Skip to main content
American Journal of Physiology - Gastrointestinal and Liver Physiology logoLink to American Journal of Physiology - Gastrointestinal and Liver Physiology
. 2019 Feb 15;316(5):G563–G573. doi: 10.1152/ajpgi.00370.2018

Gut microbiota in liver disease: too much is harmful, nothing at all is not helpful either

Phillipp Hartmann 1,*, Huikuan Chu 2,3,*, Yi Duan 3,4, Bernd Schnabl 3,4,
PMCID: PMC6580239  PMID: 30767680

Abstract

The intestinal microbiome plays a major role in the pathogenesis of liver disease, with a hallmark event being dysbiosis, or an imbalance of pathobionts and beneficial bacteria with the associated deleterious effects on their host. Reducing the number of intestinal bacteria with antibiotic treatment is generally advantageous in experimental liver diseases. Complete absence of intestinal microbiota as in germ-free rodents can be protective in autoimmune hepatitis and hepatic tumors induced by chemicals, or it can exacerbate disease as in acute toxic liver injury and liver fibrosis/cirrhosis. In alcoholic liver disease, nonalcoholic fatty liver disease, and autoimmune cholangiopathies, germ-free status can be associated with worsened or improved hepatic phenotype depending on the experimental model and type of rodent. Some of the unexpected outcomes can be explained by the limitations of rodents raised in a germ-free environment including a deficient immune system and an altered metabolism of lipids, cholesterol, xenobiotics/toxins, and bile acids. Given these limitations and to advance understanding of the interactions between host and intestinal microbiota, simplified model systems such as humanized gnotobiotic mice, or gnotobiotic mice monoassociated with a single bacterial strain or colonized with a defined set of microbes, are unique and useful models for investigation of liver disease in a complex ecosystem.

Keywords: antibiotics, germ-free, humanized rodents, liver disease, microbiota

INTRODUCTION

There is a strong symbiotic relationship between gut microbiota and its host. The human body harbors ~1–10 intestinal bacteria for each human cell (60, 93). Although each person’s microbial profile is distinct, the relative abundance and composition of bacterial species are similar among healthy individuals (119). Microbiota has protective effects on the human body, as it helps with digestion and synthesis of vitamins and furthers resistance to intestinal colonization of pathogens (27, 85). On the other hand, microbiota can become pathogenic. Alterations of the intestinal microbiome have been reported to affect the pathogenesis of many diseases, including liver diseases (14). Intestinal dysbiosis [or an imbalance of pathobionts and beneficial bacteria with its associated deleterious effects on their host (40)] plays a critical role in the development of alcoholic liver disease (88), nonalcoholic fatty liver disease (NAFLD; 1, 87), nonalcoholic steatohepatitis (NASH; 1, 87), acute (toxic) liver injury, fibrosis/cirrhosis (88), hepatic tumors (13), autoimmune hepatitis (AIH; 58), and autoimmune cholangiopathies (78, 86). Highlighting the significance of intestinal dysbiosis in the pathogenesis of liver diseases, a study published in 1990 reported development of liver inflammation and fibrosis following surgical creation of a small intestinal loop causing bacterial overgrowth (57). Gut microbiota affects liver disease via changing the metabolism and caloric yield of diet; it can also perturb gut permeability with resulting bacterial translocation and exposure of the liver to microbial components with subsequent inflammation (14, 88).

Although gaining popularity again today because of the mounting evidence of the significant effect of microbiota on its host’s health, germ-free rodents were employed to study liver disease already in the 1960s (8, 30, 39, 53, 54, 75). Germ-free rodents do not harbor any microbiota from birth on as they are born by caesarean section in sterile conditions and immediately separated from their biological mothers to be raised in isolators. Because of the lack of exposure to bacteria, germ-free animals display an aberrant maturation of the immune system with an abnormal intestinal epithelial cell morphology characterized by longer villi and shorter crypts, diminished antimicrobial peptides, and underdeveloped lymphoid tissue including the spleen, thymus, lymphoid follicles, Peyer’s patches, and mesenteric lymph nodes (7, 35, 37). A disrupted gut barrier contributes to the pathogenesis of liver disease in conventional rodents; however, intestinal dysfunction does not appear to be crucial for disease in germ-free mice, as, for example, germ-free mice can show intestinal permeability similar to or even higher than that of conventional rodents on control or high-caloric diet but are protected against diet-induced obesity and NAFLD (11, 45). This can be partially explained by the absence of intestinal microbiota in germ-free mice and hence lack of translocation of bacteria-derived lipopolysaccharide (LPS) into the bloodstream with associated hepatotoxic and hepato-modulating effects (see below). By understanding these hallmark events and examining disease development in germ-free rodents, scientists can have a better idea how microbiota affects human health. For this review, we screened the PubMed library for the terms “liver germ-free,” “liver disease germ-free,” “hepatitis germ-free,” “steatohepatitis germ-free,” “hepatoma/hepatocellular carcinoma germ-free,” and “liver disease antibiotics” to obtain a large set of articles related to liver disease in germ-free animals and selected articles on the basis of their title and/or abstract. In this review, we discuss the effects of germ-free status on the development of liver diseases and possible mechanisms (Table 1). We review implications of antibiotics in experimental liver disease (Table 2) and discuss the use of “humanized” rodents (Fig. 1).

Table 1.

Phenotype of germ-free rodents in various liver diseases compared with controls

Type of Liver Disease Experimental Model Animals Phenotype of Germ-Free Animals Relative to Conventional Controls Reference
Alcoholic liver disease Single gavage of alcohol C57BL/6 mice Exacerbated Chen et al. (21)
Alcoholic liver disease Lieber-DeCarli alcohol diet over 7 days plus binge Swiss mice Protected Canesso et al. (18)
NAFLD High-carbohydrate diet (57% carbohydrates and 5% fat) C57BL/6J mice Protected Bäckhed et al. (4)
NAFLD High-fructose diet C57BL/6J mice Protected Kaden-Volynets et al. (45)
NAFLD High-fat diet C57BL/6J mice Protected Rabot et al. (79)
NAFLD Western diet C3H/HeJ mice Protected Fleissner et al. (34)
NAFLD Choline-deficient diet Fischer rats Protected/exacerbated depending on cystine and cholesterol content in diet Levenson et al. (53)
NAFLD Choline-deficient diet Lobund rats Exacerbated Levenson and Tennant (54)
HCC No intervention until euthanasia at 12 mo C3H/He mice Protected Mizutani and Mitsuoka (68)
HCC No intervention until euthanasia at 19-41 mo Lobund-Wistar rats Similar Pollard et al. (73)
HCC MAM-GlcUA Sprague-Dawley rats Protected Laqueur et al. (50)
HCC DMBA C3H mice Protected Grant and Roe (39), Roe and Grant (83)
HCC Phenobarbital C3H mice Protected Mizutani and Mitsuoka (66)
HCC DEN and CCl4 C57BL/6 mice Protected Dapito et al. (26)
AIH ConA BALB/c mice Protected Wei et al. (109)
AIH ConA Swiss Webster (TAC:SW) mice Protected Celaj et al. (20)
Autoimmune cholangiopathies
    PSC Abcb4−/− (Mdr2−/−) mice FVB/N Abcb4−/− (Mdr2−/−) mice Exacerbated Tabibian et al. (99)
    PBC NOD.c3c4 mice NOD.c3c4 mice Protected Schrumpf et al. (90)
Acute (toxic) liver injury Acetaminophen-induced liver injury C3H/HeH mice Similar Possamai et al. (76)
Acute (toxic) liver injury DMA plus sodium nitrite Carworth Farms Swiss Webster mice Exacerbated Pollard et al. (74)
Acute (toxic) liver injury DMN or DMA plus sodium nitrite Wistar rats Exacerbated Sumi and Miyakawa (95)
Acute (toxic) liver injury Chlortetracycline ICR mice Exacerbated Einheber et al. (30)
Acute (toxic) liver injury Butter yellow (3′-methyl-4-dimethylaminoazobenzene) Fischer rats Exacerbated Bauer et al. (8)
Acute (toxic) liver injury Partial hepatectomy BALB/c mice Exacerbated Cornell et al. (24)
Liver fibrosis/cirrhosis TAA or CCl4 C57BL/6 mice Exacerbated Mazagova et al. (65)
Liver fibrosis/cirrhosis CCl4 ICR mice Exacerbated Popper et al. (75)

Abcb4−/− (Mdr2−/−), ATP-binding cassette subfamily B member 4 (also known as multidrug resistance protein-2) knockout; AIH, autoimmune hepatitis; ConA, concanavalin A; CCl4, carbon tetrachloride; DEN, diethylnitrosamine; DMA, dimethylamine; DMBA, 7,12-dimethylbenz(a)anthracene; DMN, dimethylnitrosamine; HCC, hepatocellular carcinoma; MAM-GlcUA, methylazoxymethanol-β-d-glucosiduronic acid; NAFLD, nonalcoholic fatty liver disease; PBC, primary biliary cholangitis; PSC, primary sclerosing cholangitis; TAA, thioacetamide.

Table 2.

Phenotype of antibiotic-treated rodents in various liver diseases compared with controls (representative articles selected)

Type of Liver Disease Experimental Model Animals Antibiotics Phenotype of Antibiotic-Treated Animals Relative to Controls Reference
Alcoholic liver disease Lieber-DeCarli alcohol diet for 8 wk C57BL/6 mice Polymyxin B and neomycin Improved Chen et al. (22)
Alcoholic liver disease Intragastric alcohol for ≤3 wk Wistar rats Polymyxin B and neomycin Improved Adachi et al. (2)
NAFLD High-fat diet C57BL/6N mice Bacitracin, neomycin, and streptomycin Improved Jiang et al. (44)
NAFLD High-fructose diet C57BL/6J mice Polymyxin B and neomycin Improved Bergheim et al. (9)
NASH High-fat diet C57BL/6J wild-type mice cohoused with C57BL/6J ASC−/− mice Ciprofloxacin and metronidazole Improved Henao-Mejia et al. (43)
NASH High-fat diet (STHD-01) C57BL/6J mice Ceftazidime and metronidazole Improved Yamada et al. (114)
HCC DEN and CCl4 C3H/HeOuJ mice ANMV Improved Dapito et al. (26)
HCC DMBA C57BL/6 mice ANMV or vancomycin only Improved Yoshimoto et al. (115)
AIH ConA BALB/c mice ANMV Improved Celaj et al. (20)
AIH ConA C57BL/6NCrlCrlj mice Rifampicin Improved Kataoka et al. (48)
Autoimmune cholangiopathies
    PSC Abcb4−/− (Mdr2−/−) mice C57BL/6J Abcb4−/− (Mdr2−/−) mice Polymyxin B and neomycin Improved Peng et al. (72)
    PBC NOD.c3c4 mice NOD.c3c4 mice Ampicillin and neomycin Improved Schrumpf et al. (90)
Acute (toxic) liver injury Acetaminophen-induced liver injury BALB/c mice ANMV Improved Gong et al. (38)
Acute (toxic) liver injury d-Gal Crlj:CD(SD) rats Rifampicin Improved Kataoka et al. (48)
Liver fibrosis/cirrhosis Bile duct ligation Tlr4-wild-type C3H/HeOuJ mice ANMV Improved Seki et al. (91)
Liver fibrosis/cirrhosis CCl4 plus ethanol Wistar rats Ciprofloxacin Improved Zhang et al. (117)

Abcb4−/− (Mdr2−/−), ATP-binding cassette subfamily B member 4 (also known as multidrug resistance protein-2) knockout; AIH, autoimmune hepatitis; ANMV, ampicillin, neomycin, metronidazole, and vancomycin; ASC−/−, apoptosis-associated speck-like protein containing a COOH-terminal caspase recruitment domain knockout; ConA, concanavalin A; CCl4, carbon tetrachloride; DEN, diethylnitrosamine; d-Gal, d-galactosamine; DMBA, 7,12-dimethylbenz(a)anthracene; HCC, hepatocellular carcinoma; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; PBC, primary biliary cholangitis; PSC, primary sclerosing cholangitis; STHD, steatohepatitis-inducing high-fat diet; Tlr4, Toll-like receptor 4.

Fig. 1.

Fig. 1.

Use of germ-free mice in preclinical liver disease models. Germ-free mice have an immature immune system and a significantly altered metabolism related to lipids, cholesterol, xenobiotics/toxins, and bile acids. They can be colonized with only one known bacterial strain (monoassociated), a specific set of known bacterial strains, or human feces. Monoassociation and colonization with several bacteria are unique models to investigate the effect of single or multiple bacterial strains on experimental disease. Transmissibility of disease can be investigated by microbiota transfer into gnotobiotic mice. Humanized gnotobiotic mice allow the testing of microbiome-centered therapies to treat disease. Gnotobiotic rodents oftentimes have a more normalized immune system and metabolism than germ-free mice. Gnotobiotic mice can be subjected to preclinical models of liver disease to examine the impact of colonized microbes on the disease process.

ALCOHOLIC LIVER DISEASE

Chronic alcoholic liver disease may progress from simple steatosis to steatohepatitis, liver fibrosis, liver cirrhosis, and hepatocellular carcinoma (HCC; 42). Alcoholic liver disease is a leading cause of liver-related morbidity and mortality worldwide.

Factors that link ethanol to the onset and progression of liver disease are poorly understood. The susceptibility of patients with alcohol use disorder to alcoholic liver disease is variable. With similar alcohol intake, some people can develop severe forms of alcoholic hepatitis, whereas others remain healthy (103). A specific intestinal microbiota composition confers a higher risk of alcohol-induced liver injury (33), underlying the key role of gut microbiota in alcoholic liver disease. Intestinal dysbiosis has been described in patients with alcohol use disorder (51) and cirrhosis (77), and deteriorating dysbiosis is associated with cirrhosis progression (6). Severe alcoholic hepatitis was associated with higher fecal proportions of bifidobacteria, streptococci, and enterobacteria (59). Thus, intestinal microbiota is central in the onset and progression of alcoholic liver disease.

Germ-free rodents are a tool to investigate the function of the intestinal microbiome. In a model of acute alcohol exposure that mimics binge drinking, germ-free C57BL/6 mice exhibited significantly more pronounced liver injury, inflammation, and steatosis than their conventional counterparts (21). One reason for the increased susceptibility to liver injury might be that germ-free mice have a more efficient xenobiotic metabolism compared with conventional mice (12). Hepatic and intestinal expression of genes involved in xenobiotic metabolism was overall higher in germ-free mice than in conventional mice, including alcohol dehydrogenase, aldehyde dehydrogenase, and cytochrome P450 superfamily members (12, 21). Although the detailed mechanism is still unknown, it is possible that the induction of these genes resulted from accumulation of constitutive androstane receptor ligands such as bilirubin, bile acids, and steroid hormones due to the absence of intestinal bacteria metabolizing these molecules in germ-free mice (12). Upregulated genes were associated with more rapid ethanol elimination from the blood and increased production of toxic alcohol metabolites in the liver, further exacerbating liver injury in germ-free mice (21). Besides, elevated baseline levels of inflammatory mediators such as macrophage markers CD68 and F4/80 were also likely to contribute to increased liver damage in germ-free mice (21). Moreover, the increased hepatic steatosis observed in germ-free C57BL/6 mice was associated with upregulated expression of genes involved in fatty acid and triglyceride synthesis, including sterol regulatory element-binding protein-1 (SREBP-1), stearoyl-coenzyme A desaturase 1 (SCD-1), fatty acid synthase (FASN), and acetyl-CoA carboxylase-1 (ACC-1; 21).

On the other hand, germ-free Swiss mice were found to exhibit reduced liver injury after chronic alcohol consumption relative to their conventional controls (18). This was accompanied by a (not significant) reduction in gut permeability to ~50% and a significantly lower hepatic neutrophil accumulation in germ-free mice compared with conventional mice after ethanol administration (18).

In summary, germ-free C57BL/6 mice exhibit higher susceptibility to acute alcohol-induced liver injury relative to conventional mice, which may be related to a more efficient xenobiotic metabolism, elevated baseline levels of inflammatory mediators, and/or upregulated expression of genes involved in fatty acid and triglyceride synthesis. In contrast, germ-free Swiss mice appear to be resistant to liver damage secondary to alcohol compared with their conventionally raised counterparts. The discrepancy between these two studies could also be due to different mouse strains. Multiple rodent studies in alcoholic liver disease have shown that strains can differ significantly with regard to liver injury, immune response (TNF-α), alcohol-metabolizing enzymes [alcohol dehydrogenase, aldehyde dehydrogenase, and cytochrome P450 2E1 (CYP2E1)], endoplasmic reticulum stress [78-kDa glucose-regulated protein (GRP78) and C/EBP-homologous protein (CHOP)], and fat metabolism [nuclear SREBP-1 (nSREBP-1); 29, 104]. Germ-free status further modulates these differences and could hence potentially confer some protection in one strain and exacerbate alcoholic liver disease in another. The different durations of the alcohol-feeding models could further explain exacerbation of liver injury in germ-free mice (after a 1-time binge) versus an alleviated hepatic phenotype (after alcohol feeding over 1 wk plus binge). Animal facility-related factors should also be considered such as slightly different diets and vivarium temperature (49). Finally, it is unclear whether the germ-free condition was maintained during the feeding of a Lieber-DeCarli diet in a chronic model of alcoholic liver disease, as it is difficult to sterilize/irradiate the liquid diet reliably and as it has to be changed frequently. Further studies are required to investigate potential mechanisms to explain the dissimilar phenotype between different mouse strains in that type of liver injury.

NONALCOHOLIC FATTY LIVER DISEASE

NAFLD is considered the hepatic manifestation of metabolic syndrome and is commonly associated with insulin resistance (15). It is defined as a spectrum of liver diseases ranging from simple steatosis to NASH, advanced fibrosis, cirrhosis, or even HCC (16).

The pathophysiology of NAFLD involves many factors, including ecological, genetic, and metabolic factors such as limited physical activity and an imbalanced diet (41, 112). Gut microbiota is a new environmental factor contributing to obesity and NAFLD (1, 28, 87, 88). Compositions of gut microbiota are disturbed in patients with NAFLD. Members of the phylum Firmicutes were detected at elevated levels in individuals with NAFLD relative to nonobese controls (80), whereas contributions of Bacteroidetes were decreased in adult patients with NASH compared with subjects with simple steatosis and healthy individuals (69). Conversely, Bacteroidetes, Proteobacteria, and Enterobacteriaceae/Escherichia coli were found to be increased in pediatric patients with NASH compared with healthy controls, whereas Firmicutes were detected at lower levels (120). Probiotics can protect against the onset of NAFLD and attenuate NAFLD (41, 81, 108). An intervention with Lactobacillus casei strain Shirota protected against experimental NASH induced by methionine-choline-deficient diet, with reduced serum LPS concentrations and suppression of inflammation and fibrosis in the liver (71). VSL#3 probiotics (a mixture of bifidobacteria, lactobacilli, and Streptococcus thermophilus) reduced high-fat diet (HFD)-induced steatosis, inflammation, and insulin resistance in a rodent model (61). Daily gavage of fecal microbiota from control C57BL/6 mice for 8 wk for the second half of a 16-wk HFD-feeding experiment protected against NASH with lower hepatic fat, inflammatory cytokines, and NAFLD Activity Score relative to mice that did not receive a fecal transplant (118). Fecal transplant was also associated with increased intestinal butyrate concentrations, increased expression of the tight junction protein zonula occludens-1 (ZO-1), and lower serum LPS concentrations (118).

Interestingly, germ-free C57BL/6J mice gained less weight (4, 5, 79) and had a lower hepatic fat concentration (4, 79) than conventional mice when given a high-carbohydrate diet (57% carbohydrates and 5% fat; 4), an HFD (60% fat; 79), or a Western diet (41% carbohydrates and 41% fat; 5), suggesting that those germ-free mice are protected against obesity and NAFLD. Similarly, germ-free C3H/HeJ mice fed a Western diet were protected from fatty liver compared with their conventional counterparts (34). Germ-free C57BL/6J mice were also found to be protected against hepatic steatosis after a high-fructose diet (45). Conversely, germ-free Fischer rats fed a choline-deficient, low-cystine, low-cholesterol diet exhibited less liver fat content than their controls but worsened hepatic steatosis relative to conventional controls after a choline-deficient, high-cystine, high-cholesterol diet (53). These findings suggest that the liver injury is ameliorated in germ-free rats in the low-cystine-low-cholesterol environment likely secondary to absent bacterial utilization of choline and methionine and is much worse in the high-cystine-high-cholesterol environment possibly because of a missing reduction of cystine/cholesterol content by bacterial metabolization and/or the lacking effects of hepatoprotective bacterial metabolites. On the other hand, more than 50 years ago, germ-free Lobund rats were shown to develop NAFLD-related liver cirrhosis due to choline-deficient diet more rapidly relative to their conventional counterparts (54).

The protective effect observed in the germ-free C57BL/6J mice might be the consequence of a lower consumption of calories and higher excretion of fecal lipids as observed in one study (79). However, the protective effect was also documented when the same number of calories was consumed and the same fecal energy content noted in germ-free and conventional mice (5). Furthermore, the beneficial outcome might also result from sufficient choline levels in germ-free mice, as they harbor no gut microbiota to regulate concentrations of choline (41, 53, 88). Additionally, the protection in germ-free rodents might be due to altered inflammasome activity with an abolished caspase-1 autocleavage and associated reduced IL-18 concentrations, indicating insufficient macrophage activity (56, 63). Consistent with this idea, fecal transplantation from conventional mice on high-carbohydrate diet (4) or HFD (52) to germ-free mice resulted in gain of fat mass (4, 52) and NAFLD (52) in colonized mice.

Moreover, germ-free status enhances insulin sensitivity with improved glucose tolerance, reduced fasting and nonfasting insulinemia, and increased phosphorylation of Akt in adipose tissue (5, 79). Germ-free mice are thought to be protected against diet-induced obesity because of increased AMP-activated protein kinase (AMPK) activity and increased fatty acid oxidation in their peripheral tissues (5). AMPK is a key regulator of energy balance, and loss/attenuation of its kinase activity leads to several metabolic disorders (70). Additionally, increased fatty acid oxidation in germ-free mice is secondary to increased intestinal levels of fasting-induced adipose factor (Fiaf) via higher expression of peroxisome proliferator-activated receptor-γ coactivator-1 (Pgc-1; 70, 107). Increased Fiaf in germ-free mice also suppresses lipoprotein lipase (LPL) resulting in reduced uptake of fatty acids and triglycerides in adipocytes (4).

Finally, alteration of cholesterol metabolism appears to be another important reason. Germ-free mice on HFD display reduced plasma cholesterol levels and higher hepatic cholesterol contents and excrete more fecal cholesterol compared with their conventional counterparts (79). This may be related to upregulated hepatic nSREBP-2, upregulated cholesterol biosynthesis genes such as the rate-limiting enzyme in cholesterol biosynthesis 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), and upregulated membrane transporters for cholesterol excretion in liver and small intestine ATP-binding cassette subfamily G members 5 and 8 (ABCG5/8; 31, 79, 82). Besides, cholesterol is converted to coprostanol by the gut microbiota in conventional rodents and in humans (25, 32, 36). The conversion of cholesterol to coprostanol therefore does not occur in germ-free mice and is another explanation for elevated levels of fecal cholesterol.

In summary, germ-free rodents are either protected from or more susceptible to NAFLD depending on their strain and on the specific composition of the diet. Germ-free C57BL/6J and C3H/HeJ mice are resistant to NAFLD, partially secondary to reduced energy uptake, increased lipid excretion, enhanced insulin sensitivity due to increased AMPK activity, upregulated intestinal expression of Fiaf, and altered cholesterol metabolism. Conversely, germ-free Fischer rats can be protected from or more susceptible to NAFLD depending on the contribution of specific dietary ingredients such as choline, cystine, and cholesterol. Germ-free Lobund rats are more prone to liver cirrhosis due to choline-deficient diet than their conventional counterparts.

LIVER CANCER/HEPATOCELLULAR CARCINOMA

The incidence/mortality of liver cancer is influenced by sex, age, race/ethnicity, location (3), level of dietary protein (94), caloric intake (101), and the intestinal microbiota (13).

Liver tumorigenesis in C3H/He male mice that were gnotobiotic, i.e., mice with only one known bacterial strain or a specific set of known bacterial strains, was promoted by a bacterial combination of E. coli, Streptococcus faecalis, and Clostridium paraputrificum (67, 68). Monoassociated mice with Bifidobacterium longum, Lactobacillus acidophilus, or Eubacterium rectale exhibited an improved cancer burden; nevertheless, significantly more rodents with liver tumors, significantly more tumor nodules per liver, and significantly higher tumor sizes were found in these mouse groups than in germ-free mice (68).

Aging conventional and germ-free Lobund-Wistar rats (19–41 mo) had a similar incidence of HCC (73). However, germ-free Sprague-Dawley rats were protected from liver cancer induced by methylazoxymethanol-β-d-glucosiduronic acid (MAM-GlcUA; 50). Similarly, germ-free C3H mice were less susceptible to the induction of hepatoma by 7,12-dimethylbenz(a)anthracene (DMBA) or phenobarbital (39, 66). This holds true in particular with regard to early development of liver tumors (before 40 wk) in germ-free C3H mice given DMBA shortly after birth; yet, germ-free mice of older age (>80 wk) lose that protective effect (83). Germ-free C57BL/6 mice were also protected against liver cancer induced by diethylnitrosamine (DEN) and carbon tetrachloride (CCl4; 26). Germ-free mice had a significantly lower number of tumor cells, with smaller tumor size and liver weight-to-body weight ratio, than conventional mice (26, 66, 67), which was secondary to absent LPS and lack of Toll-like receptor 4 (TLR4) signaling in germ-free rodents (26). Consistent with this, mice treated with broad-spectrum antibiotics as well as TLR4-inactivated mutant mice were overall protected against carcinogenesis due to DEN and CCl4 and displayed a significant increase in cleaved caspase-3-positive cells in hepatocytes, and the number of cleaved caspase-3-positive cells in these mice was inversely correlated with tumor number, tumor size, and liver weight-to-body weight ratio (26). Additionally, treatment with low-dose LPS over 12 wk led to increased tumor number, tumor size, and ratio of liver weight to body weight (26). These results indicate that the LPS-TLR4 pathway promotes hepatocarcinogenesis. This pathway, among others, induces IL-1β, which promotes inflammation, stimulates hepatic stellate cells to produce fibrogenic mediators, and activates human liver progenitor cells, eventually leading to cirrhosis and HCC (13). Besides LPS, there are also other bacterial products or pathogen-associated molecular patterns that activate the immune system but are rarely measured, such as peptidoglycan, lipoproteins, lipoteichoic acid, double-stranded RNA, and unmethylated DNA (40). Measuring these pathogen recognition receptor ligands and their downstream signaling could broaden the understanding of how gut-derived products and metabolites affect HCC onset and progression. The intestinal microbiome also impacts liver tumorigenesis by other means: Alterations of gut microbiota induced by obesity lead to increased concentrations of deoxycholic acid, which induces secretion of various inflammatory and procarcinogenic factors in the liver and thus promotes HCC development (115).

Hence, germ-free rodents were less susceptible to the induction of hepatic tumors by chemicals, which was in part secondary to an absent metabolism of gut microbiota and lack of gut-derived LPS and related TLR4 activation.

AUTOIMMUNE HEPATITIS

The etiology of AIH is poorly understood. Immune tolerance breakdown, genetic predisposition, and environmental factors may all contribute to triggering the autoimmune process (62, 116). Recently, the intestinal microbiome has been shown to be involved in AIH development in patients with decreasing fecal bifidobacteria and lactobacilli concentrations as well as increasing plasma LPS in later stages of AIH (58).

Germ-free mice are protected against experimental AIH, as leukocyte infiltration and levels of inflammatory cytokines including IFN-γ, TNF-α, IL-4, monocyte chemotactic protein-1 (MCP-1), granulocyte colony-stimulating factor (G-CSF), C-X-C motif chemokine 1 (CXCL1) [or keratinocyte chemoattractant (KC)], granulocyte-macrophage colony-stimulating factor (GM-CSF), eotaxin, macrophage inflammatory protein-1β (MIP-1b), and MIP-1a were significantly lower, whereas apoptosis was nearly undetectable in the liver of concanavalin A (ConA)-treated germ-free BALB/c mice relative to controls (109). This may be due to the deficient activation of natural killer T cells in germ-free mice, resulting from lower levels of presented bacterial glycolipid antigens compared with controls (109). Additionally, bacteria-derived systemic LPS was significantly increased in ConA-treated conventional mice but undetectable in their germ-free counterparts (109), which could be another reason for the alleviated liver damage in germ-free mice, as LPS can activate hepatic Kupffer cells and lead to liver injury (105). Similarly, germ-free Swiss Webster mice also showed lower alanine aminotransferase (ALT) levels and liver histopathological scores after ConA treatment relative to conventionalized controls, which were previously germ-free but orally gavaged with fecal contents of conventional mice (20).

In summary, germ-free mice are resistant to ConA-induced liver injury, and this may be explained by lack of bacterial translocation including LPS and deficient activation of natural killer T cells.

AUTOIMMUNE CHOLANGIOPATHIES

Primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC) are clinically heterogeneous diseases with genetic, immunological, environmental, and other contributory components (19, 55, 97, 110). In addition, intestinal microbiota also plays a pathophysiological role in PSC (64, 86, 97, 110) and PBC (64, 78, 100). Bile acid metabolism is central for the pathogenesis of PSC and PBC. Bacteria metabolize, dehydrogenate, and deconjugate primary to secondary bile acids and degrade taurine. Germ-free rodents thus have markedly lower concentrations of unconjugated and higher concentrations of taurine-conjugated bile acids in liver, kidney, heart, and plasma as well as higher free taurine levels in the liver relative to their conventional counterparts. They furthermore exhibit a markedly different metabolism with significantly altered farnesoid X receptor (FXR)/retinoid X receptor (RXR) activation, hepatic cholestasis, steroid synthesis, and fatty acid metabolism pathways (96).

In a model of experimental PSC, germ-free ATP-binding cassette subfamily B member 4 (also known as multidrug resistance protein-2) knockout [Abcb4−/− (Mdr2−/−)] mice displayed more pronounced biochemical and histological features of PSC than their conventional counterparts (99). Serum biochemical parameters, including alkaline phosphatase, aspartate aminotransferase, ALT, and bilirubin, were significantly higher in germ-free Abcb4−/− mice, with more severe fibrosis, ductular reaction, and ductopenia (99). This susceptibility is related to increased cholangiocyte senescence and the absence of ursodeoxycholic acid, which reduces insult-induced cholangiocyte senescence, as shown in vitro (99). Senescent cells can transition to a potentially pathological state referred to as senescence-associated secretory phenotype (17, 98, 102). Cells of senescence-associated secretory phenotype can, in turn, alter their microenvironment, secrete proinflammatory cytokines and growth factors, and thereby induce senescence in neighboring cells (17). Conversely, in a murine model of PBC, germ-free NOD.c3c4 mice exhibited less portal inflammation and smaller liver weights in relation to their conventionally raised counterparts while displaying a similar level of fibrosis and dilated intrahepatic bile ducts (90).

Hence, germ-free Abcb4−/− mice exhibit a worse PSC phenotype compared with conventional Abcb4−/− mice, which is likely related to an altered bile acid metabolism (96), whereas germ-free NOD.c3c4 mice show some biliary/hepatic protection relative to conventional NOD.c3c4 mice. The discrepancy could possibly be explained by the fact that the disease process in Abcb4−/− mice is driven by changes in bile composition, whereas biliary disease in NOD.c3c4 mice is mainly immune driven (90, 99). Correspondingly, there was no difference in the inflammatory markers CD4+ and CD8+ cells and IL-10 in the Abcb4−/− mouse model between the groups, but there was lower inflammation [decreased CD3+, lymphocyte antigen 6 complex, locus G-positive (Ly6G+), and galectin-3-positive (Mac-2+) cells] in germ-free NOD.c3c4 mice relative to their conventional counterparts (90, 99). Further studies are necessary to clarify the role and mechanisms of the PSC and PBC phenotypes under germ-free conditions.

ACUTE (TOXIC) LIVER INJURY AND LIVER FIBROSIS/CIRRHOSIS

Germ-free C3H/HeH mice showed not significantly different ALT levels and liver necrosis after acetaminophen-induced liver insult compared with conventional controls (76). On the other hand, studies in Carworth Farms Swiss Webster mice (74) and Wistar rats (95) showed that germ-free rodents developed more pronounced liver necrosis and higher ALT/aspartate aminotransferase levels within 48 h after administration of dimethylnitrosamine (DMN) or dimethylamine (DMA) plus sodium nitrite than conventional controls and are hence more prone to that type of acute toxic liver insult. Similarly, germ-free ICR mice subjected to treatment with the antibiotic chlortetracycline over 4 days developed a higher rise of hepatic lipids compared with their conventional counterparts (30). Treatment with butter yellow (3′-methyl-4-dimethylaminoazobenzene; ip injections twice weekly for 6 wk) resulted in more liver necrosis and hepatocyte degeneration in germ-free Fischer rats as well as fibrosis, whereas conventional rats did not show any fibrosis (8). Hepatic regeneration following partial hepatectomy was markedly reduced in germ-free BALB/c mice in relation to conventional controls, which may be due to the lack of endogenous LPS in germ-free mice, as LPS has been shown to stimulate liver regeneration (24).

If the toxic liver injury recurs, liver fibrosis can develop and is characterized by excessive deposition of extracellular matrix proteins. It can result from any chronic liver disease including alcoholic liver disease, NASH, viral hepatitis, cholestatic liver disease, autoimmune liver disease, and toxin-related liver disease (89, 92). In experimental toxin-induced liver fibrosis secondary to administration of thioacetamide (TAA) in drinking water over 21 wk or 12 intraperitoneal injections of CCl4 over 4 wk, germ-free C57BL/6 mice were found to be more susceptible to liver fibrosis relative to conventional mice, with more toxin-induced hepatic oxidative stress and liver cell death (65). Similarly, in studies performed more than 50 years ago, liver cirrhosis was observed in germ-free female ICR mice after 12 wk of twice weekly subcutaneous CCl4 injections, whereas none was detected in their conventional female counterparts after 14 wk (cirrhosis was present in both germ-free and conventional male mice after 12 wk of treatment; 75). Exposure to some microbial products and metabolites might hence preserve liver homeostasis. The more pronounced liver damage in germ-free mice may be due to lower levels of P450 enzymes such as the phase I xenobiotic-metabolizing enzyme CYP26A1 (possibly allowing an accumulation of toxic products, increased oxidative stress, and resulting hepatotoxicity) and absent indole-3-propionic acid (IPA; 65). IPA is a deamination product of dietary tryptophan, is a powerful antioxidant, and is neuroprotective and hepatoprotective (23, 47). Production of IPA depends on the presence of the gut microflora, in particular Clostridium sporogenes (111).

In experimental cirrhosis with hepatic encephalopathy due to gavage with CCl4, germ-free C57BL/6 mice exhibited less systemic inflammation, neuroinflammation, and microglial/glial activation in relation to their conventional counterparts (46). However, they showed a significantly higher increase in serum ammonia levels after cirrhosis development compared with germ-free controls than did conventional mice with cirrhosis compared with conventional controls (46). The elevated ammonia level may be due to upregulated small bowel glutaminase activity (46, 84), which has been shown to affect the occurrence of hepatic encephalopathy in humans (84). The reason for the increased small bowel glutaminase activity in germ-free mice is still unclear but could be related to disinhibition due to absent microbial metabolites.

Overall, the majority of germ-free rodents appear to be more susceptible to acute (toxic) liver injury and liver fibrosis/cirrhosis, which could be due to lack of metabolism of the toxic substances by the gut microbiome, a different intestinal absorption, hepatic uptake, or microsomal activation (95). The different phenotype could also be due to absent LPS in germ-free mice, as it was already shown decades ago (106) that LPS has multiple effects on the liver such as modulation of bile secretion and hepatic blood flow and could thereby result in a different resistance of the liver to toxins.

ANTIBIOTICS VERSUS GERM-FREE STATUS IN EXPERIMENTAL LIVER DISEASE

The complete absence of microbes confers sometimes protection, sometimes exacerbation of certain liver diseases. Does reducing the number of intestinal microbes using antibiotics lead to the same liver findings as reported with a complete absence of microbiota in germ-free mice? Although antibiotics can be associated with hepatotoxicity (10, 30), antibiotic treatment generally results in an improved hepatic phenotype in a multitude of experimental liver diseases including alcoholic liver disease (2, 22), NAFLD (9, 44), NASH (43, 114), HCC (26, 115), AIH (20, 48), PSC (72), PBC (90), toxic liver injury (38, 48), and liver fibrosis/cirrhosis (91, 117; Table 2).

Thus, chronic overstimulation via toxins, metabolites, and TLR ligands present in conventional rodents is harmful. However, no exposure to bacteria and their components and metabolites, as seen in germ-free animals, might not be helpful either.

GNOTOBIOTIC MICE AS MODEL TO STUDY HUMAN MICROBIOTA

Germ-free animals are somewhat artificial systems and might hence not produce physiologically meaningful results as exemplified by discordant findings in germ-free (Table 1) and in antibiotic-treated rodents (Table 2). How can we use them to mirror reality better? We can create gnotobiotic rodents colonized with only one known bacterial strain (monoassociated) or a specific set of known bacterial strains to examine the impact on the disease process; this allows investigation of the contribution of single or a specified set of bacteria to disease pathogenesis. The latter is sometimes hard to achieve, as humans and mice are colonized by multiple and often poorly defined microbes.

We can also “humanize” germ-free mice, or colonize them with human feces, to reproduce human disease better in rodents (Fig. 1). This could be done in cases where only suboptimal rodent disease models exist such as in alcoholic hepatitis: Germ-free mice were transplanted with human microbiota from patients with alcoholism and no hepatitis, nonsevere alcoholic hepatitis, or severe alcoholic hepatitis, which conferred susceptibility to more severe liver disease in mice transplanted with microbiota from the sickest patients (59). The link between intestinal dysbiosis and alcoholic liver disease was further strengthened by subsequent transplantation of microbiota from patients with no alcoholic hepatitis to mice previously transplanted with microbiota from patients with severe alcoholic hepatitis as these mice exhibited markedly improved ALT levels compared with mice that did not receive the subsequent fecal transplant (59). Studies as such could not be executed in humans because of ethical concerns, nor could they achieve such clinical relevance without humanizing rodents with human microbiota. Humanizing animals can also help us to understand carcinogenesis: After administration of the carcinogen azoxymethane, germ-free mice/antibiotic-treated mice were gavaged with microbiota from patients with colorectal cancer and were found to be more susceptible to tumorigenesis than mice gavaged with microbiota from healthy controls or gavaged with no microbiota (113). Again, these studies cannot be performed in humans, nor would an animal study have as much relevance for human disease as humanized models can have. Humanized models can also be used for precision medicine approaches such as precise editing of the intestinal microbiome: Mice transplanted with intestinal microbiota from patients with inflammatory bowel disease showed significantly decreased concentrations of Enterobacteriaceae after treatment with tungstate that selectively blocked metabolic pathways in Enterobacteriaceae; this was associated with markedly improved intestinal inflammation (121).

Hence, gnotobiotic or humanized models, despite being more costly than germ-free or conventional rodents, appear to be alternatives to overcome oftentimes dissimilar phenotypes between rodents and humans, in particular in microbiome research. However, posttransplant analyses of the intestinal microbiome should be performed to confirm successful transplantation, as not all bacteria are being transferred 1:1 because of a different microenvironment.

SUMMARY AND CONCLUSIONS

Germ-free rodents fulfill a central function in elucidating the role of the intestinal microbiota in the pathogenesis of liver diseases. Germ-free status can occasionally have divergent effects in different mouse or rat strains and even within the same strain can show completely opposite outcomes in different liver diseases. Antibiotic treatment is usually associated with ameliorated liver disease in preclinical models. The different phenotypes of germ-free rodents in various liver diseases in relation to conventional rodents can be explained by an immature immune system and a markedly altered metabolism with regard to lipids, cholesterol, xenobiotics, toxins, and bile acids. To advance understanding of the interactions between host and intestinal microbiota, simplified model systems such as gnotobiotic mice monoassociated with a single bacterial strain or colonized with a defined set of microbes constitute unique models for investigation of this complex ecosystem. In addition, humanized gnotobiotic mice allow us to study bacteria not present in mice and to exclude bacteria found in mice but not in humans. Humanized gnotobiotic mice will be very useful when microbiome-centered therapies are tested to treat human disease.

GRANTS

This study was supported in part by National Institutes of Health Grants R01-AA-020703, R01-AA-24726, U01-AA-021856, and U01-AA-026939 and by Award I01BX002213 from the Biomedical Laboratory Research & Development Service of the Veterans Affairs Office of Research & Development (to B. Schnabl).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

P.H. and H.C. drafted manuscript; P.H., H.C., Y.D., and B.S. edited and revised manuscript; P.H., H.C., Y.D., and B.S. approved final version of manuscript.

REFERENCES

  • 1.Abu-Shanab A, Quigley EM. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 7: 691–701, 2010. doi: 10.1038/nrgastro.2010.172. [DOI] [PubMed] [Google Scholar]
  • 2.Adachi Y, Moore LE, Bradford BU, Gao W, Thurman RG. Antibiotics prevent liver injury in rats following long-term exposure to ethanol. Gastroenterology 108: 218–224, 1995. doi: 10.1016/0016-5085(95)90027-6. [DOI] [PubMed] [Google Scholar]
  • 3.Altekruse SF, Henley SJ, Cucinelli JE, McGlynn KA. Changing hepatocellular carcinoma incidence and liver cancer mortality rates in the United States. Am J Gastroenterol 109: 542–553, 2014. doi: 10.1038/ajg.2014.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 101: 15718–15723, 2004. doi: 10.1073/pnas.0407076101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 104: 979–984, 2007. doi: 10.1073/pnas.0605374104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, White MB, Monteith P, Noble NA, Unser AB, Daita K, Fisher AR, Sikaroodi M, Gillevet PM. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol 60: 940–947, 2014. doi: 10.1016/j.jhep.2013.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bauer H, Horowitz RE, Levenson SM, Popper H. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am J Pathol 42: 471–483, 1963. [PMC free article] [PubMed] [Google Scholar]
  • 8.Bauer H, Paronetto F, Porro RF, Einheber A, Popper H. The influence of the microbial flora on liver injury and associated serum gamma-globulin elevation. A study in germ-free rats treated with 3′-methyl-4-dimethylaminoazobenzene. Lab Invest 16: 847–857, 1967. [PubMed] [Google Scholar]
  • 9.Bergheim I, Weber S, Vos M, Krämer S, Volynets V, Kaserouni S, McClain CJ, Bischoff SC. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: role of endotoxin. J Hepatol 48: 983–992, 2008. doi: 10.1016/j.jhep.2008.01.035. [DOI] [PubMed] [Google Scholar]
  • 10.Bernal W, Auzinger G, Dhawan A, Wendon J. Acute liver failure. Lancet 376: 190–201, 2010. doi: 10.1016/S0140-6736(10)60274-7. [DOI] [PubMed] [Google Scholar]
  • 11.Bindels LB, Segura Munoz RR, Gomes-Neto JC, Mutemberezi V, Martínez I, Salazar N, Cody EA, Quintero-Villegas MI, Kittana H, de Los Reyes-Gavilán CG, Schmaltz RJ, Muccioli GG, Walter J, Ramer-Tait AE. Resistant starch can improve insulin sensitivity independently of the gut microbiota. Microbiome 5: 12, 2017. doi: 10.1186/s40168-017-0230-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Björkholm B, Bok CM, Lundin A, Rafter J, Hibberd ML, Pettersson S. Intestinal microbiota regulate xenobiotic metabolism in the liver. PLoS One 4: e6958, 2009. doi: 10.1371/journal.pone.0006958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brandi G, De Lorenzo S, Candela M, Pantaleo MA, Bellentani S, Tovoli F, Saccoccio G, Biasco G. Microbiota, NASH, HCC and the potential role of probiotics. Carcinogenesis 38: 231–240, 2017. doi: 10.1093/carcin/bgx007. [DOI] [PubMed] [Google Scholar]
  • 14.Brenner DA, Paik YH, Schnabl B. Role of gut microbiota in liver disease. J Clin Gastroenterol 49, Suppl 1: S25–S27, 2015. doi: 10.1097/MCG.0000000000000391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bugianesi E, Moscatiello S, Ciaravella MF, Marchesini G. Insulin resistance in nonalcoholic fatty liver disease. Curr Pharm Des 16: 1941–1951, 2010. doi: 10.2174/138161210791208875. [DOI] [PubMed] [Google Scholar]
  • 16.Burt AD, Lackner C, Tiniakos DG. Diagnosis and assessment of NAFLD: definitions and histopathological classification. Semin Liver Dis 35: 207–220, 2015. doi: 10.1055/s-0035-1562942. [DOI] [PubMed] [Google Scholar]
  • 17.Burton DG. Cellular senescence, ageing and disease. Age (Dordr) 31: 1–9, 2009. doi: 10.1007/s11357-008-9075-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Canesso MC, Lacerda NL, Ferreira CM, Gonçalves G, Almeida D, Gamba C, Cassali G, Pedroso SH, Moreira C, Martins FS, Nicoli JR, Teixeira MM, Godard G, Vieira AT. Comparing the effects of acute alcohol consumption in germ-free and conventional mice: the role of the gut microbiota. BMC Microbiol 14: 240, 2014. doi: 10.1186/s12866-014-0240-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Carey EJ, Ali AH, Lindor KD. Primary biliary cirrhosis. Lancet 386: 1565–1575, 2015. doi: 10.1016/S0140-6736(15)00154-3. [DOI] [PubMed] [Google Scholar]
  • 20.Celaj S, Gleeson MW, Deng J, O’Toole GA, Hampton TH, Toft MF, Morrison HG, Sogin ML, Putra J, Suriawinata AA, Gorham JD. The microbiota regulates susceptibility to Fas-mediated acute hepatic injury. Lab Invest 94: 938–949, 2014. doi: 10.1038/labinvest.2014.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen P, Miyamoto Y, Mazagova M, Lee KC, Eckmann L, Schnabl B. Microbiota protects mice against acute alcohol-induced liver injury. Alcohol Clin Exp Res 39: 2313–2323, 2015. doi: 10.1111/acer.12900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chen P, Stärkel P, Turner JR, Ho SB, Schnabl B. Dysbiosis-induced intestinal inflammation activates tumor necrosis factor receptor I and mediates alcoholic liver disease in mice. Hepatology 61: 883–894, 2015. doi: 10.1002/hep.27489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chyan YJ, Poeggeler B, Omar RA, Chain DG, Frangione B, Ghiso J, Pappolla MA. Potent neuroprotective properties against the Alzheimer β-amyloid by an endogenous melatonin-related indole structure, indole-3-propionic acid. J Biol Chem 274: 21937–21942, 1999. doi: 10.1074/jbc.274.31.21937. [DOI] [PubMed] [Google Scholar]
  • 24.Cornell RP, Liljequist BL, Bartizal KF. Depressed liver regeneration after partial hepatectomy of germ-free, athymic and lipopolysaccharide-resistant mice. Hepatology 11: 916–922, 1990. doi: 10.1002/hep.1840110603. [DOI] [PubMed] [Google Scholar]
  • 25.Dam H. The formation of coprosterol in the intestine: the action of intestinal bacteria on cholesterol. Biochem J 28: 820–825, 1934. doi: 10.1042/bj0280820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 21: 504–516, 2012. doi: 10.1016/j.ccr.2012.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Debré P, Le Gall JY; Commission I (Biologie) . [Intestinal microbiota]. Bull Acad Natl Med 198: 1667–1684, 2014. [PubMed] [Google Scholar]
  • 28.Delzenne NM, Cani PD. Interaction between obesity and the gut microbiota: relevance in nutrition. Annu Rev Nutr 31: 15–31, 2011. doi: 10.1146/annurev-nutr-072610-145146. [DOI] [PubMed] [Google Scholar]
  • 29.DeNucci SM, Tong M, Longato L, Lawton M, Setshedi M, Carlson RI, Wands JR, de la Monte SM. Rat strain differences in susceptibility to alcohol-induced chronic liver injury and hepatic insulin resistance. Gastroenterol Res Pract 2010: 312790, 2010. doi: 10.1155/2010/312790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Einheber A, Rosen H, Wren RE, Beaudry NN. The role of microbial flora in the hepatotoxicity of chlortetracycline in vivo: a study with germfree mice. Biochem Pharmacol 15: 1093–1104, 1966. doi: 10.1016/0006-2952(66)90274-7. [DOI] [PubMed] [Google Scholar]
  • 31.Eisa-Beygi S, Ekker M, Moon TW, Macdonald RL, Wen XY. Developmental processes regulated by the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) pathway: highlights from animal studies. Reprod Toxicol 46: 115–120, 2014. doi: 10.1016/j.reprotox.2014.04.001. [DOI] [PubMed] [Google Scholar]
  • 32.Eyssen HJ, Parmentier GG, Compernolle FC, De Pauw G, Piessens-Denef M. Biohydrogenation of sterols by Eubacterium ATCC 21,408: Nova species. Eur J Biochem 36: 411–421, 1973. doi: 10.1111/j.1432-1033.1973.tb02926.x. [DOI] [PubMed] [Google Scholar]
  • 33.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. Fecal microbiota manipulation prevents dysbiosis and alcohol-induced liver injury in mice. J Hepatol 66: 806–815, 2017. doi: 10.1016/j.jhep.2016.11.008. [DOI] [PubMed] [Google Scholar]
  • 34.Fleissner CK, Huebel N, Abd El-Bary MM, Loh G, Klaus S, Blaut M. Absence of intestinal microbiota does not protect mice from diet-induced obesity. Br J Nutr 104: 919–929, 2010. doi: 10.1017/S0007114510001303. [DOI] [PubMed] [Google Scholar]
  • 35.Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science 352: 539–544, 2016. doi: 10.1126/science.aad9378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gérard P, Lepercq P, Leclerc M, Gavini F, Raibaud P, Juste C. Bacteroides sp. strain D8, the first cholesterol-reducing bacterium isolated from human feces. Appl Environ Microbiol 73: 5742–5749, 2007. doi: 10.1128/AEM.02806-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Glimstedt G. Bakterienfreie Meerschweinchen: Aufzucht, Lebensfähigkeit und Wachstum, nebst Untersuchungen über das lymphatische Gewebe. In: Acta Pathologica et Microbiologica Scandinavica Supplementum Lund, Sweden: Ohlssons, 1936, vol. 30, p. 295. [Google Scholar]
  • 38.Gong S, Lan T, Zeng L, Luo H, Yang X, Li N, Chen X, Liu Z, Li R, Win S, Liu S, Zhou H, Schnabl B, Jiang Y, Kaplowitz N, Chen P. Gut microbiota mediates diurnal variation of acetaminophen induced acute liver injury in mice. J Hepatol 69: 51–59, 2018. doi: 10.1016/j.jhep.2018.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Grant GA, Roe FJ. Influence of germ-free status on hepatoma induction by 7,12-dimethylbenz(a)anthracene in C3H mice. Nature 222: 1282–1283, 1969. doi: 10.1038/2221282a0. [DOI] [PubMed] [Google Scholar]
  • 40.Hartmann P, Chen WC, Schnabl B. The intestinal microbiome and the leaky gut as therapeutic targets in alcoholic liver disease. Front Physiol 3: 402, 2012. doi: 10.3389/fphys.2012.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hartmann P, Schnabl B. Risk factors for progression of and treatment options for NAFLD in children. Clin Liver Dis (Hoboken) 11: 11–15, 2018. doi: 10.1002/cld.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hartmann P, Seebauer CT, Schnabl B. Alcoholic liver disease: the gut microbiome and liver cross talk. Alcohol Clin Exp Res 39: 763–775, 2015. doi: 10.1111/acer.12704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482: 179–185, 2012. doi: 10.1038/nature10809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jiang C, Xie C, Li F, Zhang L, Nichols RG, Krausz KW, Cai J, Qi Y, Fang ZZ, Takahashi S, Tanaka N, Desai D, Amin SG, Albert I, Patterson AD, Gonzalez FJ. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest 125: 386–402, 2015. doi: 10.1172/JCI76738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kaden-Volynets V, Basic M, Neumann U, Pretz D, Rings A, Bleich A, Bischoff SC. Lack of liver steatosis in germ-free mice following hypercaloric diets. Eur J Nutr 1–13, 2018. doi: 10.1007/s00394-018-1748-4. [DOI] [PubMed] [Google Scholar]
  • 46.Kang DJ, Betrapally NS, Ghosh SA, Sartor RB, Hylemon PB, Gillevet PM, Sanyal AJ, Heuman DM, Carl D, Zhou H, Liu R, Wang X, Yang J, Jiao C, Herzog J, Lippman HR, Sikaroodi M, Brown RR, Bajaj JS. Gut microbiota drive the development of neuroinflammatory response in cirrhosis in mice. Hepatology 64: 1232–1248, 2016. doi: 10.1002/hep.28696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Karbownik M, Reiter RJ, Garcia JJ, Cabrera J, Burkhardt S, Osuna C, Lewiński A. Indole-3-propionic acid, a melatonin-related molecule, protects hepatic microsomal membranes from iron-induced oxidative damage: relevance to cancer reduction. J Cell Biochem 81: 507–513, 2001. doi:. [DOI] [PubMed] [Google Scholar]
  • 48.Kataoka K, Kono Y, Sugimoto M, Furuichi Y, Shichiri M, Tanaka Y. Hepatocyte-protective and anti-oxidant effects of rifampicin on human chronic hepatitis C and murine acute hepatocyte disorder. Exp Ther Med 1: 1041–1047, 2010. doi: 10.3892/etm.2010.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kurnath P, Dearing MD. Warmer ambient temperatures depress liver function in a mammalian herbivore. Biol Lett 9: 20130562, 2013. doi: 10.1098/rsbl.2013.0562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Laqueur GL, Matsumoto H, Yamamoto RS. Comparison of the carcinogenicity of methylazoxymethanol-β-d-glucosiduronic acid in conventional and germfree Sprague-Dawley rats. J Natl Cancer Inst 67: 1053–1055, 1981. doi: 10.1093/jnci/67.5.1053. [DOI] [PubMed] [Google Scholar]
  • 51.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. Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci USA 111: E4485–E4493, 2014. doi: 10.1073/pnas.1415174111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.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. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 62: 1787–1794, 2013. doi: 10.1136/gutjnl-2012-303816. [DOI] [PubMed] [Google Scholar]
  • 53.Levenson SM, Nagler AL, Geever EF, Seifter E. Acute choline deficiency in germfree, conventionalized and open-animal-room rats: effects of neomycin, chlortetracycline, vitamin B12 and coprophagy prevention. J Nutr 95: 247–270, 1968. doi: 10.1093/jn/95.2.247. [DOI] [PubMed] [Google Scholar]
  • 54.Levenson SM, Tennant B. Some metabolic and nutritional studies with germfree animals. Fed Proc 22: 109–119, 1963. [PubMed] [Google Scholar]
  • 55.Levy C, Lindor KD. Primary sclerosing cholangitis: epidemiology, natural history, and prognosis. Semin Liver Dis 26: 22–30, 2006. doi: 10.1055/s-2006-933560. [DOI] [PubMed] [Google Scholar]
  • 56.Levy M, Thaiss CA, Zeevi D, Dohnalová L, Zilberman-Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, Pevsner-Fischer M, Shapiro H, Christ A, Harmelin A, Halpern Z, Latz E, Flavell RA, Amit I, Segal E, Elinav E. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell 163: 1428–1443, 2015. doi: 10.1016/j.cell.2015.10.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lichtman SN, Sartor RB, Keku J, Schwab JH. Hepatic inflammation in rats with experimental small intestinal bacterial overgrowth. Gastroenterology 98: 414–423, 1990. doi: 10.1016/0016-5085(90)90833-M. [DOI] [PubMed] [Google Scholar]
  • 58.Lin R, Zhou L, Zhang J, Wang B. Abnormal intestinal permeability and microbiota in patients with autoimmune hepatitis. Int J Clin Exp Pathol 8: 5153–5160, 2015. [PMC free article] [PubMed] [Google Scholar]
  • 59.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. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut 65: 830–839, 2016. doi: 10.1136/gutjnl-2015-310585. [DOI] [PubMed] [Google Scholar]
  • 60.Luckey TD. Introduction to intestinal microecology. Am J Clin Nutr 25: 1292–1294, 1972. doi: 10.1093/ajcn/25.12.1292. [DOI] [PubMed] [Google Scholar]
  • 61.Ma X, Hua J, Li Z. Probiotics improve high fat diet-induced hepatic steatosis and insulin resistance by increasing hepatic NKT cells. J Hepatol 49: 821–830, 2008. doi: 10.1016/j.jhep.2008.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mackay IR. Autoimmune hepatitis: what must be said. Exp Mol Pathol 93: 350–353, 2012. doi: 10.1016/j.yexmp.2012.11.001. [DOI] [PubMed] [Google Scholar]
  • 63.Mariathasan S, Newton K, Monack DM, Vucic D, French DM, Lee WP, Roose-Girma M, Erickson S, Dixit VM. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430: 213–218, 2004. doi: 10.1038/nature02664. [DOI] [PubMed] [Google Scholar]
  • 64.Mattner J. Impact of microbes on the pathogenesis of primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC). Int J Mol Sci 17: 1864, 2016. doi: 10.3390/ijms17111864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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. Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice. FASEB J 29: 1043–1055, 2015. doi: 10.1096/fj.14-259515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mizutani T, Mitsuoka T. Effect of dietary phenobarbital on spontaneous hepatic tumorigenesis in germfree C3H/He male mice. Cancer Lett 39: 233–237, 1988. doi: 10.1016/0304-3835(88)90065-1. [DOI] [PubMed] [Google Scholar]
  • 67.Mizutani T, Mitsuoka T. Effect of intestinal bacteria on incidence of liver tumors in gnotobiotic C3H/He male mice. J Natl Cancer Inst 63: 1365–1370, 1979. doi: 10.1093/jnci/63.6.1365. [DOI] [PubMed] [Google Scholar]
  • 68.Mizutani T, Mitsuoka T. Inhibitory effect of some intestinal bacteria on liver tumorigenesis in gnotobiotic C3H/He male mice. Cancer Lett 11: 89–95, 1980. doi: 10.1016/0304-3835(80)90098-1. [DOI] [PubMed] [Google Scholar]
  • 69.Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK, Fischer SE, McGilvray ID, Allard JP. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 58: 120–127, 2013. doi: 10.1002/hep.26319. [DOI] [PubMed] [Google Scholar]
  • 70.Novikova DS, Garabadzhiu AV, Melino G, Barlev NA, Tribulovich VG. AMP-activated protein kinase: structure, function, and role in pathological processes. Biochemistry (Mosc) 80: 127–144, 2015. doi: 10.1134/S0006297915020017. [DOI] [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. Lactobacillus casei strain Shirota protects against nonalcoholic steatohepatitis development in a rodent model. Am J Physiol Gastrointest Liver Physiol 305: G911–G918, 2013. doi: 10.1152/ajpgi.00225.2013. [DOI] [PubMed] [Google Scholar]
  • 72.Peng ZW, Rothweiler S, Wei G, Ikenaga N, Liu SB, Sverdlov DY, Vaid KA, Longhi MS, Kuang M, Robson SC, Popov YV. The ectonucleotidase ENTPD1/CD39 limits biliary injury and fibrosis in mouse models of sclerosing cholangitis. Hepatol Commun 1: 957–972, 2017. doi: 10.1002/hep4.1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pollard M, Luckert PH, Snyder D. Prevention of prostate cancer and liver tumors in L-W rats by moderate dietary restriction. Cancer 64: 686–690, 1989. doi:. [DOI] [PubMed] [Google Scholar]
  • 74.Pollard M, Sharon N, Chang CF. Synthesis of hepatotoxic agents in germfree and conventional mice which had been fed NaNO2 and dimethylamine. Proc Soc Exp Biol Med 140: 1073–1076, 1972. doi: 10.3181/00379727-140-36614. [DOI] [PubMed] [Google Scholar]
  • 75.Popper H, Bauer H, Horowitz R, Paronetto F; Mount Sinai Hospital New York . Histopathology of Germfree Animals. Arlington, VA: Armed Services Technical Information Agency, 1963, 15 pp. (Publ. AD-403868) [Google Scholar]
  • 76.Possamai LA, McPhail MJ, Khamri W, Wu B, Concas D, Harrison M, Williams R, Cox RD, Cox IJ, Anstee QM, Thursz MR. The role of intestinal microbiota in murine models of acetaminophen-induced hepatotoxicity. Liver Int 35: 764–773, 2015. doi: 10.1111/liv.12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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. Alterations of the human gut microbiome in liver cirrhosis. Nature 513: 59–64, 2014. doi: 10.1038/nature13568. [DOI] [PubMed] [Google Scholar]
  • 78.Quigley EM. Primary biliary cirrhosis and the microbiome. Semin Liver Dis 36: 349–353, 2016. doi: 10.1055/s-0036-1594006. [DOI] [PubMed] [Google Scholar]
  • 79.Rabot S, Membrez M, Bruneau A, Gérard P, Harach T, Moser M, Raymond F, Mansourian R, Chou CJ. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J 24: 4948–4959, 2010. doi: 10.1096/fj.10-164921. [DOI] [PubMed] [Google Scholar]
  • 80.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. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 11: 868–875.e1-3, 2013. doi: 10.1016/j.cgh.2013.02.015. [DOI] [PubMed] [Google Scholar]
  • 81.Ren T, Huang C, Cheng M. Dietary blueberry and bifidobacteria attenuate nonalcoholic fatty liver disease in rats by affecting SIRT1-mediated signaling pathway. Oxid Med Cell Longev 2014: 469059, 2014. doi: 10.1155/2014/469059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Repa JJ, Berge KE, Pomajzl C, Richardson JA, Hobbs H, Mangelsdorf DJ. Regulation of ATP-binding cassette sterol transporters ABCG5 and ABCG8 by the liver X receptors α and β. J Biol Chem 277: 18793–18800, 2002. doi: 10.1074/jbc.M109927200. [DOI] [PubMed] [Google Scholar]
  • 83.Roe FJ, Grant GA. Inhibition by germ-free status of development of liver and lung tumours in mice exposed neonatally to 7,12-dimethylbenz(a)anthracene: implications in relation to tests for carcinogenicity. Int J Cancer 6: 133–144, 1970. doi: 10.1002/ijc.2910060117. [DOI] [PubMed] [Google Scholar]
  • 84.Romero-Gómez M. Role of phosphate-activated glutaminase in the pathogenesis of hepatic encephalopathy. Metab Brain Dis 20: 319–325, 2005. doi: 10.1007/s11011-005-7913-5. [DOI] [PubMed] [Google Scholar]
  • 85.Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, Tuohy K. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 57: 1–24, 2018. doi: 10.1007/s00394-017-1445-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sabino J, Vieira-Silva S, Machiels K, Joossens M, Falony G, Ballet V, Ferrante M, Van Assche G, Van der Merwe S, Vermeire S, Raes J. Primary sclerosing cholangitis is characterised by intestinal dysbiosis independent from IBD. Gut 65: 1681–1689, 2016. doi: 10.1136/gutjnl-2015-311004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Saltzman ET, Palacios T, Thomsen M, Vitetta L. Intestinal microbiome shifts, dysbiosis, inflammation, and non-alcoholic fatty liver disease. Front Microbiol 9: 61, 2018. doi: 10.3389/fmicb.2018.00061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 146: 1513–1524, 2014. doi: 10.1053/j.gastro.2014.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Schnabl B, Scholten D, Brenner DA. What is the potential role of antifibrotic agents for the treatment of liver disease? Nat Clin Pract Gastroenterol Hepatol 5: 496–497, 2008. doi: 10.1038/ncpgasthep1200. [DOI] [PubMed] [Google Scholar]
  • 90.Schrumpf E, Kummen M, Valestrand L, Greiner TU, Holm K, Arulampalam V, Reims HM, Baines J, Bäckhed F, Karlsen TH, Blumberg RS, Hov JR, Melum E. The gut microbiota contributes to a mouse model of spontaneous bile duct inflammation. J Hepatol 66: 382–389, 2017. doi: 10.1016/j.jhep.2016.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, Schwabe RF. TLR4 enhances TGF-β signaling and hepatic fibrosis. Nat Med 13: 1324–1332, 2007. doi: 10.1038/nm1663. [DOI] [PubMed] [Google Scholar]
  • 92.Seki E, Schnabl B. Role of innate immunity and the microbiota in liver fibrosis: crosstalk between the liver and gut. J Physiol 590: 447–458, 2012. doi: 10.1113/jphysiol.2011.219691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 164: 337–340, 2016. doi: 10.1016/j.cell.2016.01.013. [DOI] [PubMed] [Google Scholar]
  • 94.Silverstone H, Tannenbaum A. Proportion of dietary protein and the formation of spontaneous hepatomas in the mouse. Cancer Res 11: 442–446, 1951. [PubMed] [Google Scholar]
  • 95.Sumi Y, Miyakawa M. Susceptibility of germ-free rats to the hepatotoxic effects of dimethylnitrosamine or dimethylamine plus sodium nitrite administered orally. Cancer Res 43: 2942–2946, 1983. [PubMed] [Google Scholar]
  • 96.Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, Nicholson JK, Holmes E. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc Natl Acad Sci USA 108, Suppl 1: 4523–4530, 2011. doi: 10.1073/pnas.1006734107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Tabibian JH, Lindor KD. Primary sclerosing cholangitis: a review and update on therapeutic developments. Expert Rev Gastroenterol Hepatol 7: 103–114, 2013. doi: 10.1586/egh.12.80. [DOI] [PubMed] [Google Scholar]
  • 98.Tabibian JH, O’Hara SP, Splinter PL, Trussoni CE, LaRusso NF. Cholangiocyte senescence by way of N-ras activation is a characteristic of primary sclerosing cholangitis. Hepatology 59: 2263–2275, 2014. doi: 10.1002/hep.26993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Tabibian JH, O’Hara SP, Trussoni CE, Tietz PS, Splinter PL, Mounajjed T, Hagey LR, LaRusso NF. Absence of the intestinal microbiota exacerbates hepatobiliary disease in a murine model of primary sclerosing cholangitis. Hepatology 63: 185–196, 2016. doi: 10.1002/hep.27927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tang R, Wei Y, Li Y, Chen W, Chen H, Wang Q, Yang F, Miao Q, Xiao X, Zhang H, Lian M, Jiang X, Zhang J, Cao Q, Fan Z, Wu M, Qiu D, Fang JY, Ansari A, Gershwin ME, Ma X. Gut microbial profile is altered in primary biliary cholangitis and partially restored after UDCA therapy. Gut 67: 534–541, 2018. doi: 10.1136/gutjnl-2016-313332. [DOI] [PubMed] [Google Scholar]
  • 101.Tannenbaum A, Silverstone H. The influence of the degree of caloric restriction on the formation of skin tumors and hepatomas in mice. Cancer Res 9: 724–727, 1949. [PubMed] [Google Scholar]
  • 102.Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest 123: 966–972, 2013. doi: 10.1172/JCI64098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Teli MR, Day CP, Burt AD, Bennett MK, James OF. Determinants of progression to cirrhosis or fibrosis in pure alcoholic fatty liver. Lancet 346: 987–990, 1995. doi: 10.1016/S0140-6736(95)91685-7. [DOI] [PubMed] [Google Scholar]
  • 104.Tsuchiya M, Ji C, Kosyk O, Shymonyak S, Melnyk S, Kono H, Tryndyak V, Muskhelishvili L, Pogribny IP, Kaplowitz N, Rusyn I. Interstrain differences in liver injury and one-carbon metabolism in alcohol-fed mice. Hepatology 56: 130–139, 2012. doi: 10.1002/hep.25641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tsutsui H, Nishiguchi S. Importance of Kupffer cells in the development of acute liver injuries in mice. Int J Mol Sci 15: 7711–7730, 2014. doi: 10.3390/ijms15057711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Utili R, Abernathy CO, Zimmerman HJ. Endotoxin effects on the liver. Life Sci 20: 553–568, 1977. doi: 10.1016/0024-3205(77)90458-1. [DOI] [PubMed] [Google Scholar]
  • 107.Vega RB, Huss JM, Kelly DP. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol 20: 1868–1876, 2000. doi: 10.1128/MCB.20.5.1868-1876.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wagnerberger S, Spruss A, Kanuri G, Stahl C, Schröder M, Vetter W, Bischoff SC, Bergheim I. Lactobacillus casei Shirota protects from fructose-induced liver steatosis: a mouse model. J Nutr Biochem 24: 531–538, 2013. doi: 10.1016/j.jnutbio.2012.01.014. [DOI] [PubMed] [Google Scholar]
  • 109.Wei Y, Zeng B, Chen J, Cui G, Lu C, Wu W, Yang J, Wei H, Xue R, Bai L, Chen Z, Li L, Iwabuchi K, Uede T, Van Kaer L, Diao H. Enterogenous bacterial glycolipids are required for the generation of natural killer T cells mediated liver injury. Sci Rep 6: 36365, 2016. doi: 10.1038/srep36365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Weismüller TJ, Wedemeyer J, Kubicka S, Strassburg CP, Manns MP. The challenges in primary sclerosing cholangitis: aetiopathogenesis, autoimmunity, management and malignancy. J Hepatol 48, Suppl 1: S38–S57, 2008. doi: 10.1016/j.jhep.2008.01.020. [DOI] [PubMed] [Google Scholar]
  • 111.Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci USA 106: 3698–3703, 2009. doi: 10.1073/pnas.0812874106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, Landt CL, Harrison SA. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology 140: 124–131, 2011. doi: 10.1053/j.gastro.2010.09.038. [DOI] [PubMed] [Google Scholar]
  • 113.Wong SH, Zhao L, Zhang X, Nakatsu G, Han J, Xu W, Xiao X, Kwong TN, Tsoi H, Wu WK, Zeng B, Chan FK, Sung JJ, Wei H, Yu J. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology 153: 1621–1633.e6, 2017. doi: 10.1053/j.gastro.2017.08.022. [DOI] [PubMed] [Google Scholar]
  • 114.Yamada S, Kamada N, Amiya T, Nakamoto N, Nakaoka T, Kimura M, Saito Y, Ejima C, Kanai T, Saito H. Gut microbiota-mediated generation of saturated fatty acids elicits inflammation in the liver in murine high-fat diet-induced steatohepatitis. BMC Gastroenterol 17: 136, 2017. doi: 10.1186/s12876-017-0689-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.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. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499: 97–101, 2013. [Erratum in Nature 506: 396, 2014.] doi: 10.1038/nature12347. [DOI] [PubMed] [Google Scholar]
  • 116.Yuksel M, Wang Y, Tai N, Peng J, Guo J, Beland K, Lapierre P, David C, Alvarez F, Colle I, Yan H, Mieli-Vergani G, Vergani D, Ma Y, Wen L. A novel “humanized mouse” model for autoimmune hepatitis and the association of gut microbiota with liver inflammation. Hepatology 62: 1536–1550, 2015. doi: 10.1002/hep.27998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhang M, Song G, Minuk GY. Effects of hepatic stimulator substance, herbal medicine, selenium/vitamin E, and ciprofloxacin on cirrhosis in the rat. Gastroenterology 110: 1150–1155, 1996. doi: 10.1053/gast.1996.v110.pm8613004. [DOI] [PubMed] [Google Scholar]
  • 118.Zhou D, Pan Q, Shen F, Cao HX, Ding WJ, Chen YW, Fan JG. Total fecal microbiota transplantation alleviates high-fat diet-induced steatohepatitis in mice via beneficial regulation of gut microbiota. Sci Rep 7: 1529, 2017. doi: 10.1038/s41598-017-01751-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhou L, Foster JA. Psychobiotics and the gut-brain axis: in the pursuit of happiness. Neuropsychiatr Dis Treat 11: 715–723, 2015. doi: 10.2147/NDT.S61997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57: 601–609, 2013. doi: 10.1002/hep.26093. [DOI] [PubMed] [Google Scholar]
  • 121.Zhu W, Winter MG, Byndloss MX, Spiga L, Duerkop BA, Hughes ER, Büttner L, de Lima Romão E, Behrendt CL, Lopez CA, Sifuentes-Dominguez L, Huff-Hardy K, Wilson RP, Gillis CC, Tükel Ç, Koh AY, Burstein E, Hooper LV, Bäumler AJ, Winter SE. Precision editing of the gut microbiota ameliorates colitis. Nature 553: 208–211, 2018. doi: 10.1038/nature25172. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Gastrointestinal and Liver Physiology are provided here courtesy of American Physiological Society

RESOURCES