Elsevier

Autoimmunity Reviews

Volume 14, Issue 3, March 2015, Pages 183-191
Autoimmunity Reviews

Review
The intestinal microbiota and microenvironment in liver,☆☆

https://doi.org/10.1016/j.autrev.2014.10.013Get rights and content

Abstract

The intestinal microbiome plays a significant role in the development of autoimmune diseases, in particular, inflammatory bowel diseases. But the interplay between the intestinal tract and the liver may explain the increased association with autoimmune liver diseases and inflammatory bowel diseases. The gut–liver axis involves multiple inflammatory cell types and cytokines, chemokines and other molecules which lead to the destruction of normal liver architecture. Triggers for the initiation of these events are unclear, but appear to include multiple environmental factors, including pathogenic or even commensal microbial agents. The variation in the gut microbiome has been cited as a major factor in the pathogenesis of autoimmune liver disease and even other autoimmune diseases. The unique positioning of the liver at the juncture of the peripheral circulation and the portal circulation augments the interaction between naïve T cells and other hepatic cells and leads to the disruption in the development of tolerance to commensal bacteria and other environmental agents. Finally, the innate immune system and in particular toll-like receptors play a significant role in the pathogenesis of autoimmune liver disease.

Introduction

The liver and the gastrointestinal tract are intimately connected in the context of metabolic activity and immune responses, primarily resulting from their close anatomical and physiological relationship [1]. The liver has a dual blood supply. A quarter of the blood supply is derived from the systemic circulation which reaches the liver through the hepatic artery. The other three quarters are gut derived nutrient-rich blood that enters the liver through the portal vein. The term “gut–liver axis” has been coined to reflect the immunological phenomenon linking the two in health and disease [2].

Microbes exist in the gut environment and play a significant role in digestion, and are a part of the mucosal immune system that helps shape our ability to distinguish safe and danger signals [3], [4]. Since the liver receives blood from both systemic circulation and the intestines, microbes in the intestines may also affect the immune environment in the liver. Portal venous blood returning from the intestines contains the products of digestion, along with antigens and microbial products that originate from the bacteria in the small and large intestines [5]. The exposure of liver cells to antigens, and to microbial products derived from the intestinal bacteria, results in a distinctive local immune environment that modulates immune tolerance in the liver [6], [7], [8].

To establish this tolerogenic environment, hepatic immune cells including Kupffer cells, natural killer cells, dendritic cells and lymphocytes, together with other nonparenchymal cells including endothelial cells and stellate cells orchestrate a controlled and organized response to these potentially highly inflammatory factors from the intestines [9], [10]. However, this tolerance is quite metastable and can be reversed by the right combination of signals, resulting in active local immunity [5]. For example, changes in the composition of the microbiome or alterations in gut permeability can promote translocation of microbes into the portal circulation that delivers blood directly to the liver [2]. These gut-derived microbial components represent danger signals for the host cells in liver, activate the inflammatory cascade in immune cells and modulate the function of liver parenchymal cells [9].

There is a growing body of evidence showing that the intestinal microbiome possesses critical functions in liver physiology and metabolic homeostasis [11]. Gut-derived bacterial products aggravate hepatic fibrosis [12], [13], whereas, gut sterilization helps prevent hepatic fibrosis after bile-duct ligation [12]. Moreover, disruption of the intestinal mucosal barrier facilitates bacterial translocation and promotes the progression of liver fibrosis [13]. Prior studies also suggested that the gut microbiota may affect the progression of non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) [14], [15]. This dysbiosis in the digestive tract results in the exacerbation of hepatic steatosis and obesity [16].

The relationship between intestinal homeostasis and autoimmune liver diseases is summarized in this review by discussing the potential involvement of intestinal homeostasis in the pathogenesis of autoimmune liver diseases [17], [18], [19], the understanding of which will ultimately steer the development of novel clinical treatments.

Section snippets

Gut microbiota participates in the initiation and maintenance of autoimmune liver diseases by modulating the innate immune system

As the first line of defense against pathogens, the innate immune system mediates interactions between the hosts and their intestinal microflora [20]. Pattern recognition receptors widely expressed on innate immune cells and parenchymal cell are essential for the recognition and clearance of commensal and pathogenic microflora [21]. The conflict between the innate immune system and gut microflora is not only necessary to repel the invasion of pathogenic microorganisms, but also critical to

Molecular mimicry and production of autoantibodies

Serologically, PBC is characterized by the presence of anti-mitochondrial antibodies (AMAs) in 90–95% of patients [56]. The presence of autoantibodies may precede the occurrence of symptoms by many years [57]. AMAs react with mitochondrial antigens of the 2-oxoacid dehydrogenase complex (2-OADC), including an epitope on the E2 subunit of the pyruvate dehydrogenase enzyme complex (PDC-E2) and the E2 subunit of functionally related 2-oxo-acid dehydrogenase complexes, branched chain 2-oxo-acid

The association of intestinal status with inflammatory cytokines in the context of autoimmune liver diseases

Tumor necrosis factor (TNF), a major factor in the immune response to bacterial infection, contributes to the pathogenesis of several autoimmune diseases, including rheumatoid arthritis (RA) and Crohn's disease [93], [94], [95], [96]. Proven as the most efficient therapy for RA thus far, TNF antagonists have attracted considerable attention in the treatment of autoimmune diseases [97]. Likewise, circulating levels of TNF was most significantly correlated with hepatobiliary injury in autoimmune

Conclusion

Autoimmune liver diseases are complex diseases arising from the interaction of both environmental factors and genetic background. In the last few years, the incidence of autoimmune liver diseases has been increasing [135]. The reasons for this observation are unclear, but may reflect a change in our lifestyles and environments, which may include changes in the microbiome to which we are exposed. Since the immune-modulating capabilities of microorganisms in the gut have become a significant

Take-home messages

  • Autoimmune liver diseases are complex diseases arising from the interaction of both environmental factors and genetic background.

  • Gut microbiota participates in the initiation and maintenance of autoimmune liver diseases by modulating the innate immune system.

  • Multiple cell types and cytokine molecules may be involved in the pathogenesis of autoimmune liver disease.

  • The gut–liver axis allows greater interaction between the peripheral circulation and the liver, leading to an association between

References (143)

  • A. Karrar et al.

    Biliary epithelial cell antibodies link adaptive and innate immune responses in primary sclerosing cholangitis

    Gastroenterology

    (2007)
  • K. Harada et al.

    Lipopolysaccharide activates nuclear factor-kappaB through toll-like receptors and related molecules in cultured biliary epithelial cells

    Lab Invest

    (2003)
  • K. Harada et al.

    Molecular identification of bacterial 16S ribosomal RNA gene in liver tissue of primary biliary cirrhosis: is Propionibacterium acnes involved in granuloma formation?

    Hepatology

    (2001)
  • A.P. Wang et al.

    Hepatic expression of toll-like receptor 4 in primary biliary cirrhosis

    J Autoimmun

    (2005)
  • Y.H. Chuang et al.

    Increased killing activity and decreased cytokine production in NK cells in patients with primary biliary cirrhosis

    J Autoimmun

    (2006)
  • G.M. Barton et al.

    Control of adaptive immune responses by Toll-like receptors

    Curr Opin Immunol

    (2002)
  • A. Geremia et al.

    Innate and adaptive immunity in inflammatory bowel disease

    Autoimmun Rev

    (2014)
  • R. Singh et al.

    Status of bacterial colonization, Toll-like receptor expression and nuclear factor-kappa B activation in normal and diseased human livers

    Clin Immunol

    (2011)
  • Y. Honda et al.

    Altered expression of TLR homolog RP105 on monocytes hypersensitive to LPS in patients with primary biliary cirrhosis

    J Hepatol

    (2007)
  • C.L. Bowlus et al.

    The diagnosis of primary biliary cirrhosis

    Autoimmun Rev

    (2014)
  • J.A. Talwalkar et al.

    Natural history of pruritus in primary biliary cirrhosis

    Clin Gastroenterol Hepatol

    (2003)
  • H. Kita et al.

    Analysis of TCR antagonism and molecular mimicry of an HLA-A0201-restricted CTL epitope in primary biliary cirrhosis

    Hepatology

    (2002)
  • Y. Saeki et al.

    Infection–immunity liaison: pathogen-driven autoimmune-mimicry (PDAIM)

    Autoimmun Rev

    (2014)
  • D. Rigante et al.

    The cryptic interplay between systemic lupus erythematosus and infections

    Autoimmun Rev

    (2014)
  • K. Kikuchi et al.

    Bacterial CpG induces hyper-IgM production in CD27(+) memory B cells in primary biliary cirrhosis

    Gastroenterology

    (2005)
  • Y. Moritoki et al.

    B cells and autoimmune liver diseases

    Autoimmun Rev

    (2006)
  • M.W. Laass et al.

    Diagnosis and classification of Crohn's disease

    Autoimmun Rev

    (2014)
  • B. Eksteen et al.

    Gut homing receptors on CD8 T cells are retinoic acid dependent and not maintained by liver dendritic or stellate cells

    Gastroenterology

    (2009)
  • M. Iwata et al.

    Retinoic acid imprints gut-homing specificity on T cells

    Immunity

    (2004)
  • S. Manicassamy et al.

    Retinoic acid-dependent regulation of immune responses by dendritic cells and macrophages

    Semin Immunol

    (2009)
  • B.S. Holder et al.

    Retinoic acid stabilizes antigen-specific regulatory T-cell function in autoimmune hepatitis type 2

    J Autoimmun

    (2014)
  • S. Ben-Horin et al.

    Optimizing anti-TNF treatments in inflammatory bowel disease

    Autoimmun Rev

    (2014)
  • F. Atzeni et al.

    Immunogenicity and autoimmunity during anti-TNF therapy

    Autoimmun Rev

    (2013)
  • F. Atzeni et al.

    Rheumatic manifestations in inflammatory bowel disease

    Autoimmun Rev

    (2014)
  • G. Fiorino et al.

    Paradoxical immune-mediated inflammation in inflammatory bowel disease patients receiving anti-TNF-alpha agents

    Autoimmun Rev

    (2014)
  • W.Z. Mehal

    The gut–liver axis: a busy two-way street

    Hepatology

    (2012)
  • G. Szabo et al.

    Gut–liver axis and sensing microbes

    Dig Dis

    (2010)
  • C.J. Robinson et al.

    From structure to function: the ecology of host-associated microbial communities

    Microbiol Mol Biol Rev

    (2010)
  • P. Gillevet et al.

    Quantitative assessment of the human gut microbiome using multitag pyrosequencing

    Chem Biodivers

    (2010)
  • I.N. Crispe

    The liver as a lymphoid organ

    Annu Rev Immunol

    (2009)
  • R.Y. Calne et al.

    Induction of immunological tolerance by porcine liver allografts

    Nature

    (1969)
  • V. Racanelli et al.

    The liver as an immunological organ

    Hepatology

    (2006)
  • G. Son et al.

    Contribution of gut bacteria to liver pathobiology

    Gastroenterol Res Pract

    (2010)
  • A.W. Thomson et al.

    Antigen-presenting cell function in the tolerogenic liver environment

    Nat Rev Immunol

    (2010)
  • E. Seki et al.

    TLR4 enhances TGF-beta signaling and hepatic fibrosis

    Nat Med

    (2007)
  • A. Abu-Shanab et al.

    The role of the gut microbiota in nonalcoholic fatty liver disease

    Nat Rev Gastroenterol Hepatol

    (2010)
  • J. Henao-Mejia et al.

    Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity

    Nature

    (2012)
  • F.A. Carvalho et al.

    Toll-like receptor-gut microbiota interactions: perturb at your own risk!

    Annu Rev Physiol

    (2012)
  • K.S. Michelsen et al.

    Toll-like receptors and innate immunity in gut homeostasis and pathology

    Curr Opin Hematol

    (2007)
  • C. Selmi et al.

    Innate immunity and primary biliary cirrhosis

    Curr Mol Med

    (2009)
  • Cited by (55)

    • System lupus erythematosus and the environment

      2020, Systemic Lupus Erythematosus: Basic, Applied and Clinical Aspects
    View all citing articles on Scopus

    Financial support: Financial support was provided by the National Basic Research Program of China (973 Program-2013CB944900, 2010CB945300) and the National Natural Science Foundation of China (81130058, 81430034).

    ☆☆

    There are no conflicts of interests regarding the publication of this article.

    View full text