Next Article in Journal
Extracellular Vesicles in Blood: Sources, Effects, and Applications
Next Article in Special Issue
Smectite as a Preventive Oral Treatment to Reduce Clinical Symptoms of DSS Induced Colitis in Balb/c Mice
Previous Article in Journal
Efficient In Vitro and In Vivo Anti-Inflammatory Activity of a Diamine-PEGylated Oleanolic Acid Derivative
Previous Article in Special Issue
Leaky Gut: Effect of Dietary Fiber and Fats on Microbiome and Intestinal Barrier
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Leaky Gut in Nonalcoholic Fatty Liver Disease: A Novel Therapeutic Target

by
Takaomi Kessoku
1,2,*,
Takashi Kobayashi
1,
Kosuke Tanaka
1,2,
Atsushi Yamamoto
1,
Kota Takahashi
1,
Michihiro Iwaki
1,2,
Anna Ozaki
1,
Yuki Kasai
1,
Asako Nogami
1,
Yasushi Honda
1,2,
Yuji Ogawa
1,
Shingo Kato
1,
Kento Imajo
1,
Takuma Higurashi
1,
Kunihiro Hosono
1,
Masato Yoneda
1,
Haruki Usuda
3,
Koichiro Wada
3,
Satoru Saito
1 and
Atsushi Nakajima
1
1
Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
2
Department of Palliative Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
3
Department of Pharmacology, Shimane University Faculty of Medicine, 89-1 Enyacho, Izumo, Shimane 693-8501, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(15), 8161; https://doi.org/10.3390/ijms22158161
Submission received: 28 June 2021 / Revised: 26 July 2021 / Accepted: 28 July 2021 / Published: 29 July 2021

Abstract

:
The liver directly accepts blood from the gut and is, therefore, exposed to intestinal bacteria. Recent studies have demonstrated a relationship between gut bacteria and nonalcoholic fatty liver disease (NAFLD). Approximately 10–20% of NAFLD patients develop nonalcoholic steatohepatitis (NASH), and endotoxins produced by Gram-negative bacilli may be involved in NAFLD pathogenesis. NAFLD hyperendotoxicemia has intestinal and hepatic factors. The intestinal factors include impaired intestinal barrier function (leaky gut syndrome) and dysbiosis due to increased abundance of ethanol-producing bacteria, which can change endogenous alcohol concentrations. The hepatic factors include hyperleptinemia, which is associated with an excessive response to endotoxins, leading to intrahepatic inflammation and fibrosis. Clinically, the relationship between gut bacteria and NAFLD has been targeted in some randomized controlled trials of probiotics and other agents, but the results have been inconsistent. A recent randomized, placebo-controlled study explored the utility of lubiprostone, a treatment for constipation, in restoring intestinal barrier function and improving the outcomes of NAFLD patients, marking a new phase in the development of novel therapies targeting the intestinal barrier. This review summarizes recent data from studies in animal models and randomized clinical trials on the role of the gut–liver axis in NAFLD pathogenesis and progression.

1. Introduction

Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome and the leading cause of chronic liver disease in pediatric and adult populations living in industrialized countries. NAFLD encompasses steatosis and nonalcoholic steatohepatitis (NASH), and it is characterized by perivenular and lobular inflammation. Progression to fibrosis and cirrhosis are the primary complications of NAFLD [1]. Based on a recent meta-analysis, one in four people in Europe, the United States, and Asia have NAFLD [2]. The “multiple parallel hit” hypothesis may explain the pathogenesis and progression of NAFLD. Especially in recent years, there has been increasing interest in gut–liver axis dysfunction (dysbiosis, bacterial overgrowth, and changes in intestinal permeability) associated with the progression of NAFLD; therefore, gut–liver axis dysfunction is considered important as a possible alternative therapeutic target for patients who are unable to benefit from lifestyle changes, healthy eating, and promotion of physical activity [3,4]. In NASH, chronic inflammation is triggered by hepatocyte fat accumulation, followed by exposure to inflammatory cytokines, insulin resistance, oxidative stress, lipotoxicity mainly from free fatty acids (FFAs), and gut-derived endotoxins. Here, we focused on gut-derived endotoxins and reviewed the most recent data regarding the gut–liver axis and its role in the pathogenesis and progression of NAFLD. We also reviewed experimental studies in animal models and preliminary results from several randomized clinical trials (RCTs). The objectives of our review were to (1) appraise the pathophysiology of the gut–liver axis focusing on endotoxins and (2) delineate novel therapeutic perspectives via intestinal permeability.

2. Hepatic Inflammation and NAFLD

The inflammatory state associated with the metabolic syndrome is unique because it is not accompanied by signs of infection or autoimmunity. Furthermore, there is no major tissue damage. In addition, the dimension of inflammatory activation is not large and is often referred to as low-grade chronic inflammation. Other researchers have named this inflammatory condition metainflammation, which corresponds to metabolically induced inflammation [5], or parainflammation, which is defined as an intermediate state between basal and inflammatory states [6]. Whatever the terminology, the inflammatory processes that characterize metabolic syndrome have unique features and mechanisms that are not fully understood [7]. Metabolic syndrome, especially NASH, is caused by metainflammation. Indeed, chronic mild inflammation is an important factor in the pathogenesis of NASH [8,9].
Since the portal vein is the direct venous outflow of the intestine, the liver is continuously exposed to factors that have an intestinal origin, including bacteria and bacterial components. The liver is an important site for bacterial phagocytosis and clearance because it contains the largest population of tissue macrophages. Experimental data demonstrate that exposure of membrane components of Gram-negative bacteria and bacterial products to proinflammatory mediators such as LPS results in the activation of Kupffer cells. Kupffer cells, the resident macrophages of the liver, are the nitrogenous species that contribute to liver damage in the presence of proinflammatory cytokines and are a major source of inflammatory mediators such as ROS [10]. Through pattern recognition receptors, including Toll-like receptors (TLRs), the innate immune system recognizes conserved pathogen-associated molecular patterns (PAMPs) [11]. Although healthy livers have a low TLR mRNA level and a high resistance to TLR ligands from the constant exposure of the microflora, TLR-mediated signaling plays a major role in liver physiology and pathophysiology [12]. LPS is a potent activator of innate immune responses as it can bind to the TLR4 complex. TLR4 is expressed by Kupffer cells, hepatic stellate cells, hepatocytes, biliary epithelial cells, sinusoidal endothelial cells, and hepatic dendritic cells, which are consequently responsive to LPS [13]. There is a positive correlation among hepatic dysfunction, increased bacterial transfer, and LPS. Furthermore, in conditions with hepatic dysfunction such as cirrhosis, the clearance of LPS from circulation is reduced [13]. Cytoplasmic screening and selective recruitment of signaling adapter proteins target downstream of TLR4 signals through interactions between Toll/IL-1 receptor (TIR) domains [14,15,16]. Therefore, TLR4 activation involves the bone marrow differentiation factor 88 (MyD88), an adapter protein containing the TIR domain, or the MyD88 adapter-like factor, which is associated with the nuclear factor kappa B (NF-κB) and AP-1 transcription factor. There is substantial evidence that TLR4-mediated intracellular events exacerbate liver damage in fatty liver [17,18].
Kupffer cells produce tumor necrosis factor-α (TNF-α) and interleukin (IL)-10 in response to physiological concentrations of LPS [19,20,21]. TNF-α is an inflammatory cytokine that activates various signaling cascades, including many pathways that are described below, as important inhibitors of insulin action. TNF-α is overexpressed in obese rodents and in human adipose tissue and liver. Furthermore, it has a decreased concentration when the body is undergoing weight loss. Geoffrey et al. [22] explored the administration of a methionine–choline-deficient (MCD) diet to TNF-α and NF-κB knockout animals. Activation of TNF-α and NF-κB is essential for hepatic inflammatory recruitment in steatohepatitis. Furthermore, NF-κB activation occurs independently of TNF-α. Other studies using the MCD dietary model have reported that curcumin, which blocks oxidative stress-mediated NF-κB activation, provides protection [23], but TNF-α antiserum reduces liver injury in rats administered the MCD diet [24]. Tomita, et al. [25] demonstrated that TNF-receptor knockout mice were protected against liver fibrosis in their MCD experiments.
Alternatively, activated macrophages, including Kupffer cells, which are also referred to as M2, as opposed to the classical M1 or pro-inflammatory phenotype, represent another important pathway in resolving the inflammatory response. The coordinated program of alternative activation is primarily stimulated by the Th2 cytokines, IL-4 and IL-13, and is characterized by cell-surface expression of M2 signature genes, such as mannose receptor, arginase-1, and dectin-1 [26]. There is evidence that adiposity promotes Th1 polarization of cytokine balance, favoring congenital or classical activation of macrophages in NAFLD. Thus, in experimental and human NAFLD, the pool of hepatic natural killer T cells (NKT) is reduced, while the pool of hepatic tissue Th1 cytokines such as TNF-α, IL-12, IL-18, and interferon-γ is elevated [27,28,29,30].

3. Endotoxins and NAFLD

Fatty liver is caused by excessive calories due to overeating, obesity, and lack of exercise. The mechanism by which a fatty liver develops includes exposure to inflammatory cytokines, IR, oxidative stress, lipotoxicity mainly due to FFAs, and genetic predisposition. Nevertheless, exposure of the liver to endotoxins from intestinal bacteria is important.
Endotoxin levels in the blood are high in NASH patients [31]. In recent years, it was reported that serum endotoxin levels increased when volunteers were fed a high-fat Western diet, referred to as metabolic endotoxemia [32]. Gram-negative bacilli in the intestinal microflora are considered the largest source of stable endotoxins. As the number of anaerobic Gram-negative rods increases due to the deterioration of the intestinal environment, the amount of LPS, the so-called endotoxin, also increases. Although it is unlikely that all of these endotoxins enter the portal vein from the intestine due to the presence of an elaborate immune system and a functional intestinal barrier, it is still likely that some of them reach the liver and serve as a stable source of endotoxins. When enterobacteria from the intestine invade the portal vein, the first target organ is the hepatic vasculature. This suggests that Enterobacteriaceae-derived endotoxins are important in the inflammatory response that leads to the development of NASH. Endotoxins are PAMPs, which are members of a group of receptors called TLRs and nucleotide-binding oligomerization domain receptors (NLRs).
In particular, TLR4 is expressed in the plasma membrane of hepatocytes and Kupffer cells. When endotoxin stimulates TLR4, signaling molecules such as NF-kB are activated. This leads to the production of inflammatory cytokines (IL-1b and IL-18), which result in liver injury. In addition, antimicrobial therapy reduces hepatic damage in NASH. Animal studies have shown that TLR4-deficient mice did not develop NASH, thereby suggesting that intestinal bacteria play an important role in the emergence of NASH. In terms of elucidation of the mechanism of intestinal bacterial NASH development, the following are the intestinal factors: (1) disruption of the intestinal barrier function, which results in a leaky gut, (2) qualitative and quantitative dysregulation of intestinal bacteria, referred to as small intestinal bacterial overgrowth (SIBO), and (3) increased endogenous alcohol concentration [33]. On the other hand, the increased responsiveness to endotoxin is a hepatic factor (Figure 1).

4. Intestinal Factors Associated with Endotoxins

4.1. Intestinal Permeability

The concept that increased intestinal permeability and intestinal microbiota might contribute to development of some diseases was first proposed in 1890 [34]. Crosstalk between the gut and the liver may explain changes in the hepatobiliary system associated with several inflammatory and infectious bowel diseases, such as celiac disease, and infections caused by Salmonella and Yersinia [35]. Evidence of involvement of the hepatointestinal system in the development of NASH has recently emerged [36,37,38].
Obesity increases intestinal permeability through indirectly damaging the intestinal barrier [38,39,40]. An animal study showed that a high-fat diet (HFD) may increase metabolic endotoxinemia and reduce intestinal Bifidobacteria [41,42], which can lower intestinal LPS levels and improve mucosal barrier function [39]. Mechanisms that regulate the health of the intestinal barrier may also regulate the degree of endotoxinemia [38,39,40,43]. Another potential pathway that can be involved in the absorption of LPS in the intestine is the secretion of chylomicrons from enterocytes rather than disrupting tight junctions between cells. Studies using cell culture and animal models have suggested that endotoxins are actively secreted into the blood along with chylomicrons. Furthermore, inhibition of chylomicron synthesis inhibits endotoxin secretion [44]. These data show that gut microbiota-derived endotoxins are strongly associated with the development of NASH through impaired intestinal barriers and increased chylomicron secretion of enterocytes.
Under physiological conditions, tight junction proteins, such as zonula occludens, seal apical junctions between intestinal endothelial cells. Enteric dysbiosis disrupts these tight junctions and increases mucosal permeability, which exposes intestinal mucosal cells and the liver to potential inflammatory products. The recognition of toxic substances in the liver is mediated by pattern-recognition receptors (PRRs), a group of receptors that includes TLRs and NLRs. In a meta-analysis by Luther et al. [45], patients with NAFLD exhibited an increased intestinal permeability compared with that of healthy controls (NAFLD group: 39.1%; healthy controls: 6.8%), with an odds ratio of 5.08 (95% confidence interval (CI): 1.98–13.05). Furthermore, they reported that patients with NASH had an increased intestinal permeability compared with that of the healthy controls, with an odds ratio of 7.21 (95% CI: 2.35–22.13).

4.2. SIBO

Qualitative or quantitative imbalances of complex intestinal microbiota might have serious health consequences for a macroorganism, including SIBO. The discovery of a linkage between SIBO and NAFLD [46,47,48], and the observation that endotoxin triggers liver inflammation in mice with steatosis [49], prompted the formulation of this hypothesis [50]. The prevalence of SIBO was about three times that of the controls [51]. Results consistent with those demonstrating SIBO were found in 2.5% to 22% of studies investigating small sets of clinically healthy people as controls [52,53,54,55,56,57,58,59,60]. Animal studies reported that excessive multiplication of Escherichia coli coexisted in NASH rats [61], consistent with previous studies. This suggested that SIBO is one of many factors important in the pathogenesis of NASH, as antibacterial treatment could alleviate the severity of NASH [61]. Thus, SIBO may coexist with NASH. In addition, levels of ALT increased or decreased relative to serum levels of TNF-α [61]. This strongly supported TNF-α as an important mediator for the promotion of NASH by SIBO. Generally, endotoxemia is thought to be a link between SIBO and elevated TNF-α levels [46,62]. The association between SIBO and NAFLD and the increased endotoxemia across studies highlight the role of gut microbiota in the initiation and development of metabolic liver disease [42,52]. Lichtman et al. showed that antibiotics (metronidazole and tetracycline) reduced hepatic injury in rats with surgically induced intestinal bacterial overgrowth [48]. Drenick et al. and Vanderhoof et al. also showed that antibiotics prevented and reversed hepatic steatosis and liver injury after intestinal bypass for patients with morbid obesity [63,64]. Additionally, Bergheim et al. showed that antibiotics could reduce hepatic steatosis and endotoxinemia in a fructose-induced rodent NAFLD model [65]. These findings imply a critical role for small bowel flora, suggesting that intestinal bacterial overgrowth treatment reduces ethanol and LPS levels.

4.3. Ethanol-Producing Bacteria

Gut microorganisms directly cause liver damage either by means of microbe-associated molecular patterns (MAMPs) and PAMPs, such as LPS, or by means of the products of their metabolism, such as ethanol, short-chain fatty acids, and trimethylamine [66]. Proteobacteria, particularly Enterobacteriaceae, can ferment carbohydrates to ethanol. Significant correlations between the presence of ethanol-producing bacteria, blood ethanol levels, and liver inflammation have been demonstrated, and a positive correlation was found between increased abundance of Proteobacteria/Enterobacteriaceae/Escherichia and serum alcohol levels [33]. Under adequate conditions, the amount of ethanol produced can be remarkable [67]. Aside from conferring direct toxic effects to the liver, this overproduction activates hepatic ethanol metabolic pathways and increases liver oxidative stress [68]. Zhu et al. investigated gut microbiota (GM) composition and ethanol levels in the blood of NASH, obese, and healthy children [69]. The GM composition of NASH showed slight differences in the lineage, family, and genera of Proteobacteria, Enterobacteriaceae, and Escherichia coli compared to obese patients without liver disease. Among these microbiome changes was an increase in alcohol-producing bacteria, with significantly higher ethanol levels in NAFLD patients compared with those of obese and healthy children. In addition, an increase in ethanol concentration was particularly correlated with NASH. These results suggest that the production of ethanol by GM leads to hepatotoxicity that contributes to the onset of NAFLD and the progression to NASH [69]. Increased permeability, endogenous ethanol, and systemic endotoxin levels reflect some gut–liver axis dysfunction associated with obesity and its hepatic complications. In this regard, our group has recently demonstrated that the value of the lactulose/mannitol ratio is comparable to the grade of liver damage, significantly correlating with concentrations of ethanolemia and endotoxemia. Increased permeability was a risk factor for the development of steatosis [70].

5. Hepatic Factors Associated with Endotoxins

5.1. Enhanced Response to Endotoxin

Previous studies have shown that gut microbiota-derived endotoxins may be involved in the progression of NASH from simple fat deposition to steatohepatitis [31,38,39,40,61,62,71,72,73]. Despite these findings, the impact of increased endotoxinemia on the progression of NASH is controversial. It is still unclear whether serum endotoxin levels in NASH patients are significantly higher than those in control subjects and patients with simple fat deposition. Harte et al. reported that serum endotoxin levels were elevated in NAFLD patients compared to those in healthy controls [31]. In another human study, plasma IgG levels against endotoxin were increased in biopsy-proven human NASH patients and progressively increased with NASH grade [74]. These findings suggest a relationship between chronic endotoxin exposure and human NASH severity in which increased permeability drives endotoxemia, which in turn triggers inflammatory cytokine responses and IR [42]. However, in the study by Loguercio et al. [32,75], all NAFLD patients tested were free of endotoxinemia; however, the results were inconsistent. Currently, there is general agreement that mild portal endotoxemia can be detected in healthy subjects due to gut-derived bacterial endotoxins [40]. However, the levels of portal endotoxemia observed under healthy conditions do not usually cause liver dysfunction [76]. Furthermore, Imajo et al. reported that increased levels of leptin lead to overexpression of CD14 via activation of STAT3 signaling in Kupffer cells, resulting in a hepatic hyperinflammatory response to gut-derived low-dose bacterial endotoxin and progression from simple steatosis to steatohepatitis with associated liver inflammation and fibrosis [77]. Previous studies have also shown that a high-cholesterol diet increases the sensitivity of mice to LPS without affecting plasma levels of LPS. This further supports our hypothesis [78]. CD14 is an important regulatory factor in LPS-induced inflammation and enhances the LPS effects in Kupffer cells [79,80,81,82,83,84]. Furthermore, a previous report showed that promoter polymorphisms of CD14 are a risk factor for human NASH [85]. Therefore, increased expression of CD14 is closely related to the pathogenesis of NASH. Indeed, Imajo et al. showed that CD14 mRNA expression levels were much higher in NAFLD patients, including NAFL and NASH patients, than in control subjects [77]. Thus, hepatic CD14 may serve as an important factor in the development of NASH by enhancing hepatic inflammation against gut-derived bacterial endotoxin. We also investigated the leptin-dependent increase in hepatic CD14 expression using leptin-deficient ob/ob mice and leptin receptor-deficient db/db mice. Leptin and STAT3 signaling increased the responsiveness to gut-derived, low-dose bacterial endotoxin even in the healthy liver via an increase in CD14-positive Kupffer cells, regardless of the presence of steatosis. In humans, elevated serum leptin levels are generally associated with obesity, visceral fat accumulation, and fat deposition [86,87]. Thus, enhanced expression of hepatic CD14 by leptin may increase hepatic responsiveness to gut microbiota-derived endotoxins even at low levels, resulting in the progression from simple steatosis to NASH via STAT3 signaling. Moreover, Kessoku et al. demonstrated that resveratrol, a natural polyphenol, administration-mediated improvement of inflammation and fibrosis was due to inhibition of LPS reactivity controlled by CD14 expression in Kupffer cells. These findings suggest that resveratrol could be a candidate agent for the treatment of NASH [88].

5.2. Responses to Gut-Derived Ethanol

Under normal conditions, alcohol is constantly produced in the human body [88]. The intestinal microbiota is the major source of endogenous alcohol, as indicated by increased blood alcohol level after intake of alcohol-free food [89,90,91]. This endogenously produced alcohol is immediately and almost completely removed from portal blood by liver alcohol dehydrogenases (ADHs), catalases, and the microsomal ethanol-oxidizing system. When the action of ADH is inhibited, blood alcohol levels increase [89]. Production of ethanol in the gut is also reflected by the fact that the liver and the gastrointestinal tract exhibit the highest activity of ADHs [92]. Elevated breath alcohol levels are observed in obese mice. Here, the aberrant intestinal microbiota is the source for increased alcohol production, and neomycin treatment decreases alcohol concentration [93]. As patients with NASH are generally obese, and their liver histology is the same as that observed in alcoholic liver disease, it was hypothesized that NASH patients also have elevated blood alcohol levels [93]. The alcohol hypothesis of NASH could also explain the observation of increased gut permeability [40] and, possibly, elevated serum lipopolysaccharide levels in NASH patients [94], since alcohol is known to increase gut permeability [95]. The first evidence in support of this hypothesis was that the gene expression associated with all three major pathways for ethanol catabolism in the NASH liver is significantly elevated [70]. Recently, elevated blood ethanol concentration was observed in patients with NAFLD [96]. The observation of Volynets et al. [96] provides a link between blood alcohol and NAFLD. Zhu et al. further clarified that the blood ethanol concentration of obese patients without NASH is not elevated; however, obese patients with NASH exhibit significantly elevated blood ethanol levels [33].

6. NASH/NAFLD Treatment Focused on the Gut

6.1. Probiotics and Prebiotics

Probiotics are living microorganisms that regulate the intestinal flora and promote physical health. The most common probiotics on the market are Lactobacillus, Streptococci, and Bifidobacteria. Prebiotics are indigestible carbohydrates that stimulate the growth and activity of beneficial bacteria, especially Lactobacillus and Bifidobacteria. Lactulose is an example of a prebiotic that increases the number of Bifidobacteria, whereas fructooligosaccharides, including oligofructose, and inulin increase the abundance of Lactobacillus rhamnosus G and Bifidobacteria lactis Bb12 [97].
Influencing the intestinal flora using probiotics in mice with fatty liver reduced intestinal inflammation and improved epithelial barrier function [41,98], indicating that probiotics can be a new treatment for NAFLD in humans. Regarding clinical trials, Loguercio et al. [99,100] showed that probiotics reduced liver damage and improved liver function in NAFLD patients. However, subsequent pediatric meta-analyses emphasized that probiotic treatment of patients with NAFLD and nonalcoholic steatohepatitis is not recommended due to the lack of robust evidence from RCTs. Recently, a double-blind RCT showed that administration of Lactobacillus bulgaricus and Streptococcus thermophiles reduced hepatic transaminase levels in biopsy-proven adults with NAFLD [101,102]. Another double-blind RCT study showed that obese children with NAFLD treated with Lactobacillus GG for 8 weeks demonstrated significant decreases in ALT levels [103]. Prebiotics are indigestible food ingredients that have beneficial effects on the host by selectively stimulating the growth of selected gut microbiota and altering metabolic activity. The health benefits of prebiotics are transmitted to the host by three broad mechanisms, which include improved glucose regulation, altered lipid metabolism, and selective regulation of the gut microbiota [104]. Studies involving obese rats demonstrated that prebiotic fibers improved or normalized the intestinal microbiota dysbiosis by increasing the amount of Firmicutes and decreasing the amount of Bacteroidetes phylae [105].
These promising preliminary results strongly demonstrate that probiotics and prebiotics have great potential for the prevention and treatment of NASH. However, as pointed out in a recent meta-analysis, further clinical research is needed to clarify this unique, yet cost-effective, strategy [101]. Currently, substantial experimental evidence shows the beneficial effects of probiotics and prebiotics, which may be useful for designing future clinical trials.

6.2. Fecal Microbiota Transplantation

In addition to probiotic supplementation, transplantation of fecal microbiota has been shown to reduce HFD-induced steatohepatitis through modulation of the GM. In fact, fecal microbiota transplantation from lean donors to NASH patients is under investigation in humans (NCT02469272). Despite its routine use, it is difficult to consider in current clinical practice, but the results of the study help to better define the etiological mechanisms of NAFLD [4].

6.3. Anti-LPS Immunoglobulins

A recent promising NAFLD treatment is oral supplementation with IMM-124e, an IgG-rich bovine colostrum extract from cows immunized against LPS, which improved liver fat, insulin sensitivity [106], and immune-mediated colitis in animal models [107]. One study showed improved glycemic control in a small human trial [108]. Benefits seem to be due to reduction of liver exposure to GM LPS and consequent Kupffer cells activation.

6.4. Vitamin B6

The gut microbiota plays a chief role in vitamin production. Micronutrients, especially vitamins, play a crucial role in several metabolic reactions. Vitamin B6 is one of the vital micronutrients. Vitamin B6 (VitB6) is a generic name that includes six vitamers: pyridoxine (PN), pyridoxal (PL), pyridoxamine (PM), and their respective phosphate esters pyridoxine 5′-phosphate (PNP), pyridoxal 5′-phosphate (PLP), and pyridoxamine 5′-phosphate (PMP) [3]. VitB6 intake and hepatic steatosis are negatively correlated, and patients with NAFLD have diets low in VitB6 compared with healthy individuals [12,13]. In addition, patients with NAFLD have low levels of plasma VitB6, and VitB6 administration ameliorated hepatic lipid accumulation in mice model [14]. There are no reports on the efficacy of VitB6 administration in patients with NAFLD examined in clinical trials, but a study by Kobayashi and Kessoku et al. [109] was the first to demonstrate that 12 weeks of VitB6 administration ameliorates liver lipid deposition in patients with NAFLD. Although the mechanism by which VitB6 ameliorates hepatic lipid accumulation has not been fully elucidated, an experimental study reported that one potential mechanism could be linked to homocysteine (Hcy) catabolism. PLP, a physiologically active form of VitB6, functions as a coenzyme of cysteine-b-synthase (CBS) and cystathionine-g-lyase (CGL) [19]. Because CBS and CGL contribute to Hcy catabolism, inadequate amounts of VitB6 result in the accumulation of Hcy [110]; Hcy causes protein misfolding in the endoplasmic reticulum (ER), which leads to the ER stress response. In turn, ER stress induces the activation of the transcription factor sterol response element binding protein 1c, causing de novo lipogenesis [111]. Thus, based on this mechanism, VitB6 deficiency is thought to induce hepatic lipid accumulation. Glutathione is synthesized in cells from glutamic acid, cysteine, and glycine. Cysteine and glycine are generated from methionine and serine, respectively, and glutamic acid is synthesized from α-ketoglutarate, a metabolite of glucose. Glutathione has a long history in the treatment of chronic liver disease through intravenous injection; however, Honda et al. [112] demonstrated a therapeutic effect of glutathione through oral administration in patients with NAFLD. The primary outcome of this study was a change in ALT levels.

6.5. Vitamin D

Vitamin D deficiency also seems to play a role in NAFLD, but the underlying mechanisms are not well understood. Recently, the possible involvement of vitamin D in the dysregulation of the gut–liver axis is gradually becoming apparent. Indeed, optimal vitamin D levels are essential for maintaining the integrity of intestinal permeability through the upregulation of tight junction components and mucosal proteoglycans in the ileal epithelium, and through defensins and their converting enzymes (matrix metalloproteinase 7—MMP7) by certain intestinal mucosal paneth cells. The presence of vitamin D deficiency in the mouse HFD model exacerbates leaky gut, enterotoxemia, endotoxemia, systemic inflammation, and consequently IR and hepatic lipidosis [113]. Thus, supplementation with vitamin D has been recommended [114].

6.6. Constipation Drug Lubiprostone (LUB)

LUB is a bicyclic fatty acid derived from a prostone metabolite of prostaglandin E1. It is a type 2 chloride channel activator that causes an efflux of chloride into the gastrointestinal lumen, which ultimately promotes intestinal fluid secretion [115]. Thus, LUB is normally used for treating both chronic idiopathic constipation and irritable bowel syndrome with constipation. However, there are potentially additional actions of LUB on the intestinal mucosa. For example, some experimental studies reported that LUB prevented small bowel injury induced by nonsteroidal anti-inflammatory drugs (NSAIDs) in rats [116], ameliorated increases in intestinal permeability induced by a Western diet in an atherosclerotic mouse model [117], and maintained intestinal tight junction barrier function by activating the chloride channel in Caco-2 cell line [118]. Furthermore, some clinical trials demonstrated that LUB dramatically improved the intestinal permeability induced by NSAIDs in healthy volunteers [119]. Recently, a parallel three-arm, double-blind RCT was conducted using LUB as a new therapeutic target for the treatment of NAFLD, focused on intestinal permeability. A total of 150 Japanese NAFLD patients with constipation were treated with a placebo, 12 μg, or 24 μg of LUB for 12 weeks. In the LUB groups, the urinary lactulose/mannitol ratio, an index of intestinal permeability, was improved. Liver enzymes, liver fat, and blood endotoxin levels were significantly improved [120]. In particular, in the group of patients with improved intestinal permeability, a marked decrease in liver enzymes, liver fat content, and blood endotoxin concentration was observed (Figure 2). Drugs targeting intestinal permeability may be a promising new therapeutic approach to NAFLD.

7. Conclusions

The pathogenesis and progression of NAFLD/NASH are associated with an increased susceptibility to endotoxins, qualitative and quantitative abnormalities of intestinal bacteria, and an increase in intestinal permeability. Advances obtained in the understanding of the role of the gut–liver axis in NAFLD pathogenesis and the encouraging results already obtained by gut microbiota modulation via probiotic supplementation provide a promising and safe innovative mode of therapy. However, other extensive and long-term studies are needed to better define the best probiotic strains, their doses, timing, and duration of supplementation therapy. This will serve to individualize probiotic therapy with a patient-tailored approach for modulating intestinal permeability, endotoxemia, and treating liver disease Future studies should explore the pathogenesis of NAFLD, in particular, the mechanism involving intestinal permeability, to facilitate the development of novel therapeutic approaches targeting the gut–liver axis.

Author Contributions

Conceptualization, T.K. (Takaomi Kessoku) and A.N. (Atsushi Nakajima); writing—original draft preparation, T.K. (Takaomi Kessoku) and T.K. (Takashi Kobayashi); writing—review and editing, K.T. (Kosuke Tanaka), A.Y., K.T. (Kota Takahashi), M.I., A.O., Y.K., A.N. (Asako Nogami), Y.H., Y.O., S.K., K.I., T.H., K.H., M.Y., and S.S.; supervision, H.U. and K.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Yokohama City University Hospital. No external funding was used.

Data Availability Statement

Not applicable.

Conflicts of Interest

A.N. (Atsushi Nakajima) has grants and research support from Gilead, Mylan EPD, EA Pharma, Kowa, Taisho, and Biofermin. A.N. (Atsushi Nakajima) is a consulting adviser in Gilead, Boehringer Ingelheim, BMS, Kowa, Astellas, EA Pharma, Mylan EPD. The other authors declare no conflict interest.

References

  1. Loomba, R.; Sanyal, A.J. The global NAFLD epidemic. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 686–690. [Google Scholar] [CrossRef]
  2. 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] [PubMed] [Green Version]
  3. Clemente, M.G.; Mandato, C.; Poeta, M.; Vajro, P. Pediatric non-alcoholic fatty liver disease: Recent solutions, unresolved issues, and future research directions. World J. Gastroenterol. 2016, 22, 8078–8093. [Google Scholar] [CrossRef] [PubMed]
  4. Rotman, Y.; Sanyal, A.J. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut 2017, 66, 180–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  6. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  7. Monteiro, R.; Azevedo, I. Chronic inflammation in obesity and the metabolic syndrome. Mediat. Inflamm. 2010, 2010, 289645. [Google Scholar] [CrossRef]
  8. Wellen, K.E.; Hotamisligil, G.S. Inflammation, stress, and diabetes. J. Clin. Investig. 2005, 115, 1111–1119. [Google Scholar] [CrossRef] [Green Version]
  9. Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef]
  10. Racanelli, V.; Rehermann, B. The liver as an immunological organ. Hepatology 2006, 43 (Suppl. 1), S54–S62. [Google Scholar] [CrossRef]
  11. Akira, S.; Takeda, K.; Kaisho, T. Toll-like receptors: Critical proteins linking innate and acquired immunity. Nat. Immunol. 2001, 2, 675–680. [Google Scholar] [CrossRef]
  12. Mencin, A.; Kluwe, J.; Schwabe, R.F. Toll-like receptors as targets in chronic liver diseases. Gut 2009, 58, 704–720. [Google Scholar] [CrossRef]
  13. Szabo, G.; Bala, S. Alcoholic liver disease and the gut-liver axis. World J. Gastroenterol. 2010, 16, 1321–1329. [Google Scholar] [CrossRef]
  14. O’Neill, L.A.; Bowie, A.G. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353–364. [Google Scholar] [CrossRef] [PubMed]
  15. Kagan, J.C.; Medzhitov, R. Phosphoinositide-mediated adaptor recruitment controls Toll-like receptor signaling. Cell 2006, 125, 943–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Fitzgerald, K.A.; Chen, Z.J. Sorting out Toll signals. Cell 2006, 125, 834–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Seki, E.; Brenner, D.A. Toll-like receptors and adaptor molecules in liver disease: Update. Hepatology 2008, 48, 322–335. [Google Scholar] [CrossRef] [PubMed]
  18. Szabo, G.; Dolganiuc, A.; Mandrekar, P. Pattern recognition receptors: A contemporary view on liver diseases. Hepatology 2006, 44, 287–298. [Google Scholar] [CrossRef] [PubMed]
  19. Su, G.L.; Klein, R.D.; Aminlari, A.; Zhang, H.Y.; Steinstraesser, L.; Alarcon, W.H.; Remick, D.G.; Wang, S.C. Kupffer cell activation by lipopolysaccharide in rats: Role for lipopolysaccharide binding protein and toll-like receptor 4. Hepatology 2000, 31, 932–936. [Google Scholar] [CrossRef] [PubMed]
  20. Yin, M.; Wheeler, M.D.; Kono, H.; Bradford, B.U.; Gallucci, R.M.; Luster, M.I.; Thurman, R.G. essential role of tumor necrosis factor alpha in alcohol-induced liver injury in mice. Gastroenterology 1999, 117, 942–952. [Google Scholar] [CrossRef]
  21. Santucci, L.; Fiorucci, S.; Chiorean, M.; Brunori, P.M.; Di Matteo, F.M.; Sidoni, A.; Migliorati, G.; Morelli, A. Interleukin 10 reduces lethality and hepatic injury induced by lipopolysaccharide in galactosamine-sensitized mice. Gastroenterology 1996, 111, 736–744. [Google Scholar] [CrossRef] [PubMed]
  22. Dela Peña, A.; Leclercq, I.; Field, J.; George, J.; Jones, B.; Farrell, G. NF-KappaB activation, rather than TNF, mediates hepatic inflammation in a murine dietary model of steatohepatitis. Gastroenterology 2005, 129, 1663–1674. [Google Scholar] [CrossRef]
  23. Leclercq, I.A.; Farrell, G.C.; Sempoux, C.; dela Peña, A.; Horsmans, Y. Curcumin inhibits NF-KappaB activation and reduces the severity of experimental steatohepatitis in mice. J. Hepatol. 2004, 41, 926–934. [Google Scholar] [CrossRef] [PubMed]
  24. Koca, S.S.; Bahcecioglu, I.H.; Poyrazoglu, O.K.; Ozercan, I.H.; Sahin, K.; Ustundag, B. The treatment with antibody of TNF-alpha reduces the inflammation, necrosis and fibrosis in the non-alcoholic steatohepatitis induced by methionine- and choline-deficient diet. Inflammation 2008, 31, 91–98. [Google Scholar] [CrossRef]
  25. Tomita, K.; Tamiya, G.; Ando, S.; Ohsumi, K.; Chiyo, T.; Mizutani, A.; Kitamura, N.; Toda, K.; Kaneko, T.; Horie, Y.; et al. Tumour necrosis factor alpha signalling through activation of Kupffer cells plays an essential role in liver fibrosis of non-alcoholic steatohepatitis in mice. Gut 2006, 55, 415–424. [Google Scholar] [CrossRef] [PubMed]
  26. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef] [PubMed]
  27. Li, Z.; Soloski, M.J.; Diehl, A.M. Dietary factors alter hepatic innate immune system in mice with nonalcoholic fatty liver disease. Hepatology 2005, 42, 880–885. [Google Scholar] [CrossRef]
  28. Guebre-Xabier, M.; Yang, S.; Lin, H.Z.; Schwenk, R.; Krzych, U.; Diehl, A.M. Altered hepatic lymphocyte subpopulations in obesity-related murine fatty livers: Potential mechanism for sensitization to liver damage. Hepatology 2000, 31, 633–640. [Google Scholar] [CrossRef]
  29. Kremer, M.; Hines, I.N.; Milton, R.J.; Wheeler, M.D. Favored T helper 1 response in a mouse model of hepatosteatosis is associated with enhanced T cell-mediated hepatitis. Hepatology 2006, 44, 216–227. [Google Scholar] [CrossRef]
  30. Xu, J.; Gordon, J.I. Honor thy symbionts. Proc. Natl. Acad. Sci. USA 2003, 100, 10452–10459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Harte, A.L.; da Silva, N.F.; Creely, S.J.; McGee, K.C.; Billyard, T.; Youssef-Elabd, E.M.; Tripathi, G.; Ashour, E.; Abdalla, M.S.; Sharada, H.M.; et al. Elevated endotoxin levels in non-alcoholic fatty liver disease. J. Inflamm. 2010, 7, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pendyala, S.; Walker, J.M.; Holt, P.R. A high-fat diet is associated with endotoxemia that originates from the gut. Gastroenterology 2012, 142, 1100–1101.e2. [Google Scholar] [CrossRef] [Green Version]
  33. 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] [PubMed]
  34. Bjarnason, I.; Takeuchi, K.; Bjarnason, A.; Adler, S.N.; Teahon, K. The G.U.T. of gut. Scand. J. Gastroenterol. 2004, 39, 807–815. [Google Scholar] [CrossRef]
  35. Zeuzem, S. Gut-liver axis. Int. J. Colorectal Dis. 2000, 15, 59–82. [Google Scholar] [CrossRef] [PubMed]
  36. 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. Natl. Acad. Sci. USA 2006, 103, 12511–12516. [Google Scholar] [CrossRef] [Green Version]
  37. Solga, S.F.; Diehl, A.M. Non-alcoholic fatty liver disease: Lumen-liver interactions and possible role for probiotics. J. Hepatol. 2003, 38, 681–687. [Google Scholar] [CrossRef]
  38. Farhadi, A.; Gundlapalli, S.; Shaikh, M.; Frantzides, C.; Harrell, L.; Kwasny, M.M.; Keshavarzian, A. Susceptibility to gut leakiness: A possible mechanism for endotoxaemia in non-alcoholic steatohepatitis. Liver Int. 2008, 28, 1026–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Brun, P.; Castagliuolo, I.; Di Leo, V.; Buda, A.; Pinzani, M.; Palù, G.; Martines, D. Increased intestinal permeability in obese mice: New evidence in the pathogenesis of nonalcoholic steatohepatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G518–G525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Mascianà, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef]
  41. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
  42. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sharma, R.; Young, C.; Neu, J. Molecular modulation of intestinal epithelial barrier: Contribution of microbiota. J. Biomed. Biotechnol. 2010, 2010, 305879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ghoshal, S.; Witta, J.; Zhong, J.; de Villiers, W.; Eckhardt, E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J. Lipid Res. 2009, 50, 90–97. [Google Scholar] [CrossRef] [Green Version]
  45. Luther, J.; Garber, J.J.; Khalili, H.; Dave, M.; Bale, S.S.; Jindal, R.; Motola, D.L.; Luther, S.; Bohr, S.; Jeoung, S.W.; et al. Hepatic injury in nonalcoholic steatohepatitis contributes to altered intestinal permeability. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 222–232. [Google Scholar] [CrossRef] [Green Version]
  46. Nazim, M.; Stamp, G.; Hodgson, H.J. Non-alcoholic steatohepatitis associated with small intestinal diverticulosis and bacterial overgrowth. Hepato-gastroenterology 1989, 36, 349–351. [Google Scholar]
  47. Lichtman, S.N.; Sartor, R.B.; Keku, J.; Schwab, J.H. Hepatic inflammation in rats with experimental small intestinal bacterial overgrowth. Gastroenterology 1990, 98, 414–423. [Google Scholar] [CrossRef]
  48. Lichtman, S.N.; Keku, J.; Schwab, J.H.; Sartor, R.B. Hepatic injury associated with small bowel bacterial overgrowth in rats is prevented by metronidazole and tetracycline. Gastroenterology 1991, 100, 513–519. [Google Scholar] [CrossRef]
  49. Diehl, A.M.; Li, Z.P.; Lin, H.Z.; Yang, S.Q. Cytokines and the pathogenesis of non-alcoholic steatohepatitis. Gut 2005, 54, 303–306. [Google Scholar] [CrossRef] [Green Version]
  50. Farrell, G.C.; Larter, C.Z. Nonalcoholic fatty liver disease: From steatosis to cirrhosis. Hepatology 2006, 43 (Suppl. 1), S99–S112. [Google Scholar] [CrossRef] [PubMed]
  51. Shanab, A.A.; Scully, P.; Crosbie, O.; Buckley, M.; O’Mahony, L.; Shanahan, F.; Gazareen, S.; Murphy, E.; Quigley, E.M. 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. [Google Scholar] [CrossRef]
  52. Grover, M.; Kanazawa, M.; Palsson, O.S.; Chitkara, D.K.; Gangarosa, L.M.; Drossman, D.A.; Whitehead, W.E. Small intestinal bacterial overgrowth in irritable bowel syndrome: Association with colon motility, bowel symptoms, and psychological distress. Neurogastroenterol. Motil. 2008, 20, 998–1008. [Google Scholar] [CrossRef] [Green Version]
  53. Lin, H.C. Small intestinal bacterial overgrowth: A framework for understanding irritable bowel syndrome. JAMA 2004, 292, 852–858. [Google Scholar] [CrossRef]
  54. Lupascu, A.; Gabrielli, M.; Lauritano, E.C.; Scarpellini, E.; Santoliquido, A.; Cammarota, G.; Flore, R.; Tondi, P.; Pola, P.; Gasbarrini, G.; et al. Hydrogen glucose breath test to detect small intestinal bacterial overgrowth: A prevalence case–control study in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2005, 22, 1157–1160. [Google Scholar] [CrossRef] [PubMed]
  55. Pimentel, M.; Chow, E.J.; Lin, H.C. Eradication of small intestinal bacterial overgrowth reduces symptoms of irritable bowel syndrome. Am. J. Gastroenterol. 2000, 95, 3503–3506. [Google Scholar] [CrossRef]
  56. Pimentel, M.; Chow, E.J.; Lin, H.C. Normalization of lactulose breath testing correlates with symptom improvement in irritable bowel syndrome. a double-blind, randomized, placebo controlled study. Am. J. Gastroenterol. 2003, 98, 412–419. [Google Scholar] [PubMed]
  57. Pimentel, M.; Mayer, A.G.; Park, S.; Chow, E.J.; Hasan, A.; Kong, Y. Methane production during lactulose breath test is associated with gastrointestinal disease presentation. Dig. Dis. Sci. 2003, 48, 86–92. [Google Scholar] [CrossRef] [PubMed]
  58. Pimentel, M.; Wallace, D.; Hallegua, D.; Chow, E.; Kong, Y.; Park, S.; Lin, H.C. A link between irritable bowel syndrome and fibromyalgia may be related to findings on lactulose breath testing. Ann. Rheum. Dis. 2004, 63, 450–452. [Google Scholar] [CrossRef] [Green Version]
  59. Sabaté, J.M.; Jouët, P.; Harnois, F.; Mechler, C.; Msika, S.; Grossin, M.; Coffin, B. High prevalence of small intestinal bacterial overgrowth in patients with morbid obesity: A contributor to severe hepatic steatosis. Obes. Surg. 2008, 18, 371–377. [Google Scholar] [CrossRef]
  60. Scarpellini, E.; Giorgio, V.; Gabrielli, M.; Lauritano, E.C.; Pantanella, A.; Fundarò, C.; Gasbarrini, A. Prevalence of small intestinal bacterial overgrowth in children with irritable bowel syndrome: A case–control study. J. Pediatr. 2009, 155, 416–420. [Google Scholar] [CrossRef] [PubMed]
  61. 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] [PubMed] [Green Version]
  62. Wu, W.C.; Zhao, W.; Li, S. Small intestinal bacteria overgrowth decreases small intestinal motility in the NASH rats. World J. Gastroenterol. 2008, 14, 313–317. [Google Scholar] [CrossRef] [PubMed]
  63. Drenick, E.J.; Fisler, J.; Johnson, D. Hepatic steatosis after intestinal bypass. Prevention and reversal by metronidazole, irrespective of protein calorie malnutrition. Gastroenterology 1982, 82, 535–548. [Google Scholar] [CrossRef]
  64. Vanderhoof, J.A.; Tuma, D.J.; Antonson, D.L.; Sorrell, M.F. Effect of antibiotics in the prevention of jejunoileal bypass-induced liver dysfunction. Digestion 1982, 23, 9–15. [Google Scholar] [CrossRef] [PubMed]
  65. Bergheim, I.; Weber, S.; Vos, M.; Krämer, S.; Volynets, V.; Kaserouni, S.; McClain, C.J.; Bischoff, S.C. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin. J. Hepatol. 2008, 48, 983–992. [Google Scholar] [CrossRef]
  66. 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]
  67. Dawes, E.A.; Foster, S.M. The formation of ethanol in Escherichia coli. Biochim. Biophys. Acta 1956, 22, 253–265. [Google Scholar] [CrossRef]
  68. Baker, S.S.; Baker, R.D.; Liu, W.; Nowak, N.J.; Zhu, L. Role of alcohol metabolism in non-alcoholic steatohepatitis. PLoS ONE 2010, 5, e9570. [Google Scholar] [CrossRef] [Green Version]
  69. Zhu, L.; Baker, R.D.; Zhu, R.; Baker, S.S. Gut microbiota produce alcohol and contribute to NAFLD. Gut 2016, 65, 1232. [Google Scholar] [CrossRef]
  70. Guercio Nuzio, S.; Di Stasi, M.; Pierri, L.; Troisi, J.; Poeta, M.; Bisogno, A.; Belmonte, F.; Tripodi, M.; Di Salvio, D.; Massa, G.; et al. Multiple gut-liver axis abnormalities in children with obesity with and without hepatic involvement. Pediatr. Obes. 2017, 12, 446–452. [Google Scholar] [CrossRef]
  71. Zhao, L.F.; Jia, J.M.; Han, D.W. The role of enterogenous endotoxemia in the pathogenesis of non-alcoholic steatohepatitis. Zhonghua Gan Zang Bing Za Zhi Zhonghua Ganzangbing Zazhi Chin. J. Hepatol. 2004, 12, 632. [Google Scholar]
  72. Chitturi, S.; Farrell, G.C. Etiopathogenesis of nonalcoholic steatohepatitis. Semin. Liver Dis. 2001, 21, 27–41. [Google Scholar] [CrossRef]
  73. Kudo, H.; Takahara, T.; Yata, Y.; Kawai, K.; Zhang, W.; Sugiyama, T. Lipopolysaccharide triggered TNF-alpha-induced hepatocyte apoptosis in a murine non-alcoholic steatohepatitis model. J. Hepatol. 2009, 51, 168–175. [Google Scholar] [CrossRef]
  74. Verdam, F.J.; Rensen, S.S.; Driessen, A.; Greve, J.W.; Buurman, W.A. Novel evidence for chronic exposure to endotoxin in human nonalcoholic steatohepatitis. J. Clin. Gastroenterol. 2011, 45, 149–152. [Google Scholar] [CrossRef]
  75. Loguercio, C.; De Simone, T.; D’Auria, M.V.; de Sio, I.; Federico, A.; Tuccillo, C.; Abbatecola, A.M.; Del Vecchio, B.C.; Italian AISF Clinical Group. Non-alcoholic fatty liver disease: A multicentre clinical study by the Italian Association for the Study of the Liver. Dig. Liver Dis. 2004, 36, 398–405. [Google Scholar] [CrossRef]
  76. Ansell, J.; Widrich, W.; Johnson, W.; Fine, J. Endotoxin and bacteria in portal blood. Gastroenterology 1977, 73, 1190. [Google Scholar] [CrossRef]
  77. Imajo, K.; Fujita, K.; Yoneda, M.; Nozaki, Y.; Ogawa, Y.; Shinohara, Y.; Kato, S.; Mawatari, H.; Shibata, W.; Kitani, H.; et al. Hyperresponsivity to low-dose endotoxin during progression to nonalcoholic steatohepatitis is regulated by leptin-mediated signaling. Cell Metab. 2012, 16, 44–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Huang, H.; Liu, T.; Rose, J.L.; Stevens, R.L.; Hoyt, D.G. Sensitivity of mice to lipopolysaccharide is increased by a high saturated fat and cholesterol diet. J. Inflamm. 2007, 4, 22. [Google Scholar] [CrossRef] [Green Version]
  79. Roncon-Albuquerque, R., Jr.; Moreira-Rodrigues, M.; Faria, B.; Ferreira, A.P.; Cerqueira, C.; Lourenço, A.P.; Pestana, M.; von Hafe, P.; Leite-Moreira, A.F. Attenuation of the cardiovascular and metabolic complications of obesity in CD14 knockout mice. Life Sci. 2008, 83, 502–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Delude, R.L.; Savedra, R., Jr.; Zhao, H.; Thieringer, R.; Yamamoto, S.; Fenton, M.J.; Golenbock, D.T. CD14 enhances cellular responses to endotoxin without imparting ligand-specific recognition. Proc. Natl. Acad. Sci. USA 1995, 92, 9288–9292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Ferrero, E.; Jiao, D.; Tsuberi, B.Z.; Tesio, L.; Rong, G.W.; Haziot, A.; Goyert, S.M. Transgenic mice expressing human CD14 are hypersensitive to lipopolysaccharide. Proc. Natl. Acad. Sci. USA 1993, 90, 2380–2384. [Google Scholar] [CrossRef] [Green Version]
  82. Haziot, A.; Ferrero, E.; Lin, X.Y.; Stewart, C.L.; Goyert, S.M. CD14-deficient mice are exquisitely insensitive to the effects of LPS. Prog. Clin. Biol. Res. 1995, 392, 349–351. [Google Scholar]
  83. Su, G.L. Lipopolysaccharides in liver injury: Molecular mechanisms of Kupffer cell activation. Am. J. Physiol. Gastrointest. Liver Physiol. 2002, 283, G256–G265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wright, S.D.; Ramos, R.A.; Tobias, P.S.; Ulevitch, R.J.; Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef] [PubMed]
  85. Brun, P.; Castagliuolo, I.; Floreani, A.R.; Buda, A.; Blasone, L.; Palù, G.; Martines, D. Increased risk of NASH in patients carrying the C(−159)T polymorphism in the CD14 gene promoter region. Gut 2006, 55, 1212. [Google Scholar] [CrossRef] [Green Version]
  86. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996, 334, 292–295. [Google Scholar] [CrossRef]
  87. Halaas, J.L.; Gajiwala, K.S.; Maffei, M.; Cohen, S.L.; Chait, B.T.; Rabinowitz, D.; Lallone, R.L.; Burley, S.K.; Friedman, J.M. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 1995, 269, 543–546. [Google Scholar] [CrossRef] [PubMed]
  88. Kessoku, T.; Imajo, K.; Honda, Y.; Kato, T.; Ogawa, Y.; Tomeno, W.; Kato, S.; Mawatari, H.; Fujita, K.; Yoneda, M.; et al. Resveratrol ameliorates fibrosis and inflammation in a mouse model of nonalcoholic steatohepatitis. Sci. Rep. 2016, 6, 22251. [Google Scholar] [CrossRef] [Green Version]
  89. Sarkola, T.; Eriksson, C.J. Effect of 4-methylpyrazole on endogenous plasma ethanol and methanol levels in humans. Alcohol. Clin. Exp. Res. 2001, 25, 513–516. [Google Scholar] [CrossRef] [PubMed]
  90. Blomstrand, R. Observations of the formation of ethanol in the intestinal tract in man. Life Sci. II 1971, 10, 575–582. [Google Scholar] [CrossRef]
  91. Watanabe-Suzuki, K.; Seno, H.; Ishii, A.; Kumazawa, T.; Suzuki, O. Ultrasensitive method for determination of ethanol in whole blood by headspace capillary gas chromatography with cryogenic oven trapping. J. Chromatogr. B. Biomed. Sci. Appl. 1999, 727, 89–94. [Google Scholar] [CrossRef]
  92. Engeland, K.; Maret, W. Extrahepatic, differential expression of four classes of human alcohol dehydrogenase. Biol. Chem. Biophys. Res. Commun. 1993, 193, 47–53. [Google Scholar] [CrossRef]
  93. Cope, K.; Risby, T.; Diehl, A.M. Increased gastrointestinal ethanol production in obese mice: Implications for fatty liver disease pathogenesis. Gastroenterology 2000, 119, 1340–1347. [Google Scholar] [CrossRef]
  94. Ruiz, A.G.; Casafont, F.; Crespo, J.; Cayon, A.; Mayorga, M.; Estebanez, A.; Fernadez-Escalante, J.C.; Pons-Romero, F. Lipopolysaccharide-binding protein plasma levels and liver TNFalpha 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. [Google Scholar] [CrossRef] [PubMed]
  95. Rao, R.K.; Seth, A.; Sheth, P. Recent advances in alcoholic liver disease I. Role of intestinal permeability and endotoxemia in alcoholic liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 286, G881–G884. [Google Scholar] [CrossRef] [PubMed]
  96. Volynets, V.; Kuper, M.A.; Strahl, S.; Maier, I.B.; Spruss, A.; Wagnerberger, S.; Königsrainer, 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]
  97. Rafter, J.; Bennett, M.; Caderni, G.; Clune, Y.; Hughes, R.; Karlsson, P.C.; Klinder, A.; O’Riordan, M.; O’Sullivan, G.C.; Pool-Zobel, B.; et al. Dietary synbiotics reduce cancer risk factors in polypectomized and colon cancer patients. Am. J. Clin. Nutr. 2007, 85, 488–496. [Google Scholar] [CrossRef]
  98. Esposito, E.; Iacono, A.; Bianco, G.; Autore, G.; Cuzzocrea, S.; Vajro, P.; Canani, R.B.; Calignano, A.; Raso, G.M.; Meli, R. Probiotics reduce the inflammatory response induced by a high-fat diet in the liver of young rats. J. Nutr. 2009, 139, 905–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Loguercio, C.; De Simone, T.; Federico, A.; Terracciano, F.; Tuccillo, C.; Di Chicco, M.; Cartenì, M. Gut-liver axis: A new point of attack to treat chronic liver damage? Am. J. Gastroenterol. 2002, 97, 2144–2146. [Google Scholar] [CrossRef]
  100. Loguercio, C.; Federico, A.; Tuccillo, C.; Terracciano, F.; D’Auria, M.V.; De Simone, C.; Del Vecchio Blanco, C. Beneficial effects of a probiotic VSL #3 on parameters of liver dysfunction in chronic liver diseases. J. Clin. Gastroenterol. 2005, 39, 540–543. [Google Scholar]
  101. Socha, P.; Horvath, A.; Vajro, P.; Dziechciarz, P.; Dhawan, A.; Szajewska, H. Pharmacological interventions for nonalcoholic fatty liver disease in adults and in children: A systematic review. J. Pediatr. Gastroenterol. Nutr. 2009, 48, 587–596. [Google Scholar] [CrossRef] [PubMed]
  102. Aller, R.; De Luis, D.A.; Izaola, O.; Conde, R.; Gonzalez Sagrado, M.; Primo, D.; De La Fuente, B.; Gonzalez, J. Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver disease patients: A double blind randomized clinical trial. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 1090–1095. [Google Scholar] [PubMed]
  103. 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]
  104. Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B.; et al. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104 (Suppl. 2), S1–S63. [Google Scholar] [CrossRef] [Green Version]
  105. Parnell, J.A.; Reimer, R.A. Prebiotic fiber modulation of the gut microbiota improves risk factors for obesity and the metabolic syndrome. Gut Microbes 2012, 3, 29–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Adar, T.; Ben Ya’acov, A.; Lalazar, G.; Lichtenstein, Y.; Nahman, D.; Mizrahi, M.; Wong, V.; Muller, B.; Rawlin, G.; Ilan, Y. Oral administration of immunoglobulin G-enhanced colostrum alleviates insulin resistance and liver injury and is associated with alterations in natural killer T cells. Clin. Exp. Immunol. 2012, 167, 252–260. [Google Scholar] [CrossRef] [PubMed]
  107. Ben Ya’acov, A.; Lichtenstein, Y.; Zolotarov, L.; Ilan, Y. The gut microbiome as a target for regulatory T cell-based immunotherapy: Induction of regulatory lymphocytes by oral administration of anti-LPS enriched colostrum alleviates immune mediated colitis. BMC Gastroenterol. 2015, 15, 154. [Google Scholar]
  108. Mizrahi, M.; Shabat, Y.; Ben Ya’acov, A.; Lalazar, G.; Adar, T.; Wong, V.; Muller, B.; Rawlin, G.; Ilan, Y. Alleviation of insulin resistance and liver damage by oral administration of Imm124-E is mediated by increased Tregs and associated with increased serum GLP-1 and adiponectin: Results of a phase I/II clinical trial in NASH. J. Inflamm. Res. 2012, 5, 141–150. [Google Scholar]
  109. Kobayashi, T.; Kessoku, T.; Ozaki, A.; Iwaki, M.; Honda, Y.; Ogawa, Y.; Imajo, K.; Yoneda, M.; Saito, S.; Nakajima, A. Vitamin B6 efficacy in the treatment of nonalcoholic fatty liver disease: An open-label, single-arm, single-center trial. J. Clin. Biochem. Nutr. 2021, 68, 181–186. [Google Scholar] [CrossRef]
  110. Nijhout, H.F.; Gregory, J.F.; Fitzpatrick, C.; Cho, E.; Lamers, K.Y.; Ulrich, C.M.; Reed, M.C. A mathematical model gives insights into the effects of vitamin B-6 deficiency on 1-carbon and glutathione metabolism. J. Nutr. 2009, 139, 784–791. [Google Scholar] [CrossRef] [Green Version]
  111. Ai, Y.; Sun, Z.; Peng, C.; Liu, L.; Xiao, X.; Li, J. Homocysteine induces hepatic steatosis involving ER stress response in high methionine diet-fed mice. Nutrients 2017, 9, 346. [Google Scholar] [CrossRef] [Green Version]
  112. Honda, Y.; Kessoku, T.; Sumida, Y.; Kobayashi, T.; Kato, T.; Ogawa, Y.; Tomeno, W.; Imajo, K.; Fujita, K.; Yoneda, M.; et al. Efficacy of glutathione for the treatment of nonalcoholic fatty liver disease: An open-label, single-arm, multicenter, pilot study. BMC Gastroenterol. 2017, 17, 96. [Google Scholar] [CrossRef]
  113. Su, D.; Nie, Y.; Zhu, A.; Chen, Z.; Wu, P.; Zhang, L.; Luo, M.; Sun, Q.; Cai, L.; Lai, Y.; et al. Vitamin D signaling through induction of paneth cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models. Front. Physiol. 2016, 7, 498. [Google Scholar] [CrossRef]
  114. Luger, M.; Kruschitz, R.; Kienbacher, C.; Traussnigg, S.; Langer, F.B.; Schindler, K.; Würger, T.; Wrba, F.; Trauner, M.; Prager, G.; et al. Prevalence of liver fibrosis and its association with non-invasive fibrosis and metabolic markers in morbidly obese patients with vitamin D deficiency. Obes. Surg. 2016, 26, 2425–2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Fei, G.; Raehal, K.; Liu, S.; Qu, M.H.; Sun, X.; Wang, G.D.; Wang, X.Y.; Xia, Y.; Schmid, C.L.; Bohn, L.M.; et al. Lubiprostone reverses the inhibitory action of morphine on intestinal secretion in guinea pig and mouse. J. Pharmacol. Exp. Ther. 2010, 334, 333–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hayashi, S.; Kurata, N.; Yamaguchi, A.; Amagase, K.; Takeuchi, K. Lubiprostone prevents nonsteroidal anti-inflammatory drug-induced small intestinal damage by suppressing the expression of inflammatory mediators via EP4 receptors. J. Pharmacol. Exp. Ther. 2014, 349, 470–479. [Google Scholar] [CrossRef] [Green Version]
  117. Arakawa, K.; Ishigami, T.; Nakai-Sugiyama, M.; Chen, L.; Doi, H.; Kino, T.; Minegishi, S.; Saigoh-Teranaka, S.; Sasaki-Nakashima, R.; Hibi, K.; et al. Lubiprostone as a potential therapeutic agent to improve intestinal permeability and prevent the development of atherosclerosis in apolipoprotein E-deficient mice. PLoS ONE 2019, 14, e0218096. [Google Scholar] [CrossRef] [PubMed]
  118. Nighot, P.K.; Leung, L.; Ma, T.Y. Chloride channel ClC-2 enhances intestinal epithelial tight junction barrier function via regulation of caveolin-1 and caveolar trafficking of occludin. Exp. Cell Res. 2017, 352, 113–122. [Google Scholar] [CrossRef] [Green Version]
  119. Kato, T.; Honda, Y.; Kurita, Y.; Iwasaki, A.; Sato, T.; Kessoku, T.; Uchiyama, S.; Ogawa, Y.; Ohkubo, H.; Higurashi, T.; et al. Lubiprostone improves intestinal permeability in humans, a novel therapy for the leaky gut: A prospective randomized pilot study in healthy volunteers. PLoS ONE 2017, 12, e0175626. [Google Scholar] [CrossRef] [Green Version]
  120. Kessoku, T.; Imajo, K.; Kobayashi, T.; Ozaki, A.; Iwaki, M.; Honda, Y.; Kato, T.; Ogawa, Y.; Tomeno, W.; Kato, S.; et al. Lubiprostone in patients with non-alcoholic fatty liver disease: A randomised, double-blind, placebo-controlled, phase 2a trial. Lancet Gastroenterol. Hepatol. 2020, 5, 996–1007. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of NAFLD progression promoted by intestinal and hepatic side factors. SIBO; small intestinal bacterial overgrowth.
Figure 1. Mechanisms of NAFLD progression promoted by intestinal and hepatic side factors. SIBO; small intestinal bacterial overgrowth.
Ijms 22 08161 g001
Figure 2. Therapeutic effects of lubiprostone on NAFLD via targeting the intestinal barrier. LUB, lubiprostone; NAFLD, nonalcoholic fatty liver disease; SIBO, small intestinal bacterial overgrowth.
Figure 2. Therapeutic effects of lubiprostone on NAFLD via targeting the intestinal barrier. LUB, lubiprostone; NAFLD, nonalcoholic fatty liver disease; SIBO, small intestinal bacterial overgrowth.
Ijms 22 08161 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kessoku, T.; Kobayashi, T.; Tanaka, K.; Yamamoto, A.; Takahashi, K.; Iwaki, M.; Ozaki, A.; Kasai, Y.; Nogami, A.; Honda, Y.; et al. The Role of Leaky Gut in Nonalcoholic Fatty Liver Disease: A Novel Therapeutic Target. Int. J. Mol. Sci. 2021, 22, 8161. https://doi.org/10.3390/ijms22158161

AMA Style

Kessoku T, Kobayashi T, Tanaka K, Yamamoto A, Takahashi K, Iwaki M, Ozaki A, Kasai Y, Nogami A, Honda Y, et al. The Role of Leaky Gut in Nonalcoholic Fatty Liver Disease: A Novel Therapeutic Target. International Journal of Molecular Sciences. 2021; 22(15):8161. https://doi.org/10.3390/ijms22158161

Chicago/Turabian Style

Kessoku, Takaomi, Takashi Kobayashi, Kosuke Tanaka, Atsushi Yamamoto, Kota Takahashi, Michihiro Iwaki, Anna Ozaki, Yuki Kasai, Asako Nogami, Yasushi Honda, and et al. 2021. "The Role of Leaky Gut in Nonalcoholic Fatty Liver Disease: A Novel Therapeutic Target" International Journal of Molecular Sciences 22, no. 15: 8161. https://doi.org/10.3390/ijms22158161

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