Open access peer-reviewed chapter

Nonalcoholic Fatty Liver Disease in Children: Role of the Gut Microbiota

Written By

Ding-You Li, Min Yang, Sitang Gong and Shui Qing Ye

Submitted: 02 October 2015 Reviewed: 04 July 2016 Published: 02 November 2016

DOI: 10.5772/64799

From the Edited Volume

The Gut Microbiome - Implications for Human Disease

Edited by Gyula Mozsik

Chapter metrics overview

2,031 Chapter Downloads

View Full Metrics

Abstract

Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common cause of liver disease among children and adolescents in industrialized countries due to increasing prevalence of obesity. It is generally recognized that both genetic and environmental risk factors contribute to the pathogenesis of NAFLD. Convincing evidences have shown that gut microbiota alteration is associated with NAFLD pathogenesis both in patients and animal models. Bacterial overgrowth and increased intestinal permeability are evident in NAFLD patients and lead to increased delivery of gut-derived bacterial products, such as lipopolysaccharide and bacterial DNA, to the liver through portal vein and then activation of toll-like receptors (TLRs), mainly TLR4 and TLR9, and their downstream cytokines and chemokines, resulting in hepatic inflammation. Currently, the role of gut microbiota in the pathogenesis of NAFLD is still the focus of many active clinical/basic researches. Modulation of gut microbiota with probiotics or prebiotics has been targeted as a preventive or therapeutic strategy on this pathological condition. Their beneficial effects on the NAFLD have been demonstrated in animal models and limited human studies.

Keywords

  • nonalcoholic fatty liver disease (NAFLD)
  • children
  • gut microbiota
  • probiotics
  • prebiotics

1. Introduction

A growing obesity epidemic over the past three decades has become a major public health concern in developed as well as developing countries. According to the 2012 National Health and Nutrition Examination Survey [1, 2], in the United States, 35.5% of men, 35.8% of women, and 16.9% of children (2–19 years old) were considered obese. The worldwide prevalence of overweight and obesity increased from 28.8 to 36.9% in men, and from 29.8 to 38.0% in women between 1980 and 2013 [3]. Specifically, the prevalence for children increased from 16.9 to 23.8% for boys and from 16.2 to 22.6% for girls in developed countries, and from 8.1 to 12.9% for boys and from 8.4 to 13.4% for girls in developing countries as well [3].

Nonalcoholic fatty liver disease (NAFLD) has become the most common cause of liver disease in children in industrialized countries due to increasing prevalence of obesity [4]. NAFLD is defined as hepatic fat infiltration >5% of hepatocytes based on liver biopsy after excessive alcohol intake, viral, autoimmune, or drug-induced liver disease have been excluded. NAFLD is characterized by liver damage similar to that caused by alcohol but occurs in individuals that do not consume toxic quantities of alcohol. NAFLD includes a spectrum of liver diseases from simple fat infiltration (steatosis) through nonalcoholic steatohepatitis (NASH, steatosis with liver inflammation) to hepatic fibrosis and even hepatocellular carcinoma. The prevalence of NAFLD in the United States was 9.6% in normal weight children and 38% in obese ones based on liver biopsy at autopsy after accidents [5]. In the United States, the highest rates of pediatric NAFLD are in Hispanic and Asian children. In a study of 748 school children in Taiwan, the rates of NAFLD were 3% in the normal weight, 25% in the overweight, and 76% in the obese children determined by ultrasonography [6]. NAFLD in children is associated with severe obesity and metabolic syndrome, which includes abdominal obesity, type-2 diabetes, dyslipidemia, and hypertension. This chapter briefly summarizes the current understanding of the pathogenesis of NAFLD, role of gut microbiota, and potential new treatment strategies.

Advertisement

2. NAFLD pathogenesis: current understanding

Although the pathogenesis of NAFLD is not completely understood, considerable progresses have been made in recent years in explicating the mechanisms behind liver injury. As in other complex diseases, both genetic and environmental factors contribute to NAFLD development and progression. It is generally accepted that there is a genetic predisposition. In patients with NAFLD, genomic studies have identified many single nucleotide polymorphisms (SNPs) variants in genes controlling lipid metabolism, proinflammatory cytokines, fibrotic mediators, and oxidative stress. The most important one is the patatin-like phospholipase domain-containing 3 gene (PNPLA3) [7]. PNPLA3 rs738409 variant has been shown to confer susceptibility to NAFLD in obese children in different ethnic groups [8]. Other reported susceptible genes include glucokinase regulatory protein (GCKR), transmembrane 6 superfamily member 2 (TM6SF2), G-protein-coupled-receptor 120 (GPR120), farnesyl-diphosphate farnesyltransferase 1 (FDFT1), parvin beta (PARVB), sorting and assembly machinery component (SAMM50), lipid phosphate phosphatase-related protein type 4 (LPPR4), solute carrier family 38 member 8 (SLC38A8), lymphocyte cytosolic protein-1 (LCP1), group-specific component (GC), protein phosphatase 1 regulatory subunit 3b (PPP1R3B), lysophospholipase-like 1 (LYPLAL1), neurocan (NCAN), and polipoprotein C3 (APOC3) [9, 10]. To date, the strongest SNP variants associated with pediatric NAFLD are the rs738409 in the PNPLA3 gene, the 1260326 in the GCKR gene, and the rs58542926 in the TM6SF2 gene.

Day and James initially proposed a two-hit hypothesis to explain the pathogenesis of NAFLD [11]. In individuals with genetic predisposition, the “first hit” results in liver fat accumulation (steatosis) due to environmental factors (e.g., western diet and lack of physical activity), obesity, insulin resistance, or metabolic syndrome. A subsequent “second hit”, such as free fatty acids, adipokines/cytokines, oxidative stress (reactive oxygen species, lipid peroxidation), gut microbiota-derived endotoxins, mitochondrial dysfunction, and stellate cell activation, further amplify liver injury and NASH progression. A recent proposed multiple parallel hits hypothesis suggested that gut-derived and adipose tissue-derived factors may play a central role [12]. Both two-hit and multiple parallel hit hypotheses recognized that insulin resistance plays a crucial role in NAFLD pathogenesis and other factors including genetic determinants, nutritional factors, adipose tissue, and the immune system may be necessary for NAFLD manifestation and progression [1113]. A new lipotoxicity hypothesis proposes that insulin resistance facilitates an excessive flow of free fatty acids to the liver, resulting in increased production of lipotoxic intermediates and eventually NASH, through oxidative stress, mitochondrial dysfunction, adiponectin, and other complex pathways [14, 15].

It has been well established that gut microbiota has been implicated in the development of NAFLD through the gut-liver axis [1618]. An alteration of gut microbiota composition leads to bacterial overgrowth and increased intestinal permeability [1921], resulting in translocation of gut micriobiota-derived products, such as lipopolysaccharide (LPS), bacterial DNA, and peptidoglycan, which would activate liver cell surface receptors (TLR4 and 9); a cascade of signal transductions is triggered and various cytokines and chemokines, such as TNF-α, TGF-β, IL-6, IL-10, CCL2, CCL5, and CxCL8, are released, leading to hepatic inflammation and fibrosis [22].

Evidences from both human and animal studies have supported important roles of gut microbiota-derived endotoxins, especially LPS, and their downstream signal pathways in the progression of NAFLD. Patients with NAFLD had increased serum endotoxin levels, with marked increases noted in NASH and early stage fibrosis. The increase in endotoxin level is related to IL-1α and TNF-α production [2326]. In genetically obese fatty/fatty rats and obese/obese mice, Yang et al. showed that LPS contributes to the development of steatohepatitis by sensitizing TNF-α [27].

Toll-like receptors (TLRs) have been shown to play a crucial role in pathogenesis of NAFLD. Activation of TLRs and the adaptor molecule, MyD88, results in a cascade of signal transduction leading to release of various cytokines (TNF-α, TGF-β, interleukin-6 (IL-6), and IL-10) and chemokines (CCL2, CCL5, and CXCL8), which have been associated with NAFLD progression and hepatic fibrosis, as demonstrated in both human and animal studies [28]. TLRs are a class of pattern recognizing proteins that perceive bacterial and viral components. Gut microbiota is a source of TLR ligands, which can stimulate production of proinflammatory cytokines in the liver. TLRs are expressed on Kupffer cells, biliary epithelial cells, hepatocytes, hepatic stellate cells, epithelial cells, and dendritic cells in the liver. Among 13 known TLRs, TLR2, TLR4, and TLR9 have been implicated in NAFLD pathogenesis [17].

TLR4 is mainly activated by LPS, a cell component of Gram-negative bacteria. Elevated plasma and portal LPS levels are evident in human and animals with NAFLD [25, 2932]. In methionine choline deficient diet(MCDD)-induced mouse model of NASH, liver injury and inflammatory cytokine production increased after challenge with LPS [33]. Rivera et al. further demonstrated histological change typical of steatohepatitis (extensive macrovesicular steatosis and necrosis), three-fold increase of portal blood endotoxin level, and enhanced TLR4 expression in wild-type mice fed with MCDD [31]. In a mouse model of high-fat diet-induced NAFLD, TLR4 signaling is involved in free fatty-acid-induced NF-kB activation in hepatocytes through release of free high-mobility group box1 (HMGB1), which is a key molecule for the activation of the TLR4/MyD88-dependent pathway [34]. TLR4 mutant mice fed with fructose-enriched diet had significantly less hepatic steatosis and lower TNFα levels in comparison to fructose-fed wild-type mice, indicating an important role of LPS/TLR4 signaling in fructose-induced NAFLD [35]. Plasma LPS levels are also markedly elevated in children and adults with NAFLD [25, 29, 30, 32]. Thus, gut microbiota-derived LPS/TLR4 signaling pathway is crucial for the progression of NAFLD in humans as well as animal models.

TLR9 is activated by bacterial DNA CpG motif and induces proinflammatory cytokine production. In a mouse model of CDAA diet-induced NASH, Miura et al. showed hepatic inflammation and fibrosis in wild-type mice, which was suppressed in mice deficient in TLR9 or MyD88, suggesting the critical role of the TLR9/MyD88 signaling pathway in the pathogenesis of NASH [36].

Inflammasomes have been shown to be major contributors to inflammation and are upregulated in mouse models of MCDD or high-fat-induced NASH and in livers of NASH patients. Stimulation of TLR4 by LPS can further activate inflammasomes [37]. In genetic inflammasome-deficiency mice, an altered gut microbiota configuration is associated with abnormal TLR4 and TLR9 agonist accumulation in the portal circulation, resulting in elevated hepatic TNF-α expression and exacerbation of hepatic steatosis and inflammation [38].

TLR2 recognizes components from Gram-positive and Gram-negative bacteria, as well as mycoplasma and yeast. In comparison to wild-type mice, TLR2-deficiency animals are substantially protected from high-fat diet-induced adiposity, insulin resistance, hypercholesterolemia, and hepatic steatosis [39]. In contrast, increased hepatic inflammation and TNF-α mRNA expression were observed in TLR2-deficiency mice fed with MCDD [33, 40]. The conflicting results of the role of TLR2 signaling in those studies could be due to different animal models used, different gut microbial ligands involved or compensation by other TLRs.

Advertisement

3. Modulation of gut microbiota: effects of prebiotics and probiotics on NAFLD

Given the accumulating evidence of the critical role of gut microbiota in the pathogenesis of NAFLD, microbiota manipulation has been targeted as a potentially therapeutic option for this pathological condition. Possible strategies for altering gut microbiota include probiotics, prebiotics, synbiotics, antibiotics, dietary modification/supplementation, and microbiota transplantation. So far, only probiotics have been tested for the treatment of NAFLD in animal models and human subjects with promising effects.

Probiotics are live commensal microorganisms that have been shown to beneficially modulate the host’s gut microbiota. In animal models of NAFLD, VSL#3 (a probiotic mixture containing streptococcus, Bifidobacterium, and lactobacillus) improved hepatic inflammation and decreased hepatic steatosis with reduction of serum alanine aminotransferase (ALT) levels. Those changes were associated with decreased hepatic expression of TNF-mRNA and reduced activity of Jun N-terminal kinase (JNK) [4143]. In methionine choline deficient diet (MCDD)-induced NASH rats treated with probiotic mixture containing 6 or 13 bacterial strains, which were isolated from the healthy human stool samples, improved hepatic inflammation, likely in part through modulation of TNF-α activity [44]. Furthermore, the treatment of apolipoprotein E-deficiency mice with dextran sulfate sodium (DSS) induced histopathological features typical of steatohepatitis, which were prevented by 12-week VSL#3 administration, through modulation of the expression of nuclear receptors, peroxisome proliferator-activated receptor-γ, Farnesoid-X-receptors, and vitamin D receptor [45].

In human studies, Aller et al. reported that a 3-month treatment with Lactobacillus bulgaricus and Streptococcus thermophilus improved liver aminotransferases in adult patients with NAFLD [46]. Alisi et al. performed a double-blind and placebo-controlled RCT to assess the effect of VSL#3 in 44 obese children with biopsy-proven NAFLD and demonstrated that VSL#3 supplement for 4 months significantly improved hepatic steatosis and BMI [47].

Prebiotics are nondigestible dietary fibers that stimulate the growth and activity of intestinal bacteria. In genetically obese mice, supplementation with prebiotics (oligofructose, a mix of fermentable dietary fibers) decreased plasma levels of LPS and cytokines (TNF-α, IL1b, IL1α, IL6, and INFγ) and reduced gut permeability through a mechanism involving glucagon-like peptide-2 [48]. Lactulose, as a prebiotic, can promote the growth of certain intestinal bacteria such as Lactobacillus and Bifidobacterium. In a rat model of high-fat diet-induced steatohepatitis, lactulose improved hepatic inflammatory activity and decreased serum endotoxin levels [49]. Human studies with prebiotics are very limited. In an earlier clinical pilot study in patients with biopsy-proven NASH, dietary supplementation of oligofructose 16 g/day for 8 weeks significantly decreased serum aminotransferases and insulin levels [50]. There have been no randomized, controlled, double-blind, prospective clinical trials of prebiotics on NAFLD, except a randomized controlled trial protocol, which will randomize adults with confirmed NAFLD to either a 16 g/day prebiotic supplemented group or isocaloric placebo group for 24 weeks (n = 30/group) [51].

Advertisement

4. NAFLD in children

4.1. Gut microbiota and NAFLD in children

Given the important role of gut microbiota in obesity and metabolic syndrome [52, 53], it is not surprising that ever-increasing literature in recent years suggested a potential role of gut microbiota in NAFLD pathogenesis. An observation by Spencer et al. provided the initial evidence that gut microbiota and human fatty liver are closely linked [54]. In adult subjects with choline-deficient diet-induced fatty liver, gut microbiota compositions were associated with changes in liver fat in each subject during choline depletion. Subsequently, Mouzaki et al. showed that patients with NASH had a lower percentage of Bacteroidetes compared to both simple steatosis and healthy controls and higher fecal Clostridium coccoides compared to those with simple steatosis [55]. There was an inverse and diet/BMI-independent association between the presence of NASH and percentage of Bacteroidetes, suggesting a link between gut microbiota and NAFLD severity. Raman et al. reported an over-representation of Lactobacillus species and selected members of phylum Firmicutes (Lachnospiraceae; genera, Dorea, Robinsoniella, and Roseburia) in NAFLD patients [56]. A recent study identified Bacteroides as independently associated with NASH and Ruminococcus with significant fibrosis and further confirmed the association of NAFLD severity with gut dysbiosis [57].

In a pediatric cohort of 63 children, Zhu et al. determined the composition of gut bacterial communities of obese children with NASH [58]. They found that Bacteroidetes were significantly elevated (mainly Prevotella) in obese and NASH patients compared to lean healthy children and that an increased abundance of ethanol-producing Escherichia in NASH children was observed. Ethanol can promote gut permeability. A recent study by Michail et al. showed that children with NAFLD had more abundant Gammaproteobacteria and Prevotella and significantly higher levels of ethanol, with differential effects on short chain fatty acids [59]. Both studies demonstrated that the gut microbiota profile in pediatric NAFLD is different from lean healthy children, with more ethanol-producing bacteria, suggesting that endogenous alcohol production by intestinal microbiota may play a role in NAFLD pathogenesis. Engstler et al. also showed that fasting ethanol levels were positively associated with measures of insulin resistance and significantly higher in children with NAFLD than in controls [60]. Interestingly, with further animal experiments, they demonstrated that increased blood ethanol levels in children with NAFLD may result from insulin-dependent impairments of alcohol dehydrogenase activity in liver tissue rather than from an increased endogenous ethanol synthesis [60]. Taken together, human studies demonstrated significant differences in gut microbiota between normal subjects and patients with NAFLD. However, there were great variations in microbiota compositions among these human studies, likely due to patient’s age, fatty liver disease stages, study design, methods used, and observation endpoints.

4.2. Current management guidelines

All children with BMI ≥ 95th percentile or 85–94th percentile with risk factors (e.g., central obesity, metabolic syndrome, and strong family history) are recommended to have liver function test and hepatic ultrasonography [4, 61]. Since infants and children < 3 years old with fatty liver are less likely to have NAFLD, tests should be performed to exclude genetic, metabolic, syndromic, and systemic causes, such as fatty acid oxidation defects, lysosomal storage diseases, and peroxisomal disorders. In older children and teenagers, metabolic, infectious, toxic, and systemic causes should also be considered for differential diagnosis. Recommended common laboratory tests include viral hepatitis panel, α-1 antitrypsin phenotype, ceruloplasmin, antinuclear antibody, lipid profile, TSH, and celiac panel.

Ultrasonography is the only imaging technique used for NAFLD screening in children because it is safe, noninvasive, widely available, relatively inexpensive, and can detect evidence of portal hypertension. Liver biopsy is recommended to exclude other treatable disease, in cases of clinically suspected advanced liver disease, before pharmacological/surgical treatment, and as part of a structured intervention protocol or clinical research trial [4, 61].

Treatment options for children with NAFLD are limited by a small number of randomized clinical trials and insufficient information on the natural history of the condition to assess risk-benefit ratios [4, 62]. So far, weight loss, though hard to achieve, is still the cornerstone of treatment regimen. Koot et al. demonstrated that a lifestyle intervention (physical exercise, dietary change, and behavioral modification) of 6 months significantly improved hepatic steatosis and serum aminotransferases in 144 children with NAFLD [63]. A long-term follow-up study showed that the greatest decrease of NAFLD prevalence was observed in children with the greatest overweight reduction [64]. Grønbæk et al. assessed the effect of a 10-week “weight loss camp” (restricted caloric intake and moderate exercise for one hour daily) in 117 obese children and found that the children had an average weight loss of 7.1 ± 2.7 kg, with significant improvements in hepatic steatosis, transaminases, and insulin sensitivity [65].

In children with poor adherence to lifestyle changes, pharmacological interventions and dietary supplementations, including antioxidants (vitamin E), insulin sensitizers (metoformin), ursodeoxycholic acid (UDCA), omega-3 docosahexaenoic acid (DHA), and probiotics, may be tried, but no randomized clinical trials have proved their effectiveness in children with NAFLD.

Advertisement

5. Summary and future directions

The increase of pediatric NAFLD is attributed to the worldwide obesity epidemic. Current evidences suggest that both genetic and environmental risk factors play a crucial role in the pathogenesis of NAFLD in children and adolescents. Although human studies clearly showed significant differences in gut microbiota between normal subjects and patients with NAFLD, there were great variations in microbiota compositions among these studies [66]. Adult patients have altered gut microbiota with an increase in the relative proportion of Bacteroidales and Clostridiales, whereas in children with NAFLD, ethanol-producing bacteria are predominant. Bacterial overgrowth and increased intestinal permeability are evident in NAFLD patients and lead to increased delivery of gut-derived bacterial products (e.g., LPS and bacterial DNA) to the liver through portal vein and then activation of toll-like receptors (TLRs), mainly TLR4 and TLR9, and their downstream cytokines and chemokines, resulting in hepatic inflammation [17].

Given the accumulating evidence of the critical role of gut-derived microbial factors in the development and/or progression of NAFLD, modulation of gut microbiota with probiotics and/or prebiotics has been targeted as a therapeutic option. Their beneficial effects on NALFD are promising based on studies in animal models and patients including children. However, before probiotics and prebiotics become prime-time therapeutic modalities for NAFLD in children, several issues need to be addressed. First, we still do not know whether all children with NAFLD are truly associated with altered intestinal microbiota, and if so, which microbiota is involved. Second, randomized clinical trials with appropriate powers are required to assess benefits of tailored interventions with probiotics and/or prebiotics in children with NAFLD. Finally, it is clinically important to know the best types of probiotics or prebiotics to be prescribed in children with NAFLD. Nevertheless, probiotics and other integrated strategies to modify intestinal microbiota are promising to become efficacious therapeutic modalities to treat NALFD, with emerging evidence to demonstrate that prebiotics and probiotics modulate the intestinal microbiota, improve epithelial barrier function, and reduce intestinal inflammation.

References

  1. 1. Flegal KM, Carroll MD, Kit BK, Ogden CL: Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA 2012; 307:491–497. DOI: 10.1001/jama.2012.39
  2. 2. Ogden CL, Carroll MD, Kit BK, Flegal KM: Prevalence of obesity and trends in body mass index among US children and adolescents, 1999–2010. JAMA 2012; 307:483–490. DOI: 10.1001/jama.2012.40
  3. 3. Ng M, Fleming T, Robinson M, et al: Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014; 384:766–781. DOI: 10.1016/S0140-6736(14)60460-8
  4. 4. 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. DOI: 10.1097/MPG.0b013e318252a13f
  5. 5. Schwimmer JB, Deutsch R, Kahen T, Lavine JE, Stanley C, Behling C: Prevalence of fatty liver in children and adolescents. Pediatrics 2006; 118:1388–1393.
  6. 6. Huang SC, Yang YJ: Serum retinol-binding protein 4 is independently associated with pediatric NAFLD and fasting triglyceride level. J Pediatr Gastroenterol Nutr. 2013; 56: 145–150. DOI: 10.1097/MPG.0b013e3182722aee
  7. 7. Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, Boerwinkle E, Cohen JC, Hobbs HH: Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008; 40:1461–1465. DOI: 10.1038/ng.257
  8. 8. Lin YC, Chang PF, Chang MH, Ni YH: Genetic variants in GCKR and PNPLA3 confer susceptibility to nonalcoholic fatty liver disease in obese individuals. Am J Clin Nutr. 2014; 99:869–874. DOI: 10.3945/ajcn.113.079749
  9. 9. Marzuillo P, Miraglia del Giudice E, Santoro N: Pediatric fatty liver disease: role of ethnicity and genetics. World J Gastroenterol. 2014; 20:7347–7355. DOI: 10.3748/wjg.v20.i23.7347
  10. 10. Marzuillo P, Grandone A, Perrone L, Miraglia Del Giudice E: Understanding the pathophysiological mechanisms in the pediatric non-alcoholic fatty liver disease: the role of genetics. World J Hepatol. 2015; 7:1439–1443. DOI: 10.4254/wjh.v7.i11.1439
  11. 11. Day CP, James OF: Steatohepatitis: a tale of two “hits”? Gastroenterology 1998; 114:842–845.
  12. 12. Tilg H, Moschen AR: Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology 2010; 52:1836–1846. DOI: 10.1002/hep.24001
  13. 13. Yang M, Gong S, Ye SQ, Lyman B, Geng L, Chen P, Li DY: Non-alcoholic fatty liver disease in children: focus on nutritional interventions. Nutrients 2014; 6:4691–4705. DOI: 10.3390/nu6114691
  14. 14. Neuschwander-Tetri BA: Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology 2010; 52:774–788. DOI: 10.1002/hep.23719
  15. 15. Bechmann LP, Kocabayoglu P, Sowa JP, Sydor S, Best J, Schlattjan M, Beilfuss A, Schmitt J, Hannivoort RA, Kilicarslan A, Rust C, Berr F, Tschopp O, Gerken G, Friedman SL, Geier A, Canbay A: Free fatty acids repress small heterodimer partner (SHP) activation and adiponectin counteracts bile acid-induced liver injury in superobese patients with nonalcoholic steatohepatitis. Hepatology 2013; 57:1394–1406. DOI: 10.1002/hep.26225
  16. 16. Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S, Cartenì M, Nardone G: Gut-liver axis: the impact of gut microbiota on non alcoholic fatty liver disease. Nutr Metab Cardiovasc Dis. 2012; 22:471–476. DOI: 10.1016/j.numecd.2012.02.007
  17. 17. Miura K, Ohnishi H: Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World J Gastroenterol. 2014; 20:7381–7391. DOI: 10.3748/wjg.v20.i23.7381
  18. 18. Kirpich IA, Marsano LS, McClain CJ: Gut-liver axis, nutrition, and non-alcoholic fatty liver disease. Clin Biochem. 2015; 48:923–930. DOI: 10.1016/j.clinbiochem.2015.06.023
  19. 19. Wigg AJ, Roberts-Thomson IC, Dymock RB, McCarthy PJ, Grose RH, Cummins AG: The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut 2001; 48:206–211.
  20. 20. Sabaté JM, Jouët P, Harnois F, Mechler C, Msika S, Grossin M, Coffin B: High prevalence of small intestinal bacterial overgrowth in patients with morbid obesity: a contributor to severe hepatic steatosis. Obes Surg. 2008; 18:371–377. DOI: 10.1007/s11695-007-9398-2
  21. 21. Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, Mascianà R, Forgione A, Gabrieli ML, Perotti G, Vecchio FM, Rapaccini G, Gasbarrini G,Day CP, Grieco A: Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009; 49:1877–1887. DOI: 10.1002/hep.22848
  22. 22. Li DY, Yang M, Edwards S, Ye SQ: Non-alcoholic fatty liver disease: for better or worse, blame gut microbiota? JPEN J Parenter Enteral Nutr. 2013; 37:787–793. DOI: 10.1177/0148607113481623
  23. 23. Poniachik J, Csendes A, Díaz JC, Rojas J, Burdiles P, Maluenda F, Smok G, Rodrigo R, Videla LA: Increased production of IL-1alpha and TNF-alpha in lipopolysaccharide-stimulated blood from obese patients with non-alcoholic fatty liver disease. Cytokine 2006; 33:252–257.
  24. 24. Ruiz AG, Casafont F, Crespo J, Cayón A, Mayorga M, Estebanez A, Fernadez-Escalante JC, Pons-Romero F: Lipopolysaccharide-binding protein plasma levels and liver TNF-alpha gene expression in obese patients: evidence for the potential role of endotoxin in the pathogenesis of non-alcoholic steatohepatitis. Obes Surg. 2007; 17:1374–1380.
  25. 25. Harte AL, da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd EM, Tripathi G, Ashour E, Abdalla MS, Sharada HM, Amin AI, Burt AD, Kumar S, Day CP, McTernan PG: Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflamm (Lond). 2010; 7:15. DOI: 10.1186/1476-9255-7-15
  26. 26. Verdam FJ, Rensen SS, Driessen A, Greve JW, Buurman WA: Novel evidence for chronic exposure to endotoxin in human nonalcoholic steatohepatitis. J Clin Gastroenterol. 2011; 45:149–152. DOI: 10.1097/MCG.0b013e3181e12c24
  27. 27. Yang SQ, Lin HZ, Lane MD, Clemens M, Diehl AM: Obesity increases sensitivity to endotoxin liver injury: implications for the pathogenesis of steatohepatitis. Proc Natl Acad Sci U S A. 1997; 94:2557–2562.
  28. 28. Braunersreuther V, Viviani GL, Mach F, Montecucco F: Role of cytokines and chemokines in non-alcoholic fatty liver disease. World J Gastroenterol. 2012; 18:727–735.DOI: 10.3748/wjg.v18.i8.727
  29. 29. Alisi A, Manco M, Devito R, Piemonte F, Nobili V: Endotoxin and plasminogen activator inhibitor-1 serum levels associated with nonalcoholic steatohepatitis in children. J Pediatr Gastroenterol Nutr. 2010; 50:645–649. DOI: 10.1097/MPG.0b013e3181c7bdf1
  30. 30. Pendyala S, Walker JM, Holt PR: A high-fat diet is associated with endotoxemia that originates from the gut. Gastroenterology 2012; 142:1100–1101. DOI: 10.1053/j.gastro.2012.01.034
  31. 31. Rivera CA, Adegboyega P, van Rooijen N, Tagalicud A, Allman M, Wallace M: Toll-like receptor-4 signaling and Kupffer cells play pivotal roles in the pathogenesis of non-alcoholic steatohepatitis. J Hepatol. 2007; 47:571–579.
  32. 32. Sharifnia T, Antoun J, Verriere TG, Suarez G, Wattacheril J, Wilson KT, Peek RM Jr, Abumrad NN, Flynn CR: Hepatic TLR4 signaling in obese NAFLD. Am J Physiol Gastrointest Liver Physiol. 2015; 309:G270–278. DOI: 10.1152/ajpgi.00304.2014
  33. 33. Szabo G, Velayudham A, Romics L Jr, Mandrekar P: Modulation of non-alcoholic steatohepatitis by pattern recognition receptors in mice: the role of toll-like receptors 2 and 4. Alcohol Clin Exp Res. 2005; 29(11Suppl):140S–145S.
  34. 34. Li L, Chen L, Hu L, Liu Y, Sun HY, Tang J, Hou YJ, Chang YX, Tu QQ, Feng GS, Shen F, Wu MC, Wang HY: Nuclear factor high-mobility group box1 mediating the activation of Toll-like receptor 4 signaling in hepatocytes in the early stage of nonalcoholic fatty liver disease in mice. Hepatology 2011; 54:1620–1630. DOI: 10.1002/hep.24552
  35. 35. Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC, Bergheim I: Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Hepatology 2009; 50:1094–1104. DOI: 10.1002/hep.23122
  36. 36. Miura K, Kodama Y, Inokuchi S, Schnabl B, Aoyama T, Ohnishi H, Olefsky JM, Brenner DA, Seki E: Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology 2010; 139:323–334.e7. DOI: 10.1053/j.gastro.2010.03.052
  37. 37. 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. DOI: 10.1002/hep.24341
  38. 38. 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 2012; 482:179–185. DOI: 10.1038/nature10809
  39. 39. Himes RW, Smith CW: Tlr2 is critical for diet-induced metabolic syndrome in a murine model. FASEB J. 2010; 24:731–739. DOI: 10.1096/fj.09-141929
  40. 40. Rivera CA, Gaskin L, Allman M, Pang J, Brady K, Adegboyega P, Pruitt K: Toll-like receptor-2 deficiency enhances non-alcoholic steatohepatitis. BMC Gastroenterol. 2010; 10:52. DOI: 10.1186/1471-230X-10-52
  41. 41. Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, Desimone C, Song XY, Diehl AM: Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology 2003; 37:343–350.
  42. 42. Velayudham A, Dolganiuc A, Ellis M, Petrasek J, Kodys K, Mandrekar P, Szabo G: VSL#3 probiotic treatment attenuates fibrosis without changes in steatohepatitis in a diet-induced nonalcoholic steatohepatitis model in mice. Hepatology 2009; 49:989–997. DOI: 10.1002/hep.22711
  43. 43. Xu RY, Wan YP, Fang QY, Lu W, Cai W: Supplementation with probiotics modifies gut flora and attenuates liver fat accumulation in rat nonalcoholic fatty liver disease model. J. Clin Biochem Nutr. 2012; 50:72–77. DOI: 10.3164/jcbn.11-38
  44. 44. Karahan N, Işler M, Koyu A, Karahan AG, Başyığıt Kiliç G, Cırış IM, Sütçü R, Onaran I, Cam H, Keskın M: Effects of probiotics on methionine choline deficient diet-induced steatohepatitis in rats. Turk J Gastroenterol. 2012; 23:110–121.
  45. 45. Mencarelli A, Cipriani S, Renga B, Bruno A, D'Amore C, Distrutti E, Fiorucci S: VSL#3 resets insulin signaling and protects against NASH and atherosclerosis in a model of genetic dyslipidemia and intestinal inflammation. PLoS One 2012; 7:e45425. DOI: 10.1371/journal.pone.0045425
  46. 46. Aller R, De Luis DA, 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.
  47. 47. 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. DOI: 10.1111/apt.12758
  48. 48. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, Muccioli GG, Delzenne NM: Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009; 58:1091–1103. DOI: 10.1136/gut.2008.165886
  49. 49. Fan JG, Xu ZJ, Wang GL: Effect of lactulose on establishment of a rat non-alcoholic steatohepatitis model. World J Gastroenterol. 2005; 11:5053–5056.
  50. 50. Daubioul CA, Horsmans Y, Lambert P, Danse E, Delzenne NM: Effects of oligofructose on glucose and lipid metabolism in patients with nonalcoholic steatohepatitis: results of a pilot study. Eur J Clin Nutr. 2005; 59:723–726.
  51. 51. Lambert JE, Parnell JA, Eksteen B, Raman M, Bomhof MR, Rioux KP, Madsen KL, Reimer RA: Gut microbiota manipulation with prebiotics in patients with non-alcoholic fatty liver disease: a randomized controlled trial protocol. BMC Gastroenterol. 2015; 15:169. DOI: 10.1186/s12876-015-0400-5
  52. 52. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI: An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006; 444:1027–1031.
  53. 53. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S: Host-gut microbiota metabolic interactions. Science 2012; 336:1262–1267. DOI: 10.1126/science.1223813
  54. 54. Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA: Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011; 140:976–986. DOI: 10.1053/j.gastro.2010.11.049
  55. 55. Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK, Fischer SE, McGilvray ID, Allard JP: Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013; 58:120–127. DOI: 10.1002/hep.26319
  56. 56. 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. 2013; 11:868–875. e1-3. DOI: 10.1016/j.cgh.2013.02.015
  57. 57. Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo-Perez F, Guy CD, Seed PC, Rawls JF, David LA, Hunault G, Oberti F, Calès P, Diehl AM: The severity of NAFLD is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016; 63:764–775. DOI: 10.1002/hep.28356
  58. 58. Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, Gill SR: Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 2013; 57:601–609. DOI: 10.1002/hep.26093
  59. 59. Michail S, Lin M, Frey MR, Fanter R, Paliy O, Hilbush B, Reo NV: Altered gut microbial energy and metabolism in children with non-alcoholic fatty liver disease. FEMS Microbiol Ecol. 2015; 91:1–9. DOI: 10.1093/femsec/fiu002
  60. 60. Engstler AJ, Aumiller T, Degen C, Dürr M, Weiss E, Maier IB, Schattenberg JM, Jin CJ, Sellmann C, Bergheim I: Insulin resistance alters hepatic ethanol metabolism: studies in mice and children with non-alcoholic fatty liver disease. Gut. 2015. DOI: 10.1136/gutjnl-2014-308379. [Epub ahead of print]
  61. 61. Nobili V, Alkhouri N, Alisi A, Della Corte C, Fitzpatrick E, Raponi M, Dhawan A: Nonalcoholic fatty liver disease: a challenge for pediatricians. JAMA Pediatr. 2015;169:170–176. DOI: 10.1001/jamapediatrics.2014.2702
  62. 62. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ: The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 2012; 55:2005–2023. DOI: 10.1002/hep.25762
  63. 63. Koot BG, van der Baan-Slootweg OH, Tamminga-Smeulders CL, Rijcken TH, Korevaar JC, van Aalderen WM, Jansen PL, Benninga MA: Lifestyle intervention for non-alcoholic fatty liver disease: prospective cohort study of its efficacy and factors related to improvement. Arch Dis Child. 2011; 96:669–674. DOI: 10.1136/adc.2010.199760
  64. 64. Reinehr T, Schmidt C, Toschke AM, Andler W: Lifestyle intervention in obese children with non-alcoholic fatty liver disease: 2-year follow-up study. Arch Dis Child. 2009; 94:437–442. DOI: 10.1136/adc.2008
  65. 65. Grønbæk H, Lange A, Birkebæk NH, Holland-Fischer P, Solvig J, Hørlyck A, Kristensen K, Rittig S, Vilstrup H: Effect of a 10-week weight loss camp on fatty liver disease and insulin sensitivity in obese Danish children. J Pediatr Gastroenterol Nutr. 2012; 54:223–228. DOI: 10.1097/MPG.0b013e31822cdedf
  66. 66. Wieland A, Frank DN, Harnke B, Bambha K: Systematic review: microbial dysbiosis and nonalcoholic fatty liver disease. Aliment Pharmacol Ther. 2015; 42:1051–1063. DOI: 10.1111/apt.13376

Written By

Ding-You Li, Min Yang, Sitang Gong and Shui Qing Ye

Submitted: 02 October 2015 Reviewed: 04 July 2016 Published: 02 November 2016