ABSTRACT
Being overweight and obesity are the leading causes of liver disease in Western countries. Liver damage induced by being overweight can range from steatosis, harmless in its simple form, to steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Alcohol consumption is an additional major cause of liver disease. Not all individuals who are overweight or excessively consume alcohol develop nonalcoholic fatty liver diseases (NAFLD) or alcoholic liver disease (ALD) and advanced liver disease. The role of the intestinal microbiota (IM) in the susceptibility to liver disease in this context has been the subject of recent studies. ALD and NAFLD appear to be influenced by the composition of the IM, and dysbiosis is associated with ALD and NAFLD in rodent models and human patient cohorts. Several microbial metabolites, such as short-chain fatty acids and bile acids, are specifically associated with dysbiosis. Recent studies have highlighted the causal role of the IM in the development of liver diseases, and the use of probiotics or prebiotics improves some parameters associated with liver disease. Several studies have made progress in deciphering the mechanisms associated with the modulation of the IM. These data have demonstrated the intimate relationship between the IM and metabolic liver disease, suggesting that targeting the gut microbiota could be a new preventive or therapeutic strategy for these diseases.
METABOLIC LIVER DISEASES: FROM DIAGNOSIS TO CURRENT TREATMENTS
Pathogenesis of NAFLD and ALD
Excessive alcohol consumption and being obese/overweight are the leading causes of chronic liver disease in Western countries. Nonalcoholic fatty liver disease (NAFLD) encompasses all liver lesions that can be observed in overweight/obese patients, ranging from pure steatosis to steatohepatitis (nonalcoholic steatohepatitis [NASH]), fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). Alcoholic liver disease (ALD) defines liver lesions observed in patients with alcohol abuse and includes steatosis, hepatitis, fibrosis, cirrhosis, and HCC. It usually occurs when alcohol consumption is higher than 70 g/day in men and 60 g/day in women. However, only some chronic alcohol consumers develop liver injury, suggesting that factors other than excessive alcohol consumption play a role in ALD. Moreover, some patients with other liver diseases, such as chronic hepatitis C or NAFLD, also consume alcohol. Alcohol may lead to liver injury at a lower level of consumption in these patients with another underlying liver disease.
NAFLD usually occurs in patients with a metabolic syndrome. Such patients are overweight and may have hypertension, dyslipidemia, high blood glucose, or diabetes. However, as with chronic alcohol consumers, overweight or obese patients do not systematically develop NAFLD.
NAFLD and ALD encompass a wide spectrum of pathological lesions. Liver lesions in both diseases include a combination of steatosis, inflammation, hepatocyte necrosis, and fibrosis. Thus, NASH is merely a stage of NAFLD. Although the lesions are similar, the mechanisms of the two diseases share similarities, but there are also differences (1, 2).
Steatosis is an accumulation of triglycerides in the cytoplasm of hepatocytes. Steatosis is reversible after withdrawal of its cause (alcohol intake, being overweight), but if its cause remains, it may progress towards inflammation (Fig. 1). The presence of an inflammatory process in alcoholic patients with simple steatosis suggests that they will progress towards alcoholic hepatitis in the absence of alcohol withdrawal (3), whereas the transition from simple steatosis to steatohepatitis in overweight patients is less well understood (4). Another difference between the two diseases is the degree of the liver inflammation. The inflammatory infiltrate in both alcoholic and overweight patients may contain lymphocytes and polymorphonuclear neutrophils along with ballooning and necrosis of hepatocytes. Nevertheless, the intensity of this process is generally much higher in alcoholic than overweight patients. Megamitochondria is often observed in both diseases. The distinction of simple steatosis from true NASH may be difficult. The lesion is considered to be NASH when lobular inflammation and liver cell clarification/ballooning are present (5). The inflammatory process progresses towards fibrosis and cirrhosis in approximately 20% of alcoholic patients. The natural incidence of cirrhosis in overweight patients is less clear, possibly due to death from other diseases associated with being overweight, such as cardiovascular diseases. The incidence of HCC in cirrhotic livers is approximately 3 to 5% each year (6). HCC may occur without cirrhosis in overweight patients, whereas cirrhosis is an obligatory step in alcoholic patients (7). Severe alcoholic hepatitis is a particular form of ALD, characterized by acute hepatocellular insufficiency, severe histological liver injury, and a high mortality rate, as the 1-month survival of such patients is between 50 and 65%. Among patients who survive beyond 1 month, 50% die within the following year (8).
FIGURE 1.

Histology of the liver. Paraffin sections (4 μm thick) were stained with hematoxylin and eosin. Images were obtained using a Hamamatsu scanning module (Hamamatsu LX2000) and appropriate software (magnification, ×100). (A) Healthy tissue; (B) steatosis; (C) steatosis with inflammation; (D) fibrosis.
NAFLD and ALD: Clinical Aspects and Diagnosis
A diagnosis of NAFLD is clinically suspected in asymptomatic patients with elevated transaminases or radiological evidence of steatosis. Diagnosis of NAFLD may be associated with other components of metabolic syndrome, such as being overweight/obese, type 2 diabetes mellitus, hypertension, or dyslipidemia. When symptoms occur, they are nonspecific, consisting of fatigue and/or vague right-upper-quadrant abdominal pain. ALD should be suspected in all patients with excessive alcohol intake. The WHO safety threshold for alcohol consumption is approximately 20 g/day for women and 30 g/day for men, with 1 day of abstinence from alcohol each week. Higher consumption may lead to complications, but individual susceptibility to alcohol varies widely. For example, we have shown that overweight patients are more sensitive to alcohol-dependent liver toxicity, due to alcohol-induced inflammation in adipose tissue (9).
Elevated transaminases and gamma-glutamyltransferase are often the only abnormalities detected in overweight and alcoholic patients. A ratio of ALT to aspartate aminotransferase (AST) of <1 is suggestive of ALD in most cases, whereas an ALT/AST ratio of >1 suggests the presence of NAFLD. An ALT/AST ratio of <1 may also be observed when NAFLD progresses towards advanced fibrosis/cirrhosis. Transaminase values do not reflect the extent of liver injury, and a normal transaminase level does not guarantee the absence of underlying steatohepatitis or fibrosis. Conversely, increased transaminase levels are often associated with inflammation. The level of transaminases in ASH and NASH is rarely higher than twice the upper limit of the normal value (Table 1), and prothrombin time and bilirubin and albumin levels remain normal in the absence of hepatocellular insufficiency. Abdominal imaging aids in diagnosing steatosis. Abdominal ultrasound is noninvasive and can detect a steatosis involving more than 30% of hepatocytes (10). Computed tomography scan lacks sensitivity and introduces a radiation hazard. Magnetic resonance imaging (MRI) is expensive but provides the highest precision for quantifying steatosis. Nevertheless, liver imaging cannot distinguish between simple steatosis and NASH in the absence of liver dysmorphy due to cirrhosis (reviewed in reference 1).
TABLE 1.
Main differences between ALD and NAFLD
| ALD | NAFLD | |
|---|---|---|
| Clinical characteristic | Alcohol intake | Overweight |
| Metabolic syndrome | ||
| Hypertension | ||
| Biology | ALT/AST < 1 | ALT/AST > 1 in 66% of cases |
| ALT/AST < 1 usually in patients with advanced fibrosis | ||
| Dyslipidemia | ||
| Increased blood glucose | ||
| Liver biopsy | Steatosis | Steatosis |
| In patients with alcoholic hepatitis, high inflammatory process | In patients with NASH, usually low inflammatory process |
Liver biopsy remains the gold standard to assess liver injury because of the lack of good correlations between simple blood tests and histological liver lesions. Noninvasive biological and physical tests, such as transient elastography, are often used for initial screening to limit the use of liver biopsy.
The mortality rate is higher in patients with NAFLD than in the general population (11). NAFLD is associated with an increased risk of cardiovascular disease and liver-related mortality.
Nevertheless, most patients with NAFLD display only simple steatosis, without inflammation. Their risk of progression from steatosis to steatohepatitis (i.e., NASH) is very low. These patients do not generally show a progression of liver injury and increased mortality risk after 20 years of follow-up. Conversely, patients with NASH may progress after several years towards fibrosis, cirrhosis, and HCC, with increased mortality. The three major causes of NASH-related mortality are cardiovascular diseases, all-cause malignancy, and liver-related death. NASH is associated with a >10-fold-increased risk of liver-related death (2.8% versus 0.2%) and a 2-fold-increased risk of death from cardiovascular disease (15.5% versus 7.5%) (12, 13). The presence and severity of fibrosis are the strongest determinants of long-term prognosis (14). The progression of fibrosis is generally slow, taking approximately 8 years to progress from no fibrosis to low-stage fibrosis. However, some patients progress more rapidly (15), similar to what is seen with those with chronic hepatitis C. Factors leading to more-rapid progression are a body mass index (BMI) of >30 kg/m2, persistently abnormal liver enzyme levels with an AST/ALT ratio of >1, type 2 diabetes mellitus, hypertension, high triglycerides, low high-density lipoprotein (HDL) cholesterol levels, and a family history of diabetes mellitus (13, 16).
Of note, 30 to 60% of patients with NASH who develop HCC harbor advanced fibrosis without cirrhotic features (7, 17).
NAFLD: Current Treatments
Metabolic syndrome and its components are the primary cause of NAFLD. The management of NAFLD includes treatment of the risk factors that are commonly associated with metabolic syndrome through lifestyle modifications. No treatment is needed for simple steatosis, as it is not associated with increased morbidity. Thus, treatment should be considered in patients with NASH.
Dietary modifications leading to a weight loss of 7% or more may improve liver histology. A hypocaloric diet leading to a weight loss of 0.5 to 1.0 kg/week is generally recommended (18). Nevertheless, most patients do not achieve this weight loss goal.
Exercise, independent of weight loss, may also improve liver histology, decreasing liver fat content and increasing muscle mass (19). Lasting weight loss can be achieved in patients with NAFLD by combining diet and exercise for longer than 12 months (20). No specific treatment is recommended for NASH, despite several clinical trials. The best-studied treatments are insulin sensitizers, such as metformin, thiazolidinediones, and glucagon-like peptide 1 agonists (liraglutide); antioxidants, such as vitamin E; and cytoprotective agents, such as ursodeoxycholic acid (UDCA) and obeticholic acid (OCA) (21). Thiazolidinediones are peroxisome-proliferator-activated receptor gamma agonists. In addition to increasing insulin sensitivity, they promote peripheral fatty acid uptake and thus divert them from the liver towards adipose tissue. They improve steatosis, inflammation, and ballooning degeneration. No significant effect on liver fibrosis has been found. These drugs are no longer used in Europe due to their side effects: weight gain, increased risk of heart failure, and possible increased risk of bladder cancer (22). Metformin led to encouraging results in animal models, but no improvement of NASH has been observed in human clinical trials. However, metformin may decrease the risk of liver cancer and modulate the intestinal microbiota (IM), which may contribute to its beneficial effects. Metformin increases the abundance of the mucin-degrading bacterium Akkermansia, which may result in an improved metabolic profile in patients with type 2 diabetes (23). Vitamin E has shown a benefit in steatosis and inflammation, but not fibrosis, in nondiabetic patients with NAFLD (24). However, vitamin E does not improve insulin resistance in these nondiabetic patients and may increase all-cause mortality (24). UDCA may prevent hepatocyte apoptosis and downregulate inflammatory pathways. Improvement of liver transaminases following treatment with UDCA was shown in one clinical trial but not another, and there was only mild improvement of liver lesions, if any. OCA activates the farnesoid X receptor (FXR), whereas UDCA is a weak agonist of FXR. In a clinical trial, OCA improved liver transaminase levels and pathological lesions, fibrosis, hepatocellular ballooning, steatosis, and lobular inflammation. The safety of OCA must be tested, as it also increases alkaline phosphatase and low-density lipoprotein cholesterol (25). Bariatric surgery may be recommended in obese patients with a BMI of >40 kg/m2 or >35 kg/m2 with a comorbidity (26). Bariatric surgery improves liver histology and reduces mortality from both NASH-related complications and all-cause mortality in the 7 to 10 years that follow the bariatric procedure.
Overall, lifestyle modifications can provide a strong benefit to patients but are difficult to follow in the long term. Some treatments may be beneficial for patients, but the magnitude of the effects is small, in particular on liver fibrosis.
ALD: Current Treatments
Treatment of ALD relies principally on alcohol withdrawal. Nevertheless, patients with severe alcoholic hepatitis, defined by a Maddrey discriminant function of >32, must also be treated with corticosteroids, which is the reference treatment. Corticosteroids decrease liver inflammation and improve 1-month and possibly 6-month survival. In one of the largest trials to date, mortality after 28 days was 17% (45 of 269 patients) in the placebo group versus 14% (38 of 266 patients) in the prednisolone group (27). However, survival curves converged after 28 days, such that prednisolone therapy provided no benefit to patients after 90 days or 1 year. Pentoxifylline, a phosphodiesterase inhibitor, may reduce tumor necrosis factor alpha production. Its efficacy, alone or in association with corticosteroids, is controversial (28). It may increase survival relative to placebo but does not do so relative to prednisolone (29). N-Acetylcysteine is a glutathione precursor. In one trial, its addition to prednisolone increased survival of patients, but its efficacy needs to be confirmed (30). Enteral or parenteral nutritional supplementation may improve the prognosis, as ALD is often associated with protein, vitamin, and mineral deficiencies, but its impact on survival remains unclear (31). Nevertheless, nutritional support should be considered in patients with severe alcoholic hepatitis. Liver transplantation is generally proposed to patients with severe liver insufficiency after at least 6 months of abstinence. However, the mortality rate of patients with severe alcoholic hepatitis is very high (more than 50% during the first 3 months). Thus, it has been proposed to proceed rapidly to liver transplantation for patients with severe alcoholic hepatitis who do not respond to corticosteroids. In a clinical trial of 26 patients undergoing liver transplantation, the 6-month survival rate was 77%, similar to that of patients who respond to corticosteroids. After 2 years, 71% were still alive and only 3 patients resumed the consumption of alcohol (32).
Therapeutic resources for metabolic liver diseases are scarce, and the recent discovery of the involvement of the IM in NAFLD and ALD opens new therapeutic perspectives.
NAFLD AND THE GUT MICROBIOTA
NASH and Dysbiosis
Dysbiosis is defined as a microbial imbalance associated with a disease. Dysbiosis in NAFLD was first described in animal studies. For example, the severity of steatosis was associated with an increase in the Lactobacillus population in mice fed a high-fat diet (HFD) for 10 weeks (33). Since then, an increasing number of studies have investigated the gut microbiota in relation to human NAFLD, using culture-independent techniques, such as quantitative PCR (qPCR) or sequencing (Table 2). Children and adolescents (with or without NASH) were found to have a higher abundance of Bacteroidetes and a lower abundance of Firmicutes than healthy controls. Proteobacteria, Enterobacteriaceae, and Escherichia were the only phylum, family, and genus, respectively, exhibiting a significant difference between microbiomes from obese and NASH individuals and were found to be more abundant in patients with NASH (34). In adults, the comparison of 30 obese patients with clinically defined NAFLD and 30 nonobese controls revealed an overrepresentation of Lactobacillus species and selected members of the phylum Firmicutes (Lachnospiraceae, Dorea, Robinsoniella, and Roseburia) in NAFLD patients (35). Another study comprising 53 NAFLD patients and 32 healthy subjects found that Escherichia, Anaerobacter, Lactobacillus, and Clostridium cluster XI were more abundant in the gut microbiotas of NAFLD patients, whereas Alistipes, Odoribacter, Oscillibacter, and Flavonifractor were less abundant (36).
TABLE 2.
Microbiotas associated with different stages of NAFLDa
| Condition | Phylum | Family | Genus | Reference |
|---|---|---|---|---|
| NAFLD versus healthy controls | [Bacteroidetes] | Porphyromonadaceae ⇩ | Healthy (n = 30, BMI = 22)NAFLD (n = 30, BMI = 33)Raman et al., 2013 (35) | |
| [Firmicutes] | Veillonellaceae ⇧ | |||
| Lactobacillaceae ⇧ | Lactobacillus ⇧ | |||
| Ruminococcaceae ⇩ | Oscillibacter ⇩ | |||
| Lachnospiraceae ⇧ | Robinsoniella ⇧ | |||
| Roseburia ⇧ | ||||
| Dorea ⇧ | ||||
| [Proteobacteria] | Kiloniellaceae ⇧ | |||
| Pasteurellaceae ⇧ | ||||
| [Bacteroidetes] | [Rikenellaceae] | Alistipes ⇩ | Healthy (n = 32, BMI = 22)NAFLD (n = 53, BMI = 26)Jiang et al., 2015 (36) | |
| [Porphyromonadaceae] | Odoribacter ⇩ | |||
| [Firmicutes] | [Lactobacillaceae] | Lactobacillus ⇧ | ||
| [Oscillospiraceae] | Oscillibacter ⇩ | |||
| [Clostridiaceae] | Anaerobacter ⇧ | |||
| [Lachnospiraceae] | Clostridium XI ⇧ | |||
| [Streptococcaceae] | Streptococcus ⇧ | |||
| Flavonifractor ⇩ | ||||
| [Proteobacteria] | [Enterobacteriaceae] | Escherichia ⇧ | ||
| Lentisphaerae ⇩ | ||||
| NASH versus healthy controls | Bacteroidetes ⇩ | Healthy (n = 17, BMI = 26) | ||
| NASH (n = 22, BMI = 32) | ||||
| Mouzaki et al., 2013 (37) | ||||
| Bacteroidetes ⇧ | Prevotellaceae ⇧ | Prevotella ⇧ | Healthy (n = 16, BMI = 20)NASH (n = 22, BMI = 34)ChildrenZhu et al., 2013 (34) | |
| Rikenellaceae ⇩ | Allistipes ⇩ | |||
| Firmicutes ⇩ | Ruminococcaceae ⇩ | Oscillospira ⇩ | ||
| Ruminococcus ⇩ | ||||
| Lachnospiraceae ⇩ | Blautia ⇩ | |||
| Coprococcus ⇩ | ||||
| Eubacterium ⇩ | ||||
| Roseburia ⇩ | ||||
| Proteobacteria ⇧ | Enterobacteriaceae ⇧ | Escherichia coli ⇧ | Healthy (n = 22, BMI = 22)NASH (n = 16, BMI = 29)Wong et al., 2013 (38) | |
| Actinobacteria | Bifidobacteriaceae ⇩ | Bifidobacterium ⇩ | ||
| Firmicutes ⇩ | Unclassified Clostridiales ⇩ | Faecalibacterium ⇩ | ||
| [Ruminococcaceae] | Anaerosporobacter ⇩ | |||
| [Lachnospiraceae] | Allisonella ⇧ | |||
| [Veillonellaceae] | ||||
| [Bacteroidetes] | Porphyromonadaceae ⇧ | Parabacteroides ⇧ | ||
| [Proteobacteria] | Succinivibrionaceae ⇧ | |||
| NASH versus obese | Bacteroidetes ⇩ | [Lachnospiraceae] | Clostridium coccoides ⇧ | Obese (n = 11, BMI = 28) |
| NASH (n = 22, BMI = 32) | ||||
| [Firmicutes] | Mouzaki et al., 2013 (37) | |||
| Proteobacteria ⇧ | Enterobacteriaceae ⇧ | Escherichia coli ⇧ | Obese (n = 25, BMI = 33) | |
| NASH (n = 22, BMI = 34) | ||||
| Zhu et al., 2013 (34) | ||||
| NAFLD patients NASH versus no NASH | [Bacteroidetes] | Bacteroidaceae ⇧ | Bacteroides ⇧ | No NASH (n = 22, BMI = 30) |
| NASH (n = 35, BMI = 32) | ||||
| Boursier et al., 2016 (122) |
Microbiota were analyzed using 16S deep sequencing, except in the study by Mouzaki et al., in which qPCR was used. Additional taxonomic information for discriminating taxa is in brackets. Taxa in boldface are those identified in more than one study.
In a prospective, cross-sectional study, the gut microbiota composition (analyzed using qPCR) was compared between adults with biopsy-proven NAFLD (simple steatosis or NASH) and healthy controls. Patients with NASH had a lower proportion of Bacteroidetes than individuals with simple steatosis and healthy controls, even after adjusting for body mass index (37). Wong et al. investigated the microbiome composition using 16S rRNA gene sequencing in 16 biopsy-proven NASH patients and 22 healthy controls. The order Aeromonadales, the families Succinivibrionaceae and Porphyromonadaceae, and the genera Parabacteroides and Allisonella were found to be more abundant in NASH patients. Conversely, the class Clostridia, the order Clostridiales, and the genera Anaerosporobacter and Faecalibacterium were less abundant in NASH patients (38). Differences in fecal microbiota between nonobese adults with and without NAFLD have also been found, characterized by a higher abundance of Bacteroidetes and a lower abundance of Firmicutes in nonobese patients with NAFLD (39).
The effect of a choline-deficient diet on the composition of the human gut microbiome and the development of fatty liver has been studied by Spencer et al. They found that a baseline microbial composition correlated with the development of fatty liver in response to choline deficiency. The abundance of Gammaproteobacteria negatively correlated with the development of steatosis, whereas the abundance of Erysipelotrichia was positively associated, suggesting that baseline levels of these populations may predict susceptibility to fatty liver due to choline deficiency (40).
Altogether, these human studies revealed taxonomic differences between the gut microbiotas of healthy controls and patients with NASH. However, the results were heterogeneous, even contradictory, despite the identification of dysbiosis at the phylum or genus levels. Factors that may explain these differences include differences in the study design and methods of gut microbiota analysis, the variety of clinical endpoints, or changes of the gut microbiota due to diet or medications. The specific phyla or genera that are the causative agents in NASH development remain unclear.
A Causative Role for the Gut Microbiota in NAFLD
Patients suffering from NAFLD harbor a gut microbiota different from that of healthy controls, but the previously described association studies do not indicate whether this dysbiosis is a cause or a consequence of liver disease. Strategies have been developed in animal models to prove the causality of the gut microbiota in disease development. These strategies aim to transmit a phenotype from one mouse to another by a microbiota transfer that can be performed via cohousing of conventional mice or fecal transplant to germ-free or antibiotic-treated mice. These microbiota transfer strategies have already proven the causative role of the gut bacteria in several diseases including obesity and inflammatory bowel disease (41, 42). It was first demonstrated that cohousing germ-free mice with their conventional counterparts leads to increased glycogenesis in the liver of ex-germ-free mice prior to triggering increases in hepatic triglyceride synthesis. Colonization was also associated with altered hepatic Cyp8b1 expression and changes in bile acid (BA) composition. The hepatic triglyceride, glucose, and glycogen levels were strongly associated with the Coriobacteriaceae family (phylum Actinobacteria) (43). Henao-Mejia et al. further established the causative role of the gut microbiota in the development of liver disease using mouse models deficient in the proinflammatory multiprotein complex called the inflammasome (44). These inflammasome-deficient mice exhibited exacerbated NAFLD phenotypes when fed a methionine choline-deficient diet or HFD. This was associated with changes in the composition of the gut microbiota, especially an increase in the proportion of bacteria belonging to the Porphyromonadaceae family. Strikingly, cohousing of wild-type mice with these inflammasome-deficient mice resulted in increased glucose intolerance and obesity, as well as hepatic steatosis and liver inflammation, in the wild-type mice, indicating that dysbiosis itself can induce NAFLD progression. Another study conducted in mouse models demonstrated that the susceptibility to develop NAFLD can be transmitted by fecal transplant to recipient germ-free mice (45). They first observed that conventionally raised mice displayed various levels of weight gain, glucose intolerance, and steatosis when given an HFD for 16 weeks. They selected two donor mice for microbiota transplant based on their opposite responses to the HFD. Although the mice were the same weight, one displayed low fasting glycemia and slight steatosis (“nonresponder”), and the other displayed insulin resistance and marked steatosis (“responder”). Two groups of germ-free mice were transplanted with the gut microbiota of the two selected mice by oral gavage of donor cecal content. Only the mice transplanted with the responder microbiota developed fasting hyperglycemia, hyperinsulinemia, and liver steatosis after being fed an HFD and showed increased expression of genes involved in lipogenesis. This suggests that differences in susceptibility to develop NAFLD may be controlled primarily by the gut microbiota. Lachnospiraceae and Barnesiella were found to be overrepresented in the gut microbiotas of mice with glucose intolerance and steatosis, whereas the group of mice that did not develop features of NAFLD had a higher population of Bacteroides vulgatus (45).
Mechanisms Linking the Gut Microbiota to NAFLD
Several mechanisms have been proposed to link the gut microbiota to NAFLD, including small intestinal bowel overgrowth (SIBO), gut leakiness and resulting endotoxemia and inflammation, endogenous ethanol production, or BA and choline metabolism.
SIBO is a condition in which colonic bacteria colonize the small bowel due to altered intestinal motility. The diagnosis of bacterial overgrowth is performed using various techniques including breath testing and the culture of an aspirate from the jejunum, whereas culture-independent techniques, such as qPCR and sequencing, have been rarely applied. Most controlled trials using breath testing revealed a higher prevalence of SIBO in NAFLD patients than in healthy subjects (reviewed in reference 46). Moreover, SIBO correlated with the severity of steatosis but not NASH (47). However, no difference was found in bacterial numbers between patients suffering from NAFLD or NASH and healthy controls in a study using qPCR (37), suggesting that the use of molecular techniques may revise the current view on the role of SIBO in NAFLD.
Numerous studies have shown that the IM tunes the homeostasis of the gut barrier. A disruption of gut barrier integrity, characterized by the disruption of tight junctions and increased permeability, has been found in biopsy-proven NAFLD patients (47). Similarly, increased gut permeability has been observed in children with NAFLD and was found to correlate with the severity of the disease (48). This loss of barrier function leads to increased amounts of microbial products reaching the liver via portal circulation (49). Among these microbial products, lipopolysaccharide (LPS), a component of the bacterial cell wall of Gram-negative bacteria, binds to Toll-like receptor 4 (TLR4), triggering not only inflammation but also production of the extracellular matrix by hepatic stellate cells, leading to fibrosis (Fig. 2) (50, 51). In addition to TLR4, TLR2 and TLR9 may also be involved in NAFLD development, as TLR2- and TLR9-deficient mice display less steatosis and inflammation than their wild-type counterparts. In addition to increased gut permeability, HFD results in increased intestinal LPS absorption through high chylomicron production by intestinal epithelial cells, leading to low-grade endotoxemia (52). Animal studies suggest that endotoxemia alone may induce NASH, and higher endotoxin levels have been observed in NASH patients than in subjects with steatosis (53, 54). However, endotoxemia is not always higher in NASH (55), suggesting that LPS is not the only component that triggers the cascade of increased proinflammatory cytokines, insulin resistance, and fibrosis.
FIGURE 2.

Intestinal microbiota in liver disease. Diet and alcohol influence the composition of the gut bacteria. Dysbiosis is associated with changes in bacterial metabolites such as SCFA and BAs. The gut barrier is also altered, leading to increased endotoxemia (LPS). Acetaldehyde is specifically produced by the gut bacteria in ALD. Modifications of BAs and activation of their receptors, FXR and TGR5, participate in the development of liver lesions.
Endogenous ethanol production has also been proposed as a mechanism linking the gut microbiota to NAFLD (56). Indeed, gut bacteria, including the Enterobacteriaceae family, carry out mixed-acid fermentation, a major product of which is ethanol. This microbially produced ethanol reaches the liver via the portal vein and is converted to acetate and acetaldehyde. Acetate is a fatty acid substrate, whereas acetaldehyde triggers the production of reactive oxygen species. Moreover, ethanol alters gut permeability, leading to increased endotoxemia. Thus, endogenous ethanol production may be another component that triggers the proinflammatory cascade. Higher ethanol levels have been detected in patients with histology-proven NAFLD than in healthy controls (57) and in non-alcohol-consuming adolescents with NASH than in obese or healthy controls (34). This was associated with an increased abundance of Escherichia bacteria, ethanol-producing members of the Enterobacteriaceae family.
Choline, a phospholipid component of the cell membrane, is an essential nutrient required for lipid metabolism and neurotransmitter synthesis. Diets deficient in choline lead to triglyceride accumulation in the liver and reduced hepatic secretion of very-low-density lipoproteins, resulting in liver steatosis, which is reversed after choline supplementation (58). The gut microbiota can reduce choline bioavailability, thus mimicking a choline-deficient diet. Different gut bacteria can convert choline to methylamines, which may further induce inflammation when absorbed by the liver. Indeed, the high capacity of the gut microbiota to metabolize choline into methylamines explains the high susceptibility to develop NAFLD in response to an HFD in mouse strain 129S6 (59). However, it is not currently known whether bacterial choline metabolism contributes to NAFLD susceptibility in humans.
Primary BAs (in humans, cholic acid [CA] and chenodeoxycholic acid [CDCA]) are synthesized from cholesterol in the liver and conjugated to either taurine or glycine (60). They are then excreted into the small intestine, where their deconjugation and dehydroxylation are dependent on the gut bacterial species and are therefore modified when dysbiosis occurs (61). More than 95% of the BAs secreted in bile are reabsorbed in the distal ileum and return to the liver, taking part in the enterohepatic cycle. The main function of BAs is to assist the absorption of dietary lipids and lipid-soluble nutrients. However, they are also signaling molecules through the activation of receptors, such as the FXR or Takeda G protein-coupled receptor 5 (TGR5). Thus, they may modulate the inflammatory response and lipid, glucose, energy, and drug metabolisms, as well as their own biosynthesis (62). FXR-deficient mice develop features of NAFLD, including increased steatosis and inflammation. Beneficial effects of BAs on glucose and lipid metabolism, as well as on NAFLD, have been recently reported. OCA, a synthetic FXR ligand, improves liver injuries in biopsy-proven NASH patients (25). However, less than 50% of the patients receiving OCA showed histological improvement, whereas transaminases and glucose intolerance returned to baseline levels when the treatment ceased. Dyslipidemia was a side effect of OCA administration, preventing its use as an adjuvant treatment for NAFLD. The fraction of BAs that escape enterohepatic circulation, approximately 5% (i.e., 200 to 800 mg daily in humans), pass into the colon, where they undergo bacterial metabolism leading to the production of over 20 different secondary BAs in adult human feces (63). The capacity to activate the FXR and TGR5 depends on the type of BA. The primary BA, CDCA, is the most potent, whereas secondary BAs activate the FXR and TGR5 to a much lesser extent. The metabolism of the gut microbiota may thus modulate FXR and TGR5 activation, and subsequently NAFLD susceptibility, by altering the composition of the BA pool.
Specific BAs produced by the gut microbiota can also directly affect liver health. For example, the dehydroxylation of CDCA leads to formation of lithocholic acid, which is toxic to liver cells (64), and high levels of deoxycholic acid (DCA) are associated with an increased risk of liver cancer (65). Some studies also showed that DCA can impair gut barrier function (66). Conversely, UDCA, produced by few intestinal bacteria through the epimerization of the 7α-hydroxyl group of CDCA (67), is thought to be chemopreventive and is used to treat cholesterol gallstones and primary biliary cirrhosis. UDCA is also able to reverse the effect of DCA on the gut barrier (66). Finally, the gut microbiota may also modulate host lipid metabolism and NAFLD development through the metabolism of BAs by changing their emulsification and absorption properties, which may affect fatty acid storage in the liver.
Probiotics, Prebiotics, and Symbiotics as Therapeutic Approaches
Prebiotics and probiotics are known modulators of the gut microbiota and have demonstrated beneficial effects in both animal models of NAFLD and humans. Several probiotic strains have been found to be effective in different experimental rodent models of NAFLD. For example, supplementation with Lactobacillus rhamnosus GG reduced steatosis in fructose-induced NAFLD (68). This was associated with restored gut barrier function and decreased expression of proinflammatory cytokines in the liver. Similarly, Bacteroides uniformis CECT 7771 and Bifidobacterium pseudocatenulatum CECT 7765 improved steatosis and immune defense mechanisms in HFD-fed mice (69, 70), whereas Lactobacillus casei strain Shirota was shown to protect against NASH induced by a methionine- and choline-deficient diet (71). Also, Lactobacillus paracasei F19 reduced steatosis and inflammation in a rat model of ischemia-reperfusion (72). Finally, administration of VSL#3, a preparation composed of eight bacterial strains, improved liver steatosis and insulin resistance in HFD-fed mice (73) and reduced liver inflammation and serum ALT in leptin-deficient (ob/ob) mice (74). Few randomized, prospective clinical trials have been performed in humans to assess the effect of probiotic administration on NAFLD. However, a meta-analysis of four trials involving 134 NAFLD/NASH patients provided evidence that probiotic therapies may be effective in improving NAFLD markers, despite the use of different bacterial strains and administration protocols (75). In particular, the use of probiotics was associated with lower plasma aminotransferase and total cholesterol levels, lower systemic inflammation, and improved insulin resistance.
Prebiotics, which can promote the growth of beneficial bacteria, may also improve markers of NAFLD. For example, fructo-oligosaccharide (FOS) administration reduced endotoxemia and steatosis in HFD-fed mice, probably via the restoration of Bifidobacteria and Akkermansia muciniphila populations (76, 77). Similarly, an HFD supplemented with fungal chitin-glucan was found to decrease steatosis as well as fat mass development and hyperglycemia in mice. These beneficial effects correlated with the abundance of Roseburia spp., which was decreased by the HFD and restored by the prebiotics. These changes in gut microbiota composition, due to prebiotic treatment, are thought to improve gut barrier integrity through mechanisms including the production of endogenous glucagon-like peptide 2 and endocannabinoids.
Finally, symbiotics, which consist of a mix of probiotics and prebiotics, have been recently used in patients with NAFLD. A 24-week treatment with Bifidobacterium longum and FOS (78), together with lifestyle modifications, reduced endotoxemia and serum transaminase and tumor necrosis factor alpha levels, as well as steatosis and liver inflammation (79). However, these parameters also improved in the placebo group. Similarly, a 28-week treatment with a mixture of seven bacterial strains associated with FOS had beneficial effects on hepatic inflammation and overall liver function (80).
These encouraging results strongly suggest that the use of pro-, pre-, and symbiotics may be an effective addition in the treatment of NAFLD.
ALCOHOL LIVER DISEASE
Microbiota, a Key Player in ALD
In 1995, Adachi et al. reported that antibiotics protected rats against alcohol-induced liver injury by lowering Kupffer cell activation, without linking antibiotic use to gut bacteria (81). ALD is also associated with elevated plasma endotoxin levels in alcoholic patients and rodent models of alcohol consumption (82–84), which is further explained by increased intestinal permeability (85). The disruption of the gut barrier is now largely understood (86–88) and is explained by the lower expression of several proteins of the tight junctions (89). Moreover, ethanol consumption modulates the glycosylation of mucin, which modifies the protective mucus layer and, potentially, the adherent bacterial species (90). Using germ-free mice, it was shown that the alcohol-associated increase of intestinal permeability is not induced by alcohol, per se, but at least in part by acetaldehyde produced by bacterial ethanol metabolism (91). These data do not exclude that other bacterial metabolites may be involved in the disruption of tight junctions (92). Thus, the IM participates in the disruption of the gut barrier, which may impair liver homeostasis by increasing the level of bacterial products or bacterial translocation into the blood and lymph nodes. This translocation may be particularly deleterious in the case of dysbiosis. Indeed, dysbiosis has already been described in alcohol-fed rodents (93–95). Alcohol intake in rats was associated with a decrease in the abundance of lactic acid bacteria, especially those of the genera Lactobacillus, Pediococcus, Leuconostoc, and Lactococcus (95). An increase in the abundance of Proteobacteria and Actinobacteria and decreases in Bacteroidetes and Firmicutes were described in alcohol-fed mice. Moreover, these modifications were accompanied by a decrease in the bacterial diversity (93). In alcoholic patients, dysbiosis of the colon IM was associated with a decrease in the abundance of Bacteroidetes and an increase in Enterobacteriaceae and Proteobacteria (96). These studies show that dysbiosis is associated with alcohol-induced liver lesions, but the causal role of the IM has only recently been demonstrated.
Patients with severe alcoholic hepatitis (sAH) harbor an IM different from that of alcoholic patients without liver lesions (noAH) (97). The propensity of alcohol-induced inflammation was shown to be transmissible from patients to mice via the transplantation of the IM. Germ-free mice that received the IM of an alcoholic patient with sAH developed more-severe liver lesions than mice that received the IM of an alcoholic patient without AH. They also developed more-pronounced disruption of the intestinal barrier, associated with visceral inflammation. In conventional mice, efficient IM transplantation of a human stool sample from an alcoholic patient with noAH reverted the development of liver lesions in alcohol-fed mice that initially received the IM of an alcoholic patient with sAH (97). Altogether, these findings show that individual susceptibility to ALD is dependent on the IM and provide strong evidence for a causal role of the IM in ALD.
The connections between the microbiota and their host in the development of ALD have been established, but the mechanisms that are involved in the deleterious role mediated by the IM remain unclear (98). However, metabolites produced by bacteria, such as short-chain fatty acids (SCFA) and, more importantly, volatile organic compounds (VOC) or BAs seem to be key players in ALD.
Alcohol consumption is associated with a specific mixture of VOC in stools (99). Among these VOC, SCFA propionate and isobutyrate, involved in intestinal epithelial cell homeostasis and gut barrier integrity, are less abundant in the stools of alcoholics than in those of nonalcoholic healthy individuals. BA profiles are also altered by alcohol intake in rats (100). In humans, alcohol intake induces an increase in total stool BA levels, specifically lithocholic acid and DCA, and more generally an increase in the secondary/primary BA ratio (101). However, these studies compared alcoholic and nonalcoholic individuals, and alcohol as a specific carbohydrate substrate could itself induce marked changes.
Analysis of the fecal metabolome from alcohol-fed mice has highlighted the involvement of BAs in ALD (97). Mice transplanted with the IM of a patient with severe alcoholic hepatitis developed more-severe liver lesions than those transplanted with the IM of an alcoholic patient without liver disease. Comparison of the fecal metabolites allowed the identification of 13 biomarkers related to alcohol-induced liver toxicity. The most discriminating molecules were BA derivatives and hydroxygenated/oxygenated fatty acids. Higher amounts of the primary BA, CDCA, were found in fecal samples of mice without liver lesions than in those of mice with alcohol-induced liver lesions. The secondary BA, UDCA, was more abundant in the fecal metabolome of mice that received the MI of the alcoholic patient without liver disease than of those that received the MI of the patient with ALD.
The synthesis of BAs is dependent on a negative-feedback loop following FXR activation (102, 103). This cycle also involves several transporters that take part in the secretion of BAs from the liver to the gallbladder and BA reabsorption from the ileum. Specific BAs play a role in gut barrier disruption. UDCA and tauroursodeoxycholic acid attenuate alcohol-induced liver lesions by preventing liver damage (104). UDCA has hepatoprotective properties and was a discriminant metabolite between patients with and without alcohol-induced liver lesions (97). DCA impairs the gut barrier function in the jejunum and colon, and UDCA has a protective effect on the colon against DCA-induced barrier dysfunction (66). Moreover, mice deficient for the BA receptor FXR showed an increase of liver lesions, suggesting that the disruption of BA homeostasis was involved in alcohol-induced liver lesions (105). Thus, the dysregulation of the enterohepatic cycle by IM dysbiosis may be involved in alcohol-induced liver lesions, but further studies are needed to better understand this relationship (Fig. 2).
Disruption of the gut barrier is a prerequisite for ALD, resulting in bacterial translocation (106). The mucus layer and antimicrobial peptides are essential for protecting the intestinal epithelium against bacteria, and their alteration is associated with ALD. The production of mucin by goblet cells specifically decreases in mice with alcohol-induced liver lesions depending on the IM (97). Surprisingly, mice deficient for mucin production were protected against alcohol-induced liver lesions. This deficiency was associated with a large increase in defensin production, as well as lectins Reg3β and Reg3γ, increasing the killing of commensal bacteria and preventing bacterial overgrowth (107). Moreover, defensin, Reg3β, and Reg3γ levels decreased in mice fed alcohol in a mouse model of ALD (95), and mice deficient for Reg3β and Reg3γ showed increased adherent bacteria and a high level of bacterial translocation, promoting alcohol-induced liver lesions (108). Conversely, mice that overexpressed Reg3γ were protected from alcohol-induced hepatic toxicity, associated with decreased adherent bacteria and a low-level bacterial translocation. Moreover, mice harboring a specific IM, that were protected from the hepatic toxicity of alcohol, also expressed high levels of defensins (109). These data suggest that the control of bacterial overgrowth and adherent bacterial content is essential to the protective mechanisms against alcohol-induced liver toxicity.
Recent data have emphasized the role played by the IM in the sensing of nutrients and hormones via the gastrointestinal nervous system (110–112). The increase in intestinal permeability and dysbiosis in alcoholic patients was associated with higher scores of depression, anxiety, and alcohol craving (113). Alcoholic patients with gut permeability harbored a lower abundance of genera belonging to the Ruminococcaceae family (Ruminococcus, Faecalibacterium, Subdoligranulum, Oscillibacter, and Anaerofilum), as well as clostridia, than patients with less gut permeability. Conversely, the abundance of Dorea (Lachnospiraceae), Blautia, and Megasphaera was increased in these patients. VOC produced by these members of the IM were different in alcoholic patients harboring dysbiosis and gut leakiness, especially several indole and phenol species. This suggests that metabolites produced by the IM are involved not only in cell dysfunction and liver disease but also in the psychological symptoms of alcohol dependence (113).
Prebiotics, Probiotics, or Fecal Microbiota Transfer To Treat ALD
Several probiotics have been tested in rodent models and several human trials to improve ALD progression. The first use of probiotics in ALD was performed in a rodent model of alcohol intake by using Lactobacillus sp. strain GG (Lactobacillus GG), which improved gut leakiness and liver inflammation (114–116). The addition of oat fiber or a supernatant of Lactobacillus GG induced similar results, suggesting that bacterial products were partially involved in the protective mechanisms (94, 117, 118). VSL#3, a mixture of eight probiotic strains, also improved liver lesions in ALD in humans and rodents (119, 120). Several other probiotic strains have been tested and improved liver lesions in rodents. They are well summarized in a recent review (121). Prebiotic use was also tested and showed that FOS improved rodent alcohol-induced liver damage in a mouse model of alcoholic liver disease (95).
Altogether, these studies highlight the promise of controlling the IM by pre- or probiotic treatments, but further studies are needed to decipher the mechanisms involved in bacterium-related gut and liver damage.
ADVANCED LIVER DISEASE
Fibrosis
The composition and functions of the gut microbiota are altered in patients with fibrosis (122). Patients with a fibrosis score of 2 or more harbor more Bacteroides and Ruminococcus and fewer Prevotella bacteria than patients with a fibrosis score of 0 or 1. Moreover, genes related to carbohydrate, lipid, and amino acid metabolism are overrepresented in the microbiome of patients with stage 2 fibrosis or greater, indicating a shift in the metabolic functions of the gut microbiota (122).
The bile duct ligation mouse model in combination with an HFD was used to evaluate the consequences of fibrosis on the IM. These mice developed higher hepatic stellate cell (major producers of the fibrotic matrix) activity, resulting in increased liver fibrosis. This was associated with dysbiosis, characterized by a marked increase in the abundance of Enterobacteriaceae (Gram negative) and the complete disappearance of Bifidobacterium (Gram positive) (123). The authors further selected the Gram-negative and Gram-positive fractions of the gut microbiota from the fibrogenic HFD/bile duct ligation mice and transplanted them into control mice. Mice receiving the Gram-negative fraction displayed increased liver injury, showing that dysbiosis may contribute not only to steatosis and inflammation but also to fibrogenesis in the liver (123).
Fibrosis has also been induced by injections of carbon tetrachloride (CCl4), showing that the IM was modified during the process of fibrosis development. There was a specific decrease in the abundance of Clostridium spp. and, more generally, an increase in the ratio of aerobic to anaerobic bacteria, which correlated with a higher fibrosis score (124). Bacterial DNA found in the mesenteric lymph nodes, reflecting disruption of the gut barrier, was modified in rats with chemically induced fibrosis (125). Moreover, the absence of an IM worsened fibrosis development in germ-free mice, but the immaturity of their immune system may have also played a role in the evolution of the liver damage (126). Probiotic treatments, such as use of Saccharomyces cerevisiae subsp. boulardii, were associated with a slower progression of liver fibrosis (127), and VSL#3 prevented bacterial translocation (128). However, it is not possible to conclude whether these treatments played a role early in the inflammation stage to limit the progression to fibrosis.
Cirrhosis
Patients with cirrhosis have distinct fecal microbial communities relative to healthy individuals. Members of the phylum Bacteroidetes were less abundant in cirrhotic patients (24 hepatitis B virus [HBV] and 12 alcohol-related cirrhosis patients), whereas Proteobacteria and Fusobacteria were more abundant (129) (Table 3). Among cirrhotic patients, the abundance of Prevotellaceae was greater in alcoholic patients than those infected with HBV. Moreover, the prevalence of potentially pathogenic bacteria, such as Enterobacteriaceae and Streptococcaceae, and the reduction of beneficial populations, such as Lachnospiraceae, in patients with cirrhosis may affect prognosis (129). In a more recent study, the functional diversity of the IM was shown to be significantly lower in patients with hepatitis B-related cirrhosis than in controls (130). Moreover, 54% of the cirrhotic patient enriched taxonomically assigned species are of buccal origin, suggesting an invasion of the gut from the mouth in liver cirrhosis (130).
TABLE 3.
Comparison of healthy microbiotas and microbiotas associated with cirrhosisa
| Phylum | Family | Genus | Reference |
|---|---|---|---|
| Bacteroidetes ⇩ | Healthy (n = 24)Cirrhosis (n = 36: HBV = 24, Alc = 12)Chen et al., 2011 (129) | ||
| [Firmicutes] | Veillonellaceae ⇧ | ||
| Streptococcaceae ⇧ | |||
| Lachnospiraceae ⇩ | |||
| Proteobacteria ⇧ | Enterobacteriaceae ⇧ | ||
| Fusobacteria ⇧ | |||
| [Bacteroidetes] | Rikenellaceae ⇩ | Healthy (n = 14)Cirrhosis (n = 47)Kakiyama et al., 2013 (101) | |
| [Firmicutes] | Veillonellaceae ⇧ | Blautia ⇩ | |
| Ruminococcaceae ⇩ | |||
| Lachnospiraceae ⇩ | |||
| [Proteobacteria] | Enterobacteriaceae ⇧ | ||
| [Bacteroidetes] | Porphyromonadaceae ⇩ | Healthy (n = 25)Cirrhosis (n = 175: HCV = 75, Alc = 31, others = 71)Bajaj et al., 2014 (132, 137) | |
| [Firmicutes] | Veillonellaceae ⇩ | ||
| Ruminococcaceae ⇩ | |||
| Lachnospiraceae ⇩ | |||
| Clostridiales XIV ⇩ | |||
| Enterococcaceae ⇧ | |||
| Staphylococcaceae ⇧ | |||
| [Proteobacteria] | Enterobacteriaceae ⇧ | ||
| Bacteroidetes ⇩ | [Bacteroidaceae] | Bacteroides ⇩ | Healthy (n = 83)Cirrhosis (n = 98: HBV = 79, Alc = 10, others = 9)Qin et al., 2014 (130) |
| [Prevotellaceae] | Prevotella ⇧ | ||
| [Rikenellaceae] | Alistipes ⇩ | ||
| [Firmicutes] | [Veillonellaceae] | Veillonella ⇧ | |
| [Streptococcaceae] | Streptococcus ⇧ | ||
| [Clostridiaceae] | Clostridium ⇧ | ||
| [Lachnospiraceae] | Eubacterium ⇩ | ||
| Proteobacteria ⇧ | |||
| Fusobacteria ⇧ |
Microbiota were analyzed using 16S deep sequencing, except in the study of Qin et al., which used metagenomics. Additional taxonomic information for discriminating taxa is in brackets. Taxa in boldface are those identified in more than one study.
The gut microbiome has been assessed in cirrhotic patients and compared to that of healthy individuals by metagenomic studies. The modified metabolic pathways showed an enrichment of genes involved in gluconeogenesis and lipid metabolism in the IM of cirrhotic patients and a decrease of genes involved in BA metabolism (131). Another study revealed specific clusters representing cognate bacterial species; 28 were enriched in cirrhotic patients and 38 in control individuals. Among bacterial genes, those involved in denitrification, assimilation or dissimilation of nitrate, gamma-aminobutyric acid biosynthesis, heme biosynthesis, and phosphotransferase systems were found to be associated with liver cirrhosis (130). Genetic and functional biomarkers specific for liver cirrhosis were revealed by a comparison with those for type 2 diabetes and inflammatory bowel disease. Fifteen biomarkers were specifically associated with cirrhosis. However, generalization of these results is difficult, as the number of patients was low and the cause of cirrhosis was restricted mainly to HBV infection (130).
Most cirrhotic patients are asymptomatic until they develop decompensated cirrhosis. The survival rate of cirrhotic patients is dependent on the associated complications, including episodes of hepatic encephalopathy (HE), ascites, or infections, which have been associated with specific bacterial communities (132).
In cirrhotic patients, HE was associated with a specific dysbiosis: a high level of Enterobacteriaceae, already associated with infection in decompensated cirrhosis, was also associated with HE (133). Alcaligenaceae, bacteria that are specifically involved in urea metabolism, were also more abundant in HE patients. In a prospective clinical trial, VSL#3 was found to be efficient in preventing hepatic encephalopathy in cirrhotic patients, suggesting that the dysbiosis in HE may have a causative role (134). Conversely, this treatment did not affect the portal pressure (135).
A cirrhosis dysbiosis ratio (CDR) has been developed to evaluate the severity of cirrhosis (132). The CDR is a ratio between the abundance of Clostridiales (Clostridium cluster XIV), Ruminococcaceae, and Lachnospiraceae and that of Bacteroidaceae and Enterobacteriaceae. Healthy patients have a CDR of approximately 2, decreasing to 0.9 in patients with compensated cirrhosis and to 0.3 in cirrhotic patients with an infection. Enterobacteriaceae is the most prevalent family modified in the CDR and is specifically associated with the complications of cirrhosis due to the production of potent endotoxins. The level of fecal Bacteroidaceae and Clostridium cluster XIV may predict the risk of hospitalization in patients with cirrhosis, independently of other classical clinical predictors, such as the MELD (model for end-stage liver disease) score, HE, or the intake of proton pump inhibitors (136). These data prove the relationship between dysbiosis and the severity of cirrhosis. Moreover, a randomized clinical trial using Lactobacillus GG in cirrhotic patients demonstrated that altering the composition of the IM through the use of probiotics improved dysbiosis and endotoxemia (137).
Viable bacteria have been found in the ascites of patients with decompensated cirrhosis, including several species of Propionibacterium, Pseudomonas, and Staphylococcus recognized as commonly colonizing the skin (138). Thus, access to the peritoneal cavity is not limited to the translocation of bacteria from the gut. The quantity of bacteria in the ascites is not sufficient to induce spontaneous bacterial peritonitis, but there is a relationship between the bacterial species and the severity of the disease (138).
Bacterial metabolites are related to the abundance of the bacterial species and may have beneficial or deleterious effects. Secondary BAs are exclusively produced by the IM. Cirrhosis has been recently shown to be associated with the abundance of specific bacterial families and to be linked with a decrease in the amount of secondary BAs in the feces (101, 132). Moreover, BA levels can discriminate between patients with severe alcoholic hepatitis associated with cirrhosis and alcoholic patients without liver lesions (97). Further studies are needed to decipher the spectra of metabolites that mediate the deleterious effects of bacterial dysbiosis.
Hepatocellular Carcinoma
HCC occurs in the presence of chronic liver inflammation, typically in patients with cirrhosis (139). To understand the contribution of NAFLD or ALD to the development of HCC, other causes of liver disease need to be excluded, especially HBV or HCV infection. TLR4 deficiency resulted in decreased promotion of HCC but did not affect HCC initiation in chemically induced HCC in adult mice, either by diethylnitrosamine (DEN) alone or by DEN and chronic CCL4 injections (140, 141). Sterilization of the gut by antibiotherapy decreased both HCC initiation and progression. Accordingly, germ-free mice showed an approximately 80% reduction of HCC (140). Altogether, these data show that the inhibition of the LPS/TLR4 pathway prevents HCC progression. However, opposite results were obtained in another model of HCC, using TLR4-deficient mice in which HCC was induced by the unique injection of DEN, 15 days after birth (142). These discrepancies can be explained by the high frequency of DNA mutations in this DEN model relative to DEN injections in adult rodents associated with the development of a chronic liver inflammation. In humans, only indirect evidence has been published and showed that TLR2, -4, and -9 overexpression was associated with a poor prognosis for HCC progression (143).
The role of the IM was recently assessed in obesity-associated HCC. HCC was chemically induced in mice using DMBA [7,12-dimethylbenz(a)anthracene] at the neonatal stage. The association of DMBA and HFD induced tumor development in the mice (65). The analysis of serum metabolites showed an increase in DCA levels in mice that developed tumors. The production of the secondary BA DCA by the IM was associated with the presence of Clostridium species that harbored a 7α-dehydroxylase function to metabolize primary BAs (144). DCA levels were lowered using UDCA or by chemical inhibition of 7α-dehydroxylase. Moreover, vancomycin, which targets Gram-positive bacteria, especially Clostridium, also decreased serum DCA levels. Under these conditions, lowering serum DCA levels reduced HCC development.
These findings provide new evidence that the IM could be involved in the initiation and progression of HCC.
CONCLUSION
The complex community of the intestinal bacteria influences both the normal and pathological states of the liver, and we are only beginning to understand the mechanisms involved in this relationship. In dysbiosis of the gut microbiota associated with metabolic or alcoholic liver diseases, the changes in bacterial gene expression or metabolite production may be more important than the changes in bacterial composition. Understanding the clinical significance of gut microbiota dysbiosis in the context of liver disease remains a challenge and must consider the possible association between host genetic or/and epigenetic changes that favor liver injury. Further studies are needed, combining metagenomics, metatranscriptomics, and metabolomics with longitudinal studies, along with standardization of these techniques, to better comprehend the role of microbiota changes in the development of liver disease.
Many studies already strongly suggest that targeting the IM through the use of pro-, pre-, and symbiotics, and possibly fecal transfer, may be an effective treatment for NAFLD and ALD. However, larger studies incorporating liver biopsies, standardized dose administration and duration, and an analysis of the impact on the gut microbiota are clearly needed. This should help to open new treatment strategies based on the modulation of the gut microbiota.
Contributor Information
Anne-Marie Cassard, INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud, Université Paris-Saclay, 92140 Clamart, France.
Philippe Gérard, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.
Gabriel Perlemuter, INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud, Université Paris-Saclay, 92140 Clamart, France; AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France.
Robert Allen Britton, Baylor College of Medicine, Houston, TX.
Patrice D. Cani, Université catholique de Louvain, Brussels, Belgium
REFERENCES
- 1.Perlemuter G, Bigorgne A, Cassard-Doulcier AM, Naveau S. 2007. Nonalcoholic fatty liver disease: from pathogenesis to patient care. Nat Clin Pract Endocrinol Metab 3:458–469. 10.1038/ncpendmet0505 [PubMed] [DOI] [PubMed] [Google Scholar]
- 2.Voican CS, Perlemuter G, Naveau S. 2011. Mechanisms of the inflammatory reaction implicated in alcoholic hepatitis: 2011 update. Clin Res Hepatol Gastroenterol 35:465–474. 10.1016/j.clinre.2011.01.017 [DOI] [PubMed] [Google Scholar]
- 3.Voican CS, Njiké-Nakseu M, Boujedidi H, Barri-Ova N, Bouchet-Delbos L, Agostini H, Maitre S, Prévot S, Cassard-Doulcier AM, Naveau S, Perlemuter G. 2015. Alcohol withdrawal alleviates adipose tissue inflammation in patients with alcoholic liver disease. Liver Int 35:967–978. 10.1111/liv.12575 [PubMed] [DOI] [PubMed] [Google Scholar]
- 4.Hardy T, Oakley F, Anstee QM, Day CP. 2016. Nonalcoholic fatty liver disease: pathogenesis and disease spectrum. Annu Rev Pathol 11:451–496. 10.1146/annurev-pathol-012615-044224 [PubMed] [DOI] [PubMed] [Google Scholar]
- 5.Bedossa P. 2013. Current histological classification of NAFLD: strength and limitations. Hepatol Int 7(Suppl 2):765–770. 10.1007/s12072-013-9446-z [PubMed] [DOI] [PubMed] [Google Scholar]
- 6.European Association for the Study of the Liver--European Organisation for Research and Treatment of Cancer. 2012. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol 56:908–943. 10.1016/j.jhep.2011.12.001 [PubMed] [DOI] [PubMed] [Google Scholar]
- 7.Paradis V, Zalinski S, Chelbi E, Guedj N, Degos F, Vilgrain V, Bedossa P, Belghiti J. 2009. Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: a pathological analysis. Hepatology 49:851–859. 10.1002/hep.22734 [PubMed] [DOI] [PubMed] [Google Scholar]
- 8.Dugum M, McCullough A. 2015. Diagnosis and management of alcoholic liver disease. J Clin Transl Hepatol 3:109–116. 10.14218/JCTH.2015.00008 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Naveau S, Cassard-Doulcier AM, Njiké-Nakseu M, Bouchet-Delbos L, Barri-Ova N, Boujedidi H, Dauvois B, Balian A, Maitre S, Prévot S, Dagher I, Agostini H, Grangeot-Keros L, Emilie D, Perlemuter G. 2010. Harmful effect of adipose tissue on liver lesions in patients with alcoholic liver disease. J Hepatol 52:895–902. 10.1016/j.jhep.2010.01.029 [PubMed] [DOI] [PubMed] [Google Scholar]
- 10.Saadeh S, Younossi ZM, Remer EM, Gramlich T, Ong JP, Hurley M, Mullen KD, Cooper JN, Sheridan MJ. 2002. The utility of radiological imaging in nonalcoholic fatty liver disease. Gastroenterology 123:745–750. 10.1053/gast.2002.35354 [PubMed] [DOI] [PubMed] [Google Scholar]
- 11.Musso G. 2011. The Finnish Diabetes Risk Score (FINDRISC) and other non-invasive scores for screening of hepatic steatosis and associated cardiometabolic risk. Ann Med 43:413–417. 10.3109/07853890.2011.579152 [PubMed] [DOI] [PubMed] [Google Scholar]
- 12.Ekstedt M, Franzén LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, Kechagias S. 2006. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology 44:865–873. 10.1002/hep.21327 [PubMed] [DOI] [PubMed] [Google Scholar]
- 13.Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, Angulo P. 2005. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology 129:113–121. 10.1053/j.gastro.2005.04.014 [PubMed] [DOI] [PubMed] [Google Scholar]
- 14.Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, Mills PR, Keach JC, Lafferty HD, Stahler A, Haflidadottir S, Bendtsen F. 2015. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 149:389–397.e10. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McPherson S, Hardy T, Henderson E, Burt AD, Day CP, Anstee QM. 2015. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: implications for prognosis and clinical management. J Hepatol 62:1148–1155. 10.1016/j.jhep.2014.11.034 [PubMed] [DOI] [PubMed] [Google Scholar]
- 16.Ratziu V, Giral P, Charlotte F, Bruckert E, Thibault V, Theodorou I, Khalil L, Turpin G, Opolon P, Poynard T. 2000. Liver fibrosis in overweight patients. Gastroenterology 118:1117–1123. 10.1016/S0016-5085(00)70364-7 [PubMed] [DOI] [PubMed] [Google Scholar]
- 17.Ertle J, Dechêne A, Sowa JP, Penndorf V, Herzer K, Kaiser G, Schlaak JF, Gerken G, Syn WK, Canbay A. 2011. Non-alcoholic fatty liver disease progresses to hepatocellular carcinoma in the absence of apparent cirrhosis. Int J Cancer 128:2436–2443. 10.1002/ijc.25797 [PubMed] [DOI] [PubMed] [Google Scholar]
- 18.Promrat K, Kleiner DE, Niemeier HM, Jackvony E, Kearns M, Wands JR, Fava JL, Wing RR. 2010. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology 51:121–129. 10.1002/hep.23276 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kistler KD, Brunt EM, Clark JM, Diehl AM, Sallis JF, Schwimmer JB, NASH CRN Research Group. 2011. Physical activity recommendations, exercise intensity, and histological severity of nonalcoholic fatty liver disease. Am J Gastroenterol 106:460–468, quiz 469. 10.1038/ajg.2010.488 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wu T, Gao X, Chen M, van Dam RM. 2009. Long-term effectiveness of diet-plus-exercise interventions vs. diet-only interventions for weight loss: a meta-analysis. Obes Rev 10:313–323. 10.1111/j.1467-789X.2008.00547.x [PubMed] [DOI] [PubMed] [Google Scholar]
- 21.Hardy T, Anstee QM, Day CP. 2015. Nonalcoholic fatty liver disease: new treatments. Curr Opin Gastroenterol 31:175–183. 10.1097/MOG.0000000000000175 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mackenzie TA, Zaha R, Smith J, Karagas MR, Morden NE. 2016. Diabetes pharmacotherapies and bladder cancer: a medicare epidemiologic study. Diabetes Ther 7:61–73. 10.1007/s13300-016-0152-4 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, Bae JW. 2014. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63:727–735. 10.1136/gutjnl-2012-303839 [PubMed] [DOI] [PubMed] [Google Scholar]
- 24.Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR, NASH Clinical Research Network. 2010. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med 362:1675–1685. 10.1056/NEJMoa0907929 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van Natta ML, Abdelmalek MF, Chalasani N, Dasarathy S, Diehl AM, Hameed B, Kowdley KV, McCullough A, Terrault N, Clark JM, Tonascia J, Brunt EM, Kleiner DE, Doo E, NASH Clinical Research Network. 2015. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet 385:956–965. 10.1016/S0140-6736(14)61933-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lassailly G, Caiazzo R, Buob D, Pigeyre M, Verkindt H, Labreuche J, Raverdy V, Leteurtre E, Dharancy S, Louvet A, Romon M, Duhamel A, Pattou F, Mathurin P. 2015. Bariatric surgery reduces features of nonalcoholic steatohepatitis in morbidly obese patients. Gastroenterology 149:379–388; quiz e315–376. [DOI] [PubMed] [Google Scholar]
- 27.Thursz MR, Forrest EH, Ryder S, STOPAH Investigators. 2015. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 373:282–283. [PubMed] [DOI] [PubMed] [Google Scholar]
- 28.Mathurin P, Louvet A, Duhamel A, Nahon P, Carbonell N, Boursier J, Anty R, Diaz E, Thabut D, Moirand R, Lebrec D, Moreno C, Talbodec N, Paupard T, Naveau S, Silvain C, Pageaux GP, Sobesky R, Canva-Delcambre V, Dharancy S, Salleron J, Dao T. 2013. Prednisolone with vs without pentoxifylline and survival of patients with severe alcoholic hepatitis: a randomized clinical trial. JAMA 310:1033–1041. 10.1001/jama.2013.276300 [PubMed] [DOI] [PubMed] [Google Scholar]
- 29.Thursz MR, Richardson P, Allison M, Austin A, Bowers M, Day CP, Downs N, Gleeson D, MacGilchrist A, Grant A, Hood S, Masson S, McCune A, Mellor J, O’Grady J, Patch D, Ratcliffe I, Roderick P, Stanton L, Vergis N, Wright M, Ryder S, Forrest EH, STOPAH Trial. 2015. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 372:1619–1628. 10.1056/NEJMoa1412278 [PubMed] [DOI] [PubMed] [Google Scholar]
- 30.Nguyen-Khac E, Thevenot T, Piquet MA, Benferhat S, Goria O, Chatelain D, Tramier B, Dewaele F, Ghrib S, Rudler M, Carbonell N, Tossou H, Bental A, Bernard-Chabert B, Dupas JL, AAH-NAC Study Group. 2011. Glucocorticoids plus N-acetylcysteine in severe alcoholic hepatitis. N Engl J Med 365:1781–1789. 10.1056/NEJMoa1101214 [PubMed] [DOI] [PubMed] [Google Scholar]
- 31.Thursz M, Morgan TR. 2016. Treatment of severe alcoholic hepatitis. Gastroenterology 150:1823–1834. 10.1053/j.gastro.2016.02.074 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mathurin P, Moreno C, Samuel D, Dumortier J, Salleron J, Durand F, Castel H, Duhamel A, Pageaux GP, Leroy V, Dharancy S, Louvet A, Boleslawski E, Lucidi V, Gustot T, Francoz C, Letoublon C, Castaing D, Belghiti J, Donckier V, Pruvot FR, Duclos-Vallée JC. 2011. Early liver transplantation for severe alcoholic hepatitis. N Engl J Med 365:1790–1800. 10.1056/NEJMoa1105703 [PubMed] [DOI] [PubMed] [Google Scholar]
- 33.Zeng H, Liu J, Jackson MI, Zhao FQ, Yan L, Combs GF Jr. 2013. Fatty liver accompanies an increase in lactobacillus species in the hind gut of C57BL/6 mice fed a high-fat diet. J Nutr 143:627–631. 10.3945/jn.112.172460 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR. 2013. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57:601–609. 10.1002/hep.26093 [PubMed] [DOI] [PubMed] [Google Scholar]
- 35.Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, Greenwood R, Sikaroodi M, Lam V, Crotty P, Bailey J, Myers RP, Rioux KP. 2013. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 11:868–875.e1–3. [PubMed] [DOI] [PubMed] [Google Scholar]
- 36.Jiang W, Wu N, Wang X, Chi Y, Zhang Y, Qiu X, Hu Y, Li J, Liu Y. 2015. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep 5:8096. 10.1038/srep08096 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK, Fischer SE, McGilvray ID, Allard JP. 2013. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 58:120–127. 10.1002/hep.26319 [PubMed] [DOI] [PubMed] [Google Scholar]
- 38.Wong VW, Tse CH, Lam TT, Wong GL, Chim AM, Chu WC, Yeung DK, Law PT, Kwan HS, Yu J, Sung JJ, Chan HL. 2013. Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis—a longitudinal study. PLoS One 8:e62885. 10.1371/journal.pone.0062885 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang B, Jiang X, Cao M, Ge J, Bao Q, Tang L, Chen Y, Li L. 2016. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non-alcoholic fatty liver disease. Sci Rep 6:32002. 10.1038/srep32002 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA. 2011. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 140:976–986. 10.1053/j.gastro.2010.11.049 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gérard P. 2016. Gut microbiota and obesity. Cell Mol Life Sci 73:147–162. 10.1007/s00018-015-2061-5 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang ZK, Yang YS, Chen Y, Yuan J, Sun G, Peng LH. 2014. Intestinal microbiota pathogenesis and fecal microbiota transplantation for inflammatory bowel disease. World J Gastroenterol 20:14805–14820. 10.3748/wjg.v20.i40.14805 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Claus SP, Ellero SL, Berger B, Krause L, Bruttin A, Molina J, Paris A, Want EJ, de Waziers I, Cloarec O, Richards SE, Wang Y, Dumas ME, Ross A, Rezzi S, Kochhar S, Van Bladeren P, Lindon JC, Holmes E, Nicholson JK. 2011. Colonization-induced host-gut microbial metabolic interaction. mBio 2:e00271-10. 10.1128/mBio.00271-10 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, Camporez JP, Shulman GI, Gordon JI, Hoffman HM, Flavell RA. 2012. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482:179–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, Martin P, Philippe C, Walker F, Bado A, Perlemuter G, Cassard-Doulcier AM, Gérard P. 2013. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 62:1787–1794. 10.1136/gutjnl-2012-303816 [PubMed] [DOI] [PubMed] [Google Scholar]
- 46.Ferolla SM, Armiliato GN, Couto CA, Ferrari TC. 2014. The role of intestinal bacteria overgrowth in obesity-related nonalcoholic fatty liver disease. Nutrients 6:5583–5599. 10.3390/nu6125583 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, Mascianà R, Forgione A, Gabrieli ML, Perotti G, Vecchio FM, Rapaccini G, Gasbarrini G, Day CP, Grieco A. 2009. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 49:1877–1887. 10.1002/hep.22848 [PubMed] [DOI] [PubMed] [Google Scholar]
- 48.Giorgio V, Miele L, Principessa L, Ferretti F, Villa MP, Negro V, Grieco A, Alisi A, Nobili V. 2014. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig Liver Dis 46:556–560. 10.1016/j.dld.2014.02.010 [PubMed] [DOI] [PubMed] [Google Scholar]
- 49.Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, Waget A, Delmée E, Cousin B, Sulpice T, Chamontin B, Ferrières J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, Burcelin R. 2007. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56:1761–1772. 10.2337/db06-1491 [PubMed] [DOI] [PubMed] [Google Scholar]
- 50.Poggi M, Bastelica D, Gual P, Iglesias MA, Gremeaux T, Knauf C, Peiretti F, Verdier M, Juhan-Vague I, Tanti JF, Burcelin R, Alessi MC. 2007. C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia 50:1267–1276. 10.1007/s00125-007-0654-8 [PubMed] [DOI] [PubMed] [Google Scholar]
- 51.Ye D, Li FY, Lam KS, Li H, Jia W, Wang Y, Man K, Lo CM, Li X, Xu A. 2012. Toll-like receptor-4 mediates obesity-induced non-alcoholic steatohepatitis through activation of X-box binding protein-1 in mice. Gut 61:1058–1067. 10.1136/gutjnl-2011-300269 [PubMed] [DOI] [PubMed] [Google Scholar]
- 52.Laugerette F, Vors C, Géloën A, Chauvin MA, Soulage C, Lambert-Porcheron S, Peretti N, Alligier M, Burcelin R, Laville M, Vidal H, Michalski MC. 2011. Emulsified lipids increase endotoxemia: possible role in early postprandial low-grade inflammation. J Nutr Biochem 22:53–59. 10.1016/j.jnutbio.2009.11.011 [PubMed] [DOI] [PubMed] [Google Scholar]
- 53.Harte AL, da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd EM, Tripathi G, Ashour E, Abdalla MS, Sharada HM, Amin AI, Burt AD, Kumar S, Day CP, McTernan PG. 2010. Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflamm (Lond) 7:15. 10.1186/1476-9255-7-15 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Verdam FJ, Rensen SS, Driessen A, Greve JW, Buurman WA. 2011. Novel evidence for chronic exposure to endotoxin in human nonalcoholic steatohepatitis. J Clin Gastroenterol 45:149–152. 10.1097/MCG.0b013e3181e12c24 [PubMed] [DOI] [PubMed] [Google Scholar]
- 55.Yuan J, Baker SS, Liu W, Alkhouri R, Baker RD, Xie J, Ji G, Zhu L. 2014. Endotoxemia unrequired in the pathogenesis of pediatric nonalcoholic steatohepatitis. J Gastroenterol Hepatol 29:1292–1298. 10.1111/jgh.12510 [PubMed] [DOI] [PubMed] [Google Scholar]
- 56.de Medeiros IC, de Lima JG. 2015. Is nonalcoholic fatty liver disease an endogenous alcoholic fatty liver disease? - A mechanistic hypothesis. Med Hypotheses 85:148–152. 10.1016/j.mehy.2015.04.021 [PubMed] [DOI] [PubMed] [Google Scholar]
- 57.Volynets V, Küper MA, Strahl S, Maier IB, Spruss A, Wagnerberger S, Königsrainer A, Bischoff SC, Bergheim I. 2012. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci 57:1932–1941. 10.1007/s10620-012-2112-9 [PubMed] [DOI] [PubMed] [Google Scholar]
- 58.Buchman AL, Dubin MD, Moukarzel AA, Jenden DJ, Roch M, Rice KM, Gornbein J, Ament ME. 1995. Choline deficiency: a cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology 22:1399–1403. [PubMed] [PubMed] [Google Scholar]
- 59.Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, Fearnside J, Tatoud R, Blanc V, Lindon JC, Mitchell SC, Holmes E, McCarthy MI, Scott J, Gauguier D, Nicholson JK. 2006. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci USA 103:12511–12516. 10.1073/pnas.0601056103 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hofmann AF, Hagey LR, Krasowski MD. 2010. Bile salts of vertebrates: structural variation and possible evolutionary significance. J Lipid Res 51:226–246. 10.1194/jlr.R000042 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ridlon JM, Kang DJ, Hylemon PB. 2006. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47:241–259. 10.1194/jlr.R500013-JLR200 [PubMed] [DOI] [PubMed] [Google Scholar]
- 62.Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. 2009. Bile acids as regulatory molecules. J Lipid Res 50:1509–1520. 10.1194/jlr.R900007-JLR200 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gérard P. 2013. Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens 3:14–24. 10.3390/pathogens3010014 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hofmann AF. 2004. Detoxification of lithocholic acid, a toxic bile acid: relevance to drug hepatotoxicity. Drug Metab Rev 36:703–722. 10.1081/DMR-200033475 [PubMed] [DOI] [PubMed] [Google Scholar]
- 65.Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, Honda K, Ishikawa Y, Hara E, Ohtani N. 2013. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499:97–101. 10.1038/nature12347 [PubMed] [DOI] [PubMed] [Google Scholar]
- 66.Stenman LK, Holma R, Forsgård R, Gylling H, Korpela R. 2013. Higher fecal bile acid hydrophobicity is associated with exacerbation of dextran sodium sulfate colitis in mice. J Nutr 143:1691–1697. 10.3945/jn.113.180810 [PubMed] [DOI] [PubMed] [Google Scholar]
- 67.Lepercq P, Gérard P, Béguet F, Raibaud P, Grill JP, Relano P, Cayuela C, Juste C. 2004. Epimerization of chenodeoxycholic acid to ursodeoxycholic acid by Clostridium baratii isolated from human feces. FEMS Microbiol Lett 235:65–72. 10.1111/j.1574-6968.2004.tb09568.x [PubMed] [DOI] [PubMed] [Google Scholar]
- 68.Ritze Y, Bárdos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A, Bischoff SC. 2014. Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice. PLoS One 9:e80169. 10.1371/journal.pone.0080169 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cano PG, Santacruz A, Trejo FM, Sanz Y. 2013. Bifidobacterium CECT 7765 improves metabolic and immunological alterations associated with obesity in high-fat diet-fed mice. Obesity (Silver Spring) 21:2310–2321. 10.1002/oby.20330 [PubMed] [DOI] [PubMed] [Google Scholar]
- 70.Gauffin Cano P, Santacruz A, Moya Á, Sanz Y. 2012. Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity. PLoS One 7:e41079. 10.1371/journal.pone.0041079 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Okubo H, Sakoda H, Kushiyama A, Fujishiro M, Nakatsu Y, Fukushima T, Matsunaga Y, Kamata H, Asahara T, Yoshida Y, Chonan O, Iwashita M, Nishimura F, Asano T. 2013. Lactobacillus casei strain Shirota protects against nonalcoholic steatohepatitis development in a rodent model. Am J Physiol Gastrointest Liver Physiol 305:G911–G918. 10.1152/ajpgi.00225.2013 [PubMed] [DOI] [PubMed] [Google Scholar]
- 72.Nardone G, Compare D, Liguori E, Di Mauro V, Rocco A, Barone M, Napoli A, Lapi D, Iovene MR, Colantuoni A. 2010. Protective effects of Lactobacillus paracasei F19 in a rat model of oxidative and metabolic hepatic injury. Am J Physiol Gastrointest Liver Physiol 299:G669–G676. 10.1152/ajpgi.00188.2010 [PubMed] [DOI] [PubMed] [Google Scholar]
- 73.Ma X, Hua J, Li Z. 2008. Probiotics improve high fat diet-induced hepatic steatosis and insulin resistance by increasing hepatic NKT cells. J Hepatol 49:821–830. 10.1016/j.jhep.2008.05.025 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, Desimone C, Song XY, Diehl AM. 2003. Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology 37:343–350. 10.1053/jhep.2003.50048 [PubMed] [DOI] [PubMed] [Google Scholar]
- 75.Ma YY, Li L, Yu CH, Shen Z, Chen LH, Li YM. 2013. Effects of probiotics on nonalcoholic fatty liver disease: a meta-analysis. World J Gastroenterol 19:6911–6918. 10.3748/wjg.v19.i40.6911 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, de Vos WM, Cani PD. 2013. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA 110:9066–9071. 10.1073/pnas.1219451110 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pachikian BD, Essaghir A, Demoulin JB, Catry E, Neyrinck AM, Dewulf EM, Sohet FM, Portois L, Clerbaux LA, Carpentier YA, Possemiers S, Bommer GT, Cani PD, Delzenne NM. 2013. Prebiotic approach alleviates hepatic steatosis: implication of fatty acid oxidative and cholesterol synthesis pathways. Mol Nutr Food Res 57:347–359. 10.1002/mnfr.201200364 [PubMed] [DOI] [PubMed] [Google Scholar]
- 78.Foschini MP, Macchia S, Losi L, Dei Tos AP, Pasquinelli G, Di Tommaso L, Del Duca S, Roncaroli F, Dal Monte PR. 1998. Identification of mitochondria in liver biopsies. A study by immunohistochemistry, immunogold and Western blot analysis. Virchows Arch 433:267–273. 10.1007/s004280050246 [DOI] [PubMed] [Google Scholar]
- 79.Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, Mastrojeni S, Malaguarnera G, Mistretta A, Li Volti G, Galvano F. 2012. Bifidobacterium longum with fructo-oligosaccharides in patients with non alcoholic steatohepatitis. Dig Dis Sci 57:545–553. 10.1007/s10620-011-1887-4 [PubMed] [DOI] [PubMed] [Google Scholar]
- 80.Eslamparast T, Poustchi H, Zamani F, Sharafkhah M, Malekzadeh R, Hekmatdoost A. 2014. Synbiotic supplementation in nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled pilot study. Am J Clin Nutr 99:535–542. 10.3945/ajcn.113.068890 [PubMed] [DOI] [PubMed] [Google Scholar]
- 81.Adachi Y, Moore LE, Bradford BU, Gao W, Thurman RG. 1995. Antibiotics prevent liver injury in rats following long-term exposure to ethanol. Gastroenterology 108:218–224. 10.1016/0016-5085(95)90027-6 [DOI] [PubMed] [Google Scholar]
- 82.Bode C, Kugler V, Bode JC. 1987. Endotoxemia in patients with alcoholic and non-alcoholic cirrhosis and in subjects with no evidence of chronic liver disease following acute alcohol excess. J Hepatol 4:8–14. 10.1016/S0168-8278(87)80003-X [DOI] [PubMed] [Google Scholar]
- 83.Mathurin P, Deng QG, Keshavarzian A, Choudhary S, Holmes EW, Tsukamoto H. 2000. Exacerbation of alcoholic liver injury by enteral endotoxin in rats. Hepatology 32:1008–1017. 10.1053/jhep.2000.19621 [DOI] [PubMed] [Google Scholar]
- 84.Nanji AA, Khettry U, Sadrzadeh SM, Yamanaka T. 1993. Severity of liver injury in experimental alcoholic liver disease. Correlation with plasma endotoxin, prostaglandin E2, leukotriene B4, and thromboxane B2. Am J Pathol 142:367–373. [PubMed] [PMC free article] [PubMed] [Google Scholar]
- 85.Keshavarzian A, Fields JZ, Vaeth J, Holmes EW. 1994. The differing effects of acute and chronic alcohol on gastric and intestinal permeability. Am J Gastroenterol 89:2205–2211. [PubMed] [PubMed] [Google Scholar]
- 86.Parlesak A, Schäfer C, Schütz T, Bode JC, Bode C. 2000. Increased intestinal permeability to macromolecules and endotoxemia in patients with chronic alcohol abuse in different stages of alcohol-induced liver disease. J Hepatol 32:742–747. 10.1016/S0168-8278(00)80242-1 [DOI] [PubMed] [Google Scholar]
- 87.Rao R. 2009. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 50:638–644. 10.1002/hep.23009 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Elamin EE, Masclee AA, Dekker J, Jonkers DM. 2013. Ethanol metabolism and its effects on the intestinal epithelial barrier. Nutr Rev 71:483–499. 10.1111/nure.12027 [PubMed] [DOI] [PubMed] [Google Scholar]
- 89.Zhong W, Li Q, Xie G, Sun X, Tan X, Sun X, Jia W, Zhou Z. 2013. Dietary fat sources differentially modulate intestinal barrier and hepatic inflammation in alcohol-induced liver injury in rats. Am J Physiol Gastrointest Liver Physiol 305:G919–G932. 10.1152/ajpgi.00226.2013 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Grewal RK, Mahmood A. 2009. Ethanol induced changes in glycosylation of mucins in rat intestine. Ann Gastroenterol 22:178–183. [Google Scholar]
- 91.Ferrier L, Bérard F, Debrauwer L, Chabo C, Langella P, Buéno L, Fioramonti J. 2006. Impairment of the intestinal barrier by ethanol involves enteric microflora and mast cell activation in rodents. Am J Pathol 168:1148–1154. 10.2353/ajpath.2006.050617 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. 2011. Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 141:769–776. 10.3945/jn.110.135657 [PubMed] [DOI] [PubMed] [Google Scholar]
- 93.Bull-Otterson L, Feng W, Kirpich I, Wang Y, Qin X, Liu Y, Gobejishvili L, Joshi-Barve S, Ayvaz T, Petrosino J, Kong M, Barker D, McClain C, Barve S. 2013. Metagenomic analyses of alcohol induced pathogenic alterations in the intestinal microbiome and the effect of Lactobacillus rhamnosus GG treatment. PLoS One 8:e53028. 10.1371/journal.pone.0053028 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Mutlu E, Keshavarzian A, Engen P, Forsyth CB, Sikaroodi M, Gillevet P. 2009. Intestinal dysbiosis: a possible mechanism of alcohol-induced endotoxemia and alcoholic steatohepatitis in rats. Alcohol Clin Exp Res 33:1836–1846. 10.1111/j.1530-0277.2009.01022.x [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yan AW, Fouts DE, Brandl J, Stärkel P, Torralba M, Schott E, Tsukamoto H, Nelson KE, Brenner DA, Schnabl B. 2011. Enteric dysbiosis associated with a mouse model of alcoholic liver disease. Hepatology 53:96–105. 10.1002/hep.24018 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mutlu EA, Gillevet PM, Rangwala H, Sikaroodi M, Naqvi A, Engen PA, Kwasny M, Lau CK, Keshavarzian A. 2012. Colonic microbiome is altered in alcoholism. Am J Physiol Gastrointest Liver Physiol 302:G966–G978. 10.1152/ajpgi.00380.2011 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Llopis M, Cassard AM, Wrzosek L, Boschat L, Bruneau A, Ferrere G, Puchois V, Martin JC, Lepage P, Le Roy T, Lefèvre L, Langelier B, Cailleux F, González-Castro AM, Rabot S, Gaudin F, Agostini H, Prévot S, Berrebi D, Ciocan D, Jousse C, Naveau S, Gérard P, Perlemuter G. 2016. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut 65:830–839. 10.1136/gutjnl-2015-310585 [PubMed] [DOI] [PubMed] [Google Scholar]
- 98.Dorrestein PC, Mazmanian SK, Knight R. 2014. Finding the missing links among metabolites, microbes, and the host. Immunity 40:824–832. 10.1016/j.immuni.2014.05.015 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Couch RD, Dailey A, Zaidi F, Navarro K, Forsyth CB, Mutlu E, Engen PA, Keshavarzian A. 2015. Alcohol induced alterations to the human fecal VOC metabolome. PLoS One 10:e0119362. 10.1371/journal.pone.0119362 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Xie G, Zhong W, Li H, Li Q, Qiu Y, Zheng X, Chen H, Zhao X, Zhang S, Zhou Z, Zeisel SH, Jia W. 2013. Alteration of bile acid metabolism in the rat induced by chronic ethanol consumption. FASEB J 27:3583–3593. 10.1096/fj.13-231860 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, Takei H, Muto A, Nittono H, Ridlon JM, White MB, Noble NA, Monteith P, Fuchs M, Thacker LR, Sikaroodi M, Bajaj JS. 2013. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 58:949–955. 10.1016/j.jhep.2013.01.003 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Jones ML, Martoni CJ, Ganopolsky JG, Labbé A, Prakash S. 2014. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin Biol Ther 14:467–482. 10.1517/14712598.2014.880420 [PubMed] [DOI] [PubMed] [Google Scholar]
- 103.Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. 2009. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 89:147–191. 10.1152/physrev.00010.2008 [PubMed] [DOI] [PubMed] [Google Scholar]
- 104.Manley S, Ding W. 2015. Role of farnesoid X receptor and bile acids in alcoholic liver disease. Acta Pharm Sin B 5:158–167. 10.1016/j.apsb.2014.12.011 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wu WB, Chen YY, Zhu B, Peng XM, Zhang SW, Zhou ML. 2015. Excessive bile acid activated NF-kappa B and promoted the development of alcoholic steatohepatitis in farnesoid X receptor deficient mice. Biochimie 115:86–92. 10.1016/j.biochi.2015.05.014 [PubMed] [DOI] [PubMed] [Google Scholar]
- 106.Fouts DE, Torralba M, Nelson KE, Brenner DA, Schnabl B. 2012. Bacterial translocation and changes in the intestinal microbiome in mouse models of liver disease. J Hepatol 56:1283–1292. 10.1016/j.jhep.2012.01.019 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hartmann P, Chen P, Wang HJ, Wang L, McCole DF, Brandl K, Stärkel P, Belzer C, Hellerbrand C, Tsukamoto H, Ho SB, Schnabl B. 2013. Deficiency of intestinal mucin-2 ameliorates experimental alcoholic liver disease in mice. Hepatology 58:108–119. 10.1002/hep.26321 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wang L, Fouts DE, Stärkel P, Hartmann P, Chen P, Llorente C, DePew J, Moncera K, Ho SB, Brenner DA, Hooper LV, Schnabl B. 2016. Intestinal REG3 lectins protect against alcoholic steatohepatitis by reducing mucosa-associated microbiota and preventing bacterial translocation. Cell Host Microbe 19:227–239. 10.1016/j.chom.2016.01.003 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ferrere G, Wrzosek L, Cailleux F, Turpin W, Puchois V, Spatz M, Ciocan D, Rainteau D, Humbert L, Hugot C, Gaudin F, Noordine ML, Robert V, Berrebi D, Thomas M, Naveau S, Perlemuter G, Cassard AM. 2017. Fecal microbiota manipulation prevents dysbiosis and alcohol-induced liver injury in mice. J Hepatol 66:806–815. [PubMed] [DOI] [PubMed] [Google Scholar]
- 110.Little TJ, Feinle-Bisset C. 2011. Effects of dietary fat on appetite and energy intake in health and obesity--oral and gastrointestinal sensory contributions. Physiol Behav 104:613–620. 10.1016/j.physbeh.2011.04.038 [PubMed] [DOI] [PubMed] [Google Scholar]
- 111.Mithieux G. 2014. Crosstalk between gastrointestinal neurons and the brain in the control of food intake. Best Pract Res Clin Endocrinol Metab 28:739–744. 10.1016/j.beem.2014.03.004 [PubMed] [DOI] [PubMed] [Google Scholar]
- 112.Bauer KC, Huus KE, Finlay BB. 2016. Microbes and the mind: emerging hallmarks of the gut microbiota-brain axis. Cell Microbiol 18:632–644. 10.1111/cmi.12585 [PubMed] [DOI] [PubMed] [Google Scholar]
- 113.Leclercq S, Matamoros S, Cani PD, Neyrinck AM, Jamar F, Stärkel P, Windey K, Tremaroli V, Bäckhed F, Verbeke K, de Timary P, Delzenne NM. 2014. Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci USA 111:E4485–E4493. 10.1073/pnas.1415174111 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Forsyth CB, Farhadi A, Jakate SM, Tang Y, Shaikh M, Keshavarzian A. 2009. Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis. Alcohol 43:163–172. 10.1016/j.alcohol.2008.12.009 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Nanji AA, Khettry U, Sadrzadeh SM. 1994. Lactobacillus feeding reduces endotoxemia and severity of experimental alcoholic liver (disease). Proc Soc Exp Biol Med 205:243–247. 10.3181/00379727-205-43703 [PubMed] [DOI] [PubMed] [Google Scholar]
- 116.Keshavarzian A, Choudhary S, Holmes EW, Yong S, Banan A, Jakate S, Fields JZ. 2001. Preventing gut leakiness by oats supplementation ameliorates alcohol-induced liver damage in rats. J Pharmacol Exp Ther 299:442–448. [PubMed] [PubMed] [Google Scholar]
- 117.Wang Y, Kirpich I, Liu Y, Ma Z, Barve S, McClain CJ, Feng W. 2011. Lactobacillus rhamnosus GG treatment potentiates intestinal hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury. Am J Pathol 179:2866–2875. 10.1016/j.ajpath.2011.08.039 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wang Y, Liu Y, Kirpich I, Ma Z, Wang C, Zhang M, Suttles J, McClain C, Feng W. 2013. Lactobacillus rhamnosus GG reduces hepatic TNFα production and inflammation in chronic alcohol-induced liver injury. J Nutr Biochem 24:1609–1615. 10.1016/j.jnutbio.2013.02.001 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chang B, Sang L, Wang Y, Tong J, Zhang D, Wang B. 2013. The protective effect of VSL#3 on intestinal permeability in a rat model of alcoholic intestinal injury. BMC Gastroenterol 13:151. 10.1186/1471-230X-13-151 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Loguercio C, Federico A, Tuccillo C, Terracciano F, D’Auria MV, De Simone C, Del Vecchio Blanco C. 2005. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J Clin Gastroenterol 39:540–543. 10.1097/01.mcg.0000165671.25272.0f [PubMed] [DOI] [PubMed] [Google Scholar]
- 121.Zhong W, Zhou Z. 2014. Alterations of the gut microbiome and metabolome in alcoholic liver disease. World J Gastrointest Pathophysiol 5:514–522. 10.4291/wjgp.v5.i4.514 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo-Perez F, Guy CD, Seed PC, Rawls JF, David LA, Hunault G, Oberti F, Calès P, Diehl AM. 2016. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 63:764–775. 10.1002/hep.28356 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.De Minicis S, Rychlicki C, Agostinelli L, Saccomanno S, Candelaresi C, Trozzi L, Mingarelli E, Facinelli B, Magi G, Palmieri C, Marzioni M, Benedetti A, Svegliati-Baroni G. 2014. Dysbiosis contributes to fibrogenesis in the course of chronic liver injury in mice. Hepatology 59:1738–1749. 10.1002/hep.26695 [PubMed] [DOI] [PubMed] [Google Scholar]
- 124.Gómez-Hurtado I, Santacruz A, Peiró G, Zapater P, Gutiérrez A, Pérez-Mateo M, Sanz Y, Francés R. 2011. Gut microbiota dysbiosis is associated with inflammation and bacterial translocation in mice with CCl4-induced fibrosis. PLoS One 6:e23037. 10.1371/journal.pone.0023037 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cuenca S, Sanchez E, Santiago A, El Khader I, Panda S, Vidal S, Camilo Nieto J, Juárez C, Sancho F, Guarner F, Soriano G, Guarner C, Manichanh C. 2014. Microbiome composition by pyrosequencing in mesenteric lymph nodes of rats with CCl4-induced cirrhosis. J Innate Immun 6:263–271. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Mazagova M, Wang L, Anfora AT, Wissmueller M, Lesley SA, Miyamoto Y, Eckmann L, Dhungana S, Pathmasiri W, Sumner S, Westwater C, Brenner DA, Schnabl B. 2015. Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice. FASEB J 29:1043–1055. 10.1096/fj.14-259515 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Li M, Zhu L, Xie A, Yuan J. 2015. Oral administration of Saccharomyces boulardii ameliorates carbon tetrachloride-induced liver fibrosis in rats via reducing intestinal permeability and modulating gut microbial composition. Inflammation 38:170–179. 10.1007/s10753-014-0019-7 [PubMed] [DOI] [PubMed] [Google Scholar]
- 128.Sánchez E, Nieto JC, Boullosa A, Vidal S, Sancho FJ, Rossi G, Sancho-Bru P, Oms R, Mirelis B, Juárez C, Guarner C, Soriano G. 2015. VSL#3 probiotic treatment decreases bacterial translocation in rats with carbon tetrachloride-induced cirrhosis. Liver Int 35:735–745. 10.1111/liv.12566 [PubMed] [DOI] [PubMed] [Google Scholar]
- 129.Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B, Li L. 2011. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology 54:562–572. 10.1002/hep.24423 [PubMed] [DOI] [PubMed] [Google Scholar]
- 130.Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, Guo J, Le Chatelier E, Yao J, Wu L, Zhou J, Ni S, Liu L, Pons N, Batto JM, Kennedy SP, Leonard P, Yuan C, Ding W, Chen Y, Hu X, Zheng B, Qian G, Xu W, Ehrlich SD, Zheng S, Li L. 2014. Alterations of the human gut microbiome in liver cirrhosis. Nature 513:59–64. 10.1038/nature13568 [PubMed] [DOI] [PubMed] [Google Scholar]
- 131.Wei X, Yan X, Zou D, Yang Z, Wang X, Liu W, Wang S, Li X, Han J, Huang L, Yuan J. 2013. Abnormal fecal microbiota community and functions in patients with hepatitis B liver cirrhosis as revealed by a metagenomic approach. BMC Gastroenterol 13:175. 10.1186/1471-230X-13-175 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, White MB, Monteith P, Noble NA, Unser AB, Daita K, Fisher AR, Sikaroodi M, Gillevet PM. 2014. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol 60:940–947. 10.1016/j.jhep.2013.12.019 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Bajaj JS, Ridlon JM, Hylemon PB, Thacker LR, Heuman DM, Smith S, Sikaroodi M, Gillevet PM. 2012. Linkage of gut microbiome with cognition in hepatic encephalopathy. Am J Physiol Gastrointest Liver Physiol 302:G168–G175. 10.1152/ajpgi.00190.2011 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Lunia MK, Sharma BC, Sharma P, Sachdeva S, Srivastava S. 2014. Probiotics prevent hepatic encephalopathy in patients with cirrhosis: a randomized controlled trial. Clin Gastroenterol Hepatol 12:1003–1008.e1. [PubMed] [DOI] [PubMed] [Google Scholar]
- 135.Jayakumar S, Carbonneau M, Hotte N, Befus AD, St Laurent C, Owen R, McCarthy M, Madsen K, Bailey RJ, Ma M, Bain V, Rioux K, Tandon P. 2013. VSL#3 ® probiotic therapy does not reduce portal pressures in patients with decompensated cirrhosis. Liver Int 33:1470–1477. [DOI] [PubMed] [Google Scholar]
- 136.Bajaj JS, Betrapally NS, Hylemon PB, Thacker LR, Daita K, Kang DJ, White MB, Unser AB, Fagan A, Gavis EA, Sikaroodi M, Dalmet S, Heuman DM, Gillevet PM. 2015. Gut microbiota alterations can predict hospitalizations in cirrhosis independent of diabetes mellitus. Sci Rep 5:18559. 10.1038/srep18559 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, Puri P, Sterling RK, Luketic V, Stravitz RT, Siddiqui MS, Fuchs M, Thacker LR, Wade JB, Daita K, Sistrun S, White MB, Noble NA, Thorpe C, Kakiyama G, Pandak WM, Sikaroodi M, Gillevet PM. 2014. Randomised clinical trial: Lactobacillus GG modulates gut microbiome, metabolome and endotoxemia in patients with cirrhosis. Aliment Pharmacol Ther 39:1113–1125. 10.1111/apt.12695 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Rogers GB, van der Gast CJ, Bruce KD, Marsh P, Collins JE, Sutton J, Wright M. 2013. Ascitic microbiota composition is correlated with clinical severity in cirrhosis with portal hypertension. PLoS One 8:e74884. 10.1371/journal.pone.0074884 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sherman M. 2010. Epidemiology of hepatocellular carcinoma. Oncology 78(Suppl 1):7–10. 10.1159/000315223 [PubMed] [DOI] [PubMed] [Google Scholar]
- 140.Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, Caviglia JM, Khiabanian H, Adeyemi A, Bataller R, Lefkowitch JH, Bower M, Friedman R, Sartor RB, Rabadan R, Schwabe RF. 2012. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 21:504–516. 10.1016/j.ccr.2012.02.007 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yu LX, Yan HX, Liu Q, Yang W, Wu HP, Dong W, Tang L, Lin Y, He YQ, Zou SS, Wang C, Zhang HL, Cao GW, Wu MC, Wang HY. 2010. Endotoxin accumulation prevents carcinogen-induced apoptosis and promotes liver tumorigenesis in rodents. Hepatology 52:1322–1333. 10.1002/hep.23845 [PubMed] [DOI] [PubMed] [Google Scholar]
- 142.Wang Z, Yan J, Lin H, Hua F, Wang X, Liu H, Lv X, Yu J, Mi S, Wang J, Hu ZW. 2013. Toll-like receptor 4 activity protects against hepatocellular tumorigenesis and progression by regulating expression of DNA repair protein Ku70 in mice. Hepatology 57:1869–1881. 10.1002/hep.26234 [PubMed] [DOI] [PubMed] [Google Scholar]
- 143.Eiro N, Altadill A, Juarez LM, Rodriguez M, Gonzalez LO, Atienza S, Bermudez S, Fernandez-Garcia B, Fresno-Forcelledo MF, Rodrigo L, Vizoso FJ. 2014. Toll-like receptors 3, 4 and 9 in hepatocellular carcinoma: relationship with clinicopathological characteristics and prognosis. Hepatology Res 44:769–778. [PubMed] [DOI] [PubMed] [Google Scholar]
- 144.Wells JE, Hylemon PB. 2000. Identification and characterization of a bile acid 7alpha-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7alpha-dehydroxylating strain isolated from human feces. Appl Environ Microbiol 66:1107–1113. 10.1128/AEM.66.3.1107-1113.2000 [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
