Next Article in Journal
Comparison of the Effects of Fasting Glucose, Hemoglobin A1c, and Triglyceride–Glucose Index on Cardiovascular Events in Type 2 Diabetes Mellitus
Next Article in Special Issue
Fat and Sugar—A Dangerous Duet. A Comparative Review on Metabolic Remodeling in Rodent Models of Nonalcoholic Fatty Liver Disease
Previous Article in Journal
Effect of Lactobacillus plantarum TWK10 on Exercise Physiological Adaptation, Performance, and Body Composition in Healthy Humans
Previous Article in Special Issue
Atorvastatin and Vitamin E Accelerates NASH Resolution by Dietary Intervention in a Preclinical Guinea Pig Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Probiotics in Non-alcoholic Fatty Liver Disease: Does Gut Microbiota Matter?

Department of Medicine, Division of Gastroenterology and Hepatology, Thomas Jefferson University, Philadelphia, PA 19107, USA
*
Author to whom correspondence should be addressed.
Submission received: 11 October 2019 / Revised: 14 November 2019 / Accepted: 15 November 2019 / Published: 19 November 2019

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the hepatic consequence of metabolic syndrome, which often also includes obesity, diabetes, and dyslipidemia. The connection between gut microbiota (GM) and NAFLD has attracted significant attention in recent years. Data has shown that GM affects hepatic lipid metabolism and influences the balance between pro/anti-inflammatory effectors in the liver. Although studies reveal the association between GM dysbiosis and NAFLD, decoding the mechanisms of gut dysbiosis resulting in NAFLD remains challenging. The potential pathophysiology that links GM dysbiosis to NAFLD can be summarized as: (1) disrupting the balance between energy harvest and expenditure, (2) promoting hepatic inflammation (impairing intestinal integrity, facilitating endotoxemia, and initiating inflammatory cascades with cytokines releasing), and (3) altered biochemistry metabolism and GM-related metabolites (i.e., bile acid, short-chain fatty acids, aromatic amino acid derivatives, branched-chain amino acids, choline, ethanol). Due to the hypothesis that probiotics/synbiotics could normalize GM and reverse dysbiosis, there have been efforts to investigate the therapeutic effect of probiotics/synbiotics in patients with NAFLD. Recent randomized clinical trials suggest that probiotics/synbiotics could improve transaminases, hepatic steatosis, and reduce hepatic inflammation. Despite these promising results, future studies are necessary to understand the full role GM plays in NAFLD development and progression. Additionally, further data is needed to unravel probiotics/synbiotics efficacy, safety, and sustainability as a novel pharmacologic approaches to NAFLD.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is defined by an excessive accumulation of fat in the liver tissue, when other causes of secondary hepatic fat accumulation disorders, including significant alcohol consumption, hereditary disorders, and steatogenic medication, have been ruled out [1]. NAFLD is characterized by a large spectrum of liver disease, from isolated steatosis to steatohepatitis, and can progress to cause hepatic fibrosis formation and cirrhosis. Among the NAFLD spectrum, nonalcoholic steatohepatitis (NASH) is the more severe form and presents with hepatic steatosis, lobular inflammation, and cell injury (i.e., hepatocyte ballooning) on histology [1]. NAFLD has been recognized as one of the leading causes of chronic liver disease in the world, and its prevalence, along with a global epidemic of metabolic syndrome, obesity, and insulin resistance, has risen sharply across continents [2]. A recent systemic review conducted by Younossi et al. estimated that the prevalence of NAFLD has reached 25% globally [3]. Patients with NASH are at risk of progressing to cirrhosis of the liver and/or developing hepatocellular carcinoma (HCC). The 5-year cumulative incidence of HCC, in the context of NASH fibrosis/cirrhosis, varies from 4% to 12.3% [4,5,6,7], and from 2002 to 2017 the proportion of NASH-related HCC increased 8.5-fold, rising from 2.1% to 17.9% [8].
Despite the rising prevalence of NAFLD, no definitive pharmacological treatment has been approved by the Food and Drug Administration or the European Medicines Agency. Lifestyle changes include physical activity, weight loss, and diet modification are currently the main strategies to mitigate the NAFLD epidemic. Therefore, it remains essential, though challenging, to identify new therapeutic strategies for NAFLD.
It is widely accepted that the onset and progression of NAFLD are linked to inflammation, oxidative stress, insulin resistance, dyslipidemia, and obesity [9,10]. This process is driven by the interaction of environmental, diet, and genetic factors via biochemical and immunological alterations in lipid and glucose metabolism [11]. Despite decades of research on the pathogenesis of NAFLD, the pathophysiology remains unclear and most likely with multiple mechanisms. One potential mechanism is the direct connection via the portal vein between the intestines and liver (gut-gut microbiota-liver axis). It is through this process that microbiota colonized in the intestine can modulate metabolic processes that directly or indirectly impact metabolic syndrome and its associated co-morbidities [12].
The gut microbiota (GM) has been estimated to have more than 1000 bacterial species and 100 trillion bacteria with 150-fold more genes than the human genome that colonizes in the human intestinal tract [13,14]. The two major phyla in the human intestine are Firmicutes (Lactobacillus, Peptoniphilus, Ruminococcus, Clostridium, and Eubacteria) and Bacteroidetes (Bacteroides, Prevotella). Other less abundant phyla include Actinobacteria (Bifidobacterium), Proteobacteria, and Verrumicrobia [15]. However, the composition and abundance of GM varies due to considerable heterogeneity between individuals and underlying conditions such as age, gender, diet, pregnancy, hormonal changes, travel, infection, and medication such as antibiotics and proton pump inhibitors [16,17,18,19]. Dysbiosis is defined as an imbalance between healthy and disease-promoting microorganisms; itis manifested through changes of diversity and fluctuation in the relative abundance of certain microorganisms [20]. The balance and homeostasis of GM is critical for maintaining health and protecting against diseases in the host.
There are a growing number of studies revealing the association between GM dysbiosis and metabolic syndrome, obesity, type 2 diabetes, NAFLD [21,22,23,24]. Zhu et al. utilized 16S ribosomal RNA sequencing and concluded that NASH patients have a distinct composition and ratio of Firmicutes, Bacteroidetes, and Actinobacteria at the level of phylum, family, and genus compared with the healthy group. NASH patients also had an increased abundance of alcohol-producing bacteria which could increase serum alcohol levels and oxidative stress, resulting in liver injury [23]. Gut microbiome changes have also been found among pediatric NAFLD subjects. Del Chierico et al. compared NAFLD, NASH, and obese pediatric patients with healthy controls and found that NAFLD patients had an increased abundance of Anaerococcus (Actinobacteria), Ruminococcus (Firmicutes), Peptoniphilus (Firmicutes), Dorea (Firmicutes), Bradyrhizobium (Proteobacteria), and Propionibacterium acnes (Actinobacteria) but a reduced abundance of Rikenellaceae (Bacteroidetes) and Oscillospira (Firmicutes). There are no significant differences in terms of microbiome composition among the NAFLD, NASH, and obese groups [25]. Furthermore, Boursier et al. identified that GM can be used as predictors for NAFLD severity and progression. The abundance of Bacteroides was higher in both NASH and fibrosis patients, and Ruminococcus abundance was higher in fibrosis patients. Through multivariate analysis Boursier et al. concluded that Bacteroides is independently associated with NASH and Ruminococcus is linked to fibrosis [26].
Despite these studies revealing an association between GM dysbiosis and NAFLD, whether gut dysbiosis is a causative factor that results in NAFLD remains unclear. Thus, further clarification is necessary to investigate the causative relationship and potential pathogenesis links between NAFLD and dysbiosis.

2. Pathogenesis: The Links between NAFLD and Microbiome

The pathophysiology of NAFLD/NASH progression is complicated, and the “multiple hit” hypothesis formulates the idea that multiple insults work together to facilitate the progression of disease. Some of these insults include insulin resistance, genetic and epigenetic factors, nutritional supplement, and gut microbiota [27]. Among the numerous pathophysiology mechanisms that result in NAFLD/NASH, the GM has been considered to be one of the vital contributors and has received increasing attention in recent years [28]. The liver receives portal vein circulation and is exposed to nutritional supply as well as GM derived metabolites from the gut system. The gut-liver axis characterized by a functional, bidirectional interaction between the gastrointestinal tract and liver owing to the close anatomy. The GM and liver have very complicated interactive relationships and mediated by a complex metabolic and immunologic network [29]. The primary mechanism can be summarized as changing energy harvest mode, inflammatory cytokines and related signaling pathways, altered biochemistry metabolism and GM-related metabolites (bile acid, short-chain fatty acids, aromatic amino acid derivatives, branched-chain amino acids, and ethanol; see Figure 1).

2.1. Interplay with Intestinal Microbiota and the Host Immune System

Many factors such as obesity, diet, alcohol intake infection, and medication, can affect the microbiome and result in impaired intestinal integrity, intestinal bacterial overgrowth, bacterial translocation, and lipopolysaccharide (LPS) releasing. The subsequent endotoxemia enters the liver through the portal circulation which induces inflammatory cytokines releasing, resulting in liver injury that may contribute to NAFLD [30]. Miele et al. indicated that NAFLD patients have a reduced expression of ZO-1 (zona occludens 1), a major tight junction protein, which results in higher intestinal permeability [31]. Volynets et al. also confirmed that intestinal permeability and endotoxin levels in plasma were significantly higher in NAFLD patients [32]. Similar results were also found in the pediatric population when Giorgio et al. reported that the severity of intestinal permeability correlates with the severity of NAFLD [33]. NAFLD patients also presented with higher rates of small intestinal bacterial overgrowth (SIBO) which correlated with the severity of steatosis [31]. Impaired integrity of the gut barrier, bacterial overgrowth, and bacterial translocation could also allow bacterial endotoxin to pass to the liver. Lipopolysaccharides (LPS) stimulate Kupffer cells via activating toll-like receptors (TLRs). These TLRs are pattern recognition receptors that recognize pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and are inactive in healthy liver cells. Once the intestinal barrier is disrupted and endotoxemia circulates via the liver-gut through the gut-liver axis resulting in increased levels of PAMPs and DAMPs that bind with TLRs (TLR2, TLR4, TLR5, TLR9, etc.), the inflammatory cascade of releasing inflammatory cytokines (TNF-α, IL-8, IL1β) could be initiated, and lipid accumulation and cell death in hepatocytes could be stimulated leading to NAFLD, NASH, and cirrhosis [34,35,36,37,38]. Dysbiosis also plays a critical role in the weakening of mucosal immunity in a NAFLD host. Jiang et al. showed that, in the context of dysbiosis, CD4- and CD8-positive cell numbers are decreased in the duodenal mucosa lamina propria and levels of TNF-α, IL-6, and IFN-γ are increased in the NAFLD group [39].

2.2. Crosstalk between Intestinal Microbiota and Metabolite

2.2.1. Bile Acids

Bile acid homeostasis is regulated by GM. The microbiome is involved in the synthesis of primary bile acids, cholic acid and chenodeoxycholic acid, by regulating the expression of bile acid synthesis enzymes. GM also affects other bile acid metabolism processes, includes conjugation in the liver, reabsorption in the terminal ileum, deconjugation in the small intestine, conversion to secondary bile acids (lithocholic acid and deoxycholic acid) in the colon, and enterohepatic circulation, by affecting related enzymes or transporters expression or activity [20]. The disruption of any step in bile acid metabolism could have pathophysiologic impact. Altered bile acid metabolism results in metabolic and immune effects that contribute to NAFLD [20,40,41,42]. Bile acids can regulate metabolism and inflammation through the farnesoid X receptor (FXR) and transmembrane G protein-coupled receptor 5 (TGR5). FXR is predominantly activated by primary bile acids whereas TGR5 is predominantly activated by secondary bile acids [43,44].
In terms of metabolic effect, FXR inhibits de novo lipogenesis, increases fatty acid oxidation, regulates the gene expression involved in triglyceride homeostasis that reduces steatosis and downregulates gluconeogenesis. FXR also upregulates hepatic glycogen synthesis and regulates the expression of glucose transporter 4 (GLUT-4) and glucagon-like peptide 1 (GLP-1) which affects the insulin sensitivity that is closely linked to NAFLD [43]. TGR5 also affects glucose hemostasis via inducing GLP-1 secretion which increases energy expenditure and attenuates diet-induced obesity [45].
In terms of immunologic effect, bile acids help maintain intestinal barrier integrity to protect against the GM-related inflammatory cascades in the liver [43]. Bile acids can also decrease hepatic inflammation and fibrosis via FXR and TGR5 signaling pathways [46]. A few bile acid analogs have functioned as FXR or TGR5 agonists (INT-767, WAY-362450) showing that they can reduce pro-inflammatory gene expression and increase monocyte and IL-10 production to improve hepatic inflammation, steatosis, and fibrosis [47,48].

2.2.2. Short-Chain Fatty Acids (SCFAs)

SCFAs are primarily composed of acetate, propionate, and butyrate, which are microbial fermentation products that are found in the colon when a human lacks the enzymatic capacity to digest certain foods (mainly complex carbohydrates and small portions of amino acids). Most SCFAs are absorbed in the colon; however, 5–10% of SCFAs are not utilized and are excreted in the feces [20,49]. Like bile acids, SCFAs also have metabolic and immunologic effects related to NAFLD.
From a metabolic perspective, SCFAs reduce hepatic cholesterol and fatty acid synthesis while increasing hepatic lipid oxidation [50,51,52] SCFAs predominantly act on the G protein-coupled receptors GPR-41 and GPR-43, which are widely distributed in the gut enteroendocrine L cells, white adipose tissue, skeletal muscles, and the liver [20]. These L cells release glucagon-like peptides that directly act on hepatocytes by activating genes involved in fatty acid β-oxidation and insulin sensitivity which are both closely linked to NAFLD [53]. In contrast to long-chain fatty acids (LCFAs) that need carnitine shuttle to reach the mitochondrial interior, SCFAs could permeate the inner mitochondrial membrane in carnitine-independent mode. Besides, SCFAs exert weaker protonophoric effects in mitochondria and cause less disruption to the electron transport in the respiratory chain compared with LCFAs. These unique features make SCFAs important in lipogenesis, fatty acids oxidation, and ROS genesis [54].
From an immune perspective, SCFAs have an immune regulatory effect and play a complicated role in Treg cell differentiation via inhibiting the histone deacetylase and GPR-43 pathway. SCFAs have anti-inflammatory effects by reducing migration and proliferation of immune cells (T cells, neutrophil, macrophage, monocyte cells), reducing many types of pro-inflammatory cytokines (tumor necrosis factor-alpha, monocyte chemotactic protein-1, etc), and up-regulating anti-inflammatory cytokine prostaglandin E2 [55,56,57,58].

2.2.3. Aromatic Amino Acid Derivatives and Branched-Chain Amino Acids

A novel class of bacterial metabolites derived from aromatic amino acids (AAA) includes tryptophan, phenylalanine, and tyrosine have recently received attention and have been considered as factors in development of NAFLD [59,60].
Tryptophan-derived bacterial metabolites are composed of indole, indole-3-propionic acid, indole-3-acetic acid, indole-3-aldehyde, tryptamine, and 3-methylindole. Among them, indole is the main component [61]. These compounds maintain intestinal integrity, reduce bacterial translocation, prevent the release of microbiota-derived components, and limit inflammatory cascades [59,60]. Oral administration of indole can reduce the expression of key genes and proteins in the LPS-induced pro-inflammatory signaling pathway, and it has also been shown to prevent LPS-induced alterations of the cholesterol metabolism in mouse models [62]. Indole-3-acetate is also found to have similar protective hepatic effects that attenuate inflammatory responses and reduce lipid accumulation in hepatocytes via the aryl hydrocarbon receptor (AhR) pathway [63].
Phenylalanine-derived bacterial metabolites are composed of phenylacetic acid, phenylpropionic acid, and benzoic acid [64]. Hoyles et al. found that plasma phenylacetic acid (PAA) levels are positively associated with steatosis severity in a morbidly obese women cohort (n = 56) and mice treated with PAA for 2 weeks had significantly increased triglyceride accumulation. PAA was also associated with altered gene expression involved in lipid and glucose metabolism [65].
Branched-chain amino acids (BCAA) including valine, leucine, and isoleucine, are another class of metabolites that received attention in a recent hepatic steatosis study. There is crosstalk about the relationship between microbiome, host gene expression and BCAA hepatic metabolism [65]. The BCAA biosynthesis upregulated in the context of obesity and insulin resistance [66,67]. There is also a positive correlation between hepatic steatosis and plasma and urine BCAA levels. PAA can also significantly increase hepatic BCAA utilization which can synergetically facilitate hepatic lipid accumulation [65].

2.2.4. Choline

Choline metabolism is disrupted in the context of dysbiosis and deficiency in choline has been associated with NAFLD. Dysbiosis has the ability to metabolize choline to trimethylamine (TMA) which is further oxidized by hepatic flavin containing monooxygenases and converted to trimethylamine-N-oxide (TMAO) [68]. This process has two effects: (1) choline levels are reduced; (2) TMAO levels are increased.
Choline plays critical role in the process of very-low-density lipoprotein VLDL synthesis and facilitates hepatic lipid exportation. Thus choline deficiency results in triglyceride accumulation in hepatocytes [69]. A small human case study indicated that long-term total parenteral nutrition (TPN) without choline supplementation induced hepatic steatosis. After receiving parenteral nutrition solutions containing choline chloride for 6 weeks, the hepatic steatosis of subjects (4/4) resolved ultimately [70]. TMAO also increases insulin resistance, promotes inflammation and oxidative stress [71,72]. A clinical study revealed that the higher serum levels of TMAO correlated with greater severity of NAFLD [68].

2.2.5. Microbial Synthesis of Ethanol

NAFLD patients have been noted to have a higher level of serum ethanol, even with the absence of alcohol consumption. Nair et al. indicated the presence of ethanol in the breath of obese females with NASH without alcohol ingestion [73]. Zhu et al. indicated that NASH patients exhibited significantly elevated blood ethanol levels compared with their lean or obese non-NASH counterparts, along with an increased abundance of ethanol-producing bacteria [23]. Similarly, Volynets et al. showed that NAFLD patients have an increased formation of endogenous ethanol in the context of dysbiosis [74].
The accumulation of endogenous alcohol from an increased abundance of alcohol-producing bacteria subsequently results in significant amounts of free radicals and reactive oxygen that cause mitochondrial dysfunction, hepatic cellular inflammation and injury [23,75,76]. Other mechanisms that ethanol plays a role in and that contribute to NAFLD development include, downregulating tight junction expression, up-regulating de novo lipogenesis, decreasing fatty acid oxidation, and reducing very low density lipoprotein (VLDL) exportation from the liver [20].

2.3. Energy Extraction and Consumption Balance Disrupted by GM

GM is thought to alter the energy influx and expenditure which may contribute to NAFLD development. Turnbaugh et al. conducted a study with a cohort of 154 obese and lean twins and found that in obese humans, the GM-enriched phosphotransferase system is involved in the microbial processing of carbohydrates, which yields an enhanced capacity to extract calories from the diet [77]. Additionally, the microbiome also altered the host’s energy expenditure via metabolic products such as bile acids and SCFAs. As mentioned earlier, dysbiosis can alter the level of bile acids and SCFAs and ultimately affect energy extraction and expenditure. It has been found that bile acids can increase whole-body energy expenditure and energy consumption via the activation of brown adipose tissue [78]. SCFAs themselves can provide an energy source for different cell types and tissues. Butyrate can be utilized by colonocytes as an energy substrate, whereas acetate and propionate can be utilized as substrates in the process of glucose and fatty acid synthesis, respectively [79]. Canfora et al. infused SCFA mixtures (either acetate, butyrate, or propionate) into the colon of overweight/obese men. The SCFAs mixture increased fat oxidation, energy expenditure, and attenuated fasting free glycerol concentrations compared with the placebo group [80].

3. Clinical Application of Probiotics and Synbiotics in NAFLD/NASH Patients

Given the correlation between dysbiosis and liver damage, the growing burden of NAFLD, and lacking effective pharmacologic interventions, restructuring GM to reverse dysbiosis seems like a potential therapeutic strategy. Probiotics are non-pathogenic, live microorganisms that provide a health benefit to the host by modifying GM when delivered in sufficient quantities [81]. The most frequently mentioned probiotics in recent clinical trials are Lactobacilli, Streptococci, and Bifidobacteria. Prebiotics are non-digestible carbohydrates that can be fermented by bacteria and subsequently change GM composition and activity to promote health benefits [82]. Synbiotics refer to the combination of prebiotics and probiotics.
Probiotics and synbiotics can normalize GM and reverse dysbiosis, which could potentially benefit NAFLD patients [83]. Multiple experimental trials have shown probiotics to have significant therapeutic effects in fatty liver mice models. The trials showed that administrating probiotics to high-fat-diet fed mice could prevent the onset of hepatic steatosis and improve hepatic steatosis and fibrosis. The protective effects have multiple mechanisms including reducing hepatic lipid deposition, decreasing endotoxemia, reducing oxidative stress, anti-inflammatory effects through the modulation of nuclear factor kappa B (NF-κB) and tumor necrosis factor (TNF), and antifibrotic effects through the modification of transforming growth factor-beta (TGF-β) and collagen expression [84,85,86,87,88]. These preclinical studies suggest that probiotics therapy could be a potential pharmacologic intervention for NAFLD patients, though the animal model have their limitations. For example, mice are germ-free animals and the intestine bacterial composite and abundance are different from that of a human.
Most human clinical trials conducted to study the therapeutic effect of probiotics/synbiotics in NAFLD patients have been conducted over the past 10 years and are small scale with mixed clinical outcomes. However, the overall outcome indicates that probiotics/synbiotics could be a promising therapeutic strategy for the NAFLD population. Table 1 summarizes 26 randomized controlled trials (RCT) that compared probiotics and/or synbiotics versus placebo in NAFLD patients from January 2010to June 2019 in [69,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113].

3.1. Biochemistry Evaluation

Most of the clinical trials in Table 1 include serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) level changes to assess the effects of probiotics or synbiotics on liver function. A double-blind RCT enrolled 30 patients with NAFLD, thehe intervention group received 3 months of a combination of Lactobacillus bulgaricus and Streptococcus thermophilus (500 million per day) and had significant AST and ALT improvement [89]. Similarly, in a 72 NAFLD patients double-blinded RCT, Nabvi et al. utilized probiotic yogurt (300 g/day) containing Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12 as an intervention for 8 weeks. They concluded that probiotics could significantly improve AST and ALT in NAFLD patients when compared with the placebo group [96]. Shavakhi et al. [92] used probiotics capsules (Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, and Streptococcus thermophilus) plus metformin 1000mg per day as an intervention.
When compared with the control group who only take metformin 1000mg per day, it was found that the probiotics with metformin synergically improved liver aminotransferases in NASH patients. A few other RCTs that utilized different species of probiotics also revealed that probiotics could improve either AST or ALT, or both transaminases in NAFLD patients [69,91,93,97,101,105,110,112]. Four recent synbiotics RCTs utilized a combination of probiotics (Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, and Streptococcus thermophilus) plus prebiotics (either fructooligosaccharide or inulin) and concluded that synbiotics supplements can also benefit liver function [95,103,106,111].
Similar to the adult population, pediatric clinical trials also revealed that probiotics and synbiotics could improve AST and ALT in NAFLD patients. Vajro et al. enrolled 20 pediatric obese NAFLD patients and offered the intervention group a Lactobacillus rhamnosus strain GG (12 billion CFU/day). The data indicated a significant decrease in ALT after an 8 weeks trial [90]. Similarly, in a triple blinded RCT, Famouri et al. enrolled 64 obese children with sonographic NAFLD and the interventional group received probiotics capsules (Lactobacillus acidophilus, Bifidobacterium lactis, Bifidobacterium bifidum, and Lactobacillus rhamnosus). The primary outcome indicated that the probiotics group had a significantly lower mean value of AST and ALT, as well as lower levels of cholesterol, triglycerides, and LDL-C after a 12 week intervention [104]. Miccheli et al. enrolled 31 pediatric NAFLD patients with a VSL#3 (Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus casei, and Lactobacillus bulgaricus) intervention for 4 months. They concluded that, compared with the placebo group, AST but not ALT significantly improved [97].
However, not all of the completed trials support the notion that probiotics or synbiotics can improve liver enzyme levels. For example, Ahn et al. published a trial in 2019 that enrolled 68 adult obese NAFLD patients. The interventional group was treated with a multispecies probiotics mixture (Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus paracasei, Pediococcus pentosaceus, Bifidobacterium lactis, and Bifidobacterium breve) for 12 weeks. Results revealed that probiotics improved the intrahepatic fat (IHF) fraction measured by MRI-PDFF but there was no significant improvement of ALT or AST when compared with the placebo group [113]. Alisi et al. conducted a RCT in 2014, enrolling 22 NAFLD children. The interventional group was treated with probiotics capsules (Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus casei, and Lactobacillus bulgaricus) for 4 months. However, the trial failed to reveal any significant liver enzyme improvement when compared with the placebo group [94]. Some synbiotics trials have also failed to show liver enzyme improvement. For instance, Ferolla et al. conducted an RCT of 50 NASH patients and indicated that synbiotics (Lactobacillus reuteri + inulin) neither improved liver function nor steatosis [99].
These mixed results also occurred with other serum markers including Gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), fasting serum glucose (FBS) and inflammatory markers [69,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113]. Given the pattern of mixed results, Table 2 summarizes the most recently published meta-analyses of RCTs with probiotic/synbiotic treatment for NAFLD/NASH populations [114,115,116,117,118].
All of the meta-analyses listed indicated that microbial therapies are associated with a significant improvement in ALT and AST [114,115,116,117,118]. Regarding the other serum markers, Liu et al. included 15 probiotics and synbiotics RCTs involving 782 patients with NAFLD up to April 2018. They found that probiotic and synbiotic supplementation is associated with a significant reduction in TG, TC, HDL-C, LDL-C, and TNF-α levels [117]. Koutnikova et al. included 111 RCTs representing 6826 subjects with metabolic syndrome, type II DM, and NAFLD. Subgroup analysis of NAFLD patients did not indicate that probiotics significantly reduced GGT [116]. Khan et al. included 12 probiotics and synbiotics RCTs up to June 2018. They concluded that a reduction in CRP occurred in synbiotics trials. However, TNF-α, LDL-C, TG, and TC only significantly improved in the synbiotics but not probiotics group, and HDL-C and FBS did not significantly change in either group [115]. Of note, serologic markers have high variability and not directly reflect the disease progression in context of NAFLD. Thus, there are emerging studies utilized the imaging modalities to evaluate NAFLD clinical outcome outline below.

3.2. Imaging Modalities to Assess Hepatic Steatosis and Stiffness

Several trials included imaging evaluation as part of clinical outcomes to assess the effect of probiotics and synbiotics on hepatic steatosis, fibrosis, and liver stiffness. Ultrasonography (US) is the most commonly used imaging modality for liver morphology study. The research teams of Shavakhi [92], Miccheli [97], Asgharian [98], Famouri [104], and Bakhshimoghaddam et al. [107] reported that US-measured steatosis significantly improved in adult patients who took probiotics and synbiotics supplements. Alisi et al. revealed a similar efficacy of probiotics improving US-measured hepatic steatosis in pediatric NAFLD patients [94]. Manzhalii et al. conducted an RCT with 75 enrolled NASH patients on low-fat/low-calorie diets. Probiotics (Lactobacilli, Bifidobacteria, and Streptococcus thermophilus) were administered in the intervention group for 12 weeks. US-measured liver stiffness improved in the probiotics group compared with the control group [105]. However, in 2018, Wang et al. got different results in a double-blind RCT with 200 NAFLD patients. The experimental group received Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus subtilis for a 1-month duration and there was no significant difference in the severity of steatosis measured by US [112]. The likely explanation was that the intervention duration was relatively shorter. Also, as a measurement tool, US has technical limitations when it comes to accurately evaluating the severity of hepatic steatosis.
Over recent years, elastography-based imaging techniques have received increased attention in the field of non-invasive assessment for organ tissue, especially the liver. These techniques utilize soft tissue elasticity changes in various pathologies to yield qualitative and quantitative analyses and are superior to US in assessing the severity of liver steatosis, fibrosis, and cirrhosis [119]. Eslamparast et al. conducted a double-blind RCT that enrolled 52 NAFLD patients with a synbiotics (probiotics cocktail - Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus acidophilus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, Streptococcus thermophilus, and the prebiotic fructooligosaccharide) intervention for 28 weeks and the elastography-measured fibrosis score improved [95]. Mofidi et al. applied the same intervention as the Eslamparasts team, enrolling 50 NAFLD patients who took part in a transient elastography evaluation. Results indicated that synbiotics are associated with hepatic steatosis and fibrosis improvement which is consistent with the results of Eslamparasts team [106]. Kobyliak conducted two double-blind RCTs in 2018. One enrolled 58 NAFLD with type 2 diabetes patients. The intervention team received probiotics called “Symbiter” which is composed of Bifidobacterium, Lactobacillus, Lactococcus, Propionibacterium, and Acetobacter for 8 weeks and utilized shear wave elastography (SWE) to measure hepatic fibrosis. The trial concluded that probiotics can improve fatty liver index but that there is no significant change in liver stiffness graded by SWE [110]. His team conducted another double-blind RCT that enrolled 48 NAFLD with type 2 diabetes patients. The intervention team received a combination of the same probiotics mentioned above plus Omega-3 fatty acids called “Symbiter Omega”. After an 8-week intervention, results revealed similar significantly reduce liver steatosis but not liver stiffness [109].
Magnetic resonance imaging–derived proton density fat fraction (MRI-PDFF) is another novel modality that, over recent years, has been used to evaluate hepatic steatosis. The feature of accurately measuring the distribution of intrahepatic fat fraction (IHF) across the liver in seconds has led to its rising popularity in NAFLD research [120]. A few trials utilized this technique to analyze the efficacy of probiotics/synbiotics in steatosis, for example, Ahn et al. concluded that a probiotics therapy group had a reduced mean value IHF compared with the control group, however, there was no significant change in liver stiffness between the probiotics and control groups [113]. Ferolla et al. reported that MRI-PDFF-measured steatosis was reduced in the synbiotics group but there was no significant change in liver fibrosis [99]. There is only one published RCT that utilized proton-magnetic resonance spectroscopy to measure intrahepatic triglyceride content (IHTG). Wong et al. conducted a trial with 20 NASH patients. The intervention group was treated with Lactobacillus plantarum, Lactobacillus deslbrueckii, Lactobacillus acidophilus, Lactobacillus rhamnosus, and Bifidobacterium bifidum for 6 months. The primary outcome of the RCT is that IHTG decreased from the baseline of 22.6% to 14.9% in the probiotics group whereas IHTG remained static in the usual care group [93].
Two large-scale meta-analyses conducted by Sharpton et al. [118] and Liu et al. [117] indicated that probiotics/synbiotics improved liver stiffness that was measured by elastography, and probiotics/synbiotics are associated with hepatic steatosis improvement as graded by the US though analyses showed heterogeneity. Another meta-analysis conducted by Khan et al. indicated that probiotics/synbiotics are associated with a significant improvement in liver fibrosis score graded by fibroscan [115].

3.3. Histologic Evaluation

The fact that most probiotics/synbiotics intervention trials lack a liver biopsy to further assess histological changes in liver steatosis, fibrosis, cirrhosis, and inflammation is a noticeable limitation. Nevertheless, there are still two RCTs that utilized liver biopsy at the end of clinical trials to evaluate histological changes after probiotics and synbiotics interventions. Malaguarnera et al. conducted an RCT that enrolled 66 NASH patients. The study used synbiotics (Bifidobacterium longum+fructooligosaccharides) as an intervention for 24 weeks and the liver biopsy indicated hepatic steatosis improvement and reduced NASH activity in the synbiotics group when compared with the control group [91]. Duseja et al. conducted a double-blind RCT with 30 enrolled NAFLD patients. The probiotics administered were composed of Lactobacillus paracasei, Lactobacillus plantarum, Lactobacillus acidophilus, Lactobacillus delbrueckii subsp. bulgaricus, Bifidobacterium longum, Bifidobacterium infantis, Bifidobacterium breve, and Streptococcus thermophilus. After 1 year of treatment, a liver biopsy indicated significantly reduced hepatocyte ballooning, NAS score, and fibrosis compared with the control group. However, there was no significant hepatic steatosis or lobular inflammation difference between the two groups [69].

3.4. Safety and Tolerability of Probiotics in NAFLD Patients

Probiotics/synbiotics have a relatively safe profile, and historically, probiotics have been widely used in dairy products without significant safety concerns. The Agency for Healthcare Research and Quality (AHRQ) released a report in 2011 that extensively reviewed literature regarding the probiotic safety and adverse event reports. It included six probiotic genera (Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, Bacillus, and Saccharomyces) and 622 studies and concluded that the existing probiotic RCTs reveal no evidence of increased risk. However, due to the lack of assessment and systematic reporting of adverse events in probiotic intervention studies, AHRQ was not equipped to clarify the safety of probiotic interventions with confidence [121]. Though clinical trials rarely reported adverse effects or issues of safety with patients with NAFLD, lacking long-term, post-intervention follow-up in clinical trials is a setback regarding safety assessment. There are a few case reports of probiotics having adverse effects such as systemic infections (bacteremia and endocarditis), bowel ischemia, inflammatory reaction, D-lactic acidosis, and gastrointestinal side effects [122]. Although microbial therapy seems to be safe, it is necessary to evaluate probiotic- or synbiotic-associated adverse effects and investigate safety profiles for long-term use.

4. Concluding Remarks

With the rapidly growing incidence and prevalence of NAFLD and lack of effective pharmacological intervention, there is an urgent need for developing novel medications for treatment. The link between GM and NAFLD has received increased attention and a growing number of studies have revealed that the host-gut microbiome interaction and pathophysiology of dysbiosis may promote the development of NAFLD. Bacterial derivative metabolites, immune-related effects, and energy extraction/consumption balance disruption might promote NAFLD progression. Therefore, restoring dysbiosis could be a potential therapeutic strategy for NAFLD, an idea backed by multiple recent probiotics and synbiotics clinical trials in the specific context of NAFLD and NASH. The literature to date is encouraging and supports the notion that probiotics/synbiotics may improve transaminase levels, hepatic steatosis, and NAFLD activity score. To some extent, probiotics/synbiotics can also reduce proinflammatory cytokines such as TNF-α and the interleukin family (IL-1, IL-6, IL-8).
Overall, probiotics/synbiotics have a safe profile and are well-tolerated though more safety evaluations need to be completed in the future. Additionally, the sustainability of the probiotic/synbiotic protective effect on NAFLD over the long-term is still not clear. In this growing NAFLD epidemic era, further studies are necessary to more clearly elucidate the efficacy, safety, and sustainability of probiotics/synbiotics in the management of NAFLD.

Author Contributions

C.X. and D.H.-D. made substantial contributions to the conception or design of the work.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest. Halegoua-DeMarzio is a consultant for Gilead and Intercept Pharmaceuticals.

References

  1. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Diehl, A.M.; Brunt, E.M.; Cusi, K.; Charlton, M.; Sanyal, A.J. 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. [Google Scholar] [CrossRef] [PubMed]
  2. Farrell, G.C.; Wong, V.W.; Chitturi, S. NAFLD in Asia-as common and important as in the West. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 307–318. [Google Scholar] [CrossRef] [PubMed]
  3. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease. Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef]
  4. Hashimoto, E.; Yatsuji, S.; Tobari, M.; Taniai, M.; Torii, N.; Tokushige, K.; Shiratori, K. Hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. J. Gastroenterol. 2009, 44 (Suppl. 19), 89–95. [Google Scholar] [CrossRef]
  5. Kodama, K.; Tokushige, K.; Hashimoto, E.; Taniai, M.; Shiratori, K. Hepatic and extrahepatic malignancies in cirrhosis caused by nonalcoholic steatohepatitis and alcoholic liver disease. Alcohol. Clin. Exp. Res. 2013, 37 (Suppl. 1), E247–E252. [Google Scholar] [CrossRef]
  6. Paranagua-Vezozzo, D.C.; Ono, S.K.; Alvarado-Mora, M.V.; Farias, A.Q.; Cunha-Silva, M.; Franca, J.I.; Alves, V.A.; Sherman, M.; Carrilho, F.J. Epidemiology of HCC in Brazil: Incidence and risk factors in a ten-year cohort. Ann. Hepatol. 2014, 13, 386–393. [Google Scholar] [CrossRef]
  7. Yatsuji, S.; Hashimoto, E.; Tobari, M.; Taniai, M.; Tokushige, K.; Shiratori, K. Clinical features and outcomes of cirrhosis due to non-alcoholic steatohepatitis compared with cirrhosis caused by chronic hepatitis C. J. Gastroenterol. Hepatol. 2009, 24, 248–254. [Google Scholar] [CrossRef]
  8. Younossi, Z.; Stepanova, M.; Ong, J.P.; Jacobson, I.M.; Bugianesi, E.; Duseja, A.; Eguchi, Y.; Wong, V.W.; Negro, F.; Yilmaz, Y.; et al. Nonalcoholic Steatohepatitis Is the Fastest Growing Cause of Hepatocellular Carcinoma in Liver Transplant Candidates. Clin. Gastroenterol. Hepatol. 2019, 17, 748–755.e743. [Google Scholar] [CrossRef]
  9. Sangouni, A.A.; Ghavamzadeh, S. A review of synbiotic efficacy in non-alcoholic fatty liver disease as a therapeutic approach. Diabetes Metab. Syndr. 2019, 13, 2917–2922. [Google Scholar] [CrossRef]
  10. Marra, F.; Svegliati-Baroni, G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J. Hepatol. 2018, 68, 280–295. [Google Scholar] [CrossRef]
  11. Hardy, T.; Oakley, F.; Anstee, Q.M.; Day, C.P. Nonalcoholic Fatty Liver Disease: Pathogenesis and Disease Spectrum. Ann. Rev. Pathol. 2016, 11, 451–496. [Google Scholar] [CrossRef]
  12. Moran-Ramos, S.; Lopez-Contreras, B.E.; Canizales-Quinteros, S. Gut Microbiota in Obesity and Metabolic Abnormalities: A Matter of Composition or Functionality? Arch. Med. Res. 2017, 48, 735–753. [Google Scholar] [CrossRef]
  13. Chassaing, B.; Etienne-Mesmin, L.; Gewirtz, A.T. Microbiota-liver axis in hepatic disease. Hepatology 2014, 59, 328–339. [Google Scholar] [CrossRef]
  14. Kamada, N.; Nunez, G. Regulation of the immune system by the resident intestinal bacteria. Gastroenterology 2014, 146, 1477–1488. [Google Scholar] [CrossRef]
  15. Tap, J.; Mondot, S.; Levenez, F.; Pelletier, E.; Caron, C.; Furet, J.P.; Ugarte, E.; Munoz-Tamayo, R.; Paslier, D.L.; Nalin, R.; et al. Towards the human intestinal microbiota phylogenetic core. Env. Microbiol. 2009, 11, 2574–2584. [Google Scholar] [CrossRef]
  16. Naito, Y.; Kashiwagi, K.; Takagi, T.; Andoh, A.; Inoue, R. Intestinal Dysbiosis Secondary to Proton-Pump Inhibitor Use. Digestion 2018, 97, 195–204. [Google Scholar] [CrossRef]
  17. Rodriguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef]
  18. Uchiyama, K.; Naito, Y.; Takagi, T. Intestinal microbiome as a novel therapeutic target for local and systemic inflammation. Pharmcol. Ther. 2019, 199, 164–172. [Google Scholar] [CrossRef]
  19. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef]
  20. Mouzaki, M.; Loomba, R. Insights into the evolving role of the gut microbiome in nonalcoholic fatty liver disease: Rationale and prospects for therapeutic intervention. Ther. Adv. Gastroenterol. 2019, 12. [Google Scholar] [CrossRef]
  21. Mouzaki, M.; Comelli, E.M.; Arendt, B.M.; Bonengel, J.; Fung, S.K.; Fischer, S.E.; McGilvray, I.D.; Allard, J.P. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013, 58, 120–127. [Google Scholar] [CrossRef]
  22. Wigg, A.J.; Roberts-Thomson, I.C.; Dymock, R.B.; McCarthy, P.J.; Grose, R.H.; Cummins, A.G. 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. [Google Scholar] [CrossRef]
  23. Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef]
  24. Knight, R.; Callewaert, C.; Marotz, C.; Hyde, E.R.; Debelius, J.W.; McDonald, D.; Sogin, M.L. The Microbiome and Human Biology. Ann. Rev. Genom. Hum. Genet. 2017, 18, 65–86. [Google Scholar] [CrossRef]
  25. Del Chierico, F.; Nobili, V.; Vernocchi, P.; Russo, A.; De Stefanis, C.; Gnani, D.; Furlanello, C.; Zandona, 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]
  26. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef]
  27. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  28. Gupta, H.; Youn, G.S.; Shin, M.J.; Suk, K.T. Role of Gut Microbiota in Hepatocarcinogenesis. Microorganisms 2019, 7, 121. [Google Scholar] [CrossRef]
  29. Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome—A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [Google Scholar] [CrossRef]
  30. Bajaj, J.S. The role of microbiota in hepatic encephalopathy. Gut Microbes 2014, 5, 397–403. [Google Scholar] [CrossRef]
  31. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Masciana, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  32. Lavekar, A.S.; Raje, D.V.; Manohar, T.; Lavekar, A.A. Role of Probiotics in the Treatment of Nonalcoholic Fatty Liver Disease: A Meta-analysis. Euroasian J. Hepatogastroenterol. 2017, 7, 130–137. [Google Scholar] [CrossRef] [PubMed]
  33. Giorgio, V.; Miele, L.; Principessa, L.; Ferretti, F.; Villa, M.P.; Negro, V.; Grieco, A.; Alisi, A.; Nobili, V. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig. Liver Dis. 2014, 46, 556–560. [Google Scholar] [CrossRef] [PubMed]
  34. Miura, K.; Kodama, Y.; Inokuchi, S.; Schnabl, B.; Aoyama, T.; Ohnishi, H.; Olefsky, J.M.; Brenner, D.A.; Seki, E. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology 2010, 139, 323–334.e327. [Google Scholar] [CrossRef] [Green Version]
  35. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
  36. 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]
  37. Vijay-Kumar, M.; Aitken, J.D.; Carvalho, F.A.; Cullender, T.C.; Mwangi, S.; Srinivasan, S.; Sitaraman, S.V.; Knight, R.; Ley, R.E.; Gewirtz, A.T. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010, 328, 228–231. [Google Scholar] [CrossRef] [Green Version]
  38. 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, 140S–145S. [Google Scholar] [CrossRef]
  39. Jiang, W.; Wu, N.; Wang, X.; Chi, Y.; Zhang, Y.; Qiu, X.; Hu, Y.; Li, J.; Liu, Y. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci. Rep. 2015, 5, 8096. [Google Scholar] [CrossRef]
  40. Sarathy, J.; Detloff, S.J.; Ao, M.; Khan, N.; French, S.; Sirajuddin, H.; Nair, T.; Rao, M.C. The Yin and Yang of bile acid action on tight junctions in a model colonic epithelium. Physiol. Rep. 2017, 5, e13294. [Google Scholar] [CrossRef]
  41. Wegner, K.; Just, S.; Gau, L.; Mueller, H.; Gerard, P.; Lepage, P.; Clavel, T.; Rohn, S. Rapid analysis of bile acids in different biological matrices using LC-ESI-MS/MS for the investigation of bile acid transformation by mammalian gut bacteria. Anal. Bioanal. Chem. 2017, 409, 1231–1245. [Google Scholar] [CrossRef]
  42. Wahlstrom, A.; Sayin, S.I.; Marschall, H.U.; Backhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chavez-Talavera, O.; Tailleux, A.; Lefebvre, P.; Staels, B. Bile Acid Control of Metabolism and Inflammation in Obesity, Type 2 Diabetes, Dyslipidemia, and Nonalcoholic Fatty Liver Disease. Gastroenterology 2017, 152, 1679–1694.e1673. [Google Scholar] [CrossRef] [PubMed]
  44. Arab, J.P.; Karpen, S.J.; Dawson, P.A.; Arrese, M.; Trauner, M. Bile acids and nonalcoholic fatty liver disease: Molecular insights and therapeutic perspectives. Hepatology 2017, 65, 350–362. [Google Scholar] [CrossRef] [PubMed]
  45. Thomas, C.; Gioiello, A.; Noriega, L.; Strehle, A.; Oury, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.; Pruzanski, M.; et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009, 10, 167–177. [Google Scholar] [CrossRef] [Green Version]
  46. Liu, H.; Pathak, P.; Boehme, S.; Chiang, J.Y. Cholesterol 7alpha-hydroxylase protects the liver from inflammation and fibrosis by maintaining cholesterol homeostasis. J. Lipid Res. 2016, 57, 1831–1844. [Google Scholar] [CrossRef] [Green Version]
  47. Zhang, S.; Wang, J.; Liu, Q.; Harnish, D.C. Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in murine model of non-alcoholic steatohepatitis. J. Hepatol. 2009, 51, 380–388. [Google Scholar] [CrossRef]
  48. 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] [Green Version]
  49. 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]
  50. Wright, R.S.; Anderson, J.W.; Bridges, S.R. Propionate inhibits hepatocyte lipid synthesis. Proc. Soc. Exp. Biol. Med. 1990, 195, 26–29. [Google Scholar] [CrossRef]
  51. Demigne, C.; Morand, C.; Levrat, M.A.; Besson, C.; Moundras, C.; Remesy, C. Effect of propionate on fatty acid and cholesterol synthesis and on acetate metabolism in isolated rat hepatocytes. Br. J. Nutr. 1995, 74, 209–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Svegliati-Baroni, G.; Saccomanno, S.; Rychlicki, C.; Agostinelli, L.; De Minicis, S.; Candelaresi, C.; Faraci, G.; Pacetti, D.; Vivarelli, M.; Nicolini, D.; et al. Glucagon-like peptide-1 receptor activation stimulates hepatic lipid oxidation and restores hepatic signalling alteration induced by a high-fat diet in nonalcoholic steatohepatitis. Liver Int. 2011, 31, 1285–1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Schonfeld, P.; Wojtczak, L. Short- and medium-chain fatty acids in energy metabolism: The cellular perspective. J. Lipid Res. 2016, 57, 943–954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ohira, H.; Tsutsui, W.; Fujioka, Y. Are Short Chain Fatty Acids in Gut Microbiota Defensive Players for Inflammation and Atherosclerosis? J. Atheroscler. Thromb. 2017, 24, 660–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  58. Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef]
  59. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [Green Version]
  60. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef] [Green Version]
  61. Jin, U.H.; Lee, S.O.; Sridharan, G.; Lee, K.; Davidson, L.A.; Jayaraman, A.; Chapkin, R.S.; Alaniz, R.; Safe, S. Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Mol. Pharm. 2014, 85, 777–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Beaumont, M.; Neyrinck, A.M.; Olivares, M.; Rodriguez, J.; de Rocca Serra, A.; Roumain, M.; Bindels, L.B.; Cani, P.D.; Evenepoel, P.; Muccioli, G.G.; et al. The gut microbiota metabolite indole alleviates liver inflammation in mice. FASEB J. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Krishnan, S.; Ding, Y.; Saedi, N.; Choi, M.; Sridharan, G.V.; Sherr, D.H.; Yarmush, M.L.; Alaniz, R.C.; Jayaraman, A.; Lee, K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018, 23, 1099–1111. [Google Scholar] [CrossRef]
  64. Russell, W.R.; Duncan, S.H.; Scobbie, L.; Duncan, G.; Cantlay, L.; Calder, A.G.; Anderson, S.E.; Flint, H.J. Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Mol. Nutr. Food Res. 2013, 57, 523–535. [Google Scholar] [CrossRef]
  65. Hoyles, L.; Fernandez-Real, J.M.; Federici, M.; Serino, M.; Abbott, J.; Charpentier, J.; Heymes, C.; Luque, J.L.; Anthony, E.; Barton, R.H.; et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 2018, 24, 1070–1080. [Google Scholar] [CrossRef] [PubMed]
  66. Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef] [PubMed]
  67. Shoaie, S.; Ghaffari, P.; Kovatcheva-Datchary, P.; Mardinoglu, A.; Sen, P.; Pujos-Guillot, E.; de Wouters, T.; Juste, C.; Rizkalla, S.; Chilloux, J.; et al. Quantifying Diet-Induced Metabolic Changes of the Human Gut Microbiome. Cell Metab. 2015, 22, 320–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Chen, Y.M.; Liu, Y.; Zhou, R.F.; Chen, X.L.; Wang, C.; Tan, X.Y.; Wang, L.J.; Zheng, R.D.; Zhang, H.W.; Ling, W.H.; et al. Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci. Rep. 2016, 6, 19076. [Google Scholar] [CrossRef]
  69. Duseja, A.; Acharya, S.K.; Mehta, M.; Chhabra, S.; Shalimar; Rana, S.; Das, A.; Dattagupta, S.; Dhiman, R.K.; Chawla, Y.K. High potency multistrain probiotic improves liver histology in non-alcoholic fatty liver disease (NAFLD): A randomised, double-blind, proof of concept study. BMJ Open Gastroenterol. 2019, 6, e000315. [Google Scholar] [CrossRef] [Green Version]
  70. Buchman, A.L.; Dubin, M.D.; Moukarzel, A.A.; Jenden, D.J.; Roch, M.; Rice, K.M.; Gornbein, J.; Ament, M.E. Choline deficiency: A cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology 1995, 22, 1399–1403. [Google Scholar]
  71. Gao, X.; Liu, X.; Xu, J.; Xue, C.; Xue, Y.; Wang, Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J. Biosci. Bioeng. 2014, 118, 476–481. [Google Scholar] [CrossRef] [PubMed]
  72. Dumas, M.E.; Barton, R.H.; Toye, A.; Cloarec, O.; Blancher, C.; Rothwell, A.; Fearnside, J.; Tatoud, R.; Blanc, V.; Lindon, J.C.; et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc. Nat. Acad. Sci. USA 2006, 103, 12511–12516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nair, S.; Cope, K.; Risby, T.H.; Diehl, A.M. Obesity and female gender increase breath ethanol concentration: Potential implications for the pathogenesis of nonalcoholic steatohepatitis. Am. J. Gastroenterol. 2001, 96, 1200–1204. [Google Scholar] [CrossRef]
  74. Volynets, V.; Kuper, M.A.; Strahl, S.; Maier, I.B.; Spruss, A.; Wagnerberger, S.; Konigsrainer, A.; Bischoff, S.C.; Bergheim, I. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig. Dis. Sci. 2012, 57, 1932–1941. [Google Scholar] [CrossRef] [PubMed]
  75. Begriche, K.; Igoudjil, A.; Pessayre, D.; Fromenty, B. Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it. Mitochondrion 2006, 6, 1–28. [Google Scholar] [CrossRef]
  76. Elamin, E.; Masclee, A.; Troost, F.; Dekker, J.; Jonkers, D. Cytotoxicity and metabolic stress induced by acetaldehyde in human intestinal LS174T goblet-like cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G286–G294. [Google Scholar] [CrossRef] [Green Version]
  77. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [Green Version]
  78. Broeders, E.P.; Nascimento, E.B.; Havekes, B.; Brans, B.; Roumans, K.H.; Tailleux, A.; Schaart, G.; Kouach, M.; Charton, J.; Deprez, B.; et al. The Bile Acid Chenodeoxycholic Acid Increases Human Brown Adipose Tissue Activity. Cell Metab. 2015, 22, 418–426. [Google Scholar] [CrossRef] [Green Version]
  79. Boets, E.; Deroover, L.; Houben, E.; Vermeulen, K.; Gomand, S.V.; Delcour, J.A.; Verbeke, K. Quantification of in Vivo Colonic Short Chain Fatty Acid Production from Inulin. Nutrients 2015, 7, 8916–8929. [Google Scholar] [CrossRef] [Green Version]
  80. Canfora, E.E.; van der Beek, C.M.; Jocken, J.W.E.; Goossens, G.H.; Holst, J.J.; Olde Damink, S.W.M.; Lenaerts, K.; Dejong, C.H.C.; Blaak, E.E. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: A randomized crossover trial. Sci. Rep. 2017, 7, 2360. [Google Scholar] [CrossRef]
  81. Degnan, F.H. The US Food and Drug Administration and probiotics: Regulatory categorization. Clin. Infect. Dis. 2008, 46 (Suppl. 2), S133–S136; discussion S144–S151. [Google Scholar] [CrossRef]
  82. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  83. Olveira, G.; Gonzalez-Molero, I. An update on probiotics, prebiotics and synbiotics in clinical nutrition. Endocrinol. Nutr. 2016, 63, 482–494. [Google Scholar] [CrossRef] [PubMed]
  84. Liang, Y.; Liang, S.; Zhang, Y.; Deng, Y.; He, Y.; Chen, Y.; Liu, C.; Lin, C.; Yang, Q. Oral Administration of Compound Probiotics Ameliorates HFD-Induced Gut Microbe Dysbiosis and Chronic Metabolic Inflammation via the G Protein-Coupled Receptor 43 in Non-alcoholic Fatty Liver Disease Rats. Probiotics Antimicrob. Proteins 2019, 11, 175–185. [Google Scholar] [CrossRef] [PubMed]
  85. Cortez-Pinto, H.; Borralho, P.; Machado, J.; Lopes, M.T.; Gato, I.V.; Santos, A.M.; Guerreiro, A.S. Microbiota Modulation with Synbiotic Decreases Liver Fibrosis in a High Fat Choline Deficient Diet Mice Model of Non-Alcoholic Steatohepatitis (NASH). GE Port. J. Gastroenterol. 2016, 23, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Xin, J.; Zeng, D.; Wang, H.; Ni, X.; Yi, D.; Pan, K.; Jing, B. Preventing non-alcoholic fatty liver disease through Lactobacillus johnsonii BS15 by attenuating inflammation and mitochondrial injury and improving gut environment in obese mice. Appl. Microbiol. Biotechnol. 2014, 98, 6817–6829. [Google Scholar] [CrossRef] [PubMed]
  87. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Li, Z.; Yang, S.; Lin, H.; Huang, J.; Watkins, P.A.; Moser, A.B.; Desimone, C.; Song, X.Y.; Diehl, A.M. Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology 2003, 37, 343–350. [Google Scholar] [CrossRef]
  89. 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. Pharm. Sci. 2011, 15, 1090–1095. [Google Scholar]
  90. 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] [Green Version]
  91. 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 in patients with non alcoholic steatohepatitis. Dig. Dis. Sci. 2012, 57, 545–553. [Google Scholar] [CrossRef] [PubMed]
  92. 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]
  93. Wong, V.W.; Won, G.L.; Chim, A.M.; Chu, W.C.; Yeung, D.K.; Li, K.C.; Chan, H.L. Treatment of nonalcoholic steatohepatitis with probiotics. A proof-of-concept study. Ann. Hepatol. 2013, 12, 256–262. [Google Scholar] [CrossRef]
  94. 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. Pharm. 2014, 39, 1276–1285. [Google Scholar] [CrossRef]
  95. Eslamparast, T.; Poustchi, H.; Zamani, F.; Sharafkhah, M.; Malekzadeh, R.; Hekmatdoost, 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]
  96. 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]
  97. Miccheli, A.; Capuani, G.; Marini, F.; Tomassini, A.; Pratico, G.; Ceccarelli, S.; Gnani, D.; Baviera, G.; Alisi, A.; Putignani, L.; et al. Urinary (1)H-NMR-based metabolic profiling of children with NAFLD undergoing VSL#3 treatment. Int. J. Obes (Lond) 2015, 39, 1118–1125. [Google Scholar] [CrossRef]
  98. Asgharian, A.; Askari, G.; Esmailzade, A.; Feizi, A.; Mohammadi, V. The Effect of Symbiotic Supplementation on Liver Enzymes, C-reactive Protein and Ultrasound Findings in Patients with Non-alcoholic Fatty Liver Disease: A Clinical Trial. Int. J. Prev. Med. 2016, 7, 59. [Google Scholar] [CrossRef]
  99. Ferolla, S.M.; Couto, C.A.; Costa-Silva, L.; Armiliato, G.N.; Pereira, C.A.; Martins, F.S.; Ferrari Mde, L.; 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] [Green Version]
  100. 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]
  101. Abdel Monem, S.M. Probiotic Therapy in Patients with Nonalcoholic Steatohepatitis in Zagazig University Hospitals. Euroasian J. Hepatogastroenterol. 2017, 7, 101–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Behrouz, V.; Jazayeri, S.; Aryaeian, N.; Zahedi, M.J.; Hosseini, F. Effects of Probiotic and Prebiotic Supplementation on Leptin, Adiponectin, and Glycemic Parameters in Non-alcoholic Fatty Liver Disease: A Randomized Clinical Trial. Middle East. J. Dig. Dis. 2017, 9, 150–157. [Google Scholar] [CrossRef] [Green Version]
  103. Ekhlasi, G.; Zarrati, M.; Agah, S.; Hosseini, A.F.; Hosseini, S.; Shidfar, S.; Soltani Aarbshahi, S.S.; Razmpoosh, E.; Shidfar, F. Effects of synbiotics and vitamin E supplementation on blood pressure, nitric oxide and inflammatory factors in non-alcoholic fatty liver disease. EXCLI J. 2017, 16, 278–290. [Google Scholar] [CrossRef]
  104. 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]
  105. Manzhalii, E.; Virchenko, O.; Falalyeyeva, T.; Beregova, T.; Stremmel, W. Treatment efficacy of a probiotic preparation for non-alcoholic steatohepatitis: A pilot trial. J. Dig. Dis. 2017, 18, 698–703. [Google Scholar] [CrossRef]
  106. 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] [Green Version]
  107. Bakhshimoghaddam, F.; Shateri, K.; Sina, M.; Hashemian, M.; Alizadeh, M. Daily Consumption of Synbiotic Yogurt Decreases Liver Steatosis in Patients with Nonalcoholic Fatty Liver Disease: A Randomized Controlled Clinical Trial. J. Nutr. 2018, 148, 1276–1284. [Google Scholar] [CrossRef] [Green Version]
  108. Javadi, L.; Khoshbaten, M.; Safaiyan, A.; Ghavami, M.; Abbasi, M.M.; Gargari, B.P. Pro- and prebiotic effects on oxidative stress and inflammatory markers in non-alcoholic fatty liver disease. Asia Pac. J. Clin. Nutr. 2018, 27, 1031–1039. [Google Scholar] [CrossRef]
  109. Kobyliak, N.; Abenavoli, L.; Falalyeyeva, T.; Mykhalchyshyn, G.; Boccuto, L.; Kononenko, L.; Kyriienko, D.; Komisarenko, I.; Dynnyk, O. Beneficial effects of probiotic combination with omega-3 fatty acids in NAFLD: A randomized clinical study. Minerva Med. 2018, 109, 418–428. [Google Scholar] [CrossRef]
  110. Kobyliak, N.; Abenavoli, L.; Mykhalchyshyn, G.; Kononenko, L.; Boccuto, L.; Kyriienko, D.; Dynnyk, O. A Multi-strain Probiotic Reduces the Fatty Liver Index, Cytokines and Aminotransferase levels in NAFLD Patients: Evidence from a Randomized Clinical Trial. J. Gastrointestin. Liver Dis. 2018, 27, 41–49. [Google Scholar] [CrossRef] [Green Version]
  111. Sayari, S.; Neishaboori, H.; Jameshorani, M. Combined effects of synbiotic and sitagliptin versus sitagliptin alone in patients with nonalcoholic fatty liver disease. Clin. Mol. Hepatol. 2018, 24, 331–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wang, W.; Shi, L.P.; Shi, L.; Xu, L. Efficacy of probiotics on the treatment of non-alcoholic fatty liver disease. Zhonghua Nei Ke Za Zhi 2018, 57, 101–106. [Google Scholar] [CrossRef] [PubMed]
  113. Ahn, S.B.; Jun, D.W.; Kang, B.K.; Lim, J.H.; Lim, S.; Chung, M.J. Randomized, Double-blind, Placebo-controlled Study of a Multispecies Probiotic Mixture in Nonalcoholic Fatty Liver Disease. Sci. Rep. 2019, 9, 5688. [Google Scholar] [CrossRef] [Green Version]
  114. Loman, B.R.; Hernandez-Saavedra, D.; An, R.; Rector, R.S. Prebiotic and probiotic treatment of nonalcoholic fatty liver disease: A systematic review and meta-analysis. Nutr. Rev. 2018, 76, 822–839. [Google Scholar] [CrossRef]
  115. Khan, M.Y.; Mihali, A.B.; Rawala, M.S.; Aslam, A.; Siddiqui, W.J. The promising role of probiotic and synbiotic therapy in aminotransferase levels and inflammatory markers in patients with nonalcoholic fatty liver disease—A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2019, 31, 703–715. [Google Scholar] [CrossRef]
  116. Koutnikova, H.; Genser, B.; Monteiro-Sepulveda, M.; Faurie, J.M.; Rizkalla, S.; Schrezenmeir, J.; Clement, K. Impact of bacterial probiotics on obesity, diabetes and non-alcoholic fatty liver disease related variables: A systematic review and meta-analysis of randomised controlled trials. BMJ Open 2019, 9, e017995. [Google Scholar] [CrossRef]
  117. Liu, L.; Li, P.; Liu, Y.; Zhang, Y. Efficacy of Probiotics and Synbiotics in Patients with Nonalcoholic Fatty Liver Disease: A Meta-Analysis. Dig. Dis. Sci. 2019. [Google Scholar] [CrossRef]
  118. Sharpton, S.R.; Maraj, B.; Harding-Theobald, E.; Vittinghoff, E.; Terrault, N.A. Gut microbiome-targeted therapies in nonalcoholic fatty liver disease: A systematic review, meta-analysis, and meta-regression. Am. J. Clin. Nutr. 2019. [Google Scholar] [CrossRef]
  119. Sigrist, R.M.S.; Liau, J.; Kaffas, A.E.; Chammas, M.C.; Willmann, J.K. Ultrasound Elastography: Review of Techniques and Clinical Applications. Theranostics 2017, 7, 1303–1329. [Google Scholar] [CrossRef]
  120. Idilman, I.S.; Aniktar, H.; Idilman, R.; Kabacam, G.; Savas, B.; Elhan, A.; Celik, A.; Bahar, K.; Karcaaltincaba, M. Hepatic steatosis: Quantification by proton density fat fraction with MR imaging versus liver biopsy. Radiology 2013, 267, 767–775. [Google Scholar] [CrossRef]
  121. Hempel, S.; Newberry, S.; Ruelaz, A.; Wang, Z.; Miles, J.N.; Suttorp, M.J.; Johnsen, B.; Shanman, R.; Slusser, W.; Fu, N.; et al. Safety of probiotics used to reduce risk and prevent or treat disease. Evid. Rep. Technol. Assess. (Full Rep.) 2011, 200, 1–645. [Google Scholar]
  122. Doron, S.; Snydman, D.R. Risk and safety of probiotics. Clin. Infect. Dis. 2015, 60 (Suppl. 2), S129–S134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The roles gut microbiota play in liver steatosis. LPS: lipopolysaccharide; SCFAs: short-chain fatty acids; AAA: aromatic amino acids; BCAA: branched-chain amino acids; EtOH: ethanol; FXR: farnesoid X receptor; TGR5: transmembrane G protein-coupled receptor 5; GPR: G protein-coupled receptor; TMAO: trimethylamine-N-oxide; VLDL-C: very-low-density lipoprotein cholesterol; ROS: Reactive oxygen species; TNF-α: tumor necrosis factor-alfa; IFN- γ: Interferon-gamma; IL-1β: interleukin 1beta; IL-6: interleukin 6; IL-8: interleukin 8; TLR: toll-like receptor; NAFLD: Non-alcoholic fatty liver disease.
Figure 1. The roles gut microbiota play in liver steatosis. LPS: lipopolysaccharide; SCFAs: short-chain fatty acids; AAA: aromatic amino acids; BCAA: branched-chain amino acids; EtOH: ethanol; FXR: farnesoid X receptor; TGR5: transmembrane G protein-coupled receptor 5; GPR: G protein-coupled receptor; TMAO: trimethylamine-N-oxide; VLDL-C: very-low-density lipoprotein cholesterol; ROS: Reactive oxygen species; TNF-α: tumor necrosis factor-alfa; IFN- γ: Interferon-gamma; IL-1β: interleukin 1beta; IL-6: interleukin 6; IL-8: interleukin 8; TLR: toll-like receptor; NAFLD: Non-alcoholic fatty liver disease.
Nutrients 11 02837 g001
Table 1. Summary of major randomized controlled trials using probiotics or synbiotics therapy for NAFLD/NASH.
Table 1. Summary of major randomized controlled trials using probiotics or synbiotics therapy for NAFLD/NASH.
StudyDesign/Population
(N = Included in the Trial)
Bacterial SpeciesDurationMain Outcome Related to NAFLD
SerologyImaging or Biopsy
Aller et al. [89], 2011Double-blind RCT/NAFLD
(N = 28)
Lactobacillus bulgaricus
Streptococcus thermophilus
3 months(↓) ALT, AST, GGT (-) Anthropometric parameters and cardiovascular risk factorsN/A
Vajro et al. [90], 2011Double-blind RCT/NAFLD obese children
(N = 20)
Lactobacillus rhamnosus8 weeks(↓) ALT
(-) BMI, TNF-α, peptidoglycanpolysaccharide antibody
N/A
Malaguarnera et al. [91], 2012RCT/NASH
(N = 66)
Bifidobacterium longum
+
Prebiotics (Fructo-oligosaccharides)
24 weeks(↓) AST, LDL-C, CRP, TNF-α, endotoxin, HOMA-IR
(-) ALT, bilirubin, HDL-C, TC, TG, glucose, insulin, C-peptide, BMI
The liver biopsy indicated that steatosis and NASH activity improved
Shavakhi et al. [92], 2013Double-blind RCT/NASH on metformin
(N = 64)
Lactobacillus acidophilus
Lactobacillus casei
Lactobacillus rhamnosus
Lactobacillus bulgaricus
Bifidobacterium breve
Bifidobacterium longum
Streptococcus thermophilus
6 months(↓) ALT, AST, TG, TC, BMI
(-) FBS
Grading of steatosis based on US measurement improved
Wong et al. [93], 2013RCT/NASH
(N = 20)
Lactobacillus plantarum, Lactobacillus deslbrueckii
Lactobacillus acidophilus Lactobacillus rhamnosus Bifidobacterium bifidum
6 months(↓) ALT
(-) AST, BMI, waist circumference, glucose, and lipid levels
Proton-magnetic resonance spectroscopy-measured that intrahepatic triglyceride content (IHTG) improved
Alisi et al. [94], 2014Double-blind RCT/NAFJD children
(N = 44)
Streptococcus thermophilus
Bifidobacterium breve
Bifidobacterium longum Bifidobacterium infantis Lactobacillus acidophilus Lactobacillus plantarum Lactobacillus casei Lactobacillus bulgaricus
4 months(↓) BMI
(↑) GLP-1, activated GLP-1
(-) ALT, triglycerides, HOMA
Grading of steatosis based on US measurement improved
Eslamparast et al. [95], 2014Double-blind RCT/NAFLD with lifestyle modification (N = 52)Lactobacillus casei
Lactobacillus rhamnosus
Lactobacillus acidophilus
Lactobacillus bulgaricus
Bifidobacterium breve
Bifidobacterium longum
Streptococcus thermophilus
+ prebiotic
(fructo-oligosaccharide)
28 weeks(↓) ALT, AST, CRP, TNF-ɑ, NF-κB p65
(-) BMI
Transient elastography- measured fibrosis score improved
Nabvi et al. [96], 2014Double-blind RCT/NAFLD (N = 72)Lactobacillus acidophilus
Bifidobacterium lactis
8 weeks(↓) ALT, AST, TC, LDL-C
(-) Glucose, TG, HDL-C
N/A
Miccheli et al. [97], 2015Triple-blind RCT/NAFLD children
(N = 31)
Streptococcus thermophilus
Bifidobacterium breve
Bifidobacterium longum Bifidobacterium infantis Lactobacillus acidophilus Lactobacillus plantarum Lactobacillus casei Lactobacillus bulgaricus
4 months(↓) AST,
total and active GLP-1, BMI
(-) ALT, TG, TC, HDL-C, LDL-C, glucose, insulin
Grading of steatosis based on US measurement improved
Asgharian et al. [98], 2016Double-blind RCT/NAFLD (N = 80)Lactobacillus casei
Lactobacillus acidophilus
Lactobacillus rhamnosus
Lactobacillus bulgaricus
Bifidobacterium breve
Bifidobacterium longum
Streptococcus thermophilus
+ prebiotic (fructo-oligosaccharide)
8 weeksPrevent ASL and ALT elevation
(-) CRP, BMI
Grading of steatosis based on US measurement improved
Ferolla et al. [99], 2016RCT/NASH
(N = 50)
Lactobacillus reuteri
+
prebiotic (inulin)
3 months(↓) BMI
(-) AST, ALT, ALP, GGT, TC, TG, HDL-C, VDL-C, LPS, and intestinal permeability measured by lactulose/mannitol urinary excretion
MRI-PDFF- measured steatosis improved but liver fibrosis had no significant change
Sepideh et al. [100], 2016Double-blind RCT/NAFLD (N = 42)Lactobacillus casei
Lactobacillus acidophilus
Lactobacillus rhamnosus
Lactobacillus bulgaricus
Bifidobacterium breve
Bifidobacterium longum
Streptococcus thermophilus
8 weeks(↓) IL-6, FBS, insulin, insulin resistance
(-) TNF-alpha
N/A
Abdel et al. [101], 2017RCT/NASH with obesity
(N = 30)
Lactobacillus acidophilus1 month(↓) ALT, AST
(-) TG, TC, FBS Bilirubin, HDL-C
N/A
Behrouz et al. [102], 2017Double-blind RCT/NAFLD
(N = 89)
Lactobacillus casei
Lactobacillus rhamnosus
Lactobacillus acidophilus
Bifidobacterium longum
Bifidobacterium breve
12 weeks(↓) Leptin, insulin, and HOMA-IR
(-) Adiponectin, FBS
N/A
Ekhlasi et al. [103], 2017Double-blind RCT/NAFLD with Vitamin E (N = 60)Lactobacillus casei
Lactobacillus rhamnosus Stretococcus thermophilus
Bifidobacterium breve Lactobacillus acidophilus
Bifidobacterium longum Lactobacillus bulgaricus
+
prebiotic (fructo-oligosaccharide)
8 weeks(↓) ALT, AST, ALP, sytolic BP, malondialdehyde, TNF-alpha
(-) Diastolic Blood Pressure, nitric oxide, BMI
N/A
Famouri et al. [104], 2017Triple-Blind RCT/NAFLD obese children (N = 64)Lactobacillus acidophilus
Bifidobacterium lactis
Bifidobacterium bifidum
Lactobacillus rhamnosus
12 weeks(↓) ALT, AST, cholesterol, triglycerides, LDL-C, and waist circumference
(-) BMI, weight
Grading of steatosis based on US measurement improved
Manzhalii et al. [105], 2017RCT/NASH with low-fat/low-calorie diet
(N = 75)
Lactobacilli
Bifidobacteria
Streptococcus thermophilus
12 weeks(↓) ALT, BMI and cholesterol
(-) GGT
Liver stiffness based on US measurement improved
Mofidi et al. [106], 2017Double-blind RCT/NAFLD
(N = 50)
Lactobacillus acidophilus
Lactobacillus casei
Lactobacillus rhamnosus
Lactobacillus bulgaricus
Bifidobacterium breve
Bifidobacterium longum
Streptococcus thermophilus
+ prebiotic
(fructo-oligosaccharide)
28 weeks(↓) AST, ALT, GGT, glucose, triglyceride, Total cholesterol, CRP, NF-κB p65
(-) HDL-C, LDL-C, TNF-α
Transient elastography-measured hepatic steatosis and fibrosis improved
Bakhshimoghaddam et al. [107], 2018RCT/NAFLD (N = 102)Bifidobacterium animalis
+ prebiotic (inulin)
24 weeks(↓) AST, ALT, GGT, ALP, TG, TCGrading of steatosis based on US measurement improved
Javadi et al. [108], 2018Double-blind RCT/NAFLD (N = 75)Bifidobacterium longum
Lactobacillus acidophilus
+ prebiotic (inulin)
3 months(↓) CRP, TNF-α, BMI
(↑) TAC
(-) IL-6, MDA
N/A
Kobyliak et al. [109], 2018Double-blind RCT/NAFLD with type II DM
(N = 48)
Bifidobacterium
Lactobacillus
Lactococcus
Propionibacterium
Acetobacter
+ omega-3 fatty acids
8 weeks(↓) GGT, TG, TC, VLDL-C, TNF-α, IL-6
(-) AST, ALT, LDL-C, HDL-C, INF-γ, IL-1β, IL-8
Shear Wave Elastography-measured fatty liver index improvement but no significant change in liver stiffness
Kobyliak et al. [110], 2018Double-blind RCT/NAFLD with type II DM (N = 58)Bifidobacterium
Lactobacillus
Lactococcus
Propionibacterium
Acetobacter
8 weeks(↓) AST, GTT, TNF-α, IL-6
(-) ALT, TC, TG, VLDL-C, HDL-C, IL-1β, IL-8, IFN- γ
Shear Wave Elastography-measured fatty liver index improvement but no significant change in liver stiffness
Sayari et al. [111], 2018RCT/NAFLD with sitagliptin (N = 138)Lactobacillus casei Lactobacillus rhamnosus
Lactobacillus acidophilus
Lactobacillus bulgaricus Bifidobacterium breve Bifidobacterium longum
Streptococcus thermophilus
+ prebiotic
(fructo-oligosaccharide)
16 weeks(↓) AST, TC, LDL-C, FBS
(-) ALT, HDL-C, TG, BMI
N/A
Wang et al. [112], 2018Double-blind RCT/NAFLD (N = 200)Bifidobacterium Lactobacillus
Enterococcus
Bacillus subtilis
1 month(↓) AST, ALT, TC, TG, glucose, VLDL-C, TNF-α
(↑) High molecular weight adiponectin
(-) HDL-C
Grading of steatosis based on US measurement showed no significant difference
Ahn et al. [113], 2019Double-blind RCT/NAFLD with obesity
(N = 68)
Lactobacillus acidophilus
Lactobacillus rhamnosus
Lactobacillus paracasei
Pediococcus pentosaceus Bifidobacterium lactis
Bifidobacterium breve
12 weeks(↓) TG
(-) AST, ALT, TC, HDL-C, glucose, insulin, TNF-α, IL-6, LPS
MRI-PDFF-measured intrahepatic fat fraction (IHF) and mean IHF reduced compared with control, but no significant change in liver stiffness
Duseja et al. [69], 2019Double-blind RCT/NAFLD (N = 30)Lactobacillus paracasei
Lactobacillus plantarum
Lactobacillus acidophilus
Lactobacillus delbrueckii
subsp. bulgaricus
Bifidobacterium longum
Bifidobacterium infantis
Bifidobacterium breve
Streptococcus thermophilus
1 year(↓) ALT, ALP, leptin, TNF-α, and LPS
(-) AST, Bilirubin, Adiponectin, IL-1β, IL-6
The biopsy indicated hepatocyte ballooning, NAS score and fibrosis improved, but not steatosis or lobular inflammation
NAFLD: Non-alcoholic fatty liver disease; NASH: Nonalcoholic steatohepatitis; ALT: alanine transaminase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; ALP: alkaline phosphatase; LDL-C: low-density lipoprotein cholesterol; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FBS: fasting serum glucose; GLP-1: glucagon-like peptide 1; BMI: body mass index; LPS: Lipopolysaccharides; CRP: C reactive protein; TNF-α: tumor necrosis factor-alfa; NF-κB p65: nuclear factor kappa-light-chain-enhancer of activated B cells subunit p65; IFN- γ: Interferon-gamma; IL-1β: interlukin 1beta; IL-6: interleukin 6; IL-8: interleukin 8; MDA: malondialdehyde; TAC: total antioxidant capacity; NAS: NAFLD activity score; HOMA-IR: homeostasis model assessment of insulin resistance; US: ultrasonography; MRI-PDFF: magnetic resonance imaging–derived proton density fat fraction; IHF: intrahepatic fat fraction.
Table 2. Summary of recent meta-analysis of RCTs with probiotics/synbiotics therapy for NAFLD/NASH patients.
Table 2. Summary of recent meta-analysis of RCTs with probiotics/synbiotics therapy for NAFLD/NASH patients.
StudyPopulationStudy PeriodConclusions
Loman et al. [114], 2018Included 25 studies (most are RCT): Among them, 9 studies used prebiotics, 11 studies used probiotics, and 7 studies used synbiotics. 1309 patients were included.Up to December 14, 2017Microbial therapies significantly reduced AST and ALT, but not CRP.
The results of serum cholesterol and LDL-C are mixed among prebiotics, probiotics, and synbiotics.
Khan et al. [115], 2019Included 12 probiotics/synbiotics RCTs for NAFLD. 748 patients were included.Up to June 10, 2018Probiotics/synbiotics were associated with a significant improvement in ALT, AST, and liver fibrosis score graded by fibroscan. There was a reduction in CRP with synbiotics. The TNF-α, LDL-C, TG, and TC significantly improved with synbiotics but not with probiotics in a subgroup analysis. There were no significant changes in HDL-C, HOMA-IR, or FBS in either group.
Koutnikova et al. [116], 2019Included 105 articles with 111 RCTs representing 6826 subjects (includes metabolic syndrome, type II DM, and NAFLD patients). Among them, about 658 patients had NAFLD.January 1990 to June 2018In subjects with fatty liver diseases, probiotics reduced AST and ALT, but not GGT.
Liu et al. [117], 2019Included 15 probiotics and synbiotic RCTs, involving 782 patients with NAFLD.Up to April 2018Probiotics and synbiotics supplementation could significantly improve AST, ALT, TG, TC, HDL-C, LDL-C, homeostasis model assessment-insulin resistance, TNF-α, liver steatosis, and liver stiffness. However, probiotics and synbiotics could not ameliorate BMI, waist circumference, or FBS.
Sharpton et al. [118], 2019Included 21 RCTs (1252 participants) with NAFLD. 9 trials evaluated probiotics and 12 trials evaluated synbiotics.January 1, 2005 to December 1, 2018Probiotics/synbiotics could improve AST and ALT. Probiotics/synbiotics were also associated with hepatic steatosis improvement when graded with ultrasound. Last, probiotics/synbiotics were associated with liver stiffness improvement when measured by elastography, although analyses showed heterogeneity.
Probiotics, but not synbiotics, were associated with a significant reduction in body mass index.
NAFLD: Non-alcoholic fatty liver disease;DM: diabetes mellitus; ALT: alanine transaminase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; LDL-C: low-density lipoprotein cholesterol; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FBS: fasting serum glucose; BMI: body mass index; CRP: C reactive protein; TNF-α: tumor necrosis factor-alfa; HOMA-IR: homeostasis model assessment of insulin resistance.

Share and Cite

MDPI and ACS Style

Xie, C.; Halegoua-DeMarzio, D. Role of Probiotics in Non-alcoholic Fatty Liver Disease: Does Gut Microbiota Matter? Nutrients 2019, 11, 2837. https://0-doi-org.brum.beds.ac.uk/10.3390/nu11112837

AMA Style

Xie C, Halegoua-DeMarzio D. Role of Probiotics in Non-alcoholic Fatty Liver Disease: Does Gut Microbiota Matter? Nutrients. 2019; 11(11):2837. https://0-doi-org.brum.beds.ac.uk/10.3390/nu11112837

Chicago/Turabian Style

Xie, Chencheng, and Dina Halegoua-DeMarzio. 2019. "Role of Probiotics in Non-alcoholic Fatty Liver Disease: Does Gut Microbiota Matter?" Nutrients 11, no. 11: 2837. https://0-doi-org.brum.beds.ac.uk/10.3390/nu11112837

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