Gut microbiota and non-alcoholic fatty liver disease
 
Paraskevas Gkolfakis, George Dimitriadis and Konstantinos Triantafyllou
Athens, Greece
 
Author Affiliations: Hepatogastroenterology Unit, Second Department of Internal Medicine and Research Institute, Attikon University General Hospital, Medical School, Athens University, 124 62 Athens, Greece (Gkolfakis P, Dimitriadis G and Triantafyllou K)
Corresponding Author: Konstantinos Triantafyllou, MD, Hepatogastroenterology Unit, Second Department of Internal Medicine and Research Institute, Attikon University General Hospital, Medical School, Athens University, Rimini 1, 124 62 Athens, Greece (Tel: +302105832087; Email: ktriant@med.uoa.gr)
 
© 2015, Hepatobiliary Pancreat Dis Int. All rights reserved.
doi: 10.1016/S1499-3872(15)60026-1
Published online October 7, 2015.
 
 
Acknowledgment: We acknowledge Dr. AD Sioulas' critical evaluation of the manuscript.
Contributors: GP searched the literature and drafted the manuscript. DG reviewed the draft. TK conceived the idea and reviewed the draft. All authors approved the final version. TK is the guarantor.
Funding: None.
Ethical approval: Not needed.
Competing interest: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
 
 
BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a common disorder with poorly understood pathogenesis. Beyond environmental and genetic factors, cumulative data support the causative role of gut microbiota in disease development and progression.
 
DATA SOURCE: We performed a PubMed literature search with the following key words: "non-alcoholic fatty liver disease", "non-alcoholic steatohepatitis", "fatty liver", "gut microbiota" and "microbiome", to review the data implicating gut microbiota in NAFLD development and progression.
 
RESULTS: Recent metagenomic studies revealed differences in the phylum and genus levels between patients with fatty liver and healthy controls. While bacteroidetes and firmicutes remain the dominant phyla among NAFLD patients, their proportional abundance and genera detection vary among different studies. New techniques indicate a correlation between the methanogenic archaeon (methanobrevibacter smithii) and obesity, while the bacterium akkermanshia municiphila protects against metabolic syndrome. Among NAFLD patients, small intestinal bacterial overgrowth detected by breath tests might induce gut microbiota and host interactions, facilitating disease development.
 
CONCLUSIONS: There is evidence that gut microbiota participates in NAFLD development through, among others, obesity induction, endogenous ethanol production, inflammatory response triggering and alterations in choline metabolism. Further studies with emerging techniques are needed to further elucidate the microbiome and host crosstalk in NAFLD pathogenesis.
 
(Hepatobiliary Pancreat Dis Int 2015;14:572-581)
 
KEY WORDS: non-alcoholic fatty liver disease; non-alcoholic steatohepatitis; gut microbiota; 16S rRNA sequencing; archaea
 
 
Introduction
Non-alcoholic fatty liver disease (NAFLD), the liver manifestation of metabolic syndrome, is the main cause of liver enzymes abnormalities in Western countries.[1, 2] NAFLD definition requires lack of ongoing or recent excessive alcohol consumption (>20 g/d for men and >10 g/d for women, respectively) and exclusion of other causes of liver steatosis.[3] Histologic evidence of steatosis or in the absence of histology, γ-GT and/or aminotransferases elevations as well as compatible sonographic findings suffice for diagnosis. The spectrum of the disease encompasses liver steatosis, non-alcoholic steatohepatitis (NASH), NASH-related cirrhosis and hepatocellular carcinoma.[4] Moreover, NAFLD patients are at greater risk to develop cardiovascular diseases[5] and their overall mortality is higher than that of a matched general population.[6]
 
Obesity, diabetes mellitus and hypertriglyceridemia are the main risk factors for the development and progression of NAFLD.[7] Given the mounting prevalence of overweighed and obese individuals (65% and 30%, respectively in the USA),[8] it is apparent that NAFLD and its consequences represent major public health issues. NAFLD prevalence ranges from 3% to 30%,[7, 9] depending on the used diagnostic methods (biochemical markers, radiology, histology). NAFLD and NASH prevalence among an urban USA population is estimated to 20%[10] and 4%,[2] respectively. However, epidemiological data vary in special populations: Hispanics show higher prevalence than non-Hispanics, whereas non-Hispanics black individuals[11] as well as populations from Alaska[12] and American-Indians,[13] exhibit significantly lower prevalence (0.6%-2%) of NAFLD.
 
A "two hits" hypothesis about NAFLD pathogenesis has been proposed.[14] The first hit is increased triglycerides accumulation in the liver, whereas a second one (e.g. oxidative stress) induces liver parenchyma inflammation leading to NASH. Recently, investigators have proposed the "multiple parallel" hits hypothesis:[15, 16] inflammation may either precede or follow simple steatosis with multiple factors, namely lipotoxicity, increased oxidative stress, mitochondrial dysfunction and iron overload acting in parallel to promote NASH.[16] Moreover, a genetic predisposition to the disease is possible since a mutation in the patatin-like phospholipase domain-containing 3 gene has been recognized to strongly predict liver fat accumulation and disease progression.[17]
 
The aim of this review is to highlight key issues on gut microbiota-host crosstalk regarding the pathogenesis of NAFLD and NASH. We present herein the yield of a PubMed search from 1995 to 2014 using the key words "non-alcoholic fatty liver disease", "non-alcoholic steatohepatitis", "fatty liver", "gut microbiota" and "microbiome".
 
 
Gut microbiome
Gut microbiota is a group of commensal microorganisms that live synergistically with the host. They process complex, otherwise indigestible, polysaccharides to short-chain fatty acids, thus providing extra energy for the host. They also participate in the synthesis of vitamins (e.g. vitamin K) and in the development and maintenance of the immunity at the intestinal lumen level.[18, 19]
 
The adult type gut microbiota is made up from bacteria, viruses, protozoa, archaea, eukaryotes, yeasts and parasites. It counts more than 1014 cells,[20] more than 100 different bacterial species;[18] their genome counts up to 300 000 genes,[21] 100 times the number of the human genome. Among them, bacteria predominate with the Gram-positive short-chain fatty acids-producing firmicutes and the Gram-negative hydrogen-producing bacteroidetes being the main phyla, followed by proteobacteria, actinobacteria, bifidobacteria, etc.[22, 23] Based on the abundant genera, two basic enterotypes are recognized:[24] Enterotype 1, where bacteroides spp. dominate and enterotype 2, with abundance of prevotella spp.. The existence of a third enterotype--enterotype H[25]--with abundance of both bacteroides spp. and prevotella spp. has also been proposed.
 
 
Evaluating gut microbiota in NAFLD
In order to reveal microbiota composition, culture-dependent and culture-independent techniques have been implicated.[26] Traditional culture allows detection and semi-quantification of many bacterial groups. Nevertheless, since a large amount of gut bacteria requires special conditions (e.g. anaerobic environment) to grow, a loss of 80% of the detectable bacteria is anticipated.[22] Trying to overcome this problem, culture-independent techniques have been developed. Apart from the widely used quantitative real-time polymerase chain reaction (qRT-PCR), additional techniques based on the diversity in the sequence of the bacterial 16S ribosomal RNA (16S rRNA) gene are now utilized: either the entire or conserved regions of the 16S rRNA gene are amplified, the results are compared with the sequence-containing libraries, thus accurate bacterial species identification or the partial 16S rRNA sequencing (pyrosequencing) provides information about the number, nature and abundance of the diverse bacterial species[27] without need of an ex vivo bacterial culture or DNA cloning.
 
Furthermore, the "-omics" studies are reliable methods to specify the functional species that interact with the host. Accordingly, the entire bacterial genome (metagenomics), the expressed mRNA (metatranscriptomics), the obtained proteins from the investigated microenvironment (metaproteomics), and the produced metabolites (metabolomics) are used to distinguish different phenotypes as well as, potential host-microbiome interactions.
 
Table 1 summarizes data from human studies investigating stool microbiota in NAFLD. Zhu et al[25] studied a pediatric population of 22 biopsy-proven NASH subjects, 25 obese children and 16 healthy controls using 16S rRNA sequencing in stool samples. Sequencing revealed a significant increase in bacteroidetes with decreased levels of firmicutes among obese and NASH individuals. Actinobacteria were significantly decreased in NASH patients, whereas proteobacteria counts showed a gradual increase from healthy to obese and NASH patients. Authors also provided evidence of increased ethanol levels in NASH children, postulating a possible role of abundant ethanol-producing bacteria (such as Escherichia) in NASH progression.
 
The increased bacteroidetes/firmicutes ratio detected by Zhu et al[25] was not confirmed by Raman et al,[28] who studied 30 obese adult patients with clinical, biochemical and radiological suspicion of NAFLD and 30 healthy controls using multi-tag pyrosequencing in a single stool sample per patient. Although differences in family and genera level were detected, the authors could not demonstrate a significant difference in the phylum level between the two groups. Similarly, using qRT-PCR in stool samples of 33 patients with biopsy-proven NAFLD (22 with NASH) and 17 healthy controls, Mouzaki et al[29]revealed significantly lower counts of Bacteroides in NASH patients than patients with simple steatosis and healthy controls. Firmicutes, proteobacteria and actinobacteria counts were not different significantly among the groups, whereas archaea were detectable only in 9/50 subjects (2 with simple steatosis, 2 with NASH and 5 healthy controls).
 
Trying to clarify the impact of pro- and prebiotics on gut microbiota composition, Wong et al[30] evaluated biopsy-proven NASH patients and controls. Participants provided stool samples for microbial counts measurements using 16S rRNA sequencing. Bacteroidetes were the dominant phyla in both groups (NASH and healthy volunteers), followed by firmicutes, which showed a notable presence in healthy controls.
 
Finally, the effect of choline-deficient diet, a mechanism implicated in NAFLD pathogenesis, was studied by Spencer et al.[31] Stool samples were collected in predefined time points and 16S rRNA pyrosequencing was used for microbiota specification. The authors did not find any common change in the taxons' abundance in the different time points of the study. However, the dramatically fall to zero in Gammaproteobacteria (phylum Proteobacteria) counts and the strong correlation of baseline levels of Gammaproteobacteria to liver fat enrichment during the choline-deficient diet postulated that Gammaproteobacteria levels may predict the possibility of an individual to develop fatty liver when exposed to choline-deficient diet.
 
Taken together, the above studies provide different evidence regarding NAFLD/NASH and gut microbiota composition. Bacteroidetes/firmicutes ratio is increased in NASH children, while no change is revealed in NAFLD/NASH adults compared to healthy controls. This discrepancy at the phylum level raises questions about a potential role of microbiota in NAFLD/NASH development. When focusing at the genera, a wide diversity regarding flora composition can be detected. This finding can be attributed to different methods used or even different studied populations; however, it is unlikely to extract safe conclusions regarding a potential causative role of special phyla and genera in NAFLD/NASH development.
 
 
Potential mechanisms that implicate gut microbiota in the pathogenesis of NAFLD
Accumulating data from preclinical and clinical studies suggest a critical role of gut microbiota in NAFLD pathogenesis, mainly through predisposition to obesity, metabolism alterations (insulin resistance) and promoted liver inflammation. Moreover, many bacterial byproducts, such as ethanol, might demonstrate hepatotoxicity via stimulation of Kupffer cells to produce and exude nitric acid and cytokines[32] (Fig.).
 
Obesity
More than a decade ago, investigators showed that germ-free mice gained 42% less weight in comparison with animals that hosted gut microbiota, despite the fact that they consumed more calories.[33] Moreover, when cecal microbiota was transplanted from normal to germ-free mice, the animals gained 57% more weight without any change in consumed calories. The same group emphasized their results by showing that germ-free mice were unable to gain weight while maintaining a high-calorie diet.[34]
 
Turnbaugh et al[35] transplanted gut microbiota from obese ob/ob mice and lean ob/+ or +/+ mice to lean germ-free mice, respectively. Animals that received microbiota from obese donors showed a higher fat gain than those that received microbiota from lean donors. Obesity-associated gut microbiota was able to extract more energy from the diet via digesting otherwise indigestible polysaccharides into short-chain fatty acids. Further supporting the aforementioned hypothesis, the total fecal short-chain fatty acids concentration from 33 obese human individuals was 20% higher than that from 30 lean volunteers.[36]
 
A potential role of specific gut microbiota in obesity and subsequent NAFLD development has been proposed. Obese mice hosted 50% less bacteroidetes and more firmicutes as compared with lean controls, whereas archaea counts were significantly increased in the obese animals.[37]
 
The decreased bacteroidetes/firmicutes ratio was also detected in obese human studies. Overweighed and obese children at the age of 7 had less bacteroidetes and more S. aureus in stools collected at the age of 6 and 12 months of age.[38] A study of 12 obese individuals revealed more bacteroidetes and less firmicutes, but this ratio was reversed when they followed a fat or carbohydrate restricted diet for 1 year.[39] On the contrary, firmicutes and archaea showed lower counts in obese patients after bariatric surgery.[40] Enterotype 1 has been associated with the long-term consumption of animal proteins and saturated fat, whereas diet based on carbohydrates predisposes to Enterotype 2.[41] These data underline that the role of gut microbiota diversity pends to be further clarified in obesity.
 
In contrast, a recently described bacterium[42] Akkermansia municiphila has been associated with non-obese phenotype. Pregnant women who gained excess weight during pregnancy[43] as well as obese and overweight preschool children[44] had low fecal concentrations of A. municiphila. Similarly, A. municiphila counts were markedly increased in obese mice that underwent Roux-Y gastric bypass surgery.[45]
 
Experimental models provided evidence for a modulating role of A. municiphila in weight gain, type 2 diabetes mellitus and eventually NAFLD. "Treatment" of mice under high-fat-diet with A. municiphila led to induction of Tregs cells in the visceral adipose tissue. This resulted in the attenuation of adipose tissue inflammation and increased glucose tolerance, effects that resemble the anti-diabetic function of metformin.[46] Similarly, Everard et al[47] showed that A. municiphila administration in type 2 diabetes mellitus mice led to a gradual reversion of insulin resistance, adipose tissue inflammation, fat gain and endotoxemia.
 
Ethanol
Endogenous ethanol is a metabolite of many gut microbiota species. After its absorption, it reaches the liver via the portal vein.[48] Alcohol dehydrogenase catalyzes its oxidation in the liver, leading to acetate and acetaldehyde formation.[49] The first is a substrate for fatty acids synthesis, and the second produces reactive oxygen species. Therefore, ethanol metabolism induces triglycerides accumulation in the liver[50] and hepatic oxidative stress, thus fulfilling both steps of the "two hits" hypothesis.
 
Increased ethanol levels have been detected in obese patients[51] as well as, in non-alcohol consuming children with NASH,[25] indicating its causative role in NAFLD/NASH development. Moreover, significantly increased expression of the ethanol-metabolizing enzymes alcohol dehydrogenase, catalase and aldehyde dehydrogenase has been detected in NASH livers.[52] Ethanol may also promote NAFLD by increasing gut mucosal permeability[53] that induces endotoxemia:[54] five days ciprofloxacin administration in NASH patients with ethanol detected in their blood resulted in immeasurable levels of ethanol,[55] pointing to a close relationship of endogenous ethanol production and gut microbiota.
 
Lipopolysaccharide-endotoxemia-Toll-like receptors
Endotoxin is part of the Gram-negative bacterial cell membrane. Lipopolysaccharide (LPS), the active component of endotoxin, binds to the LPS-binding protein and its CD14 receptor[56] to form a complex that interacts with Toll-like receptors (TLRs) and activates inflammatory cascade.[57]
 
Genetically obese mice develop steatohepatitis after infusion of low doses of LPS.[58] Furthermore, LPS injected in NAFLD mice further promotes liver injury by enhancement of produced proinflammatory cytokines,[59] whereas high-fat-diet induces increased plasma circulating LPS.[60] Diet-induced NAFLD in rodents is also associated with increased levels of LPS.[60, 61] In human studies, subjects with NAFLD experience significantly higher levels of circulating endotoxin,[62, 63] with marked increases in early fibrosis, when compared with healthy controls.
 
TLRs activation leads to the translocation of NF-κB in the nucleus and induction of proinflammatory genes transcription, such as TNF-α, IL-1β, IL-6 and IL-12.[64] IL-1β augments triglycerides accumulation in the hepatocytes by enhancement of diacyloglycerol transferase that converts diglycerides into triglycerides.[65] Moreover, TNF-α inhibits insulin receptors as well as insulin-receptors-substrate-1, leading to increased levels of circulating insulin and therefore to insulin resistance.[62] Consequently, influx of fatty acids derived from adipose tissue is facilitated.[66]
 
Recently, inflammasomes deficiencies have been implicated in NAFLD development. Inflammasomes are cytoplasmic multiprotein complexes that regulate the cleavage of pro-IL-1β and pro-IL-18 into the respective cytokines, through activation of caspase-1.[48] Genetically deficient for components of the inflammasome (NLRP3 and NLRP6) mice had higher levels of LPS and bacterial DNA in the portal circulation. The former binds to TLR-4 and the latter to TLR-9, leading to increased expression of TNF-α in the liver, thus promoting liver inflammation and steatosis.[67]
 
To summarize, NAFLD and NASH are characterized by increased levels of endotoxin. Endotoxin, one among other components of the second hit, in the "two hits" hypothesis triggers a cascade to increased levels of proinflammatory cytokines, insulin resistance and triglycerides production, thus facilitating NAFLD development and progression. Moreover, TLRs- and inflammasome-deficient mice showed either NAFLD or steatosis worsening, strengthening the role of the endotoxin-path in NAFLD pathogenesis.
 
Choline
Choline, a phospholipid component of cell membrane, plays a crucial role in lipid transport from the liver.[68] Gut microbiota are implicated in choline metabolism by producing enzymes that catalyze choline into methylamines, which potentially induce inflammation when absorbed by the liver.[69] Metabolomics analysis in 129S6 mouse strain animals with high-fat-diet induced steatosis revealed a significant decrease in circulating phosphatidylcholine and increased urine excretion of its metabolites, thus supporting the presence of a gut microbiota phenotype that induces choline deficiency-mediated liver injury.[70] Apart from that, choline deficiency contributes to triglycerides accumulation in the liver and decreased liver secretion of very-low-density lipoprotein,[48] whereas choline deficient diet resulted in liver steatosis that reversed after choline substitution in animals.[71] Acknowledging the fact that the data derive from a deficient disease and therefore the model might not be suitable enough to explain NAFLD development, decreased choline levels and increased levels of toxic choline metabolites might represent the gut microbiota choline-deficiency-mediated mechanism in NAFLD development.
 
Dysbiosis
Investigators compared metabolic and histologic changes in mice receiving microbiota from donors with different metabolic phenotypes.[72] Mice receiving intestinal microbiota from donors with hyperglycemia and increased levels of proinflamamtory cytokines developed hyperglycemia, increased levels of insulin and macrovesicular steatosis, whereas mice receiving microbiota from normoglycemic donors did not show similar metabolic and histologic changes. Dysbiosis, namely increased counts of firmicutes in the former and increased population of Bacteroides vugatus in the latter receivers, was detected by means of 16S rRNA pyrosequencing techniques.
 
 
Small intestinal bacterial overgrowth - intestinal permeability
Small intestinal bacterial overgrowth (SIBO) is the presence of increased number of colonic type anaerobic and/or aerobic bacteria in the small intestine.[23] In combination with intestinal permeability, SIBO has been proposed as a pathogenetic mechanism of NAFLD/NASH development. Ectopic microbiota in SIBO may cause loose junctions in the small intestinal epithelium facilitating the flow of bacterial byproducts in the portal vein system and their absorption from the liver, where they manifest their toxic activities. There is no gold standard technique to diagnose SIBO yet, since even aspirate culture suffers from certain methodological caveats. Breath testing is the current diagnostic test for SIBO, whereas culture independent techniques like qRT-PCR and deep sequencing have also been used in this setting.[23]
 
The correlation of NAFLD and SIBO has been investigated in experimental[73] and human studies[53, 55, 74-79] (Table 2). Increased prevalence of SIBO in NAFLD/NASH humans[74, 78] and in a rat NASH model[73] has been indirectly documented by measuring the orocecal transit time.
 
In obese bariatric surgery patients, SIBO detected using hydrogen breath testing has been revealed as an independent factor for severe steatosis in liver biopsy.[76] Wigg et al[79] detected SIBO in 50% of NASH patients, in whom higher levels of TNF-α were also identified and lactulose breath test detected SIBO in 78% of NASH patients in association with TLR-4 expression and plasma levels of IL-8.[77] In contrast to the aforementioned data, among 20 NAFLD patients SIBO was identified in only 3 using glucose breath test.[53] Using the lactulose/mannitol test that assesses intestinal permeability, a higher index of intestinal permeability as calculated by the lactulose/mannitol urine excretion ratio was demonstrated in NAFLD patients.[53] Similarly, Miele et al[75] detected SIBO in 60% of 35 biopsy-proven NAFLD patients and a significant correlation between the severity of steatosis and increased intestinal permeability was evident. Moreover, patients with NAFLD and increased intestinal permeability had a 2-fold higher prevalence of SIBO in comparison with patients with normal intestinal permeability. Authors could not support a causative role of SIBO in the progression of NAFLD, since they did not reveal correlation between increased intestinal permeability and/or SIBO with steatohepatitis.
 
 
The role of archaea
Methanogenic archaea scavenge hydrogen (H) and ammonia (NH4) to produce methane (CH4). Hydrogen utilization optimizes microenvironmental intestinal lumen conditions that facilitate short-chain fatty acids production and salvage energy for the host by gut flora.[80] Methanobrevibacter smithii (M. smithii) is the predominant methanogenic archaeon detected in 70% of healthy individuals.[81] A causative role of M. smithii for obesity development has been proposed in a mouse model. Colonization either with M. smithii and Bacteroides thetaiotaomicron or B. thetaiotaomicron alone led to increased weight gain only in subjects colonized with M. smithii.[82]
 
Methane detection in breath test is associated with increased levels of M. smithii in stools,[83] setting this cheap and non-invasive method as a guide for further studies. Indeed, when methane-positive obese subjects were compared with obese methane-negative controls, they showed a 6.7 kg/m2 body-mass-index (BMI) increase.[84] Moreover, when methane and hydrogen were measured in 792 subjects using lactulose breath test, methane-positive and hydrogen-positive (M+/H+) individuals showed increased BMI and increased percent body fat compared with the other groups (M-/H-, M+/H-, M-/H+).[85] It was therefore proposed that hydrogen derived from increased microbiota fermentation acts as a tank providing fuel to hydrogen-required metabolism of methanogens. In order to strengthen the hypothesis that M. smithii influences obesity development, Mathur et al[86] correlated the location and extent of M. smithii colonization with weight gain in rats. Using qRT-PCR, investigators showed that M. smithii counts were higher in the small intestine (highest in the ileum), whereas total bacterial numbers were the lowest in the small intestine and the highest in the left colon and cecum. Of interest, mice fed high-fat-diet had a significantly increased M. smithii concentration in the duodenum, ileum and cecum, whereas no difference was found in the total bacterial counts. Furthermore, animals with all five intestinal parts colonized with M. smithii had higher body weight than those with limited colonization.
 
In conclusion, these studies provide evidence for the first time that methanogenic archaea counts, especially M. smithii that colonize not only the colon but also the small intestine correlate with diet-induced weight gain.
 
 
Probiotics as a novel therapeutic approach
Probiotics are "live microorganisms that, when administered in adequate amounts confer a health benefit on the host".[87] Studies in mice and rodents revealed a potential protective role of two Lactobacillus strains: Lactobacillus rhamnosus GG[88] and Lactobacillus casei Shirota,[89] in diet-induced NAFLD/NASH by demonstrating biochemical and histological attenuation of liver steatosis. Moreover, probiotics altered gut microbiota and prevented NAFLD progression in animals.[90] In humans, few low-quality randomized clinical trials investigated the potential therapeutic role for probiotics in patients with fatty liver. Wong et al[30] randomized 20 biopsy-proven NASH patients to receive either a combination of prebiotic and probiotic formula (containing Lactobacillus spp. and Bifidobacterium spp.) or prebiotic alone. Participants provided stool samples for microbial counts measurements using 16S rRNA sequencing, at baseline and at month six. Measurement of intrahepatic triglyceride content by spectroscopy was used to assess liver steatosis. Decreased counts of firmicutes and increased counts of Bacteroidetes were observed at 6 months in NASH patients experiencing lower levels of intrahepatic triglycerides content.
 
Moreover, despite different probiotics mixtures (Lactobacillus bulgaricus and Streptococcus thermophilus vs placebo,[91] Lactobacillus GG vs placebo,[92] Bifidobacterium longum with fructooligosaccharides vs placebo,[93] lepicol probiotic formula vs nothing[94]) and a variety of administration periods, a meta-analysis[95] provided evidence that probiotics might have a positive effect on biochemical markers in patients with NAFLD and NASH. More specifically, the use of probiotics was associated with a significant reduction of ALT, AST, cholesterol, TNF-α and homeostasis model assessment of insulin resistance. The amelioration of these parameters using probiotics creates promises for further scientific evaluation.
 
 
Perspectives
Gut microbiota is currently studied to elucidate its role in the pathophysiology of NAFLD. Excessive energy harvest, ethanol production, LPS-TLRs interaction and choline metabolism alterations are the main principal mechanisms implicated so far. Progress in molecular DNA-based techniques allows a more detailed characterization of this microbiota. While the gold standard for bacterial enumeration remains the qRT-PCR,[23] one of its disadvantages is that unknown species cannot be identified. In these terms, development of metagenomic techniques (16S rRNA, multi-tag sequencing) opens a promising window to investigate the microbiome. The aforementioned studies aimed to reveal a potential correlation between gut microbiota composition and NAFLD development or progression to NASH. Despite interesting findings regarding abundant phyla and genera, the results show wide discordance and heterogeneity. Factors that could explain these differences include selection criteria for the study populations, variable methodologies in defining NAFLD, unadjusted diet-induced manipulation of microbiota, medications intake that may alter gut microbiota, such as proton pump inhibitors,[96] as well as the DNA-based techniques' inability to distinguish whether the genetic material comes from alive and active or dead and inactive microorganisms.
 
Trying to temper these difficulties, RNA-, mRNA- and protein-based techniques (metagenomics, metatranscriptomics and metaproteomics) have been developed. Despite their limited availability, a future combination and standardization of these techniques might lead to deeper comprehension of the microbiota role in NAFLD.
 
Another issue that should be highlighted is that in the vast majority of the studies, only stool samples were examined. Isolate stool sample analysis may underestimate or even cloak the contribution of small intestinal flora and small bacterial overgrowth in NAFLD pathophysiology.[97] Advanced, culture-independent techniques applied directly on duodenal and small intestinal content and biofilm may uncover a potential region-dependent interaction between liver steatosis and gut microbiota. Moreover, in both experimental models and in humans, the role of archaea, yeasts and viruses in NAFLD pathogenesis remains largely uninvestigated.
 
Finally, there is limited evidence for microbiota manipulation with probiotics as prevention or therapy of NAFLD.
 
 
References
1 Bugianesi E, McCullough AJ, Marchesini G. Insulin resistance: a metabolic pathway to chronic liver disease. Hepatology 2005;42:987-1000. PMID: 16250043
2 Bhala N, Jouness RI, Bugianesi E. Epidemiology and natural history of patients with NAFLD. Curr Pharm Des 2013;19:5169-5176. PMID: 23394091
3 Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, et al. 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. PMID: 22488764
4 Duseja A, Chawla YK. Obesity and NAFLD: the role of bacteria and microbiota. Clin Liver Dis 2014;18:59-71. PMID: 24274865
5 Targher G, Bertolini L, Rodella S, Tessari R, Zenari L, Lippi G, et al. Nonalcoholic fatty liver disease is independently associated with an increased incidence of cardiovascular events in type 2 diabetic patients. Diabetes Care 2007;30:2119-2121. PMID: 17519430
6 Angulo P. Long-term mortality in nonalcoholic fatty liver disease: is liver histology of any prognostic significance? Hepatology 2010;51:373-375. PMID: 20101746
7 Bedogni G, Miglioli L, Masutti F, Tiribelli C, Marchesini G, Bellentani S. Prevalence of and risk factors for nonalcoholic fatty liver disease: the Dionysos nutrition and liver study. Hepatology 2005;42:44-52. PMID: 15895401
8 Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM. Prevalence of overweight and obesity in the United States, 1999-2004. JAMA 2006;295:1549-1555. PMID: 16595758
9 Clark JM, Brancati FL, Diehl AM. The prevalence and etiology of elevated aminotransferase levels in the United States. Am J Gastroenterol 2003;98:960-967. PMID: 12809815
10 Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011;34:274-285. PMID: 21623852
11 Browning JD, Szczepaniak LS, Dobbins R, Nuremberg P, Horton JD, Cohen JC, et al. Prevalence of hepatic steatosis in an urban population in the United States: impact of ethnicity. Hepatology 2004;40:1387-1395. PMID: 15565570
12 Fischer GE, Bialek SP, Homan CE, Livingston SE, McMahon BJ. Chronic liver disease among Alaska-Native people, 2003-2004. Am J Gastroenterol 2009;104:363-370. PMID: 19174808
13 Bialek SR, Redd JT, Lynch A, Vogt T, Lewis S, Wilson C, et al. Chronic liver disease among two American Indian patient populations in the southwestern United States, 2000-2003. J Clin Gastroenterol 2008;42:949-954. PMID: 18668704
14 Day CP, James OF. Steatohepatitis: a tale of two "hits"? Gastroenterology 1998;114:842-845. PMID: 9547102
15 Dowman JK, Tomlinson JW, Newsome PN. Pathogenesis of non-alcoholic fatty liver disease. QJM 2010;103:71-83. PMID: 19914930
16 Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology 2010;52:1836-1846. PMID: 21038418
17 Speliotes EK, Butler JL, Palmer CD, Voight BF; GIANT Consortium; MIGen Consortium; et al. PNPLA3 variants specifically confer increased risk for histologic nonalcoholic fatty liver disease but not metabolic disease. Hepatology 2010;52:904-912. PMID: 20648472
18 Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology 2009;136:65-80. PMID: 19026645
19 Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nat Rev Microbiol 2005;3:431-438. PMID: 15821725
20 Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev 2010;90:859-904. PMID: 20664075
21 Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic analysis of the human distal gut microbiome. Science 2006;312:1355-1359. PMID: 16741115
22 Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, et al. Diversity of the human intestinal microbial flora. Science 2005;308:1635-1638. PMID: 15831718
23 Triantafyllou K, Pimentel M. Small intestinal bacterial overgrowth. In: Lacy BE CM, DiBaise JK (eds), ed. Functional and motility disorders of the gastrointestinal tract. A case study approach. New York: Springer+Bussiness Media; 2015:125-136.
24 Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature 2011;473:174-180. PMID: 21508958
25 Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 2013;57:601-609. PMID: 23055155
26 Goel A, Gupta M, Aggarwal R. Gut microbiota and liver disease. J Gastroenterol Hepatol 2014;29:1139-1148. PMID: 24547986
27 Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature 2009;457:480-484. PMID: 19043404
28 Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2013;11:868-875.e1-3. PMID: 23454028
29 Mouzaki M, Comelli EM, Arendt BM, Bonengel J, Fung SK, Fischer SE, et al. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013;58:120-127. PMID: 23401313
30 Wong VW, Tse CH, Lam TT, Wong GL, Chim AM, Chu WC, et al. Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis--a longitudinal study. PLoS One 2013;8:e62885. PMID: 23638162
31 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. PMID: 21129376
32 Abu-Shanab A, Quigley EM. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2010;7:691-701. PMID: 21045794
33 Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004;101:15718-15723. PMID: 15505215
34 Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 2007;104:979-984. PMID: 17210919
35 Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature 2007;449:804-810. PMID: 17943116
36 Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010;18:190-195. PMID: 19498350
37 Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 2005;102:11070-11075. PMID: 16033867
38 Kalliomäki M, Collado MC, Salminen S, Isolauri E. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr 2008;87:534-538. PMID: 18326589
39 Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature 2006;444:1022-1023. PMID: 17183309
40 Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A 2009;106:2365-2370. PMID: 19164560
41 Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105-108. PMID: 21885731
42 Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 2004;54:1469-1476. PMID: 15388697
43 Santacruz A, Collado MC, García-Valdés L, Segura MT, Martín-Lagos JA, Anjos T, et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br J Nutr 2010;104:83-92. PMID: 20205964
44 Karlsson CL, Onnerfält J, Xu J, Molin G, Ahrné S, Thorngren-Jerneck K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring) 2012;20:2257-2261. PMID: 22546742
45 Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med 2013;5:178ra41. PMID: 23536013
46 Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014;63:727-735. PMID: 23804561
47 Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 2013;110:9066-9071. PMID: 23671105
48 Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014;146:1513-1524. PMID: 24440671
49 Sarkola T, Eriksson CJ. Effect of 4-methylpyrazole on endogenous plasma ethanol and methanol levels in humans. Alcohol Clin Exp Res 2001;25:513-516. PMID: 11329490
50 Hartmann P, Chen WC, Schnabl B. The intestinal microbiome and the leaky gut as therapeutic targets in alcoholic liver disease. Front Physiol 2012;3:402. PMID: 23087650
51 Nair S, Cope K, Risby TH, Diehl AM. Obesity and female gender increase breath ethanol concentration: potential implications for the pathogenesis of nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:1200-1204. PMID: 11316170
52 Baker SS, Baker RD, Liu W, Nowak NJ, Zhu L. Role of alcohol metabolism in non-alcoholic steatohepatitis. PLoS One 2010;5:e9570. PMID: 20221393
53 Volynets V, Küper MA, Strahl S, Maier IB, Spruss A, Wagnerberger S, et al. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci 2012;57:1932-1941. PMID: 22427130
54 Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 2009;50:638-644. PMID: 19575462
55 Sajjad A, Mottershead M, Syn WK, Jones R, Smith S, Nwokolo CU. Ciprofloxacin suppresses bacterial overgrowth, increases fasting insulin but does not correct low acylated ghrelin concentration in non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2005;22:291-299. PMID: 16097995
56 Aderem A, Ulevitch RJ. Toll-like receptors in the induction of the innate immune response. Nature 2000;406:782-787. PMID: 10963608
57 Ruiz AG, Casafont F, Crespo J, Cayón A, Mayorga M, Estebanez A, et al. 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. PMID: 18000721
58 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. PMID: 18000721
59 Imajo K, Fujita K, Yoneda M, Nozaki Y, Ogawa Y, Shinohara Y, et al. Hyperresponsivity to low-dose endotoxin during progression to nonalcoholic steatohepatitis is regulated by leptin-mediated signaling. Cell Metab 2012;16:44-54. PMID: 22768838
60 Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56:1761-1772. PMID: 17456850
61 Csak T, Velayudham A, Hritz I, Petrasek J, Levin I, Lippai D, et al. Deficiency in myeloid differentiation factor-2 and toll-like receptor 4 expression attenuates nonalcoholic steatohepatitis and fibrosis in mice. Am J Physiol Gastrointest Liver Physiol 2011;300:G433-441. PMID: 21233280
62 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. PMID: 20400911
63 Harte AL, da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd EM, et al. Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflamm (Lond) 2010;7:15. PMID: 20353583
64 Zhang G, Ghosh S. Molecular mechanisms of NF-kappaB activation induced by bacterial lipopolysaccharide through Toll-like receptors. J Endotoxin Res 2000;6:453-457. PMID: 11521070
65 Miura K, Kodama Y, Inokuchi S, Schnabl B, Aoyama T, Ohnishi H, et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology 2010;139:323-334.e7. PMID: 20347818
66 Cawthorn WP, Sethi JK. TNF-alpha and adipocyte biology. FEBS Lett 2008;582:117-131. PMID: 18037376
67 Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012;482:179-185. PMID: 22297845
68 Vance DE. Role of phosphatidylcholine biosynthesis in the regulation of lipoprotein homeostasis. Curr Opin Lipidol 2008;19:229-234. PMID: 18460912
69 Zeisel SH, Wishnok JS, Blusztajn JK. Formation of methylamines from ingested choline and lecithin. J Pharmacol Exp Ther 1983;225:320-324. PMID: 6842395
70 Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A 2006;103:12511-12516. PMID: 16895997
71 Buchman AL, Dubin MD, Moukarzel AA, Jenden DJ, Roch M, Rice KM, et al. Choline deficiency: a cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology 1995;22:1399-1403. PMID: 7590654
72 Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 2013;62:1787-1794. PMID: 23197411
73 Wu WC, Zhao W, Li S. Small intestinal bacteria overgrowth decreases small intestinal motility in the NASH rats. World J Gastroenterol 2008;14:313-317. PMID: 18186574
74 Fu XS, Jiang F. Cisapride decreasing orocecal transit time in patients with nonalcoholic steatohepatitis. Hepatobiliary Pancreat Dis Int 2006;5:534-537. PMID: 17085338
75 Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009;49:1877-1887. PMID: 19291785
76 Sabaté JM, Jouët P, Harnois F, Mechler C, Msika S, Grossin M, et al. High prevalence of small intestinal bacterial overgrowth in patients with morbid obesity: a contributor to severe hepatic steatosis. Obes Surg 2008;18:371-377. PMID: 18286348
77 Shanab AA, Scully P, Crosbie O, Buckley M, O'Mahony L, Shanahan F, et al. Small intestinal bacterial overgrowth in nonalcoholic steatohepatitis: association with toll-like receptor 4 expression and plasma levels of interleukin 8. Dig Dis Sci 2011;56:1524-1534. PMID: 21046243
78 Soza A, Riquelme A, González R, Alvarez M, Pérez-Ayuso RM, Glasinovic JC, et al. Increased orocecal transit time in patients with nonalcoholic fatty liver disease. Dig Dis Sci 2005;50:1136-1140. PMID: 15986870
79 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. PMID: 11156641
80 Gibson GR, Cummings JH, Macfarlane GT, Allison C, Segal I, Vorster HH, et al. Alternative pathways for hydrogen disposal during fermentation in the human colon. Gut 1990;31:679-683. PMID: 2379871
81 Triantafyllou K, Chang C, Pimentel M. Methanogens, methane and gastrointestinal motility. J Neurogastroenterol Motil 2014;20:31-40. PMID: 24466443
82 Samuel BS, Gordon JI. A humanized gnotobiotic mouse model of host-archaeal-bacterial mutualism. Proc Natl Acad Sci U S A 2006;103:10011-10016. PMID: 16782812
83 Pimentel M, Mayer AG, Park S, Chow EJ, Hasan A, Kong Y. Methane production during lactulose breath test is associated with gastrointestinal disease presentation. Dig Dis Sci 2003;48:86-92. PMID: 12645795
84 Basseri RJ, Basseri B, Pimentel M, Chong K, Youdim A, Low K, et al. Intestinal methane production in obese individuals is associated with a higher body mass index. Gastroenterol Hepatol (N Y) 2012;8:22-28. PMID: 22347829
85 Mathur R, Amichai M, Chua KS, Mirocha J, Barlow GM, Pimentel M. Methane and hydrogen positivity on breath test is associated with greater body mass index and body fat. J Clin Endocrinol Metab 2013;98:E698-702. PMID: 23533244
86 Mathur R, Kim G, Morales W, Sung J, Rooks E, Pokkunuri V, et al. Intestinal Methanobrevibacter smithii but not total bacteria is related to diet-induced weight gain in rats. Obesity (Silver Spring) 2013;21:748-754. PMID: 23712978
87 Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 2014;11:506-514. PMID: 24912386
88 Ritze Y, Bárdos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A, et al. Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice. PLoS One 2014;9:e80169. PMID: 24475018
89 Okubo H, Sakoda H, Kushiyama A, Fujishiro M, Nakatsu Y, Fukushima T, et al. Lactobacillus casei strain Shirota protects against nonalcoholic steatohepatitis development in a rodent model. Am J Physiol Gastrointest Liver Physiol 2013;305:G911-918. PMID: 24113768
90 Endo H, Niioka M, Kobayashi N, Tanaka M, Watanabe T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One 2013;8:e63388. PMID: 23696823
91 Aller R, De Luis DA, Izaola O, Conde R, Gonzalez Sagrado M, Primo D, et al. 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. PMID: 22013734
92 Vajro P, Mandato C, Licenziati MR, Franzese A, Vitale DF, Lenta S, et al. Effects of Lactobacillus rhamnosus strain GG in pediatric obesity-related liver disease. J Pediatr Gastroenterol Nutr 2011;52:740-743. PMID: 21505361
93 Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, et al. Bifidobacterium longum with fructo-oligosaccharides in patients with non alcoholic steatohepatitis. Dig Dis Sci 2012;57:545-553. PMID: 21901256
94 Wong VW, Won GL, Chim AM, Chu WC, Yeung DK, Li KC, et al. Treatment of nonalcoholic steatohepatitis with probiotics. A proof-of-concept study. Ann Hepatol 2013;12:256-262. PMID: 23396737
95 Ma YY, Li L, Yu CH, Shen Z, Chen LH, Li YM. Effects of probiotics on nonalcoholic fatty liver disease: a meta-analysis. World J Gastroenterol 2013;19:6911-6918. PMID: 24187469
96 Vesper BJ, Jawdi A, Altman KW, Haines GK 3rd, Tao L, Radosevich JA. The effect of proton pump inhibitors on the human microbiota. Curr Drug Metab 2009;10:84-89. PMID: 19149516
97 Lin EA, Barlow GM, Mathur R. The microbiome in non-alcoholic fatty liver disease: associations and implications. Ann Gastroenterol 2014;27:181-183. PMID: 24733426
 
Received February 15, 2015
Accepted after revision June 10, 2015