Next Article in Journal
The Fetus Can Teach Us: Oxygen and the Pulmonary Vasculature
Next Article in Special Issue
Imaging Features of Non-Alcoholic Fatty Liver Disease in Children and Adolescents
Previous Article in Journal
Trending Longitudinal Agreement between Parent and Child Perceptions of Quality of Life for Pediatric Palliative Care Patients
Previous Article in Special Issue
The Association between Non-Alcoholic Fatty Liver Disease and Cardiovascular Risk in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut–Liver Axis Derangement in Non-Alcoholic Fatty Liver Disease

1
Pediatrics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”—University of Salerno, 84081 Baronissi (Salerno), Italy
2
Pediatrics Residency Joint Programs, University of Naples Federico II, 80131 Naples, Italy & University of Salerno, 84081 Baronissi (Salerno), Italy
*
Author to whom correspondence should be addressed.
Submission received: 13 May 2017 / Revised: 18 July 2017 / Accepted: 21 July 2017 / Published: 2 August 2017
(This article belongs to the Special Issue Pediatric Nonalcoholic Fatty Liver Disease)

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the most frequent type of chronic liver disease in the pediatric age group, paralleling an obesity pandemic. A “multiple-hit” hypothesis has been invoked to explain its pathogenesis. The “first hit” is liver lipid accumulation in obese children with insulin resistance. In the absence of significant lifestyle modifications leading to weight loss and increased physical activity, other factors may act as “second hits” implicated in liver damage progression leading to more severe forms of inflammation and hepatic fibrosis. In this regard, the gut–liver axis (GLA) seems to play a central role. Principal players are the gut microbiota, its bacterial products, and the intestinal barrier. A derangement of GLA (namely, dysbiosis and altered intestinal permeability) may promote bacteria/bacterial product translocation into portal circulation, activation of inflammation via toll-like receptors signaling in hepatocytes, and progression from simple steatosis to non-alcoholic steato-hepatitis (NASH). Among other factors a relevant role has been attributed to the farnesoid X receptor, a nuclear transcriptional factor activated from bile acids chemically modified by gut microbiota (GM) enzymes. The individuation and elucidation of GLA derangement in NAFLD pathomechanisms is of interest at all ages and especially in pediatrics to identify new therapeutic approaches in patients recalcitrant to lifestyle changes. Specific targeting of gut microbiota via pre-/probiotic supplementation, feces transplantation, and farnesoid X receptor modulation appear promising.

Graphical Abstract

1. Introduction

Non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of metabolic syndrome (MetS), is the most frequent form of chronic liver disease worldwide. Liver damage due to lipid accumulation in hepatocytes is a direct consequence of obesity (OB) and insulin-resistance (IR) pandemics related to adoption of a hypercaloric Western diet (WD) rich in saturated fats, meat and refined sugars and poor in vegetables, fruits and fish, along with sedentary lifestyle [1]. Nowadays it represents a major health concern in pediatric population also, where it can affect10% of children, and up to 70% of those who are obese [2]. Probably due to different sex-specific fat metabolism patterns [3], NAFLD distribution appears to be higher in obese male (35.3%) vs. female (21.8%) adolescents [4]. The histological spectrum of NAFLD includes simple hepatic steatosis and its chronical evolution patterns: non-alcoholic steatohepatitis (NASH) with inflammation and fibrosis ending into cirrhosis [5,6]. Although the development of more severe histological pictures up to end-stage liver disease has been described generally later in life, liver transplantation may be already needed in young adulthood [7].
A “multiple-hit” hypothesis may explain NAFLD pathogenesis and progression. In particular in the last few years, a growing interest has been devoted to gut–liver axis (GLA) dysfunction (i.e., intestinal dysbiosis, bacterial overgrowth, alteration of mucosa permeability) as a relevant second hit in NAFLD progression and therefore as possible alternative therapeutic target in those patients unable to get benefits deriving from lifestyle modification, healthy diet and physical activity promotion [1,7]. Objectives of our mini-review are to appraise: (1) the pathophysiology definition of GLA components; (2) the identification of GLA derangements involved in NAFLD pathogenesis and progression; and (3) the delineation of therapeutic perspectives via GLA modulation to prevent and/or treat obesity-related liver disease.

2. Methods

A literature search was performed in the PubMed database using the following MeSH (Medical Subject Headings) terms: “NAFLD” OR “Non-alcoholic fatty liver disease” OR “NASH” OR “Non-alcoholic steatohepatitis” AND “Gut-liver axis” OR “gut microbiota” OR “probiotics” OR “intestinal permeability”. Non-English literature was excluded. Most relevant papers derived from literature search were selected from authors to writing this mini-review.

3. Gut–Liver Axis

The strong anatomical and functional interaction between the gastrointestinal tract and liver defines the term GLA. This intimate connection is already expressed in embryogenesis by the common origin from the ventral foregut endoderm [8]. GLA is characterized by bidirectional traffic. Nutrients and factors derived from gut lumen reach the liver through portal circulation; bile acids produced by hepatocytes are released in the small intestine through the biliary tract. However, this description is simplistic because GLA does not only have a nutritional function. The axis is a complex structure and the alteration, in particular, of two of its components (intestinal barrier and gut microbiota) seems to play a key role in liver damage and progression [9].
The intestinal barrier is composed of: (1) the columnar epithelium of cells bound together by an apical junction complex (tight junctions (TJs) and zonula adherens), gap junctions, and desmosomes; (2) the mucus layer on the epithelial surface consisting of mucin molecules produced by goblet cells; and (3) the antimicrobial defenses provided by Paneth cells, and gut-associated lymphoid tissue (GALT) [10].
The intestinal lumen is naturally colonized by the gut microbiota (GM), consisting of trillions of microorganisms from more than 1000 species, with a total weight of approximately 1–2 kg. As shown in Figure 1, GM has numerous health benefits, and in normal conditions, small amounts of bacterial products enter the liver via portal circulation and most of them are eliminated by hepatic macrophages called Kupffer cells (KCs). In presence of an intestinal dysbiosis and a break of intestinal barrier, it happens that increased bacteria/bacterial products translocation interrupts this immunological tolerance and promotes liver inflammation via toll-like receptor (TLR) stimulation [8].
However, also the liver may influence the microbiome according to the bidirectional definition of GLA. Bile acids in fact may have direct effects on bacteria causing membrane and/or DNA damage. Conversely, secondary molecules derived from bile acids metabolized by GM may activate specific host receptors (i.e., the farnesoid X receptor (FXR)) [11,12].
Bile contains immunoglobulin A (IgA) antibodies released by Peyer’s Patches of biliary submucosa capable of modulating the intestinal microbial load. Furthermore, IgA production is favorably influenced by normal microflora, with health benefits towards infections [13].
GM composition and GLA may therefore influence the antigenic traffic through intestinal barrier and the development of a series of diseases concerning a growing number of extra-intestinal organs [14], including hepatopathies, allergic diseases, diabetes mellitus type 1, familial Mediterranean fever, autism, cardiovascular disease, and dysfunctions in bone mass and immune response [15]. Here we will focus on the role of GLA components in NAFLD pathogenesis and progression.

4. Gut Microbiota

The GM is composed of bacteria, archea, virus, and fungi. Bacteria are dominated by four main phyla of bacteria (Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria) which represent more than 95% of the total [10]. The GM is enriched in several genes important for glycan and aminoacid metabolism, xenobiotic metabolism, methanogenesis, and biosynthesis of vitamins [16]. This may explain why GM contributes to host nutrition, bone mineral density, modulation of the immune system, xenobiotics metabolism, intestinal cell development and proliferation, and protection against pathogens [17].
The GM is specific to an individual, and is also highly resilient, promptly returning to baseline after perturbation [18,19,20]. Interestingly, despite the unique individual GM, humans share similar functional gene profiles, implying a core functional microbiome [19].
Among several mechanisms which may explain the interaction between altered GM and obesity/NAFLD there are: (1) the augmented energy extraction from diet through the GM capacity to digest complex polysaccharides with consequent fermentation to short-chain fatty acids (SCFAs); (2) the intestinal epithelium damage mediated by bacterial ethanol production; (3) the bacterial/endotoxin translocation into the portal circulation and consequent liver damage via TLR signaling; (4) the modulation of bile acid (BA) synthesis; and (5) the reduction of choline metabolism with consequent decrement of very low density lipoproteins (VLDL) liver export [21].
The first clue on the role of the microbiota in the pathogenesis of obesity and NAFLD came from Backhed’s group studies [22]. They compared body weight gain in germ-free mice and conventionally raised mice, and found that the latter group gained more weight, with increased adipose tissue and body fat percentage, which could not be explained by different diet intake [22]. In two studies it has been shown that obese and overweight adults are more likely to have a low microbial gene count (LGC) than non-obese adults. LGC subjects were found to gain more weight over time and experience increased IR, dyslipidemia and elevated inflammatory markers. However, antibiotic treatments, type of diet and some food supplements should also be considered [23,24]. Taking into account several (and sometimes contradictory) studies in general, the link between intestinal dysbiosis and obesity/NAFLD is characterized by a greater prevalence of Firmicutes and a lower prevalence of Bacteroidetes in obese vs. lean individuals, and by a lower prevalence of Bacteroidetes in NASH patients vs. obese without NASH [21].
The microbiota can modulate body weight through several mechanisms. One is the fermentation pathway of indigestible carbohydrates in SCFAs: butyrate, propionate, and acetate [25]. Overall, colonic-derived SCFAs account for 10% of harvested energy from the diet, with acetate being the main source of energy [26]. Butyrate and propionate are considered anti-obesogenic, and acetate mainly obesogenic. Interestingly, while acetate and propionate are mainly produced by the phylum Bacteroidetes, butyrate is mainly produced by Firmicutes [27]. Butyrate is a major source of energy for colonocytes, increasing intestinal health and potentially modulating gut permeability and preventing metabolic endotoxemia [28]. Butyrate also seems to positively affect insulin sensitivity through stimulation of the release of the incretins glucagon-like peptide-1 (GLP-1) and gastric inhibitory polypeptide (GIP) [29]. Both butyrate and propionate can increase the expression of the anorexigenic adipokine leptin [30]. On the other hand, acetate is the most absorbed SCFA, and is a substrate for lipogenesis and cholesterol synthesis in the liver and adipose tissue [28]. Finally, SCFAs bind to specific receptors in the gut, liver, and adipose tissue, where they seem to have anti-inflammatory and metabolic actions that protect from obesity [26]. Interestingly, supplementation of oral butyrate in mice fed a WD, partially prevented liver steatosis and inflammation, while having no effect on obesity [31].
GM can also decrease the intestinal expression of the adipose tissue lipoprotein lipase inhibitor fasting induced adipose factor (FIAF). The result is an increased uptake of fatty acids in the adipose tissue and liver, favoring expansion of the adipose tissue and hepatic steatosis. Microbiota also prevent the beneficial action of FIAF in the expression of peroxisome proliferator-activated receptor (PPAR)-1α coactivator (PGC) and fatty acid oxidation [32]. Other mechanisms by which GM promotes obesity are an increase in mucosal gut blood flow enhancing nutrient absorption [33]; inhibition of adenosine monophosphate-activated protein kinase (AMPK) in the liver and muscle, leading to peripheral fatty acids oxidation and insulin resistance; and modulation of the pattern of conjugated bile acids including their function in lipid absorption [34].
A recent meta-analytic study shows a possible association between Helicobacter pylori (H. pylori) infection and NAFLD in adults [35]. H. pylori seems to promote metabolic variations, which are considered NAFLD risk factors, including IR, systemic inflammation and dyslipidemia [36]. Furthermore, presence of H. pylori may induce gastric atrophy, with consequent acid losses predisposing small intestinal bacterial overgrowth (SIBO), leaky gut and portal endotoxin translocation. A recent cohort study of 17,028 adults without NAFLD at baseline showed a significant association of H. pylori infection and the development of NAFLD, independent of metabolic and inflammatory risk factors [37], but another study including 3663 adults showed inverse results without a significant correlation [38]. Because of these contrasting results and of therapeutic potential extensive longitudinal studies at all ages including childhood are therefore needed to confirm the pathogenetic role of H. pylori.

5. Intestinal Permeability

The liver has both an arterial and a venous blood supply, with the greatest part of hepatic blood flow coming from the gut via the portal vein. As shown in Figure 1, the liver is therefore exposed to potentially harmful substances derived from the gut, including translocated bacteria, ethanol, and endotoxins [1]. One of the key roles of the liver is to rapidly clear these substances from the circulation. TJ proteins, such as zonula occludens, normally seal the junctions between intestinal endothelial cells at their apical aspect and thus have a vital role in preventing translocation of harmful substances from the gut into the portal system [39].
Current literature increasingly supports a role for GM and its mucosal gut interaction in the development of liver steatosis, inflammation and fibrosis [40]. Indeed, it has been shown that TJ disruption in mice and NAFLD patients can increase intestinal permeability and bacterial translocation to the liver through the bloodstream [41,42].
Summing up, GM dysbiosis can damage the intestinal epithelium, increase intestinal permeability and expose the liver to harmful bacterial products [43]. Furthermore, the intestinal mucosa immune system, consisting of a complex network of innate and adaptive cell populations, may itself contribute to maintain a delicate balance with intestinal microbiota, since it establishes the tolerance mechanism on intestinal surface [44].
Recent murine model studies have been carried out to ascertain the role of the intestinal barrier in the pathogenesis and progression of NAFLD [45]. Some of these studies combined high-fat diet (HFD) plus dextran sulfate sodium (DSS)-induced colitis to impair gut barrier integrity during the generation of hepatic steatosis [46]. The degree of macrovesicular steatosis, along with lobular inflammation and hepatic focal necrosis, resulted more severe in the model in which the integrity of the intestinal barrier had been chemically altered vs. those exposed only to a HFD. Portal endotoxin levels were elevated in both models, but they were significantly higher in the DSS+HFD model, suggesting the pivotal role of bacterial translocation in NAFLD progression [47].
In human NAFLD, gut permeability and prevalence of small intestinal bacterial overgrowth correlated with the severity of steatosis but not with steatohepatitis [42]. In another human study, plasma IgG levels against endotoxin were found to be increased in biopsy proven human NASH and progressively increased with NASH grade [48]. These findings suggest a relationship between chronic endotoxin exposure and human NASH severity in which increased permeability drives endotoxemia, which in turn triggers inflammatory cytokine responses and insulin resistance [49].
Among intestinal bacteria, Escherichia coli, and Enterococcus are more efficient in extraintestinal translocation ability and especially in patients with cirrhosis are an important cause of infections. In a recent observational study, E. coli emerged as the predominant bacterium in patients with SIBO and NAFLD [50].
However, one should admit that human studies are limited in that peripheral lipopolysaccharide (LPS) levels might not reflect portal LPS levels and might also change longitudinally over time. In other words, increased gut permeability might expose the liver to deleterious levels of LPS without sufficient LPS escaping liver clearance to produce a detectable marked increase in systemic levels [51].

6. Endotoxins and Inflammation

Many microbial cell components, or pathogen-associated molecular patterns (PAMPs) including lipopolysaccharide (LPS, endotoxin), flagellin, lipoteichoic acid, and peptidoglycan may affect the physiology and pathology of their host, mediated by TLR or other pattern recognition receptors. TLR signaling is activated upon pathogen and tissue damage recognition that induces a signaling cascade leading to production of inflammatory cytokines [52]. Additionally, pathogen and damage-associated molecules may induce the formation of a cytoplasmic multi-protein complex termed the inflammasome. Inflammasome signaling has been suggested to contribute to ameliorate fatty liver, whereas its dysfunction or deficiency result in aggravated hepatic inflammatory response, liver damage, fibrosis and cell death [53,54].
The possible association between inflammasome activation and NAFLD development and progression may be explained by hepatic influx of saturated fatty acids and LPS that are abundantly found in the model of HFD mice that may induce inflammasome activation [55]. Notably, LPS has effects beyond the liver and gut. For example, chronic low doses of LPS administered subcutaneously impair fasting glucose and insulin, alter hepatic insulin sensitivity, increase visceral and subcutaneous fat, increase adipose tissue macrophage numbers and raise hepatic triglyceride content [49].
Taken together, alterations of host and gut microbiome interactions through defective inflammasome sensing, disrupted inflammatory response, and dysbiosis play a relevant role in hepatic steatosis and its progression to NASH.

7. Bacterial Ethanol

Recent studies showed that elevation of endogenously synthesized ethanol contributes to NAFLD development [56]. The role of ethanol in the GLA homeostasis has recently been proposed from the evidence that its chronic consumption was associated with impairment of intestinal barrier function, and increased permeability for bacterial endotoxins and induction of TLR-dependent signaling cascades in the liver [57]. Alcohol is constantly produced by intestinal microbiota even in the absence of an oral alcohol ingestion [58]. It has been shown that a diet rich in sugar may lead to increased blood alcohol levels, and that endogenously synthetized ethanol is eliminated by the alcohol-dehydrogenase (ADH) pathway in the liver. Moreover, it has recently been shown that pediatric and adult alcohol ingestion-free patients with NAFLD have higher blood and breath alcohol, and also acetaldehyde levels [56]. It seems that altered GM composition plays an important role in increasing fasting blood alcohol levels, even if the precise mechanisms in NAFLD development have not yet been fully understood. Hepatic ADH activity is strongly influenced by IR, a condition typical of NAFLD patients [56,59].
Zhu’s group examined GM composition and ethanol levels in the blood of NASH, obese, and healthy children [60]. Only a few differences were evident in the GM composition of NASH as compared with obese patients without liver disease, and included differences across phyla, families, and genera in Proteobacteria, Enterobacteriaceae, and E. coli, respectively. Some of these microbiome changes included more alcohol-producing bacteria, associated to a significant increase in ethanol levels in NAFLD subjects as compared to both obese and healthy children. Furthermore, increased levels of ethanol were specifically detected in correlation with NASH. All in all, these results suggest that production of ethanol by GM may serve as a hepatotoxin, contributing to development of NAFLD and its progression to NASH [60].
Summing up, increased permeability, endogenous ethanol and systemic endotoxin concentrations reflect some GLA dysfunction in obesity and its hepatic complications. In this regard, our group recently showed that the lactulose/mannitol ratio values parallels the grade of liver involvement, significantly correlated with ethanolemia and endotoxemia concentrations. Increased permeability was a risk factor for the development of steatosis [61].

8. Bile Acids and Farnesoid X Receptor

A possible role of bile acids (BA) in glucose, lipid and energy homeostasis and inflammation through activation of the FXR and G-protein coupled receptor (TGR5) has been recently proposed. Changes in BA pool size and signaling pathways may be convenient in some metabolic diseases [62].
FXR is strongly expressed in the liver and intestine, where it is a regulator of BA enterohepatic circulation. It is known to have a crucial role in hepatic de-novo lipogenesis, VLDL export and plasma trigliceryde turnover [63]. A recent human study showed low FXR protein expression in patients with NASH vs. simple NAFLD, suggesting a protective role of FXR in liver disease progression [64]. However, the FXR seems to have a tissue-specific action: (1) the intestinal FXR antagonism inhibits sterol regulatory element-binding protein-1 (SREBP-1) with positive effects on lipid metabolism; (2) conversely, its hepatic agonism increases insulin-sensitivity, reduces obesity and suppresses inflammation [11,65]. TGR5 instead binds secondary BA and stimulate GLP-1 and peptide tyrosine (TYY) secretion, playing an important role in glucose homeostasis and food intake. BAs influence the composition of GM and in turn they are chemically modified by bacterial enzymes [66]. BAs have anti-microbial properties due to their detergent actions by damaging microbial membranes and intracellular structures. Furthermore, taurine catabolic end-products of BA catabolism promote proliferation of some bacteria species, with a consequent influence of microbial gut composition [62].
The enzyme bile salt hydrolase (BSH) of Lactobacillus and Bifidobacteria spp. catalyzes the deconjugation of bile salts to generate the unconjugated cholic acid (CA) and chenodeoxycholic acids (CDCA), which then are further modified by colonic bacterial 7α-dehydroxylasis to secondary BA, such as deoxycholic acid (DCA) and lithocolic acid (LCA). The 7α-dehydroxylation is catalyzed by microbial enzymes of bile acid inducible (BAI) operon, a system biochemically and genetically characterized in many Clostridium species. Therefore, changes in bacterial gut composition which alter the BSH and BAI enzyme copies expression influence bile acid pool and its detergent and signaling properties [67].
These free BA (CA, CDCA, DCA) are less efficient in the solubilization and absorption of lipids from diet and are more largely excreted in feces than conjugated counterparts, with a consequent positive effect on total and low-density lipoprotein (LDL)-cholesterol plasma concentrations [68]. Despite this favorable effect on lipid profile, the unconjugated BA promotes the intestinal FXR signaling. This results in an increased production of ceramides triggering NAFLD through induction of fatty acid synthesis due to SREBP-1 signaling, oxidative stress and mitochondrial damage. Ceramides also impair adipose tissue function through the reduction of beige in favor of white adipocytes [69]. Studies show that the GM modulation by treatment of HFD-fed mice with antibiotics results in decreased adverse metabolic phenotypes probably due to a decrease of Lactobacillus spp. and of BSH activity as well. The decrease of the latter resulted in increased levels of tauro-β-muricholic acid (T-β-MCA), a substrate of BSH and a potent FXR antagonist. As has emerged in the literature, mice that were lacking in expression of FXR in the intestine were resistant to HFD-induced obesity, IR and NAFLD [66]. Conversely, hepatic FXR activation seems to have an anti-steatotic effect with an indirect mechanism due to improvement of and lipoprotein transport [70]. Hepatic FXR stimulation attenuates steatosis in rodents and humans, furthermore HFD-fed FXR-null mice more frequently develop fatty liver [67,68]. Role of intestinal FXR in hepato-steatosis however is still controversial, because both agonism and antagonism seems to play a protective role [71] and are also gender-specific [72]. The activation by the FXR agonist fexaramine in mice reduces weight gain and steatosis via fibroblast growth factor 15 (FGF15) signaling [73], conversely also the intestinal FXR antagonism attenuates hepatic steatosis reducing ceramides and SREBP1 signaling [69].
Moreover, in dysbiotic NASH patients the increase of fecal primary BA, primary: secondary BA fecal ratio, and plasma and hepatic BA concentrations may lead to cytotoxicity and explain NAFLD progression [62]. Indeed, in advanced liver disease and cirrhosis, a decrease of total BA inflow from the liver to the gut may cause a shift toward Firmicutes, particularly Clostridium, at the expense of other beneficial Firmicutes taxa (i.e., Lachnospiraceae, Roseburia, Rumminococcaceae and Blautia). Members of underrepresented taxa are part of normal GM, which are producers of beneficial SCFAs and are involved in conversion of primary to secondary BA. For this reasons in cirrhosis decrement of the total BA pool may unfavorably impact intestinal and systemic inflammation and worsen dysbiosis [74].
Human studies although are still scarce confirm the reciprocal influence of BA and GM and their role in NAFLD pathogenesis, assuming the role of FXR and G-protein receptor [75].

9. Fecal Biomarkers

In the last few years a large attention was dedicated to find non-invasive biomarkers of NAFLD/NASH to overcome the need for the diagnostic gold standard tool, i.e., liver biopsy. Colonic bacteria are a source of many metabolic products measurable in fecal samples by gas chromatography-mass spectrometry [76]: this approach appears therefore to provide a useful marker of GM composition and consequently may be used as a surrogate of NAFLD/NASH presence. It seems that more severe liver damage is associated: (1) with changes in the composition of GM and its metabolome; (2) with the intestinal and systemic inflammatory response; and (3) with the metabolic profile of bile acids. GM modifications are reflected in those of its metabolome, and could therefore be an excellent therapeutic target. It will be necessary to deepen the knowledge on the composition of GM and metabolome in patients with NASH in order to more precisely define altered patterns that could be useful for the diagnosis [77]. Notably, a significant increase in fecal ester compounds was already observed in NAFLD adult patients [78] and a unique fecal metabolomics profile with increased level of 2-butanone and 4-methyl-2-pentanonewas found in pediatric NASH [79]. Other extensive studies are needed to define, validate and standardize the fecal metabolomics and its usefulness in non-invasive diagnosis and staging of NAFLD/NASH. We recently showed that urinary metabolome analysis also contributes to defining a metabolic fingerprint of pediatric obesity and related NAFLD, by identifying metabolic pathways/metabolites reflecting typical obesity dietary habits and GM/GLA perturbations [80].

10. Therapeutic Prospectives

Hepatopathy of obese children is frequently recalcitrant to the first line-therapy consisting of lifestyle modifications. Therefore, the individuation of new NAFLD treatments is of extreme relevance in clinical practice especially for those patients who are unable to comply with recommended dietary and physical activity changes.
In general, NAFLD therapies target four main pathways: (1) fat accumulation and metabolic stress; (2) oxidative stress and inflammation; (3) fibrosis progression; and finally (4) gut microbiome and GLA. Pending truly effective therapies, in recent years more and more attention has been dedicated to GLA manipulation with studies conducted in animal models and in humans as well. Below we summarize those approaches which may be prove more promising in the next future. Schematically, possible actions on GLA may include GM manipulation through prebiotics (substances useful to the growth of good GM), probiotics (live microorganisms), and synbiotics (combining probiotics and prebiotics in a form of synergism), that appear to reduce intestinal inflammation and improve the epithelial barrier function [7,81,82].

10.1. Probiotics

A meta-analytic study showed in NAFLD the efficacy of probiotic therapies in terms of aminotransferases, cholesterol and tumor necrosis factor α (TNF-α) reduction and insulin resistance improvement [83]. Probiotics could improve gut microbiota composition, and downregulate serum LPS and liver TLR4, delaying the progression of liver disease [84]. As shown in Table 1, the strain more frequently used belong to genera Bifidobacteria and Lactobacillus [85,86]. Notably, Lactobacillus rhamnosus GG [87] and VSL#3 [88] showed promising results in pediatric patients with a decrease in serum alanine amino transferase (ALT) in NAFLD vs. controls, significant weight reduction, and amelioration of liver fibrosis [88]. Similar results were recently obtained in a pediatric multi-strain study using another compound of Lactobacillus acidophilus, Bifidobacterium lactis, Bifidobacterium bifidum and Lactobacillus rhamnosus [89]. Therefore, probiotics warrant consideration as a therapeutic tool to treat obese children with liver disease in addition to standard lifestyle intervention strategies.

10.2. Antibiotics

In animal models, antibiotics treatments attenuate HFD induced gut and liver inflammation [103], probably due to: (1) a decreased intestinal permeability and LPS-mediated TLR-4 signaling; and (2) an increased abundance of beneficial bacteria [104]. A number of studies carried out with antibiotic therapy in NAFLD/NASH patients have shown divergent results. The macrolide solithromycin reduces hepatocyte ballooning and inflammation in animal models, without any effect on liver fat content, through an LPS-independent mechanism [105]. Polymyxin B and neomycin reduce hepatic steatosis and endotoxin levels in animal models [106,107]. Studies on rifaximin, active on Gram-negative bacteria, show conflicting results in NASH patients ranging from a significant reduction of ALT, endotoxin and IL-10 levels [108] to the inefficacy on fat liver content and ALT levels [109]. Further studies on antibiotics as GLA-modulators are needed, especially to evaluate risks/benefits in light of recent data showing a possible increased risk of obesity due to antibiotic exposure in early life [104,110].

10.3. Fecal Microbiota Transplantation

In addition to probiotic supplementation, fecal microbiota transplantation has been shown to attenuate HFD-induced steatohepatitis, through the modulation of GM. Actually, the fecal microbiota transplantation from lean donors to NASH patients is under study in humans (NCT02469272). Despite its routine utilization it is difficult to consider in present clinical practice, the study results will be useful also to better define NAFLD pathogenetic mechanisms [7].

10.4. Farnesoid X Receptor Modulators

Last but probably not least, another group of agents is represented by those acting by FXR modulation. Recent promising research has shown the efficacy of the obeticholic acid (OCA), a semisynthetic derivate of chenodeoxycholic acid. This agent, by modulating FXR signaling, improves liver inflammation and fibrosis in NASH adult patients [111] and reduces intestinal inflammation in colitis rodent models [112]. However, patients receiving OCA show unfavorable lipid profile with increased total cholesterol and LDL and decreased high density lipoproteins (HDL), and a higher HOMA (homeostatic model assessment)-IR [112]. GS-9674, a synthetic FXR agonist with a more predictable pharmacokinetics in contrast to OCA, is now under study in human NASH [113]. Natural extracts of Astragali radix, cycloastragenol and calycosinvia FXR activation attenuates triglyceride accumulation and hepatic fibrosis in NAFLD animal models [114,115]. Other synthetic FXR agonists, GW4064, INT-767 and fexaramine, showed similar results in obese rodents [66,73,83]. Therefore, modulation of FXR signaling appears to be an emerging therapeutic molecular target for preventing NASH progression [12]. Given still conflicting reports on tissue-specific activity, gender-specificity and negative effect on lipid and glucose profiles, further extensive human studies are needed to better define its efficacy, safety and indications and to design possible selective BA receptor modulators (SBARMs) with minimal side-effects especially in pediatric age.

10.5. Anti-LPS Immunoglobulins

A recent promising therapy is oral supplementation of IMM-124e, an extract of bovine colostrum rich in IgG obtained from cows immunized versus LPS, that improved liver fat, insulin sensitivity [116] and immune-mediated colitis [117] in animal models, and improved glycemic control in a small pilot human study [118]. Benefits seem to be due to reduction of liver exposition to GM LPS and consequent Kupffer cells activation.

10.6. Vitamin D

Finally, a key role in obesity, MetS and NAFLD seems to be played also by vitamin D deficiency, although the underlying mechanism is poorly understood. Recently, the possible involvement of Vitamin D on GLA dysregulation is slowly emerging. In fact, optimal vitamin D levels are essential to maintaining integrity of intestinal permeability, through the up-regulation of TJ components and mucous proteoglycans in the ileum epithelium, and to maintain the gut microbiota in a state of eubiosis, through the expression of specific α-defensins and their converting enzyme (matrix metalloproteinase 7—MMP7) by Paneth cells of intestinal mucosa. The presence of vitamin D deficiency in murine HFD models promotes leaky gut, dysbiosis, endotoxemia, systemic inflammation and consequent IR and liver steatosis [119]. Thus, supplementation with vitamin D has been recommended [120].

11. Conclusions

Obesity and obesity-related liver disease (NAFLD/NASH) are major health concerns. At present there is no efficient treatment available for children or adults. Certainly, healthy diet and adequate physical activity levels remain the mainstay treatments in obese patients with hepatic complications, but the individuation of alternative therapeutic targets is critical especially in those with poor compliance to the prescribed lifestyle changes.
Advances obtained in the understanding of the role of GLA in NAFLD pathogenesis, and the encouraging results already obtained by GM modulation via probiotic supplementation appear a presently promising and safe innovative mode of therapy. However, other extensive and long-term studies are needed to better define which are the best probiotic strains, their doses, timing, and duration of supplementation therapy. This will serve to individualize probiotic therapy with a patient-tailored approach for modulating intestinal permeability, endotoxemia, and treating liver disease [15,43,121]. Manipulating bacterial communities by in situ microbiome engineering with high specificity and efficacy (i.e., specifically producing anti-inflammatory or antioxidants molecules) remains still a speculative way, possibly leading to wholly new therapeutic strategies [122].
Finally, FXR modulation through obeticholic acid and similar agents is an encouraging approach needing confirmation in pediatrics.

Author Contributions

The first two authors contributed equally. Marco Poeta, Luca Pierri and Pietro Vajro reviewed the literature regarding the role of the gut–liver axis in NAFLD pathogenesis and therapy. All participated in writing the paper. Pietro Vajro guided manuscript organization and reviewed the final version, which was agreed on by everyone.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Clemente, M.G.; Mandato, C.; Poeta, M.; Vajro, P. Pediatric non-alcoholic fatty liver disease: Recent solutions, unresolved issues, and future research directions. World J. Gastroenterol. 2016, 28, 22. [Google Scholar] [CrossRef] [PubMed]
  2. Schwimmer, J.B.; Deutsch, R.; Kahen, T.; Lavine, J.E.; Stanley, C.; Behling, C. Prevalence of fatty liver in children and adolescents. Pediatrics 2006, 118, 1388–1393. [Google Scholar] [CrossRef] [PubMed]
  3. Pramfalk, C.; Pavlides, M.; Banerjee, R.; McNeil, C.A.; Neubauer, S.; Karpe, F.; Hodson, L. Sex-specific differences in hepatic fat oxidation and synthesis may explain the higher propensity for NAFLD in men. J. Clin. Endocrinol. Metab. 2015, 100, 4425–4433. [Google Scholar] [CrossRef] [PubMed]
  4. Anderson, E.L.; Howe, L.D.; Jones, H.E.; Higgins, J.P.; Lawlor, D.A.; Fraser, A. The Prevalence of Non-Alcoholic Fatty Liver Disease in Children and Adolescents: A Systematic review and Meta-Analysis. PLoS ONE 2015, 10, e0140908. [Google Scholar] [CrossRef] [PubMed]
  5. Goyal, N.P.; Schwimmer, J.B. The progression and natural history of pediatric nonalcoholic fatty liver disease. Clin. Liver. Dis. 2016, 20, 325–338. [Google Scholar] [CrossRef] [PubMed]
  6. Fitzpatrick, E.; Hadzic, N. Paediatric non-alcoholic fatty liver disease: An emerging threat. Cent. Eur. J. Paediatr. 2015, 11, 1–9. [Google Scholar] [CrossRef]
  7. Rotman, Y.; Sanyal, A.J. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut 2017, 66, 180–190. [Google Scholar] [CrossRef] [PubMed]
  8. Zorn, A.M.; Wells, J.M. Vertebrate endoderm development and organ formation. Annu. Rev. Cell Dev. 2009, 25, 221–251. [Google Scholar] [CrossRef] [PubMed]
  9. Paolella, G.; Mandato, C.; Pierri, L.; Poeta, M.; Di Stasi, M.; Vajro, P. Gut-liver axis and probiotics: Their role in non-alcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 15518–15531. [Google Scholar] [CrossRef] [PubMed]
  10. Vajro, P.; Lenta, S.; Socha, P.; Dhawan, A.; McKiernan, P.; Baumann, U.; Durmaz, O.; Lacaille, F.; McLin, V.; Nobili, V. Diagnosis of nonalcoholic fatty liver disease in children and adolescents: Position paper of the ESPGHAN Hepatology Committee. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 700–713. [Google Scholar] [CrossRef] [PubMed]
  11. Zhu, Y.; Liu, H.; Zhang, M.; Guo, G.L. Fatty liver diseases, bile acids, and FXR. Acta Pharm. Sin. B 2016, 6, 409–412. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, S.G.; Kim, B.K.; Kim, K.; Fang, S. Bile Acid Nuclear Receptor Farnesoid X Receptor: Therapeutic Target for Nonalcoholic Fatty Liver Disease. Endocrinol. Metab. 2016, 31, 500–504. [Google Scholar] [CrossRef] [PubMed]
  13. Brandl, K.; Kumar, V.; Eckmann, L. Gut-liver axis at the frontier of host-microbial interactions. Am. J. Physiol.-Gastrointest. Liver Physiol. 2017, 312, G413–G419. [Google Scholar] [CrossRef] [PubMed]
  14. Sekirov, I.; Russell, S.L.; Antunes, L.C.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [PubMed]
  15. Vajro, P.; Paolella, G.; Fasano, A. Microbiota and gut-liver axis: Their influences on obesity and obesity-related liver disease. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 461–468. [Google Scholar] [CrossRef] [PubMed]
  16. Gill, S.R.; Pop, M.; Deboy, R.T.; Eckburg, P.B.; Turnbaugh, P.J.; Samuel, B.S.; Gordon, J.I.; Relman, D.A.; Fraser-Liggett, C.M.; Nelson, K.E. Metagenomic analysis of the human distal gut microbiome. Science 2006, 312, 1355–1359. [Google Scholar] [CrossRef] [PubMed]
  17. Seksik, P.; Landman, C. Understanding microbiome data: A primer for clinicians. Dig. Dis. 2015, 33, 11–16. [Google Scholar] [CrossRef] [PubMed]
  18. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed]
  19. Imajo, K.; Yoneda, M.; Ogawa, Y.; Wada, K.; Nakajima, A. Microbiota and nonalcoholic steatohepatitis. Semin. Immunopathol. 2014, 36, 115–132. [Google Scholar] [CrossRef] [PubMed]
  20. Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The long-term stability of the human gut microbiota. Science 2013, 341, 1237439. [Google Scholar] [CrossRef] [PubMed]
  21. Doulberis, M.; Kotronis, G.; Gialamprinou, D.; Kountouras, J.; Katsinelos, P. Non-alcoholic fatty liver disease: An update with special focus on the role of gut microbiota. Metabolism 2017, 71, 182–197. [Google Scholar] [CrossRef] [PubMed]
  22. Backhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef] [PubMed]
  23. Le Chatelier, E.; Nielsen, T.; Qin, J.; PriftI, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
  24. Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.; Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500, 585–588. [Google Scholar] [CrossRef] [PubMed]
  25. Chakraborti, C.K. New-found link between microbiota and obesity. World J. Gastrointest. Pathophysiol. 2015, 6, 110–119. [Google Scholar] [CrossRef] [PubMed]
  26. Brahe, L.K.; Astrup, A.; Larsen, L.H. Is butyrate the link between diet, intestinal microbiota and obesity-related metabolic diseases? Obes. Rev. 2013, 14, 950–959. [Google Scholar] [CrossRef] [PubMed]
  27. Louis, P.; Young, P.; Holtrop, G.; Flint, H.J. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA: Acetate CoA-transferase gene. Environ. Microbiol. 2010, 12, 304–314. [Google Scholar] [CrossRef]
  28. Roy, C.C.; Kien, C.L.; Bouthillier, L.; Levy, E. Short-chain fatty acids: Ready for prime time? Nutr. Clin. Pract. 2006, 21, 351–366. [Google Scholar] [CrossRef] [PubMed]
  29. Lin, H.V.; Frassetto, A.; Kowalik, E.J., Jr.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G.; et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS ONE 2012, 7, e35240. [Google Scholar] [CrossRef] [PubMed]
  30. Xiong, Y.; Miyamoto, N.; Shibata, K.; Valasek, M.A.; Motoike, T.; Kedzierski, R.M.; Yanagisawa, M. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl. Acad. Sci. USA 2004, 101, 1045–1050. [Google Scholar] [CrossRef] [PubMed]
  31. Jin, C.J.; Sellmann, C.; Engstler, A.J.; Ziegenhardt, D.; Bergheim, I. Supplementation of sodium butyrate protects mice from the development of non-alcoholic steatohepatitis (NASH). Br. J. Nutr. 2015, 114, 1745–1755. [Google Scholar] [CrossRef] [PubMed]
  32. Backhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [Google Scholar] [CrossRef] [PubMed]
  33. Ding, S.; Chi, M.M.; Scull, B.P.; Rigby, R.; Schwerbrock, N.M.; Magness, S.; Jobin, C.; Lund, P.K. High-fat diet: Bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS ONE 2010, 5, e12191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Claus, S.P.; Ellero, S.L.; Berger, B.; Krause, L.; Bruttin, A.; Molina, J.; Paris, A.; Want, E.J.; de Waziers, I.; Cloarec, O.; et al. Colonization-induced host-gut microbial metabolic interaction. MBio 2011, 2, e00271-10. [Google Scholar] [CrossRef] [PubMed]
  35. Wijarnpreecha, K.; Thongprayoon, C.; Panjawatanan, P.; Manatsathit, W.; Jaruvongvanich, V.; Ungprasert, P. Helicobacter pylori and Risk of Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-analysis. J. Clin. Gastroenterol. 2017. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, C.X.; Mao, Y.S.; Foster, P.; Zhu, Z.W.; Du, J.; Guo, C.Y. Possible association between Helicobacter pylori infection and nonalcoholic fatty liver disease. Appl. Physiol. Nutr. Metab. 2017, 42, 295–301. [Google Scholar] [CrossRef] [PubMed]
  37. Kim, T.J.; Sinn, D.H.; Min, Y.W.; Son, H.J.; Kim, J.J.; Chang, Y.; Baek, S.Y.; Ahn, S.H.; Lee, H.; Ryu, S. A cohort study on Helicobacter pylori infection associated with nonalcoholic fatty liver disease. J. Gastroenterol. 2017. [Google Scholar] [CrossRef]
  38. Baeg, M.K.; Yoon, S.K.; Ko, S.H.; Noh, Y.S.; Lee, I.S.; Choi, M.G. Helicobacter pylori infection is not associated with nonalcoholic fatty liver disease. World J. Gastroenterol. 2016, 22, 2592–2600. [Google Scholar] [CrossRef] [PubMed]
  39. Ulluwishewa, D.; Anderson, R.C.; McNabb, W.C.; Moughan, P.J.; Wells, J.M.; Roy, N.C. Regulation of tight junction permeability by intestinal bacteria and dietary components. J. Nutr. 2011, 141, 769–776. [Google Scholar] [CrossRef] [PubMed]
  40. Abdou, R.M.; Zhu, L.; Baker, R.D.; Baker, S.S. Gut Microbiota of Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2016, 61, 1268–1281. [Google Scholar] [CrossRef] [PubMed]
  41. Rahman, K.; Desai, C.; Iyer, S.S.; Thorn, N.E.; Kumar, P.; Liu, Y.; Smith, T.; Neish, A.S.; Li, H.; Tan, S.; et al. Loss of Junctional Adhesion Molecule A Promotes Severe Steatohepatitis in Mice on a Diet High in Saturated Fat, Fructose, and Cholesterol. Gastroenterology 2016, 151, 733–746. [Google Scholar] [CrossRef] [PubMed]
  42. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Mascianà, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in non alcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  43. Bluemel, S.; Williams, B.; Knight, R.; Schnabl, B. Precision medicine in alcoholic and nonalcoholic fatty liver disease via modulating the gut microbiota. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 311, G1018–G1036. [Google Scholar] [CrossRef] [PubMed]
  44. Peterson, L.W.; Artis, D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 2014, 14, 141–153. [Google Scholar] [CrossRef] [PubMed]
  45. Mao, J.W.; Tang, H.Y.; Zhao, T.; Tan, X.Y.; Bi, J.; Wang, B.Y.; Wang, Y.D. Intestinal mucosal barrier dysfunction participates in the progress of non alcoholic fatty liver disease. Int. J. Clin. Exp. Pathol. 2015, 8, 3648–3658. [Google Scholar] [PubMed]
  46. Gabele, E.; Dostert, K.; Hofmann, C.; Wiest, R.; Scholmerich, J.; Hellerbrand, C.; Obermeier, F. DSS induced colitis increases portal LPS levels and enhances hepatic inflammation and fibrogenesis in experimental NASH. J. Hepatol. 2011, 55, 1391–1399. [Google Scholar] [CrossRef] [PubMed]
  47. Luther, J.; Garber, J.J.; Khalili, H.; Dave, M.; Bale, S.S.; Jindal, R.; Motola, D.L.; Luther, S.; Bohr, S.; Jeoung, S.W.; et al. Hepatic injury in nonalcoholic steatohepatitis contributes to altered intestinal permeability. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 222–232. [Google Scholar] [CrossRef] [PubMed]
  48. Verdam, F.J.; Rensen, S.S.; Driessen, A.; Greve, J.W.; Buurman, W.A. Novel evidence for chronic exposure to endotoxin in human non alcoholic steatohepatitis. J. Clin. Gastroenterol. 2011, 45, 149–152. [Google Scholar] [CrossRef] [PubMed]
  49. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [PubMed]
  50. Kapil, S.; Duseja, A.; Sharma, B.K.; Singla, B.; Chakraborti, A.; Das, A.; Ray, P.; Dhiman, R.K.; Chawla, Y. Small intestinal bacterial overgrowth and toll-like receptor signaling in patients with non-alcoholic fatty liver disease. J. Gastroenterol. Hepatol. 2016, 31, 213–221. [Google Scholar] [CrossRef] [PubMed]
  51. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef] [PubMed]
  52. Miura, K.; Ohnishi, H. Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 7381–7391. [Google Scholar] [CrossRef] [PubMed]
  53. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef] [PubMed]
  54. Nakamoto, N.; Kanai, T. Role of toll-like receptors in immune activation and tolerance in the liver. Front. Immunol. 2014, 5, 221. [Google Scholar] [CrossRef] [PubMed]
  55. Csak, T.; Ganz, M.; Pespisa, J.; Kodys, K.; Dolganiuc, A.; Szabo, G. Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology 2011, 54, 133–144. [Google Scholar] [CrossRef] [PubMed]
  56. Engstler, A.J.; Aumiller, T.; Degen, C.; Dürr, M.; Weiss, E.; Maier, I.B.; Schattenberg, J.M.; Jin, C.J.; Sellmann, C.; Bergheim, I. Insulin resistance alters hepatic ethanol metabolism: Studies in mice and children with non-alcoholic fatty liver disease. Gut 2016, 65, 1564–1571. [Google Scholar] [CrossRef] [PubMed]
  57. Bode, C.; Bode, J.C. Effect of alcohol consumption on the gut. Best Pract. Res. Clin. Gastroenterol. 2003, 17, 575–592. [Google Scholar] [CrossRef]
  58. Blomstrand, R. Observations of the formation of ethanol in the intestinal tract in man. Life Sci. 1971, 10, 575–582. [Google Scholar] [CrossRef]
  59. Leclercq, I.A.; Field, J.; Enriquez, A.; Farrell, G.C.; Robertson, G.R. Constitutive and inducible expression of hepatic CYP2E1 in leptin-deficien TOB/OB mice. Biochem. Biophys. Res. Commun. 2000, 268, 337–344. [Google Scholar] [CrossRef] [PubMed]
  60. Zhu, L.; Baker, R.D.; Zhu, R.; Baker, S.S. Gut microbiota produce alcohol and contribute to NAFLD. Gut 2016, 65, 1232. [Google Scholar] [CrossRef] [PubMed]
  61. Guercio Nuzio, S.; di Stasi, M.; Pierri, L.; Troisi, J.; Poeta, M.; Bisogno, A.; Belmonte, F.; Tripodi, M.; di Salvio, D.; Massa, G.; et al. Multiple gut-liver axis abnormalities in children with obesity with and without hepatic involvement. Pediatr. Obes. 2016. [Google Scholar] [CrossRef] [PubMed]
  62. Chávez-Talavera, O.; Tailleux, A.; Lefebvre, P.; Staels, B. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia and NAFLD. Gastroenterology 2017, 152, 1679–1694. [Google Scholar] [CrossRef] [PubMed]
  63. Trauner, M.; Claudel, T.; Fickert, P.; Moustafa, T.; Wagner, M. Bile acids as regulators of hepatic lipid and glucose metabolism. Dig. Dis. 2010, 28, 220–224. [Google Scholar] [CrossRef] [PubMed]
  64. Aguilar-Olivos, N.E.; Carrillo-Cordova, D.; Oria-Hernandez, J.; Sanchez-Valle, V.; Ponciano-Rodriguez, G.; Ramirez-Jaramillo, M.; Chable-Montero, F.; Chavez-Tapia, N.C.; Uribe, M.; Mendez-Sanchez, N. The nuclear receptor FXR, but not LXR, up-regulates bile acid transporter expression in non-alcoholic fatty liver disease. Ann. Hepatol. 2015, 14, 487–493. [Google Scholar] [PubMed]
  65. McMahan, R.H.; Wang, X.X.; Cheng, L.L.; Krisko, T.; Smith, M.; El Kasmi, K.; Pruzanski, M.; Adorini, L.; Golden-Mason, L.; Levi, M.; et al. Bile acid receptor activation modulates hepatic monocyte activity and improves nonalcoholic fatty liver disease. J. Biol. Chem. 2013, 288, 11761–11770. [Google Scholar] [CrossRef] [PubMed]
  66. Wahlström, A.; Kovatcheva-Datchary, P.; Ståhlman, M.; Bäckhed, F.; Marschall, H.U. Crosstalk between bile acids and gut microbiota and its impact on Farnesoid X Receptor signalling. Dig. Dis. 2017, 35, 246–250. [Google Scholar] [CrossRef] [PubMed]
  67. Long, S.R.; Gahan, C.G.M.; Joyce, S.A. Interactions between gut bacteria and bile in health and disease. Mol. Asp. Med. 2017, 56, 54–65. [Google Scholar] [CrossRef] [PubMed]
  68. Begley, M.; Hill, C.; Gahan, C.G. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 2006, 72, 1729–1738. [Google Scholar] [CrossRef] [PubMed]
  69. Gonzalez, F.J.; Jiang, C.; Patterson, A.D. An Intestinal Microbiota Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016, 151, 845–859. [Google Scholar] [CrossRef] [PubMed]
  70. Li, Y.; Jadhav, K.; Zhang, Y. Bile acid receptors in non-alcoholic fatty liver disease. Biochem. Pharmacol. 2013, 86, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  71. Tanaka, N.; Aoyama, T.; Kimura, S.; Gonzalez, F.J. Targeting nuclear receptors for the treatment of fatty liver disease. Pharmacol. Ther. 2017. [Google Scholar] [CrossRef] [PubMed]
  72. Sheng, L.; Jena, P.K.; Liu, H.X.; Kalanetra, K.M.; Gonzalez, F.J.; French, S.W.; Krishnan, V.V.; Mills, D.A.; Wan, Y.Y. Gender Differences in Bile Acids and Microbiota in Relationship with Gender Dissimilarity in Steatosis Induced by Diet and FXR Inactivation. Sci. Rep. 2017, 7, 1748. [Google Scholar] [CrossRef] [PubMed]
  73. Fang, S.; Suh, J.M.; Reilly, S.M.; Yu, E.; Osborn, O.; Lackey, D.; Yoshihara, E.; Perino, A.; Jacinto, S.; Lukasheva, Y.; et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat. Med. 2015, 21, 159–165. [Google Scholar] [CrossRef] [PubMed]
  74. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B.; Bajaj, J.S. Gut microbiota, cirrhosis and alcohol regulate bile acid metabolism in the gut. Dig. Dis. 2015, 33, 338–345. [Google Scholar] [CrossRef] [PubMed]
  75. Fuchs, M. Non-alcoholic Fatty liver disease: The bile acid-activated farnesoid x receptor as an emerging treatment target. J. Lipids 2012, 2012, 934396. [Google Scholar] [CrossRef] [PubMed]
  76. Von Schönfels, W.; Patsenker, E.; Fahrner, R.; Itzel, T.; Hinrichsen, H.; Brosch, M.; Erhart, W.; Gruodyte, A.; Vollnberg, B.; Richter, K.; et al. Metabolomic tissue signature in human non-alcoholic fatty liver disease identifies protective candidate metabolites. Liver Int. 2015, 35, 207–214. [Google Scholar] [CrossRef] [PubMed]
  77. Brandl, K.L.; Schnabl, B. Intestinal microbiota and nonalcoholic steatohepatitis. Curr. Opin. Gastroenterol. 2017, 33, 128–133. [Google Scholar] [CrossRef] [PubMed]
  78. Raman, M.; Ahmed, I.; Gillevet, P.M.; Probert, C.S.; Ratcliffe, N.M.; Smith, S.; Greenwood, R.; Sikaroodi, M.; Lam, V.; Crotty, P.; et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2013, 11, 868–875. [Google Scholar] [CrossRef] [PubMed]
  79. Del Chierico, F.; Nobili, V.; Vernocchi, P.; Russo, A.; Stefanis, C.; Gnani, D.; Furlanello, C.; Zandonà, A.; Paci, P.; Capuani, G.; et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 2017, 65, 451–464. [Google Scholar] [CrossRef] [PubMed]
  80. Troisi, J.; Pierri, L.; Landolfi, A.; Marciano, F.; Bisogno, A.; Belmonte, F.; Palladino, C.; Nuzio, S.G.; Campiglia, P.; Vajro, P. Urinary Metabolomics in Pediatric Obesity and NAFLD Identifies Metabolic Pathways/Metabolites Related to Dietary Habits and Gut-Liver Axis Perturbations. Nutrients 2017, 9, 485. [Google Scholar] [CrossRef] [PubMed]
  81. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  82. Esposito, E.; Iacono, A.; Bianco, G.; Autore, G.; Cuzzocrea, S.; Vajro, P.; Canani, R.B.; Calignano, A.; Raso, G.M.; Meli, R. Probiotics reduce the inflammatory response induced by a high-fat diet in the liver of young rats. J. Nutr. 2009, 139, 905–911. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, Y.; Huang, Y.; Yan, L.; Gao, M.; Liu, D. Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm. Res. 2013, 30, 1447–1457. [Google Scholar] [CrossRef] [PubMed]
  84. Xue, L.; He, J.; Gao, N.; Lu, X.; Li, M.; Wu, X.; Liu, Z.; Jin, Y.; Liu, J.; Xu, J.; et al. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci. Rep. 2017, 7, 45176. [Google Scholar] [CrossRef] [PubMed]
  85. Vajro, P.; Mandato, C.; Veropalumbo, C.; de Micco, I. Probiotics: A possible role in treatment of adult and pediatric non alcoholic fatty liver disease. Ann. Hepatol. 2013, 12, 161–163. [Google Scholar] [PubMed]
  86. Vajro, P.; Veropalumbo, C.; D’Aniello, R.; Mandato, C. Probiotics in the treatment of non alcoholic fatty liver disease: Further evidence in obese children. Nutr. Metab. Cardiovasc. Dis. 2013, 23, e9–e10. [Google Scholar] [CrossRef] [PubMed]
  87. Vajro, P.; Mandato, C.; Licenziati, M.R.; Franzese, A.; Vitale, D.F.; Lenta, S.; Caropreso, M.; Vallone, G.; Meli, R. Effects of Lactobacillus rhamnosus strain GG in pediatric obesity-related liver disease. J. Pediatr. Gastroenterol. Nutr. 2011, 52, 740–743. [Google Scholar] [CrossRef] [PubMed]
  88. Alisi, A.; Bedogni, G.; Baviera, G.; Giorgio, V.; Porro, E.; Paris, C.; Giammaria, P.; Reali, L.; Anania, F.; Nobili, V. Randomised clinical trial: The beneficial effects of VSL#3 in obese children with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2014, 39, 1276–1285. [Google Scholar] [CrossRef] [PubMed]
  89. Famouri, F.; Shariat, Z.; Hashemipour, M.; Keikha, M.; Kelishadi, R. Effects of Probiotics on Nonalcoholic Fatty Liver Disease in Obese Children and Adolescents. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 413–417. [Google Scholar] [CrossRef] [PubMed]
  90. Loguercio, C.; Federico, A.; Tuccillo, C.; Terracciano, F.; D’Auria, M.V.; De Simone, C.; Del Vecchio Blanco, C. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J. Clin. Gastroenterol. 2005, 39, 540–543. [Google Scholar] [PubMed]
  91. Solga, S.F.; Buckley, G.; Clark, J.M.; Horska, A.; Diehl, A.M. The effect of a probiotic on hepatic steatosis. J. Clin. Gastroenterol. 2008, 42, 1117–1119. [Google Scholar] [CrossRef] [PubMed]
  92. Aller, R.; de Luis, D.A.; Izaola, O.; Conde, R.; Gonzalez Sagrado, M.; Primo, D.; de la Fuente, B.; Gonzalez, J. Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver disease patients: A double blind randomized clinical trial. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 1090–1095. [Google Scholar] [PubMed]
  93. Mykhal’chyshyn, H.P.; Bodnar, P.M.; Kobyliak, N.M. Effect of probiotics on proinflammatory cytokines level in patients with type 2 diabetes and nonalcoholic fatty liver disease. Likars’ ka Sprava 2013, 2, 56–62. [Google Scholar]
  94. Nabavi, S.; Rafraf, M.; Somi, M.H.; Homayouni-Rad, A.; Asghari-Jafarabadi, M. Effects of probiotic yogurt consumption on metabolic factors in individuals with nonalcoholic fatty liver disease. J. Dairy Sci. 2014, 97, 7386–7393. [Google Scholar] [CrossRef] [PubMed]
  95. Sepideh, A.; Karim, P.; Hossein, A.; Leila, R.; Hamdollah, M.; Mohammad, E.G.; Mojtaba, S.; Mohammad, S.; Ghader, G.; Seyed Moayed, A. Effects of multistrain probiotic supplementation on glycemic and inflammatory indices in patients with nonalcoholic fatty liver disease: A double-blind randomized clinical trial. J. Am. Coll. Nutr. 2016, 35, 500–505. [Google Scholar] [CrossRef] [PubMed]
  96. Loguercio, C.; de Simone, T.; Federico, A.; Terracciano, F.; Tuccillo, C.; Di Chicco, M.; Cartenì, M. Gut-liver axis: A new point of attack to treat chronic liver damage? Am. J. Gastroenterol. 2002, 97, 2144–2146. [Google Scholar] [CrossRef] [PubMed]
  97. Malaguarnera, M.; Vacante, M.; Antic, T.; Giordano, M.; Chisari, G.; Acquaviva, R.; Mastrojeni, S.; Malaguarnera, G.; Mistretta, A.; Li Volti, G.; et al. Bifidobacterium longum with fructo-oligosaccharides inpatients with non alcoholic steatohepatitis. Dig. Dis. Sci. 2012, 57, 545–553. [Google Scholar] [CrossRef] [PubMed]
  98. Shavakhi, A.; Minakari, M.; Firouzian, H.; Assali, R.; Hekmatdoost, A.; Ferns, G. Effect of a probiotic and metformin on liver aminotransferases in non-alcoholic steatohepatitis: A double blind randomized clinical trial. Int. J. Prev. Med. 2013, 4, 531–537. [Google Scholar] [PubMed]
  99. Wong, V.W.; Won, G.L.; Chim, A.M.; Chu, W.C.; Yeung, D.K.; Li, K.C.; Chan, L. Treatment of non alcoholic steatohepatitis with probiotics. A proof-of-concept study. Ann. Hepatol. 2013, 12, 256–262. [Google Scholar] [PubMed]
  100. Eslamparast, T.; Poustchi, H.; Zamani, F.; Sharafkhah, M.; Malekzadeh, R.; Hetmatdoost, A. Synbiotic supplementation in nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled pilot study. Am. J. Clin. Nutr. 2014, 99, 535–542. [Google Scholar] [CrossRef] [PubMed]
  101. Ferolla, S.M.; Couto, C.A.; Costa-Silva, L.; Armiliato, G.N.; Pereira, C.A.; Martins, F.S.; Ferrari, M.D.L.A.; Vilela, E.G.; Torres, H.O.; Cunha, A.S.; et al. Beneficial Effect of Synbiotic Supplementation on Hepatic Steatosis and Anthropometric Parameters, But Not on Gut Permeability in a Population with Nonalcoholic Steatohepatitis. Nutrients 2016, 8, 397. [Google Scholar] [CrossRef] [PubMed]
  102. Mofidi, F.; Poustchi, H.; Yari, Z.; Nourinayyer, B.; Merat, S.; Sharafkhah, M.; Malekzadeh, R.; Hekmatdoost, A. Synbiotic supplementation in lean patients with non-alcoholic fatty liver disease: A pilot, randomised, double-blind, placebo-controlled, clinical trial. Br. J. Nutr. 2017, 117, 662–668. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, J.; Luo, H.; Jiang, Y.; Chen, P. Dietary capsaicin and antibiotics act synergistically to reduce non-alcoholic fatty liver disease induced by high fat diet in mice. Oncotarget 2017, 8, 38161–38175. [Google Scholar] [CrossRef] [PubMed]
  104. Ianiro, G.; Tilg, H.; Gasbarrini, A. Antibiotics as deep modulators of gut microbiota: Between good and evil. Gut 2016, 65, 1906–1915. [Google Scholar] [CrossRef] [PubMed]
  105. Fernandes, P.; Hashiguchi, T.; Fujii, M. Anti-NASH effects of solithromycin in NASH-HCC mouse model. Gastroenterology 2014, 146, S145–S146. [Google Scholar] [CrossRef]
  106. Bergheim, I.; Weber, S.; Vos, M.; Kramer, S.; Volynets, V.; Kaserouni, S.; McClain, C.J.; Bischoff, S.C. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin. J. Hepatol. 2008, 48, 983–992. [Google Scholar] [CrossRef] [PubMed]
  107. Douhara, A.; Moriya, K.; Yoshiji, H.; Noguchi, R.; Namisaki, T.; Kitade, M.; Kaji, K.; Aihara, Y.; Nishimura, N.; Takeda, K.; et al. Reduction of endotoxin attenuates liver fibrosis through suppression of hepatic stellate cell activation and remission of intestinal permeability in a rat non-alcoholic steatohepatitis model. Mol. Med. Rep. 2015, 11, 1693–1700. [Google Scholar] [CrossRef] [PubMed]
  108. Gangarapu, V.; Ince, A.T.; Baysal, B.; Kayar, Y.; Kılıç, U.; Gök, Ö.; Uysal, Ö.; Şenturk, H. Efficacy of rifaximin on circulating endotoxins and cytokines in patients with nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2015, 27, 840–845. [Google Scholar] [CrossRef] [PubMed]
  109. Cobbold, J.F.L.; Atkinson, S.; Marchesi, J.R.; Smith, A.; Wai, S.N.; Stove, J.; Shojaee-Moradie, F.; Jackson, N.; Umpleby, A.M.; Fitzpatrick, J.; et al. Rifaximin in Non-Alcoholic Steatohepatitis: An Open-Label Pilot Study. Hepatol. Res. 2017. [Google Scholar] [CrossRef] [PubMed]
  110. Paolella, G.; Vajro, P. Childhood obesity, breastfeeding, intestinal microbiota, and early exposure to antibiotics: What is the link? JAMA Pediatr. 2016, 170, 735–737. [Google Scholar] [CrossRef] [PubMed]
  111. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre, randomised, placebo-controlled trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef]
  112. Gadaleta, R.M.; van Erpecum, K.J.; Oldenburg, B.; Willemsen, E.C.; Renooij, W.; Murzilli, S.; Klomp, L.W.; Siersema, P.D.; Schipper, M.E.; Danese, S.; et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 2011, 60, 463–472. [Google Scholar] [CrossRef] [PubMed]
  113. Gege, C.; Kinzel, O.; Steeneck, C.; Schulz, A.; Kremoser, C. Knocking on FXR’s door: The “hammerhead”-structure series of FXR agonists—Amphiphilic isoxazoles with potent in vitroand in vivo activities. Curr. Top. Med. Chem. 2014, 14, 2143–2158. [Google Scholar] [CrossRef] [PubMed]
  114. Gu, M.; Zhang, S.; Zhao, Y.; Huang, J.; Wang, Y.; Li, Y.; Fan, S.; Yang, L.; Ji, G.; Tong, Q.; et al. Cycloastragenol improves hepatic steatosis by activating farnesoid X receptor signalling. Pharmacol. Res. 2017, 121, 22–32. [Google Scholar] [CrossRef] [PubMed]
  115. Duan, X.; Meng, Q.; Wang, C.; Liu, Z.; Liu, Q.; Sun, H.; Sun, P.; Yang, X.; Huo, X.; Peng, J.; et al. Calycosin attenuates triglyceride accumulation and hepatic fibrosis in murine model of non-alcoholic steatohepatitis via activating farnesoid X receptor. Phytomedicine 2017, 25, 83–92. [Google Scholar] [CrossRef] [PubMed]
  116. Adar, T.; Ben Ya’acov, A.; Lalazar, G.; Lichtenstein, Y.; Nahman, D.; Mizrahi, M.; Wong, V.; Muller, B.; Rawlin, G.; Ilan, Y. Oral administration of immunoglobulin G-enhanced colostrum alleviates insulin resistance and liver injury and is associated with alterations in natural killer T cells. Clin. Exp. Immunol. 2012, 167, 252–260. [Google Scholar] [CrossRef] [PubMed]
  117. Ben Ya’acov, A.; Lichtenstein, Y.; Zolotarov, L.; Ilan, Y. The gut microbiome as a target for regulatory T cell-based immunotherapy: Induction of regulatory lymphocytes by oral administration of anti-LPS enriched colostrum alleviates immune mediated colitis. BMC Gastroenterol. 2015, 15, 154. [Google Scholar] [CrossRef]
  118. Mizrahi, M.; Shabat, Y.; Ben Ya’acov, A. Alleviation of insulin resistance and liver damage by oral administration of Imm124-E is mediated by increased Tregs and associated with increased serum GLP-1 and adiponectin: Results of a phase I/II clinical trial in NASH. J. Inflamm. Res. 2012, 5, 141–150. [Google Scholar] [CrossRef] [PubMed]
  119. Su, D.; Nie, Y.; Zhu, A.; Chen, Z.; Wu, P.; Zhang, L.; Luo, M.; Sun, Q.; Cai, L.; Lai, Y.; et al. Vitamin D signaling through induction of Paneth Cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models. Front. Physiol. 2016, 7, 498. [Google Scholar] [CrossRef] [PubMed]
  120. Luger, M.; Kruschitz, R.; Kienbacher, C.; Traussnigg, S.; Langer, F.B.; Schindler, K.; Würger, T.; Wrba, F.; Trauner, M.; Prager, G.; et al. Prevalence of liver fibrosis and its association with non-invasive fibrosis and metabolic markers in morbidly obese patients with vitamin D deficiency. Obes. Surg. 2016, 26, 2425–2432. [Google Scholar] [CrossRef] [PubMed]
  121. Putignani, L.; Alisi, A.; Nobili, V. Pediatric NAFLD: The Future role of Patient-Tailored Probiotics Therapy. J. Pediatr. Gastroenterol. Nutr. 2016, 63, S6–S8. [Google Scholar] [CrossRef] [PubMed]
  122. Sheth, R.U.; Cabral, V.; Chen, S.P.; Wang, H.H. Manipulating bacterial communities by in situ microbiome engineering. Trends Genet. 2016, 32, 189–200. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Gut–liver axis components in normal conditions (right part) and in non-alcoholic fatty liver disease (NAFLD)(left part).The presence of the dysbiotic microbiome and an altered intestinal barrier influenced by bacterial ethanol (EtOH), possibly associated with disruption of tight-junctions (TJs) (“leaky gut”), promotes the translocation of several bacterial products into the portal circulation. The interaction of bacterial products with toll-like receptors (TLRs) on the hepatic cell surface promotes the progression from simple steatosis to inflammation and fibrosis of the liver. ROS: reactive species of oxygen; NASH: non-alcoholic steatohepatitis; LPS: lipopolysaccharide; TLR: toll-like receptor.
Figure 1. Gut–liver axis components in normal conditions (right part) and in non-alcoholic fatty liver disease (NAFLD)(left part).The presence of the dysbiotic microbiome and an altered intestinal barrier influenced by bacterial ethanol (EtOH), possibly associated with disruption of tight-junctions (TJs) (“leaky gut”), promotes the translocation of several bacterial products into the portal circulation. The interaction of bacterial products with toll-like receptors (TLRs) on the hepatic cell surface promotes the progression from simple steatosis to inflammation and fibrosis of the liver. ROS: reactive species of oxygen; NASH: non-alcoholic steatohepatitis; LPS: lipopolysaccharide; TLR: toll-like receptor.
Children 04 00066 g001
Table 1. Summary of human studies with probiotics and synbiotics in non-alcoholic fatty liver disease.
Table 1. Summary of human studies with probiotics and synbiotics in non-alcoholic fatty liver disease.
ReferencePopulationStrainTimeOutcome
Probiotics
Loguercio et al., 2005 [90]Adults: 22 NAFLD, 20 alcoholic CIR, 20 HCV, 16 liver CIR (without definition)VSL#3 (Bifidobacteria, Lactobacilli, and Staphylococcus thermophilus)24 wksDecreased ALT and AST in all groups.
Solga et al., 2008 [91]4 adult patients with NAFLDVSL#3 (Bifidobacteria, Lactobacilli, and Staphylococcus thermophilus)32 wksIncreased liver steatosis.
Vajro et al., 2011 [87]20 obese children with NAFLDLactobacillus rhamnosus GG8 wksDecreased ALT and anti-peptidoglycan polysaccharide Abs.
Aller et al., 2011 [92]28 adult patients with NAFLDLactobacillus bulgaricus and Staphylococcus thermophilus12 wksDecreased ALT and GGT.
Mykhal’chyshyn et al., 2013 [93]72 adult patients with T2D and NAFLDLactobacillus, Lactococcus, Propionibacterium, Bifidobacterium, Aceticbacterium4 wksDecreased IL-6, IL-8, TNF-α, IL-1β and IFN-α.
Nabavi et al., 2014 [94]72 adult patients with NAFLDLactobacillus acidophilus La5 and Bifidobacterium breve subsp. lactis Bb128 wksDecreased ALT, AST, TC, and LDL-C.
Alisi et al., 2014 [88]44 obese children with NAFLDVSL#3 (Bifidobacteria, Lactobacilli, and Staphylococcus thermophilus )16 wksImproved fatty liver severity, decreased BMI and increased GLP1/aGLP1.
Famouri et al., 2016 [89]64 obese children with NAFLDLactobacillus acidophilus, Bifidobacterium lactis, Bifidobacterium bifidum, and Lactobacillus rhamnosus12 wksDecreased ALT; AST, TC, LDL-C, TG. Reduced waist circumference.
Sepideh et al., 2016 [95]42 adult patients with NAFLDLactobacillus casei, Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, and Staphylococcus thermophilus8 wksDecreased FBS, insulin, IR, TNF-α, and IL-6.
Synbiotics
Loguercio et al., 2002 [96]Adults: 12 HCV, 10 alcoholic cirrhosis, 10 NASHLactobacillus acidophilus, Bifidobacterium bifidum, Lactobacillus rhamnosus, Lactobacillus plantarum, Lactobacillus salivarius, Lactobacillus bulgaricus, Lactobacillus casei, Bifidobacterium lactis, Bifidobacterium breve, and FOS.8 wksNASH patients: decreased ALT, GGT, and TNF-α.
Malaguarnera et al., 2012 [97]66 adult patients with NASHBifidobacterium longum and FOS.24 wksReduced steatosis and NASH activity. Decreased AST, CRP, TNF-α and endotoxin.
Shavackhi et al., 2013 [98]64 adult patients with NAFLDLactobacillus acidophilus, Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, Staphylococcus thermophilus, and FOS.24 wksDecreased AST, ALT, TGs, TC, BMI and steatosis.
Wong et al., 2013 [99]20 adult patients with NASHLactobacillus plantarum, Lactobacillus delbrueckii spp. bulgaricus, Lactobacillus acidophilus, Lactobacillus rhamnosus, Bifidobacterium bifidum and inulin.26 wksDecreased IHTG content and AST.
Eslamparast et al., 2014 [100]52 adult patients with NAFLDLactobacillus casei, Lactobacillus rhamnosus, Staphylococcus thermophilus, Bifidobacterium breve, Lactobacillus acidophilus, Bifidobacterium longum, Lactobacillus bulgaricus, and FOS.30 wksInhibition of NF-κB and reduction of TNF-α.
Ferolla et al., 2016 [101]50 adult patients with NASHLactobacillus reuteri, guar gum and inulin.12 wksReduction in steatosis. Decreased weight, BMI and waist circumference.
Mofidi et al., 2017 [102]50 adult patients with NAFLD and normal/low BMILactobacillus casei, Lactobacillus rhamnosus, Staphylococcus thermophilus, Bifidobacterium breve, Lactobacillus acidophilus, Bifidobacterium longum and Lactobacillus bulgaricus, and FOS.28 wksReduction in steatosis and fibrosis. Decreased FBS, TGs and inflammatory mediators.
Abs: antibodies; ALT: alanine aminotransferase; AST: aspartate aminotransferase; BMI: body mass index; CIR: cirrhosis; CRP: C-reactive protein; FBS: fasting blood sugars; FOS: fructo-oligossacharides; GGT: γ-glutamyltranspeptidase; GLP1: glucagon-like peptide 1; IFN: interferon; IHTG: intrahepatic triacylglycerol; IL: interleukin; IR: insulin resistance; LDL-C: low-density lipoprotein cholesterol; NAFLD: non-alcoholic fatty liver disease; NF-κB: nuclear factor kB; T2D: type 2 diabetes; TC: total cholesterol; TGs: triglycerides; TNF-α: tumor necrosis factor α; wks: weeks.

Share and Cite

MDPI and ACS Style

Poeta, M.; Pierri, L.; Vajro, P. Gut–Liver Axis Derangement in Non-Alcoholic Fatty Liver Disease. Children 2017, 4, 66. https://0-doi-org.brum.beds.ac.uk/10.3390/children4080066

AMA Style

Poeta M, Pierri L, Vajro P. Gut–Liver Axis Derangement in Non-Alcoholic Fatty Liver Disease. Children. 2017; 4(8):66. https://0-doi-org.brum.beds.ac.uk/10.3390/children4080066

Chicago/Turabian Style

Poeta, Marco, Luca Pierri, and Pietro Vajro. 2017. "Gut–Liver Axis Derangement in Non-Alcoholic Fatty Liver Disease" Children 4, no. 8: 66. https://0-doi-org.brum.beds.ac.uk/10.3390/children4080066

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

Article Metrics

Back to TopTop