutes, and Actinobacteria (130). Spencer et al. (131), in a longitudinal experimental study in healthy women who consumed a rigorously controlled diet, showed that choline deficiency led to hepatic lipid accumulation. In addition, decreased amounts of Gammaproteobacteria and increased Erysipelotrichi were directly associated with elevated accumulation of hepatic fat. This study was the only study, to our knowledge, that examined gut microbiota composition in relation to diet and development of NAFLD in humans.
In addition to the evidence of gut dysbiosis in NAFLD, patients with NASH also have dysbiosis, including small intestinal bacterial overgrowth (23). Bacterial overgrowth enhances the number of bacteria metabolizing choline and influences hepatic lipid accumulation (125). Pyruvate produced by the gut microbiota from carbohydrate increases metabolites such as acetaldehyde and ethanol. These metabolites are toxic and associated with gut permeability (132, 133). Overall, although better designed studies are needed, there are sufficient data to support that choline and the gut microbiome together play a role in liver disease.
High-fructose dietary intake is also considered to be a significant risk factor in the development of NAFLD (134, 135). Some studies have indicated that antibiotics, prebiotics, and probiotics are protective factors in fructose-induced liver damage (136–139). These studies suggested that gut microbe manipulation may protect from the side effects of high-fructose diets on the liver. The chronic intake of fructose is associated with bacterial overgrowth, which causes increases in endotoxins in the portal vein, development of inflammatory responses in the liver, and insulin resistance in hepatocytes, resulting in increased hepatic lipid accumulation (140, 141). Ritze et al. (55) reported that the use of Lactobacillus rhamnosus GG protects against NAFLD in animals fed a high-fructose diet through increasing the proteins involved in gut integrity and decreasing inflammatory factors and endotoxins. The authors suggested that L. rhamnosus GG may increase occludin and claudin-1 (effective proteins in gut integrity), which lead to reductions in LPS and other toxins translocated to the liver.
Several recent studies reported that polyphenol compounds attenuate inflammatory markers and hepatic steatosis grade in patients with NAFLD (142–145). Some of these act through the amelioration of insulin resistance, whereas some act through the modulation of the gut microbiome (146, 147). These compounds can reduce pathogen bacteria and increase lactic acid bacteria, such as some strains of Bifidobacteria (148). The manipulation of the gut microbiota may be 1 of the mechanisms of action involved in the beneficial effects of polyphenols on NAFLD. Axling et al. (60) reported that green tea in combination with Lactobacillus plantarum (which metabolizes phenolic acids) in HFD-fed mice was able to promote growth of Lactobacillus in the intestine and to attenuate HFD-induced inflammation and hepatic TG and cholesterol accumulation. Furthermore, there was a significant inverse association between the amount of Akkermansia and hepatic TG accumulation. A. muciniphila is a mucus-degrading bacterium that contributes to the regulation of immune responses through upregulation of the expression of some genes involved in intestinal mucus integrity (149).
n–3 FAs and the intestinal microbiota in NAFLD.
A low intake of n–3 PUFAs is associated with hepatic steatosis (150). n–3 PUFAs play an important role in lipid metabolism because they can induce FA oxidation by activating PPAR-α and inhibition of lipogenesis by suppressing sterol regulatory element binding protein 1 (SREBP-1) (151). It has been shown that n–3 PUFA supplementation, and not total calories from fat, alters the gut microbial composition by enhancing numbers of Lactobacillus and Bifidobacteria spp. in adult mice (152) as well as in aged mice, which modulated both gut immunity and oxidative stress (153). In support of these data, other studies have shown that changes in gut microbiota composition by n–3 PUFA deficiency can be modulated by prebiotics such as FOS. FOS consumption was associated with increased cecal Bifidobacterium spp. and reduced Roseburia spp. in n–3 PUFA–depleted mice (58). Moreover, it has been shown that an increase in lactic acid bacteria, such as Bifidobacterium spp., is related to a reduction in cholesterol absorption (154) and an increase in bile acid deconjugation, which leads to more excretion of bile acids and elimination of hepatic cholesterol (155).
Conclusions
In summary, intestinal pathogenic bacteria contribute to NAFLD pathogenesis through the following ways: 1) by increasing LPS, which binds to TLR4 in hepatic immune cells, resulting in the activation of the NF-κB pathway and its related inflammatory pathways, leading to hepatic injury, necrosis, and finally fibrosis; 2) inhibition of FIAF, which increases the activity of LPL and lipogenesis; 3) enhancement in polysaccharide digestion and absorption, which increases the production of SCFAs and hepatic lipogenesis; and 4) conversion of choline into methylamines, which reduces choline availability and induces fat accumulation and reactive oxygen species production in the liver.
Nutritional factors and the gut microbiota have bilateral interactions that contribute to the development and/or protection from NAFLD. High-fat, high-protein, and high-fructose diets and diets that are low in n–3 FAs (Western dietary pattern) may contribute to the development of NAFLD by dysbiosis in the intestinal microbiota. On the other hand, gut microbiota manipulation by nutritional factors such as prebiotics, probiotics, and polyphenols can improve characteristics of NAFLD. Few studies have investigated the effects of nutritional factors on the gut microbiota in patients with NAFLD, and in particular, human studies are scarce. Further studies are needed to elucidate the exact mechanism(s) of action and probable therapeutic options.
Acknowledgments
ZM and AH contributed to the conception of the article, the literature search and interpretation, writing the article, and the critical revisions related to important intellectual content of the manuscript and approved the final manuscript; and DLG contributed to the final revisions of the manuscript. All authors read and approved the final manuscript.
Footnotes
Abbreviations used: ACC, acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; ASC, apoptosis-associated speck-like protein; FAS, fatty acid synthase; FIAF, fasting-induced adipocyte factor; FOS, fructo-oligosaccharide; GLP1, glucagon-like peptide 1; GPR, G-protein–coupled receptor; HFD, high-fat diet; LPL, lipoprotein lipase; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; PYY, peptide YY; RS, resistant starch; SREBP-1, sterol regulatory element binding protein 1; TLR, toll-like receptor.
References
- 1.Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011;34:274–85. [DOI] [PubMed] [Google Scholar]
- 2.Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease: pathology and pathogenesis. Annu Rev Pathol 2010;5:145–71. [DOI] [PubMed] [Google Scholar]
- 3.Angulo P. Nonalcoholic fatty liver disease. N Engl J Med 2002;346:1221–31. [DOI] [PubMed] [Google Scholar]
- 4.Bedogni G, Miglioli L, Masutti F, Tiribelli C, Marchesini G, Bellentani S. Prevalence of and risk factors for nonalcoholic fatty liver disease: the Dionysos Nutrition and Liver Study. Hepatology 2005;42:44–52. [DOI] [PubMed] [Google Scholar]
- 5.Fan JG, Saibara T, Chitturi S, Kim BI, Sung JJ, Chutaputti A. What are the risk factors and settings for non-alcoholic fatty liver disease in Asia-Pacific? J Gastroenterol Hepatol 2007;22:794–800. [DOI] [PubMed] [Google Scholar]
- 6.Wilfred de Alwis NM, Day CP. Genetics of alcoholic liver disease and nonalcoholic fatty liver disease. Semin Liver Dis 2007;27:44–54. [DOI] [PubMed] [Google Scholar]
- 7.Ghaemi A, Taleban FA, Hekmatdoost A, Rafiei A, Hosseini V, Amiri Z, Homayounfar R, Fakheri H. How much weight loss is effective on nonalcoholic fatty liver disease? Hepat Mon 2013;13:e15227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014;146:1513–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Balmer ML, Slack E, de Gottardi A, Lawson MA, Hapfelmeier S, Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, et al. The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci Transl Med 2014;6:237ra66. [DOI] [PubMed] [Google Scholar]
- 10.Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009;9:313–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology 2006;43(2, Suppl 1):S54–62. [DOI] [PubMed] [Google Scholar]
- 12.Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sima H, Hekmatdoost A, Ghaziani T, Alavian SM, Mashayekh A, Zali MR. The prevalence of celiac autoantibodies in hepatitis patients. Iran J Allergy Asthma Immunol 2010;9:157–62. [PubMed] [Google Scholar]
- 14.Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science 2005;308:1635–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 2013;57:601–9. [DOI] [PubMed] [Google Scholar]
- 16.Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, et al. Enterotypes of the human gut microbiome. Nature 2011;473(7346):174–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006;444:1027–31. [DOI] [PubMed] [Google Scholar]
- 18.Santacruz A, Collado MC, Garcia-Valdes L, Segura MT, Martin-Lagos JA, Anjos T, Martí-Romero M, Lopez RM, Florido J, Campoy C, et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br J Nutr 2010;104:83–92. [DOI] [PubMed] [Google Scholar]
- 19.Zoetendal EG, Rajilić-Stojanović M, De Vos WM. High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut 2008;57:1605–15. [DOI] [PubMed] [Google Scholar]
- 20.Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Reddy DN. Role of the normal gut microbiota. World J Gastroenterol 2015;21:8787–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012;490:55–60. [DOI] [PubMed] [Google Scholar]
- 22.Shen J, Obin MS, Zhao L. The gut microbiota, obesity and insulin resistance. Mol Aspects Med 2013;34:39–58. [DOI] [PubMed] [Google Scholar]
- 23.Wigg AJ, Roberts-Thomson IC, Dymock RB, McCarthy PJ, Grose RH, Cummins AG. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut 2001;48:206–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK, Fischer SE, McGilvray ID, Allard JP. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013;58:120–7. [DOI] [PubMed] [Google Scholar]
- 25.Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, Martin P, Philippe C, Walker F, Bado A, et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 2013;62:1787–94. [DOI] [PubMed] [Google Scholar]
- 26.Eslamparast T, Poustchi H, Zamani F, Sharafkhah M, Malekzadeh R, Hekmatdoost A. Synbiotic supplementation in nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled pilot study. Am J Clin Nutr 2014;99:535–42. [DOI] [PubMed] [Google Scholar]
- 27.Yari Z, Rahimlou M, Eslamparast T, Ebrahimi-Daryani N, Poustchi H, Hekmatdoost A. Flaxseed supplementation in non-alcoholic fatty liver disease: a pilot randomized, open labeled, controlled study. Int J Food Sci Nutr 2016;67:461–9. [DOI] [PubMed] [Google Scholar]
- 28.Eslamparast T, Eghtesad S, Poustchi H, Hekmatdoost A. Recent advances in dietary supplementation, in treating non-alcoholic fatty liver disease. World J Hepatol 2015;7:204–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shavakhi A, Minakari M, Firouzian H, Assali R, Hekmatdoost A, Ferns G. Effect of a probiotic and metformin on liver aminotransferases in non-alcoholic steatohepatitis: a double blind randomized clinical trial. Int J Prev Med 2013;4:531–7. [PMC free article] [PubMed] [Google Scholar]
- 30.Rahimlou M, Ahmadnia H, Hekmatdoost A. Dietary supplements and pediatric non-alcoholic fatty liver disease: present and the future. World J Hepatol 2015;7:2597–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palu G, Martines D. Increased intestinal permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2007;292:G518–25. [DOI] [PubMed] [Google Scholar]
- 32.Soares JB, Pimentel-Nunes P, Roncon-Albuquerque R, Leite-Moreira A. The role of lipopolysaccharide/Toll-like receptor 4 signaling in chronic liver diseases. Hepatol Int 2010;4:659–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56:1761–72. [DOI] [PubMed] [Google Scholar]
- 34.Su GL. Lipopolysaccharides in liver injury: molecular mechanisms of Kupffer cell activation. Am J Physiol Gastrointest Liver Physiol 2002;283:G256–65. [DOI] [PubMed] [Google Scholar]
- 35.Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 2004;101:15718–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 2007;104:979–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012;482:179–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011;145:745–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lee HJ, Yeon JE, Ko EJ, Yoon EL, Suh SJ, Kang K, Kim HR, Kang SH, Yoo YJ, Je J, et al. Peroxisome proliferator-activated receptor-delta agonist ameliorated inflammasome activation in nonalcoholic fatty liver disease. World J Gastroenterol 2015;21:12787–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sui YH, Luo WJ, Xu QY, Hua J. Dietary saturated fatty acid and polyunsaturated fatty acid oppositely affect hepatic NOD-like receptor protein 3 inflammasome through regulating nuclear factor-kappa B activation. World J Gastroenterol 2016;22:2533–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Volynets V, Kuper MA, Strahl S, Maier IB, Spruss A, Wagnerberger S, Königsrainer A, Bischoff SC, Bergheim I. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci 2012;57:1932–41. [DOI] [PubMed] [Google Scholar]
- 42.Kirpich IA, Feng W, Wang Y, Liu Y, Barker DF, Barve SS, McClain CJ. The type of dietary fat modulates intestinal tight junction integrity, gut permeability, and hepatic Toll-like receptor expression in a mouse model of alcoholic liver disease. Alcohol Clin Exp Res 2012;36:835–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Harris K, Kassis A, Major G, Chou CJ. Is the gut microbiota a new factor contributing to obesity and its metabolic disorders? J Obes 2012;2012:879151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Del Chierico F, Nobili V, Vernocchi P, Russo A, De Stefanis C, Gnani D, Furlanello C, Zandonà A, Paci P, Capuani G, et al. Gut microbiota profiling of pediatric NAFLD and obese patients unveiled by an integrated meta-omics based approach. Hepatology 2016 Mar 29 (Epub ahead of print; DOI: 10.1002/hep.28572). [DOI] [PubMed] [Google Scholar]
- 45.Wong VW, Tse CH, Lam TT, Wong GL, Chim AM, Chu WC, Yeung DK, Law PT, Kwan HS, Yu J, et al. Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis—a longitudinal study. PLoS One 2013;8:e62885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, Greenwood R, Sikaroodi M, Lam V, Crotty P, et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2013;11(7):868–75, e1–3. [DOI] [PubMed] [Google Scholar]
- 47.Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013;341:1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gorzelak MA, Gill SK, Tasnim N, Ahmadi-Vand Z, Jay M, Gibson DL. Methods for improving human gut microbiome data by reducing variability through sample processing and storage of stool. PLoS One 2015;10:e0134802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hekmatdoost A, Feizabadi MM, Djazayery A, Mirshafiey A, Eshraghian MR, Yeganeh SM, Sedaghat R, Jacobson K. The effect of dietary oils on cecal microflora in experimental colitis in mice. Indian J Gastroenterol 2008;27:186–9. [PubMed] [Google Scholar]
- 50.Hekmatdoost A, Mirshafiey A, Feizabadi MM, Djazayeri A. Polyunsaturated fatty acids, microflora and colitis. Ann Nutr Metab 2009;55:325. [DOI] [PubMed] [Google Scholar]
- 51.Hekmatdoost A, Wu X, Morampudi V, Innis SM, Jacobson K. Dietary oils modify the host immune response and colonic tissue damage following Citrobacter rodentium infection in mice. Am J Physiol Gastrointest Liver Physiol 2013;304:G917–28. [DOI] [PubMed] [Google Scholar]
- 52.Carmody RN, Gerber GK, Luevano JM, Gatti DM, Somes L, Svenson KL, Turnbaugh PJ. Diet dominates host genotype in shaping the murine gut microbiota. Cell Host Microbe 2015;17:72–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Saha DC, Reimer RA. Long-term intake of a high prebiotic fiber diet but not high protein reduces metabolic risk after a high fat challenge and uniquely alters gut microbiota and hepatic gene expression. Nutr Res 2014;34:789–96. [DOI] [PubMed] [Google Scholar]
- 54.Bomhof MR, Saha DC, Reid DT, Paul HA, Reimer RA. Combined effects of oligofructose and Bifidobacterium animalis on gut microbiota and glycemia in obese rats. Obesity (Silver Spring) 2014;22:763–71. [DOI] [PubMed] [Google Scholar]
- 55.Ritze Y, Bárdos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A, Bischoff SC. Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice. PLoS One 2014;9:e80169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zeng H, Liu J, Jackson MI, Zhao FQ, Yan L, Combs GF Jr. Fatty liver accompanies an increase in Lactobacillus species in the hind gut of C57BL/6 mice fed a high-fat diet. J Nutr 2013;143:627–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Park DY, Ahn YT, Park SH, Huh CS, Yoo SR, Yu R, Sung MK, McGregor RA, Choi MS. Supplementation of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 in diet-induced obese mice is associated with gut microbial changes and reduction in obesity. PLoS One 2013;8:e59470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Pachikian BD, Essaghir A, Demoulin JB, Catry E, Neyrinck AM, Dewulf EM, Sohet FM, Portois L, Clerbaux LA, Carpentier YA, et al. Prebiotic approach alleviates hepatic steatosis: implication of fatty acid oxidative and cholesterol synthesis pathways. Mol Nutr Food Res 2013;57:347–59. [DOI] [PubMed] [Google Scholar]
- 59.Cano PG, Santacruz A, Trejo FM, Sanz Y. Bifidobacterium CECT 7765 improves metabolic and immunological alterations associated with obesity in high-fat diet-fed mice. Obesity (Silver Spring) 2013;21:2310–21. [DOI] [PubMed] [Google Scholar]
- 60.Axling U, Olsson C, Xu J, Fernandez C, Larsson S, Strom K, Ahrné S, Holm C, Molin G, Berger K. Green tea powder and Lactobacillus plantarum affect gut microbiota, lipid metabolism and inflammation in high-fat fed C57BL/6J mice. Nutr Metab (Lond) 2012;9:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Neyrinck AM, Possemiers S, Druart C, Van de Wiele T, De Backer F, Cani PD, Larondelle Y, Delzenne NM. Prebiotic effects of wheat arabinoxylan related to the increase in Bifidobacteria, Roseburia and Bacteroides/Prevotella in diet-induced obese mice. PLoS One 2011;6:e20944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009;58:1091–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 2010;51:679–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, Krakoff J. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr 2011;94:58–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, Petersen KF, Kibbey RG, Goodman AL, Shulman GI. Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome. Nature 2016;534:213–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Musso G, Gambino R, Cassader M. Gut microbiota as a regulator of energy homeostasis and ectopic fat deposition: mechanisms and implications for metabolic disorders. Curr Opin Lipidol 2010;21:76–83. [DOI] [PubMed] [Google Scholar]
- 67.Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, De Backer F, Neyrinck AM, Delzenne NM. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr 2009;90:1236–43. [DOI] [PubMed] [Google Scholar]
- 68.Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am J Clin Nutr 2009;89:1751–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Puertollano E, Kolida S, Yaqoob P. Biological significance of short-chain fatty acid metabolism by the intestinal microbiome. Curr Opin Clin Nutr Metab Care 2014;17:139–44. [DOI] [PubMed] [Google Scholar]
- 70.Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016;7:189–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol 2010;12:304–14. [DOI] [PubMed] [Google Scholar]
- 72.Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, Inatomi O, Bamba S, Sugimoto M, Andoh A. Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in Crohn’s disease. Digestion 2016;93:59–65. [DOI] [PubMed] [Google Scholar]
- 73.Hosseini E, Grootaert C, Verstraete W, Van de Wiele T. Propionate as a health-promoting microbial metabolite in the human gut. Nutr Rev 2011;69:245–58. [DOI] [PubMed] [Google Scholar]
- 74.Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost F, Brummer RJ. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther 2008;27:104–19. [DOI] [PubMed] [Google Scholar]
- 75.van der Beek CM, Canfora EE, Lenaerts K, Troost FJ, Olde Damink SW, Holst JJ, Masclee AA, Dejong CH, Blaak EE. Distal, not proximal, colonic acetate infusions promote fat oxidation and improve metabolic markers in overweight/obese men. Clin Sci (Lond) 2016;130:2073–82. [DOI] [PubMed] [Google Scholar]
- 76.Brown K, Godovannyi A, Ma C, Zhang Y, Ahmadi-Vand Z, Dai C, Gorzelak MA, Chan Y, Chan JM, Lochner A, et al. Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice. ISME J 2016;10:321–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009;461:1282–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, et al. 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;105:16767–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sleeth ML, Thompson EL, Ford HE, Zac-Varghese SE, Frost G. Free fatty acid receptor 2 and nutrient sensing: a proposed role for fibre, fermentable carbohydrates and short-chain fatty acids in appetite regulation. Nutr Res Rev 2010;23:135–45. [DOI] [PubMed] [Google Scholar]
- 80.Zhu L, Baker RD, Baker SS. Gut microbiome and nonalcoholic fatty liver diseases. Pediatr Res 2015;77:245–51. [DOI] [PubMed] [Google Scholar]
- 81.Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Hamady M, Chen YY, Knight R, Ahima RS, Bushman F, Wu GD. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 2009;137(5):1716–24, e1–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008;3:213–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, Fearnside J, Tatoud R, Blanc V, Lindon JC, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci USA 2006;103:12511–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vreugdenhil AC, Rousseau CH, Hartung T, Greve JW, van't Veer C, Buurman WA. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. J Immunol 2003;170(3):1399–405. [DOI] [PubMed] [Google Scholar]
- 85.Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, Gibson GR, Delzenne NM. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007;50:2374–83. [DOI] [PubMed] [Google Scholar]
- 86.de la Serre CB, Ellis CL, Lee J, Hartman AL, Rutledge JC, Raybould HE. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest Liver Physiol 2010;299:G440–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature 2012;489:242–9. [DOI] [PubMed] [Google Scholar]
- 88.Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther 2016;43:181–96. [DOI] [PubMed] [Google Scholar]
- 89.Zhu Y, Lin X, Li H, Li Y, Shi X, Zhao F, Xu X, Li C, Zhou G. Intake of meat proteins substantially increased the relative abundance of genus lactobacillus in rat feces. PLoS One 2016;11:e0152678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wong VW, Won GL, Chim AM, Chu WC, Yeung DK, Li KC, Chan HL. Treatment of nonalcoholic steatohepatitis with probiotics: a proof-of-concept study. Ann Hepatol 2013;12:256–62. [PubMed] [Google Scholar]
- 91.Everard A, Matamoros S, Geurts L, Delzenne NM, Cani PD. Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. MBio 2014;5:e01011–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nardone G, Compare D, Liguori E, Di Mauro V, Rocco A, Barone M, Napoli A, Lapi D, Iovene MR, Colantuoni A. Protective effects of Lactobacillus paracasei F19 in a rat model of oxidative and metabolic hepatic injury. Am J Physiol Gastrointest Liver Physiol 2010;299:G669–76. [DOI] [PubMed] [Google Scholar]
- 93.Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S, Carteni M, Nardone G. Gut–liver axis: the impact of gut microbiota on nonalcoholic fatty liver disease. Nutr Metab Cardiovasc Dis 2012;22:471–6. [DOI] [PubMed] [Google Scholar]
- 94.Eslamparast T, Eghtesad S, Hekmatdoost A, Poustchi H. Probiotics and nonalcoholic fatty liver disease. Middle East J Dig Dis 2013;5:129–36. [PMC free article] [PubMed] [Google Scholar]
- 95.Kaliannan K, Wang B, Li XY, Kim KJ, Kang JX. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci Rep 2015;5:11276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R, Rowland I, Wolvers D, Watzl B, Szajewska H, Stahl B, et al. Prebiotic effects: metabolic and health benefits. Br J Nutr 2010;104(Suppl 2):S1–63. [DOI] [PubMed] [Google Scholar]
- 97.Blaut M. Relationship of prebiotics and food to intestinal microflora. Eur J Nutr 2002;41:I11–6. [DOI] [PubMed] [Google Scholar]
- 98.Tuohy KM, Probert HM, Smejkal CW, Gibson GR. Using probiotics and prebiotics to improve gut health. Drug Discov Today 2003;8:692–700. [DOI] [PubMed] [Google Scholar]
- 99.Daubioul CA, Taper HS, De Wispelaere LD, Delzenne NM. Dietary oligofructose lessens hepatic steatosis, but does not prevent hypertriglyceridemia in obese zucker rats. J Nutr 2000;130:1314–9. [DOI] [PubMed] [Google Scholar]
- 100.Birt DF, Boylston T, Hendrich S, Jane JL, Hollis J, Li L, McClelland J, Moore S, Phillips GJ, Rowling M, et al. Resistant starch: promise for improving human health. Adv Nutr 2013;4(6):587–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Keenan MJ, Zhou J, Hegsted M, Pelkman C, Durham HA, Coulon DB, Martin RJ. Role of resistant starch in improving gut health, adiposity, and insulin resistance. Adv Nutr 2015;6(2):198–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zaman SA, Sarbini SR. The potential of resistant starch as a prebiotic. Crit Rev Biotechnol 2016;36:578–84. [DOI] [PubMed] [Google Scholar]
- 103.Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol 2012;9:577–89. [DOI] [PubMed] [Google Scholar]
- 104.Martínez I, Kim J, Duffy PR, Schlegel VL, Walter J. Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLoS One 2010;5:e15046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P, Bergerat A, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J 2011;5:220–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Abell GC, Cooke CM, Bennett CN, Conlon MA, McOrist AL. Phylotypes related to Ruminococcus bromii are abundant in the large bowel of humans and increase in response to a diet high in resistant starch. FEMS Microbiol Ecol 2008;66:505–15. [DOI] [PubMed] [Google Scholar]
- 107.Ze X, Duncan SH, Louis P, Flint HJ. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J 2012;6:1535–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.O’Callaghan A, Van Sinderen D. Bifidobacteria and their role as members of the human gut microbiota. Front Microbiol 2016;7:925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Flint HJ, Duncan SH, Scott KP, Louis P. Interactions and competition within the microbial community of the human colon: links between diet and health. Environ Microbiol 2007;9:1101–11. [DOI] [PubMed] [Google Scholar]
- 110.Endo H, Niioka M, Kobayashi N, Tanaka M, Watanabe T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One 2013;8:e63388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Jin CJ, Sellmann C, Engstler AJ, Ziegenhardt D, Bergheim I. Supplementation of sodium butyrate protects mice from the development of non-alcoholic steatohepatitis (NASH). Br J Nutr 2015;114:1745–55. [DOI] [PubMed] [Google Scholar]
- 112.Macfarlane S, Macfarlane GT, Cummings JH. Review article: prebiotics in the gastrointestinal tract. Aliment Pharmacol Ther 2006;24:701–14. [DOI] [PubMed] [Google Scholar]
- 113.Griffiths EA, Duffy LC, Schanbacher FL, Qiao H, Dryja D, Leavens A, Rossman J, Rich G, Dirienzo D, Ogra PL. In vivo effects of bifidobacteria and lactoferrin on gut endotoxin concentration and mucosal immunity in Balb/c mice. Dig Dis Sci 2004;49:579–89. [DOI] [PubMed] [Google Scholar]
- 114.Hevia A, Delgado S, Sanchez B, Margolles A. Molecular players involved in the interaction between beneficial bacteria and the immune system. Front Microbiol 2015;6:1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Petrof EO, Kojima K, Ropeleski MJ, Musch MW, Tao Y, De Simone C, Chang EB. Probiotics inhibit nuclear factor-kappaB and induce heat shock proteins in colonic epithelial cells through proteasome inhibition. Gastroenterology 2004;127:1474–87. [DOI] [PubMed] [Google Scholar]
- 116.Kleessen B, Hartmann L, Blaut M. Oligofructose and long-chain inulin: influence on the gut microbial ecology of rats associated with a human faecal flora. Br J Nutr 2001;86:291–300. [DOI] [PubMed] [Google Scholar]
- 117.Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol 2007;73:1073–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and butyrate-producing colon bacteria: importance and strategies for their stimulation in the human gut. Front Microbiol 2016;7:979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ramirez-Farias C, Slezak K, Fuller Z, Duncan A, Holtrop G, Louis P. Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii. Br J Nutr 2009;101:541–50. [DOI] [PubMed] [Google Scholar]
- 120.Davis LM, Martinez I, Walter J, Goin C, Hutkins RW. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS One 2011;6:e25200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Fernando WM, Hill JE, Zello GA, Tyler RT, Dahl WJ, Van Kessel AG. Diets supplemented with chickpea or its main oligosaccharide component raffinose modify faecal microbial composition in healthy adults. Benef Microbes 2010;1:197–207. [DOI] [PubMed] [Google Scholar]
- 122.Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux J-J, Blugeon S, Bridonneau C, Furet JP, Corthier G, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA 2008;105:16731–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Zeisel SH, da Costa KA. Choline: an essential nutrient for public health. Nutr Rev 2009;67:615–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zeisel SH. Choline: an essential nutrient for humans. Nutrition 2000;16:669–71. [DOI] [PubMed] [Google Scholar]
- 125.Sherriff JL, O'Sullivan TA, Properzi C, Oddo JL, Adams LA. Choline, its potential role in nonalcoholic fatty liver disease, and the case for human and bacterial genes. Adv Nutr 2016;7(1):5–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Corbin KD, Zeisel SH. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr Opin Gastroenterol 2012;28:159–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Buchman AL, Dubin MD, Moukarzel AA, Jenden DJ, Roch M, Rice KM, Gornbein J, Ament ME. Choline deficiency: a cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology 1995;22:1399–403. [PubMed] [Google Scholar]
- 128.Noga AA, Zhao Y, Vance DE. An unexpected requirement for phosphatidylethanolamine N-methyltransferase in the secretion of very low density lipoproteins. J Biol Chem 2002;277:42358–65. [DOI] [PubMed] [Google Scholar]
- 129.Teodoro JS, Rolo AP, Duarte FV, Simoes AM, Palmeira CM. Differential alterations in mitochondrial function induced by a choline-deficient diet: understanding fatty liver disease progression. Mitochondrion 2008;8:367–76. [DOI] [PubMed] [Google Scholar]
- 130.Martínez-del Campo A, Bodea S, Hamer HA, Marks JA, Haiser HJ, Turnbaugh PJ, Balskus EP. Characterization and detection of a widely distributed gene cluster that predicts anaerobic choline utilization by human gut bacteria. MBio 2015;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011;140:976–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Nair S, Cope K, Risby TH, Diehl AM. Obesity and female gender increase breath ethanol concentration: potential implications for the pathogenesis of nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:1200–4. Erratum in: Am J Gastroenterol 2001;23:2809. [DOI] [PubMed] [Google Scholar]
- 133.Cope K, Risby T, Diehl AM. Increased gastrointestinal ethanol production in obese mice: implications for fatty liver disease pathogenesis. Gastroenterology 2000;119:1340–7. [DOI] [PubMed] [Google Scholar]
- 134.Tetri LH, Basaranoglu M, Brunt EM, Yerian LM, Neuschwander-Tetri BA. Severe NAFLD with hepatic necroinflammatory changes in mice fed trans fats and a high-fructose corn syrup equivalent. Am J Physiol Gastrointest Liver Physiol 2008;295:G987–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Ouyang X, Cirillo P, Sautin Y, McCall S, Bruchette JL, Diehl AM, Johnson RJ, Abdelmalek MF. Fructose consumption as a risk factor for non-alcoholic fatty liver disease. J Hepatol 2008;48:993–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Bergheim I, Weber S, Vos M, Kramer S, Volynets V, Kaserouni S, McClain CJ, Bischoff SC. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: role of endotoxin. J Hepatol 2008;48:983–92. [DOI] [PubMed] [Google Scholar]
- 137.Busserolles J, Gueux E, Rock E, Demigne C, Mazur A, Rayssiguier Y. Oligofructose protects against the hypertriglyceridemic and pro-oxidative effects of a high fructose diet in rats. J Nutr 2003;133:1903–8. [DOI] [PubMed] [Google Scholar]
- 138.Suganthi R, Rajamani S, Ravichandran MK, Anuradha CV. Effect of food seasoning spices mixture on biomarkers of oxidative stress in tissues of fructose-fed insulin-resistant rats. J Med Food 2007;10:149–53. [DOI] [PubMed] [Google Scholar]
- 139.Li YC, Hsieh CC. Lactoferrin dampens high-fructose corn syrup-induced hepatic manifestations of the metabolic syndrome in a murine model. PLoS One 2014;9:e97341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Sellmann C, Priebs J, Landmann M, Degen C, Engstler AJ, Jin CJ, Gärttner S, Spruss A, Huber O, Bergheim I. Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time. J Nutr Biochem 2015;26:1183–92. [DOI] [PubMed] [Google Scholar]
- 141.Spruss A, Bergheim I. Dietary fructose and intestinal barrier: potential risk factor in the pathogenesis of nonalcoholic fatty liver disease. J Nutr Biochem 2009;20:657–62. [DOI] [PubMed] [Google Scholar]
- 142.Askari F, Rashidkhani B, Hekmatdoost A. Cinnamon may have therapeutic benefits on lipid profile, liver enzymes, insulin resistance, and high-sensitivity C-reactive protein in nonalcoholic fatty liver disease patients. Nutr Res 2014;34:143–8. [DOI] [PubMed] [Google Scholar]
- 143.Faghihzadeh F, Adibi P, Hekmatdoost A. The effects of resveratrol supplementation on cardiovascular risk factors in patients with non-alcoholic fatty liver disease: a randomised, double-blind, placebo-controlled study. Br J Nutr 2015;114:796–803. [DOI] [PubMed] [Google Scholar]
- 144.Faghihzadeh F, Adibi P, Rafiei R, Hekmatdoost A. Resveratrol supplementation improves inflammatory biomarkers in patients with nonalcoholic fatty liver disease. Nutr Res 2014;34:837–43. [DOI] [PubMed] [Google Scholar]
- 145.Faghihzadeh F, Hekmatdoost A, Adibi P.. Resveratrol and liver: a systematic review. J Res Med Sci 2015;20(8):797–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Eslamparast T, Zamani F, Hekmatdoost A, Sharafkhah M, Eghtesad S, Malekzadeh R, Poustchi H. Effects of synbiotic supplementation on insulin resistance in subjects with the metabolic syndrome: a randomised, double-blind, placebo-controlled pilot study. Br J Nutr 2014;112:438–45. [DOI] [PubMed] [Google Scholar]
- 147.Ghorbani Z, Hekmatdoost A, Mirmiran P. Anti-hyperglycemic and insulin sensitizer effects of turmeric and its principal constituent curcumin. Int J Endocrinol Metab 2014;12:e18081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lee HC, Jenner AM, Low CS, Lee YK. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res Microbiol 2006;157:876–84. [DOI] [PubMed] [Google Scholar]
- 149.Derrien M, Van Baarlen P, Hooiveld G, Norin E, Muller M, de Vos WM. Modulation of mucosal immune response, tolerance, and proliferation in mice colonized by the mucin-degrader Akkermansia muciniphila. Front Microbiol 2011;2:166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Zelber-Sagi S, Nitzan-Kaluski D, Goldsmith R, Webb M, Blendis L, Halpern Z,Oren R. Long term nutritional intake and the risk for non-alcoholic fatty liver disease (NAFLD): a population based study. J Hepatol 2007;47:711–7. [DOI] [PubMed] [Google Scholar]
- 151.Araya J, Rodrigo R, Videla LA, Thielemann L, Orellana M, Pettinelli P, Poniachik J. Increase in long-chain polyunsaturated fatty acid n-6/n-3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci (Lond) 2004;106:635–43. [DOI] [PubMed] [Google Scholar]
- 152.Ghosh S, DeCoffe D, Brown K, Rajendiran E, Estaki M, Dai C, Yip A, Gibson DL. Fish oil attenuates omega-6 polyunsaturated fatty acid-induced dysbiosis and infectious colitis but impairs LPS dephosphorylation activity causing sepsis. PLoS One 2013;8:e55468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Huang EY, Leone VA, Devkota S, Wang Y, Brady MJ, Chang EB. Composition of dietary fat source shapes gut microbiota architecture and alters host inflammatory mediators in mouse adipose tissue. JPEN J Parenter Enteral Nutr 2013;37:746–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Pereira DI, Gibson GR. Cholesterol assimilation by lactic acid bacteria and bifidobacteria isolated from the human gut. Appl Environ Microbiol 2002;68:4689–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.St-Onge MP, Farnworth ER, Jones PJ. Consumption of fermented and nonfermented dairy products: effects on cholesterol concentrations and metabolism. Am J Clin Nutr 2000;71:674–81. [DOI] [PubMed] [Google Scholar]