Review Article
Gut-liver axis and fibrosis in nonalcoholic fatty liver disease: An input for novel therapies

https://doi.org/10.1016/j.dld.2012.11.010Get rights and content

Abstract

Non-alcoholic fatty liver disease is a multifactorial condition, ranging from simple steatosis to non-alcoholic steatohepatitis with or without fibrosis. In non-alcoholic fatty liver disease, alteration of gut microbiota and increased intestinal permeability increase exposure of the liver to gut-derived bacterial products: lipopolysaccharides and unmethylated CpG DNA. These products stimulate innate immune receptors, namely Toll-like receptors, which activate signalling pathways involved in liver inflammation and fibrogenesis. Currently, there are several studies on the involvement of lipopolysaccharide-activated Toll-like receptor 4 signalling in non-alcoholic fatty liver disease pathogenesis. There has been widespread interest in the study of the involvement of resident hepatic stellate cells and Kupffer cells activation in liver fibrogenesis upon TLR4 stimulation. Although the best evidence to support a role for gut microbiota in non-alcoholic fatty liver disease-induced fibrosis comes largely from animal models, data from human studies are accumulating and could lead to new therapeutic approaches. Therapeutic modulation of gut microflora may be an alternative strategy to develop an anti-fibrotic therapy.

In this review, we discuss the relevant role of gut-liver axis in non-alcoholic liver disease-associated liver fibrosis and discuss the evidence on novel anti-fibrotic therapeutic approaches.

Introduction

During the last decades non-alcoholic fatty liver disease (NAFLD) has become one of the most common forms of chronic liver disease [1]. It includes a wide spectrum of pathological liver conditions, ranging from simple hepatic fat accumulation to non-alcoholic steatohepatitis (NASH) with or without fibrosis, which can eventually progress to cirrhosis and hepatocellular carcinoma [2].

Although the complexity and timing of the pathophysiological events in NAFLD remain unclear, the “multiple-hit hypothesis” is currently the most recognized theory to explain disease development and progression. According to this theory, various genetic and environmental factors play crucial roles in the multifactorial pathogenesis of NAFLD [3]. Several factors such as genetic background, epigenetic mechanisms, environmental factors, high caloric intake and low levels of physical activity, seem to promote lipid accumulation in the liver and insulin resistance, which may lead to simple steatosis [4]. In this early stage, other triggers (i.e. lipotoxicity and gut-liver axis) may contribute to the systemic and metabolic profile that characterizes patients with steatosis. Furthermore, either adipocytokines or inflammatory signals released by adipose tissue in response to an increased intestinal permeability and consequent endotoxemia may promote oxidative stress in the liver, inducing progression to NASH [5]. During NASH progression, the described compound setting activates molecular mechanisms involved in fibrogenesis. Fibrogenesis is a wound healing response to liver injury in which liver resident specialized cells, the hepatic stellate cells (HSCs), after a pro-proliferative induction and an epithelial–mesenchymal transition (EMT), become activated and induce an excessive extracellular matrix (ECM) deposition that characterizes fibrosis [6]. In particular, HSCs acquire a myofibroblastic phenotype and produce collagens that represent the main scaffold of a fibrotic lesion [7].

So far, a number of studies have shown that the gut-liver axis is involved, not only in the pathogenesis of NAFLD, but also in the development of liver fibrosis in various ways, some unexpected [8], [9], [10]. Knowledge of interaction networks, metabolic or molecular, that link gut-liver axis to fibrosis may represent a launch platform for novel therapeutic approaches.

This review focuses on the role of gut-liver axis in NAFLD-associated liver fibrosis and on its potential as an input for new, targeted therapies.

Section snippets

Gut microbiome composition and connection with NAFLD

The human body functions in close relationship with bacteria that predominantly inhabit the gastrointestinal tract to produce essential amino acids, vitamins and digesting plant polysaccharides [11]. The majority of healthy gut microbiota comprises Bacteroidetes and Firmicutes; their ratio has been found altered in human and mice obesity [12]. Many studies have explored the possible causes of metabolic syndrome occurrence in its co-morbidities, including NAFLD [13], [14], [15].

In human studies,

Molecular signalling of gut-derived products

Microbial products such as lipopolysaccharide (LPS), lipopeptides, DNA and RNA have potentially hepatotoxic effects as they are potent inducers of inflammation [8], [34]. Proinflammatory actions of these microbial compounds are mediated through a class of specific pattern recognition receptors, named Toll-like receptors (TLRs). TLRs are evolutionary conserved type I transmembrane glycoproteins containing two domains: the extracellular ligand-binding domain, composed of several leucine-rich

Gut microbiota signalling networks in NAFLD

The liver constantly communicates with gut-derived products. Various studies sustain the idea that gut microflora may play a pivotal role in the pathogenesis of NAFLD through LPS-TLR4 signalling [49], [50]. Many studies in animals and humans suggest the involvement of gut microbiota in the development of obesity, insulin resistance and in risk factors associated with NAFLD pathogenesis [51], [52].

In genetically obese rats and mice, an increased hepatic susceptibility to endotoxin-mediated

Gut-mediated effect on hepatic proinflammatory cytokines

In liver inflammation, Kupffer cells are the primary cells that respond to LPSs in order to produce inflammatory cytokines, chemokines and reactive oxygen species (ROS) [71], [72]. In genetically obese mice, the administration of LPS induces changes in Kupffer cells function and increases liver parenchymal sensitivity to TNF-α [53]. TNF-α, which is the most important of LPS-TLR4 induced cytokines in these cells, is recognized as a mediator of hepatotoxicity, inflammation and NASH development in

Gut-mediated liver fibrosis

Hepatic fibrosis is a wound-healing response triggered primarily by the development of inflammation in response to parenchymal injury. Hepatic inflammation promotes disease progression by activating HSCs, which contribute to the formation of fibrotic scar tissue that replaces normal liver parenchyma [80]. During fibrogenesis, the liver has a remarkable regenerative capacity that leads to a recovery of its architecture in the course of acute injury. If fibrogenic damage proceeds, the liver

Gut-liver axis as target for anti-fibrotic therapy

There are currently no therapies for liver fibrosis. Overall, the data on microbiota involved in the pathogenesis of NAFLD and fibrosis sustain the hypothesis that manipulation of gut microbiota may counteract the effects of unbalanced diets on the liver and can provide new therapeutic options for NAFLD-induced fibrosis.

Probiotics have been proposed as a treatment option because their modulating effect on the gut flora could influence the gut-liver axis [98]. The use of probiotics had

Conclusion

The discovery that fibrosis in NAFLD is a dynamic process that may predispose subjects to end-stage liver disease (cirrhosis and hepatocellular carcinoma) has encouraged efforts to uncover the molecular mechanism leading to fibrogenic liver damage in order to expedite new therapeutic strategies. Several studies in the past decades have suggested the crucial role of gut-derived bacterial products in the pathogenesis of NASH-associated features including steatosis, inflammation and fibrosis.

Conflicts of interest

There is no conflict of interest to disclose for all authors.

Acknowledgement

Otilia Frasinariu was supported by research project grant from the University of Medicine and Pharmacy “Grigore T. Popa” Iasi, beneficiary of the Financing Contract POSDRU/88/1.5/S/58965 for the project” Doctoral Scholarships for increasing competitiveness in the medical and pharmaceutical field”.

References (109)

  • H.M. Kim et al.

    Crystal structure of the TLR4-MD-2 complex with bound endotoxin antagonist Eritoran

    Cell

    (2007)
  • I. Bergheim et al.

    Antibiotics protect against fructose induced hepatic lipid accumulation in mice: role of endotoxin

    Journal of Hepatology

    (2008)
  • C.A. Rivera et al.

    Toll-like receptor-4 signalling and Kupffer cells play pivotal roles in the pathogenesis of non-alcoholic steatohepatitis

    Journal of Hepatology

    (2007)
  • S. Thuy et al.

    Nonalcoholic fatty liver disease in humans is associated with increased plasma endotoxin and plasminogen activator inhibitor 1 concentrations and with fructose intake

    Journal of Nutrition

    (2008)
  • O. Takeuchi et al.

    Pattern recognition receptors and inflammation

    Cell

    (2010)
  • E. Gäbele et al.

    Role of TLR9 in hepatic stellate cells and experimental liver fibrosis

    Biochemical and Biophysical Research Communications

    (2008)
  • M. Martin-Armas et al.

    Toll-like receptor 9 (TLR9) is present in murine liver sinusoidal endothelial cells (LSECs) and mediates the effect of CpG-oligonucleotides

    Journal of Hepatology

    (2006)
  • G. Baffy

    Kupffer cells in non-alcoholic fatty liver disease: the emerging view

    Journal of Hepatology

    (2009)
  • E. Gäbele et al.

    DSS induced colitis increases portal LPS levels and enhances hepatic inflammation and fibrogenesis in experimental NASH

    Journal of Hepatology

    (2011)
  • U.E. Lee et al.

    Mechanisms of hepatic fibrogenesis

    Best Practice and Research. Clinical Gastroenterology

    (2011)
  • R.S. Lin et al.

    Endotoxemia in patients with chronic liver diseases: relationship to severity of liver diseases, presence of esophageal varices, and hyperdynamic circulation

    Journal of Hepatology

    (1995)
  • I. Grinko et al.

    Experimental biliary fibrosis correlates with increased numbers of fat-storing and Kupffer cells, and portal endotoxemia

    Journal of Hepatology

    (1995)
  • K.A. Padgett et al.

    Primary biliary cirrhosis is associated with altered hepatic microRNA expression

    Journal of Autoimmunity

    (2009)
  • Y. Sekiya et al.

    Suppression of hepatic stellate cell activation by microRNA-29b

    Biochemical and Biophysical Research Communications

    (2011)
  • A. Iacono et al.

    Probiotics as an emerging therapeutic strategy to treat NAFLD: focus on molecular and biochemical mechanisms

    Journal of Nutritional Biochemistry

    (2011)
  • Z. Li et al.

    Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease

    Hepatology

    (2003)
  • X. Ma et al.

    Probiotics improve high fat diet-induced hepatic steatosis and insulin resistance by increasing hepatic NKT cells

    Journal of Hepatology

    (2008)
  • C.P. Day

    Non-alcoholic fatty liver disease: a massive problem

    Clinical Medicine

    (2011)
  • E.M. Brunt

    Pathology of nonalcoholic fatty liver disease

    Nature Reviews Gastroenterology and Hepatology

    (2010)
  • J.C. Cohen et al.

    Human fatty liver disease: old questions and new insights

    Science

    (2011)
  • C.P. Day

    Genetic and environmental susceptibility to non-alcoholic fatty liver disease

    Digestive Diseases

    (2010)
  • H. Tilg et al.

    Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis

    Hepatology

    (2010)
  • M. Pinzani et al.

    Update on the pathophysiology of liver fibrosis

    Expert Review of Gastroenterology and Hepatology

    (2010)
  • E. Seki et al.

    Role of innate immunity and the microbiota in liver fibrossis: crosstalk between the liver and the gut

    Journal of Physiology

    (2012)
  • E. Seki et al.

    TLR4 enhances TGF-beta signalling and hepatic fibrosis

    Nature Medicine

    (2007)
  • K. Miura et al.

    Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice

    Gastroenterology

    (2010)
  • S.R. Gill et al.

    Metagenomic analysis of the human distal gut microbiome

    Science

    (2006)
  • P.B. Eckburg et al.

    Diversity of the human intestinal microbial flora

    Science

    (2005)
  • D.A. Sandoval et al.

    Medicine. The microbes made me eat it

    Science

    (2010)
  • H. Xu et al.

    Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

    Journal of Clinical Investigation

    (2003)
  • Kasturiratne A, Weerasinghe S, Dassanayake AS, et al. Influence of non-alcoholic fatty liver disease on the development...
  • P.D. Cani et al.

    Metabolic endotoxemia initiates obesity and insulin resistance

    Diabetes

    (2007)
  • T.A. Larkin et al.

    Dietary combination of soy with a probiotic or prebiotic food significantly reduces total and LDL cholesterol in mildly hypercholesterolaemic subjects

    European Journal of Clinical Nutrition

    (2009)
  • G.W. Tannock

    A special fondness for lactobacilli

    Applied and Environment Microbiology

    (2004)
  • G. Musso et al.

    Gut microbiota as a regulator of energy homeostasis and ectopic fat deposition: mechanisms and implications for metabolic disorders

    Current Opinion in Lipidology

    (2010)
  • J. Liu et al.

    Comparison of the gut microbe profiles and numbers between patients with liver cirrhosis and healthy individuals

    Current Microbiology

    (2012)
  • J. Henao-Mejia et al.

    Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity

    Nature

    (2012)
  • T. Li et al.

    Bile Acid signaling in liver metabolism and diseases

    Journal of Lipids

    (2012)
  • N. Tanaka et al.

    Disruption of phospholipid and bile acid homeostasis in mice with nonalcoholic steatohepatitis

    Hepatology

    (2012)
  • Duboc H, Rajca S, Rainteau D, et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory...
  • Cited by (81)

    • Rifaximin and lubiprostone mitigate liver fibrosis development by repairing gut barrier function in diet–induced rat steatohepatitis

      2022, Digestive and Liver Disease
      Citation Excerpt :

      In particular, LPS-mediated toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling is involved in the activation of Kupffer cells and HSCs resulting in the development of hepatic inflammation and fibrosis during NASH progression [8,9]. Therefore, a potential therapeutic strategy against NASH fibrosis involves maintaining the gut barrier function and blunting the transfer of LPS into the liver [10]. Rifaximin is a poorly absorbed antibiotic widely applied for prevention of recurrent hepatic encephalopathy in the patients with cirrhosis [11,12].

    • Translational insight into prothrombotic state and hypercoagulation in nonalcoholic fatty liver disease

      2021, Thrombosis Research
      Citation Excerpt :

      The consequent accumulation of lipotoxic metabolites and the oxidative disposal of triglycerides (involving lysosomes and β-oxidation) results in hepatic damage and immune-mediated hepatocellular injury that promotes the development of NASH [17]. Immune-mediated response in NAFLD involves not only adipocytokines but also an increased microbiota-dependent permeability of the gut and the translocation of bacterial endotoxins, which activate the pro-inflammatory cascade of toll-like receptor 4 (TLR4) in the liver [18]. The persistent inflammatory activation of the liver-resident cells, as well as the necrotic and apoptotic pathways, promote the activation of hepatic stellate cells and pro-fibrogenic signals towards a fibrotic phenotype [19].

    View all citing articles on Scopus
    View full text